Abstract
Background
The rapid increase in the incidence of head and neck squamous cell carcinoma (HNSCC) is caused by high‐risk human papillomavirus (HPV) infections. The HPV oncogenes E6 and E7 promote carcinogenesis by disrupting signaling pathways that control survival and proliferation. Although these cancers are often diagnosed with metastases, the mechanisms that regulate their dissemination are unknown.
Aims
The aim of this study was to determine whether the HPV‐16 E6 and E7 oncogenes affected the invasive and migratory properties of HNSCC cells which promote their spread and metastasis.
Methods and results
Invasiveness was determined using invadopodia assays which allow for quantitation of extracellular matrix (ECM) degradation by invadopodia which are proteolytic membrane protrusions that facilitate invasion. Using cell lines and genetic manipulations, we found that HPV inhibited invadopodia activity in aggressive cell lines which was mediated by the E6 and E7 oncogenes. Given these findings, we also tested whether HPV caused differences in the migratory ability of HNSCC cells using Transwell assays. In contrast to our invadopodia results, we found no correlation between HPV status and cell migration; however, blocking the expression of the E6 and E7 oncoproteins in a HPV‐positive (HPV+) HNSCC cell line resulted in decreased migration.
Conclusions
Our data suggest that the E6 and E7 oncoproteins are negative regulators of invadopodia activity but may promote migration in HPV+ HNSCC cells. Despite the need for ECM proteolysis to penetrate most tissues, the unique structure of the head and neck tissues in which these cancers arise may facilitate the spread of migratory cancer cells without significant proteolytic ability.
Keywords: head and neck cancer, human papillomavirus, invadopodia, invasion, migration
1. INTRODUCTION
While the overall incidence of head and neck squamous cell carcinoma (HNSCC) has been declining, there has been an alarming rise in human papillomavirus‐positive (HPV+) HNSCC cases over the last 30 years which are projected to eclipse all other types of HNSCC by 2030.1, 2, 3, 4 Despite these potentially “epidemic” levels given the link to a prevalent infectious agent,5, 6, 7 HPV+ HNSCC does not have unique treatment options despite its distinct biology.8, 9, 10 HPV+ tumors develop in the oropharyngeal region of the head and neck with localization to the tonsils and base of the tongue.11, 12, 13 The majority of HPV infections leading to HNSCC arise from the HPV‐16 strain.14, 15 Carcinogenesis caused by HPV‐16 as well as the other high‐risk strains is driven by the E6 and E7 oncoproteins which target a variety of proteins crucial for several cellular processes including apoptosis and the cell cycle.8, 10, 16, 17, 18, 19, 20
Clinically, HPV+ HNSCC is more frequently diagnosed with metastatic disease than human papillomavirus‐negative (HPV−) HNSCC.1, 2, 21 To metastasize, migrating cancer cells must breach cross‐linked and dense tissues that typically require ECM degradation.22, 23, 24 To invade these tissues, cancer cells use actin‐rich adhesive protrusions called invadopodia that localize proteinases for ECM proteolysis.25, 26, 27 Invadopodia formation correlates well with in vitro and in vivo invasive behavior of cancer cells and has thus been implicated in tumor progression.23, 28, 29 Therefore, the goal of this study was to determine whether HPV promotes invadopodia activity and migration in HNSCC cells given the frequency of metastases in HPV+ HNSCCs.
2. METHODS
2.1. Cell culture and reagents
HPV− (SCC‐25, SCC‐61, and SCC‐1) and HPV+ (SCC‐47 and SCC‐104) HNSCC cell lines were used in this study. SCC‐61 was originally obtained from the Yarbrough laboratory at our institution. The HPV status of these cells was previously verified.30 SCC‐25 and SCC‐1 with confirmed HPV− status were purchased from ATCC and Millipore Sigma, respectively. SCC‐47 and SCC‐104 with verified HPV+ status were purchased from Millipore Sigma. SCC‐61 and SCC‐25 were originally derived from aggressive tumors that had metastasized to lymph nodes.31, 32 The HPV+ HNSCC cell lines were also derived from aggressive tumors that had spread to lymph nodes.32 SCC‐61 cells were cultured as previously described using Dulbecco's modified Eagle's medium (DMEM) supplemented with 20% fetal bovine serum (FBS) (both from ThermoFisher).33, 34 SCC‐25 cells were cultured in DMEM/Ham's nutrient mixture F‐12 (DMEM/F12) (ThermoFisher) supplemented with 20% FBS and 0.4 μg/mL hydrocortisone (Millipore Sigma).31, 35, 36, 37 SCC‐1 and SCC‐47 cells were both cultured in the same medium as SCC‐25 cells but with an additional supplement of 1:100 minimum essential media nonessential amino acids (MEM NEAA) and 0.4 μg/mL hydrocortisone (both from Millipore Sigma). SCC‐104 cells were cultured in the same medium as SCC‐1 and SCC‐47 cells but supplemented with 1:100 penicillin‐streptomycin (10 000 U/mL penicillin‐10 000 μg/mL streptomycin mixture) (ThermoFisher). SCC‐25 cells were treated with lentiviral particles engineered for the simultaneous expression of the HPV‐16 E6 and E7 genes according to the manufacturer's instructions (ABM). SCC‐47 cells were treated with separate lentiviral particles for the simultaneous knockdown of the HPV‐16 E6 and E7 genes using different shRNA based on published siRNA sequences according to the manufacturer's protocol (Santa Cruz).38, 39 Transduced cells were cultured in puromycin‐containing growth medium for selection. Blank vectors were used as controls in both cell lines.
2.2. Quantitative real‐time PCR
Gene expression was measured as previously described40 using previously validated primers and the ΔΔCt method for quantitation41 with GAPDH as the internal control. In this case, mRNA was isolated with the RNeasy Mini Kit (Qiagen) and reverse transcribed using the iScript cDNA Synthesis Kit (BioRad) according to the manufacturer's instructions. The expression of E6 and E7 was measured using the SsoAdvanced Universal SYBR Green Supermix and CFX Connect Real‐Time PCR Detection System (both from BioRad). Because E6 and E7 are undetectable in SCC‐25 cells which are HPV−, we chose a cutoff value for Ct of 40 for these samples which were used as controls in the ΔΔCt calculations.
2.3. Western blotting
Western blotting was performed as previously described34 using an anti‐E6 mouse monoclonal antibody (Euromedex) and a 1:1 combination of anti‐E7 mouse monoclonal antibodies (Santa Cruz and ThermoFisher).41 In this case, 12% SDS‐PAGE gels were used, and Super Block T20 (TBS) blocking buffer (ThermoFisher) was used for blocking.41 Blots were detected using a horseradish peroxidase‐conjugated secondary antibody (PerkinElmer) which was visualized with chemiluminescence.
2.4. Invadopodia assays
Invadopodia assays were performed as previously described.34 Briefly, glass‐bottom dishes were overlaid with a 1:5 ratio of FITC‐labeled gelatin (ThermoFisher) and unlabeled gelatin (Polysciences) cross‐linked with 0.05% glutaraldehyde. Twenty‐five thousand cells were incubated for 4 or 18 hours in invadopodia medium which contains DMEM/Roswell Park Memorial Institute (RPMI) 1640, 5% NuSerum, 10% FBS, and 20 ng/mL epidermal growth factor (all from ThermoFisher).33, 34, 40, 42, 43
2.5. Immunofluorescence
Invadopodia were identified as previously described.33, 34, 43 Briefly, the punctate colocalization of F‐actin and cortactin was used as a marker and were identified with Alexa Fluor 546 phalloidin (ThermoFisher) and an anti‐cortactin mouse primary monoclonal antibody (clone 4F11 from Millipore Sigma), respectively. Fluorescent images were captured on a Nikon Ti‐E inverted microscope with a Plan Fluor 40× oil immersion objective. Invadopodia were manually counted, and ECM degradation was quantitated by thresholding the loss of FITC signal underneath the cells in Nikon Elements software (Nikon) and/or MetaMorph software (Molecular Devices).
2.6. Transwell assays
Transwell migration and invasion assays were performed as previously described.34 Briefly, 75 000 cells in serum‐free invadopodia medium were plated in uncoated and Matrigel‐coated Transwell inserts (ThermoFisher) with 5 and 8‐μm pore sizes, respectively, and incubated for 18 hours with complete invadopodia medium in the lower chamber. Cells that migrated or invaded through the permeable supports were quantitated using Hoechst (ThermoFisher) for nuclear staining in Nikon Elements and MetaMorph from fluorescent images captured using a Plan Fluor 20× objective.
2.7. Statistics
Statistics were performed as previously described using SPSS Statistics (IBM) with a P value less than .05 considered significant.33, 34, 40, 44 Briefly, all data were evaluated for normality using the Shapiro‐Wilk or Kolmogorov‐Smirnov test. Normal data were analyzed with a Student's t test for comparisons between 2 datasets or a 1‐way ANOVA with multiple t tests and Bonferroni correction for group and pairwise comparisons, respectively. Non‐normal data were analyzed with a Mann‐Whitney test for comparisons between 2 datasets or Kruskal‐Wallis test with a Tamhane post‐hoc test or Mann‐Whitney test with Bonferroni correction for group and pairwise comparisons, respectively.
3. RESULTS
Because HPV+ HNSCC has higher rates of metastasis,1, 2, 21 we first evaluated invadopodia activity in established HPV− and HPV+ HNSCC cell lines because the ability to degrade the ECM is an important component of invasion22, 23, 24 (Figure 1A). HPV− SCC‐25, SCC‐61, and SCC‐1 cells are frequently used as cell line models for studying invadopodia.31, 33, 34, 35, 36, 37, 45, 46, 47, 48, 49, 50, 51, 52, 53 HPV+ SCC‐47 and SCC‐104 cells are common cell line models for studying the effects of HPV‐16.30, 41, 54, 55, 56, 57, 58, 59, 60 We performed invadopodia assays on these cell lines and quantitated invadopodia numbers and ECM degradation of a fluorescently labeled ECM (Figure 1B‐D). Contrary to our expectations, HPV+ HNSCC cell lines degraded significantly less ECM and formed fewer actively degrading (colocalized with ECM degradation) and total (degrading and nondegrading) invadopodia when compared to HPV− HNSCC cell lines (Figure 1B‐D). However, we observed considerable variation among cell lines within each group which led us to perform individual comparisons between the HPV− and HPV+ HNSCC cell lines. SCC‐25 and SCC‐61 cells degraded more ECM and formed more degrading and total invadopodia than the SCC‐47 and SCC‐104 cells, while SCC‐1 cells were only significantly greater when compared to SCC‐104 cells (Figure 1B‐D). These differences are consistent with previous reports of invadopodia activity by these cell lines (except SCC‐104 cells which have not previously been reported to form invadopodia to our knowledge).31, 32, 35, 36, 37, 48, 50, 61
Figure 1.

HPV+ HNSCCs exhibit less invadopodia activity than HPV− HNSCCs. Representative wide‐field fluorescence images of (A) HPV− (SCC‐25, SCC‐61, SCC‐1) and HPV+ (SCC‐47, SCC‐104) HNSCC cell lines in invadopodia assays after 18‐hour incubation. Invadopodia puncta (magnified inlays of invadopodia in white boxes) were identified by colocalization of actin (orange) and cortactin (purple). Active invadopodia (ie, actively degrading) were identified by further colocalization with black degraded areas of ECM lacking FITC signal (green) while total invadopodia included active and nondegrading puncta. Quantitation of (B) the degradation area per cell, (C) active invadopodia per cell, and (D) total invadopodia per cell for the HPV− and HPV+ HNSCC cell lines. All data are presented as box and whisker plots with the black lines indicating the medians, the whiskers representing the 10th and 90th percentiles, and asterisk(s) indicating different types of comparisons with P < .05 for n = 31 to 83 cells from 3 independent experiments. * indicates comparisons between HPV− and HPV+ groups, ** indicates comparisons of SCC‐25 and SCC‐61 cell lines to the individual HPV+ HNSCC cell lines, and *** indicates a comparison between SCC‐1 and SCC‐104 cell lines. Scale bar represents 10 μm
To determine whether the differences in invadopodia activity were dependent on HPV status, we selected a representative cell line from the HPV− group, SCC‐25, and stably expressed the E6 and E7 oncogenes (Figure 2A). Overexpression of E6 and E7 in SCC‐25 cells was statistically similar to the expression of these oncogenes in HPV+ SCC‐47 cells when compared to HPV− SCC‐25 cells (Supp. Figure 1). Compared to the empty vector control (Control), E6 and E7 overexpression (E6/E7 OE) in SCC‐25 cells led to a decrease in ECM degradation as well as in the number of actively degrading but not total invadopodia (Figure 2B‐E). To further implicate HPV in mediating invadopodia activity, we also selected a representative cell line from the HPV+ group, SCC‐47, and stably knocked down the E6 and E7 oncogenes (Figure 3A). In this case, Western blots were used for confirmation because preliminary PCR results indicated that the knockdown (KD) of E6 and E7 occurred through translational repression (Figure 3A). E6 and E7 KD (E6/E7 KD) in SCC‐47 cells had the inverse effect and increased ECM degradation and the number of actively degrading but not total invadopodia when compared to the blank vector control (Figure 3B‐E).
Figure 2.

Expression of HPV E6 and E7 inhibits invadopodia activity. (A) Relative expression (fold change) in mRNA levels of E6 and E7 in HPV− SCC‐25 cells transduced with an E6/E7 lentiviral expression vector was evaluated with quantitative real‐time PCR which was performed in triplicate for each experiment and overall for 3 independent experiments. Representative wide‐field fluorescence images of (B) control and E6/E7 OE SCC‐25 cells in invadopodia assays after 4‐hour incubation. Invadopodia puncta (magnified inlays of invadopodia in white boxes) were identified by colocalization of actin (orange) and cortactin (purple). Active invadopodia (ie, actively degrading) were identified by further colocalization with black degraded areas of ECM lacking FITC signal (green), while total invadopodia included active and nondegrading puncta. Quantitation of (C) the degradation area per cell, (D) active invadopodia per cell, and (E) total invadopodia per cell for control and E6/E7 OE SCC‐25 cells. Imaging data were quantitated for n = 118 to 125 cells from 4 independent experiments. All data are presented as box and whisker plots with the black lines indicating the medians, the whiskers representing the 10th and 90th percentiles, and * indicating P < .05. Scale bar represents 5 μm
Figure 3.

Knockdown of HPV E6 and E7 promotes invadopodia activity. (A) Representative western blots of E6 (16 kDa) and E7 (20 kDa) in HPV+ SCC‐47 cells simultaneously transduced with E6 and E7 lentiviral vectors with different shRNA from 2 independent experiments which resulted in a 76% decrease for both E6 and E7, when normalized to β‐actin. Representative wide‐field fluorescence images of (B) control and E6/E7 KD SCC‐47 cells in invadopodia assays after 4‐hour incubation. Invadopodia puncta (magnified inlays of invadopodia in white boxes) were identified by colocalization of actin (orange) and cortactin (purple). Active invadopodia (ie, actively degrading) were identified by further colocalization with black degraded areas of ECM lacking FITC signal (green) while total invadopodia included active and nondegrading puncta. Quantitation of (C) the degradation area per cell, (D) active invadopodia per cell, and (E) total invadopodia per cell for control and E6/E7 KD SCC‐47 cells. Imaging data were quantitated for n = 60 cells from 3 independent experiments. All data are presented as box and whisker plots with the black lines indicating the medians, the whiskers representing the 10th and 90th percentiles, and * indicating P < .05. Scale bar represents 10 μm
Because our data indicated that E6 and E7 inhibit invadopodia activity, we determined whether HPV affected the migration and invasion of HNSCC cells which is necessary for dissemination and metastasis.62, 63, 64 We first performed Transwell migration assays with the HPV− and HPV+ HNSCC cell lines and found no significant correlation between migratory potential and HPV status (Figure 4A, B). Similar results were observed in Transwell invasion assays wherein no significant trend was observed in HPV+ versus HPV− HNSCC cell lines (Figure 4C, D). However, compared to the empty vector control, knockdown of the E6 and E7 oncoproteins (E6/E7 KD) significantly decreased cell migration and invasion in SCC‐47 cells (Figure 4E‐H). Similar assays could not be performed with SCC‐25 cells overexpressing E6 and E7 because these cells did not reliably attach to the Transwell inserts.
Figure 4.

E6 and E7 promote cell migration. Representative wide‐field fluorescence images of HPV− and HPV+ HNSCC cell lines stained with Hoechst after 18‐hour incubation in the Transwell (A) migration and (C) invasion assays with quantitation of the number of (B) migrated and (D) invaded cells. Representative wide‐field fluorescence images of control and E6/E7 KD SCC‐47 cells stained with Hoechst after 18‐hour incubation in the Transwell (E) migration and (G) invasion assays with quantitation of the number of (F) migrated and (H) invaded cells. All data are presented as box and whisker plots with the black lines indicating the medians, the whiskers representing the 10th and 90th percentiles, and asterisk(s) indicating P < .05 for n = 32 to 40 areas from 3 to 4 independent experiments. For (B), * indicates comparisons of SCC‐25 to both SCC‐61 and SCC‐47, and ** indicates comparisons of SCC‐1 and SCC‐104 to each other and to both SCC‐61 and SCC‐47. For (D), * indicates comparisons of SCC‐25 and SCC‐1 to the other HNSCC cell lines, and ** indicates comparisons of SCC‐104 to the other HNSCC cell lines. Scale bar represents 50 μm
4. DISCUSSION
High‐risk HPV drives tumorigenesis by altering the signaling pathways that control transcription, differentiation, DNA repair, apoptosis, and the cell cycle.8, 10, 16, 17, 18, 19, 20 The E6 and E7 oncoproteins target various molecules for ubiquitin‐mediated proteasomal degradation relevant for these processes thus destabilizing important regulatory mechanisms.8, 10, 20 Although E6 and E7 have different targets that contribute to carcinogenesis,16, 17, 18, 19 they are expressed together from polycistronic mRNA and may cooperate to amplify their individual effects.10, 65, 66, 67, 68 Therefore, we chose to simultaneously manipulate E6 and E7 to first understand the general effects of HPV‐16 on the invasive and migratory properties of HNSCC cells in this study. In this study, we demonstrate that HPV‐infected HNSCC cell lines have lower invadopodia activity than HPV− HNSCC cells. HPV status does not correlate with the migratory potential of a cell line; however, cell migration is compromised in HPV+ HNSCC cells with reduced expression of the E6 and E7 oncoproteins.
Tissues have long been thought to represent formidable barriers to migrating cancer cells thus necessitating ECM proteolysis for invasion69, 70, 71 which is facilitated by invadopodia.25, 26, 27 However, more recent work suggests that migrating cancer cells can invade in a nonproteolytic manner by taking advantage of variations and defects in epithelial and stromal tissues as well as physically deforming and pushing through porous ECMs.69, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82 While Matrigel is routinely used for assessing proteolytic invasion, it lacks the native cross‐links of a true basement membrane and is readily traversed by many cell types exhibiting a nonproteolytic mode of migration.70, 83, 84 Thus, Matrigel may not represent a formidable structural barrier that necessitates the use of proteolytic machinery for invasion by some types of migrating cancer cells which would be consistent with our findings that showed similar results for Transwell migration and invasion, particularly for the HPV+ HNSCC cells which exhibited significantly less invadopodia activity. Furthermore, we also found a decrease in Transwell invasion with E6 and E7 inhibition despite an increase in invadopodia formation and activity. However, the reduction in migration likely accounts for this discrepancy which would limit the ability of cells to move through the Matrigel. Alternatively, cells may use other mechanisms to mediate ECM degradation such as the release of extracellular vesicles containing proteolytic enzymes. However, the secretion of exosomes is enhanced by invadopodia,37 and microvesicle secretion is promoted by compliant matrices with thicknesses far greater than the thin coating found in Transwells.85
Although the molecular mechanisms that regulate the spread of HPV+ HNSCC remain to be explored, E6 and E7 have also been shown to increase the migration of cervical cancer cells which is thought to occur by decreasing RhoA activity.86, 87, 88, 89, 90 Rho GTPases are overexpressed in HNSCC,91, 92, 93, 94 and RhoA controls actomyosin contractility to mediate cell migration.95, 96, 97 Inhibition of the Rho pathway can promote cell migration in some cell types by reducing substrate adhesion88, 98, 99 which is normally reinforced by large contractile forces.100, 101, 102 Furthermore, RhoA signaling can promote invadopodia activity.103, 104, 105 While E6 can deregulate miRNA expression to affect migration, these mechanisms are mediated by p5386, 87, 88, 89, 90, 106 which is inactivated in SCC‐25 cells.107, 108 Therefore, HPV may exert its effects on Rho through additional mechanisms which alter downstream signaling important for migration and invadopodia activity.
One critical effector of Rho is Rho‐associated kinase (ROCK) which phosphorylates nonmuscle myosin II (NM II) to regulate actomyosin contractility.95, 96, 97 ROCK expression is also associated with tumor stage and progression and promotes migration and invasion in HNSCC cells.91, 92, 93, 94 We have shown that the ROCK isoforms differentially regulate invadopodia activity through contractile and noncontractile mechanisms in HNSCC cells.34 More specifically, we found that actomyosin contractility regulates invadopodia formation and ECM degradation as well as migration through the ROCK1/NM II pathway.34 Furthermore, we also found that signaling through ROCK2 promotes invadopodia maturation independently of contractile force generation through LIM kinase (LIMK)34 which is mediated by RhoC.109, 110, 111, 112 Although further studies are required, E6 and E7 may interfere with these ROCK‐dependent signaling pathways by altering the activity of the Rho family of GTPases to affect the migratory and invasive properties of HNSCC cells. Such roles for E6 and/or E7 would be consistent with our findings that these oncogenes increase migration but inhibit the number of mature invadopodia capable of degradation (ie, active) but not the total number of invadopodia formed. Thus, these data suggest that HPV does not interfere with invadopodia formation but maturation which requires sustained actin polymerization for stability and ECM degradation.113, 114
In addition to their distinct biological characteristics, HPV+ HNSCC tumors also have unique morphological features including little stromal desmoplasia.115, 116, 117, 118 This ECM normally stiffens tumors which promotes aggressiveness by activating mechanical signaling pathways that drive malignant cellular behavior.119, 120, 121 Tumor mechanical properties and related signaling pathways are emerging as significant factors in HPV− HNSCC. For example, these tumors are more rigid than surrounding normal tissues,122, 123, 124, 125, 126, 127 and their stromal density correlates with tumor stage, poor prognoses, and reduced survival rates.128, 129, 130, 131 Furthermore, molecules involved in ECM cross‐linking, mechanotransduction, and invasion also correlate with cancer risk, tumor progression, and poor survival.132, 133, 134, 135, 136, 137, 138, 139 In addition, our work has shown that ECM rigidity mimicking tumor mechanical properties promotes invadopodia activity.33, 34, 40, 42, 71, 140 Despite the lack of tumor‐associated ECM, HPV+ HNSCCs are more frequently diagnosed with metastases.1, 2, 21 While our data indicate that E6 and E7 can alter the relative migratory potential of HNSCC cells, we also found a reduction in proteolytic activity which calls into question the means by which these tumors spread given the importance of ECM degradation for invasion of tissue barriers.22, 23, 24
HPV+ HNSCC often arises in the lymphoid tissues of the palatine and lingual tonsils which are characterized by tubular and branched crypts composed of discontinuous basement membrane and incomplete connective tissue.115, 116, 117, 118 While these tissues are designed to facilitate immunological responses between infiltrating immune cells and incoming pathogens, their structure and lack of stromal desmoplasia may also provide little resistance to the spread of cancer cells.116, 117, 118 Furthermore, tonsillar tissue drains fluids through lymphatic vessels to surrounding lymph nodes in the neck which are the most common sites of metastases for HPV+ HNSCCs.141, 142, 143 Therefore, these tissues may facilitate the movement of migratory cells to promote tumor progression. This notion would be consistent with the clinical findings that metastases beyond the head and neck occur over much longer time periods for HPV+ versus HPV− HNSCCs144, 145, 146, 147 which would require penetration of intact tissues in other bodily regions.
5. CONCLUSIONS
In this study, we have shown that the HPV‐16 oncoproteins E6 and E7 alter the migratory potential of HPV+ HNSCC cells while inhibiting invadopodia activity. Current models describing the effects of high‐risk HPV do not address the roles of E6 and E7 on the migration and invasion of HNSCC cells. While the molecular mechanisms still need to be elucidated, our previous studies as well as work by others suggest that these oncoproteins may interfere with mechanical signaling pathways important for force generation and invadopodia dynamics. Inhibition of these pathways may drive a migratory phenotype capable of effective invasion of oropharyngeal tissues that present a permissive environment with little need for significant ECM degradation.
CONFLICT OF INTEREST
The authors have no conflict of interest to report.
AUTHORS’ CONTRIBUTIONS
All authors had full access to the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Conceptualization, C.N.K., A.P.; Methodology, C.N.K., R.J.J., A.P.; Investigation, C.N.K., R.J.J., A.P.; Formal Analysis, C.N.K., R.J.J., A.P.; Resources, C.N.K., R.J.J., A.P.; Writing ‐ Original Draft, C.N.K., R.J.J., A.P.; Writing ‐ Review & Editing, C.N.K., A.P.; Visualization, C.N.K., R.J.J., A.P; Supervision, R.J.J., A.P.; Funding Acquisition, A.P.
Supporting information
Fig. S1. Relative expression (fold change) in mRNA levels of E6 and E7 in HPV− SCC‐25 and HPV+ SCC‐47 cells evaluated with quantitative real‐time PCR which was performed in triplicate for each experiment and overall for 3 independent experiments. Expression levels of E6 and E7 in HPV+ SCC‐47 cells were statistically similar to those in the SCC‐25 E6/E7 OE in Figure 2A. All data are presented as box and whisker plots with the black lines indicating the medians, the whiskers representing the 10th and 90th percentiles, and * indicating p < 0.05.
ACKNOWLEDGEMENTS
We would like to thank Natalia Isaeva of Yale University, Christian Hübbers of the University of Cologne, Toni Brand of the University of California Santa Barbara, and Brandee Brown for helpful discussions regarding cell lines and reagents including the difficulties of detecting E6 and E7 with Western blots given the limited number of available antibodies. We would also like to thank James Lewis, Jr., for insightful discussions regarding HPV+ HNSCC tumors. Research reported in this publication was supported by the Research Scholar Grant RSG‐15‐226‐01‐CSM from the American Cancer Society to A.P.
Kahue CN, Jerrell RJ, Parekh A. Expression of human papillomavirus oncoproteins E6 and E7 inhibits invadopodia activity but promotes cell migration in HPV‐positive head and neck squamous cell carcinoma cells. Cancer Reports. 2018;1:e1125. 10.1002/cnr2.1125
[Correction added on 14 Sept 2018 after first online publication: The percentage decrease for both E6 and E7 proteins relative to β‐actin was corrected to 76% in the legend for figure 3.]
REFERENCES
- 1. Chaturvedi AK, Engels EA, Anderson WF, Gillison ML. Incidence trends for human papillomavirus‐related and ‐unrelated oral squamous cell carcinomas in the United States. J Clin Oncol. 2008;26(4):612‐619. [DOI] [PubMed] [Google Scholar]
- 2. Chaturvedi AK, Engels EA, Pfeiffer RM, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. 2011;29(32):4294‐4301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Gillison ML, Chaturvedi AK, Anderson WF, Fakhry C. Epidemiology of human papillomavirus‐positive head and neck squamous cell carcinoma. J Clin Oncol. 2015;33(29):3235‐3242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Weatherspoon DJ, Chattopadhyay A, Boroumand S, Garcia I. Oral cavity and oropharyngeal cancer incidence trends and disparities in the United States: 2000‐2010. Cancer Epidemiol. 2015;39(4):497‐504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Syrjanen S. Human papillomavirus (HPV) in head and neck cancer. J Clin Virol. 2005;32(Suppl 1):S59‐S66. [DOI] [PubMed] [Google Scholar]
- 6. Nasman A, Attner P, Hammarstedt L, et al. Incidence of human papillomavirus (HPV) positive tonsillar carcinoma in Stockholm, Sweden: An epidemic of viral‐induced carcinoma? Int J Cancer. 2009;125(2):362‐366. [DOI] [PubMed] [Google Scholar]
- 7. Nasman A, Nordfors C, Holzhauser S, et al. Incidence of human papillomavirus positive tonsillar and base of tongue carcinoma: A stabilisation of an epidemic of viral induced carcinoma? Eur J Cancer. 2015;51(1):55‐61. [DOI] [PubMed] [Google Scholar]
- 8. Dok R, Nuyts S. HPV positive head and neck cancers: Molecular pathogenesis and evolving treatment strategies. Cancer. 2016;8(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Hennessey PT, Westra WH, Califano JA. Human papillomavirus and head and neck squamous cell carcinoma: Recent evidence and clinical implications. J Dent Res. 2009;88(4):300‐306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Rautava J, Syrjanen S. Biology of human papillomavirus infections in head and neck carcinogenesis. Head Neck Pathol. 2012;6(Suppl 1):S3‐S15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Gillison ML, Koch WM, Capone RB, et al. Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst. 2000;92(9):709‐720. [DOI] [PubMed] [Google Scholar]
- 12. Gillison ML. Current topics in the epidemiology of oral cavity and oropharyngeal cancers. Head Neck. 2007;29(8):779‐792. [DOI] [PubMed] [Google Scholar]
- 13. D'Souza G, Kreimer AR, Viscidi R, et al. Case‐control study of human papillomavirus and oropharyngeal cancer. N Engl J Med. 2007;356(19):1944‐1956. [DOI] [PubMed] [Google Scholar]
- 14. Gillison ML, Castellsague X, Chaturvedi A, et al. Eurogin roadmap: comparative epidemiology of HPV infection and associated cancers of the head and neck and cervix. Int J Cancer. 2014;134(3):497‐507. [DOI] [PubMed] [Google Scholar]
- 15. Sepiashvili L, Bruce JP, Huang SH, O'Sullivan B, Liu FF, Kislinger T. Novel insights into head and neck cancer using next‐generation “omic” technologies. Cancer Res. 2015;75(3):480‐486. [DOI] [PubMed] [Google Scholar]
- 16. Pim D, Banks L. Interaction of viral oncoproteins with cellular target molecules: Infection with high‐risk vs low‐risk human papillomaviruses. APMIS. 2010;118(6–7):471‐493. [DOI] [PubMed] [Google Scholar]
- 17. Roman A, Munger K. The papillomavirus E7 proteins. Virology. 2013;445(1–2):138‐168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Vande Pol SB, Klingelhutz AJ. Papillomavirus E6 oncoproteins. Virology. 2013;445(1–2):115‐137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Ganti K, Broniarczyk J, Manoubi W, et al. The human papillomavirus E6 PDZ binding motif: From life cycle to malignancy. Viruses. 2015;7(7):3530‐3551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Sano D, Oridate N. The molecular mechanism of human papillomavirus‐induced carcinogenesis in head and neck squamous cell carcinoma. Int J Clin Oncol. 2016;21(5):819‐826. [DOI] [PubMed] [Google Scholar]
- 21. Corey AS, Hudgins PA. Radiographic imaging of human papillomavirus related carcinomas of the oropharynx. Head Neck Pathol. 2012;6(Suppl 1):S25‐S40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Wolf K, Friedl P. Extracellular matrix determinants of proteolytic and non‐proteolytic cell migration. Trends Cell Biol. 2011;21(12):736‐744. [DOI] [PubMed] [Google Scholar]
- 23. Bravo‐Cordero JJ, Hodgson L, Condeelis J. Directed cell invasion and migration during metastasis. Curr Opin Cell Biol. 2012;24(2):277‐283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Friedl P, Alexander S. Cancer invasion and the microenvironment: Plasticity and reciprocity. Cell. 2011;147(5):992‐1009. [DOI] [PubMed] [Google Scholar]
- 25. Weaver AM. Invadopodia: Specialized cell structures for cancer invasion. Clin Exp Metastasis. 2006;23(2):97‐105. [DOI] [PubMed] [Google Scholar]
- 26. Caldieri G, Ayala I, Attanasio F, Buccione R. Cell and molecular biology of invadopodia. Int Rev Cell Mol Biol. 2009;275:1‐34. [DOI] [PubMed] [Google Scholar]
- 27. Genot E, Gligorijevic B. Invadosomes in their natural habitat. Eur J Cell Biol. 2014;93(10–12):367‐379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Buccione R, Orth JD, McNiven MA. Foot and mouth: Podosomes, invadopodia and circular dorsal ruffles. Nature Reviews. 2004;5(8):647‐657. [DOI] [PubMed] [Google Scholar]
- 29. Weaver AM. Invadopodia. Curr Biol. 2008;18(9):R362‐R364. [DOI] [PubMed] [Google Scholar]
- 30. Gubanova E, Brown B, Ivanov SV, et al. Downregulation of SMG‐1 in HPV‐positive head and neck squamous cell carcinoma due to promoter hypermethylation correlates with improved survival. Clin Cancer Res. 2012;18(5):1257‐1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Clark ES, Whigham AS, Yarbrough WG, Weaver AM. Cortactin is an essential regulator of matrix metalloproteinase secretion and extracellular matrix degradation in invadopodia. Cancer Res. 2007;67(9):4227‐4235. [DOI] [PubMed] [Google Scholar]
- 32. Lin CJ, Grandis JR, Carey TE, et al. Head and neck squamous cell carcinoma cell lines: Established models and rationale for selection. Head Neck. 2007;29(2):163‐188. [DOI] [PubMed] [Google Scholar]
- 33. Jerrell RJ, Parekh A. Cellular traction stresses mediate extracellular matrix degradation by invadopodia. Acta Biomater. 2014;10(5):1886‐1896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Jerrell RJ, Parekh A. Matrix rigidity differentially regulates invadopodia activity through ROCK1 and ROCK2. Biomaterials. 2016;84:119‐129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Clark ES, Brown B, Whigham AS, Kochaishvili A, Yarbrough WG, Weaver AM. Aggressiveness of HNSCC tumors depends on expression levels of cortactin, a gene in the 11q13 amplicon. Oncogene. 2009;28(3):431‐444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Hoshino D, Jourquin J, Emmons SW, et al. Network analysis of the focal adhesion to invadopodia transition identifies a PI3K‐PKCalpha invasive signaling axis. Sci Signal. 2012;5(241):ra66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Hoshino D, Kirkbride KC, Costello K, et al. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 2013;5(5):1159‐1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Niu XY, Peng ZL, Duan WQ, Wang H, Wang P. Inhibition of HPV 16 E6 oncogene expression by RNA interference in vitro and in vivo. Int J Gynecol Cancer. 2006;16(2):743‐751. [DOI] [PubMed] [Google Scholar]
- 39. Chang JT, Kuo TF, Chen YJ, et al. Highly potent and specific siRNAs against E6 or E7 genes of HPV16‐ or HPV18‐infected cervical cancers. Cancer Gene Ther. 2010;17(12):827‐836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Parekh A, Ruppender NS, Branch KM, et al. Sensing and modulation of invadopodia across a wide range of rigidities. Biophys J. 2011;100(3):573‐582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Brand TM, Hartmann S, Bhola NE, et al. Human papillomavirus regulates HER3 expression in head and neck Cancer: implications for targeted HER3 therapy in HPV(+) patients. Clin Cancer Res. 2017;23(12):3072‐3083. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 42. Alexander NR, Branch KM, Parekh A, et al. Extracellular matrix rigidity promotes invadopodia activity. Curr Biol. 2008;18(17):1295‐1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Jerrell RJ, Parekh A. Polyacrylamide gels for invadopodia and traction force assays on cancer cells. J Vis Exp. 2015;95:52343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Jerrell RJ, Leih MJ, Parekh A. The ROCK isoforms differentially regulate the morphological characteristics of carcinoma cells. Small GTPases. 2017;1‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Clark ES, Weaver AM. A new role for cortactin in invadopodia: Regulation of protease secretion. Eur J Cell Biol. 2008;87(8–9):581‐590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Ammer AG, Kelley LC, Hayes KE, et al. Saracatinib impairs head and neck squamous cell carcinoma invasion by disrupting Invadopodia function. J Cancer Sci Ther. 2009;1(2):52‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Kelley LC, Ammer AG, Hayes KE, et al. Oncogenic Src requires a wild‐type counterpart to regulate invadopodia maturation. J Cell Sci. 2010;123(Pt 22):3923‐3932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Branch KM, Hoshino D, Weaver AM. Adhesion rings surround invadopodia and promote maturation. Biol Open. 2012;1(8):711‐722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Hayes KE, Walk EL, Ammer AG, Kelley LC, Martin KH, Weed SA. Ableson kinases negatively regulate invadopodia function and invasion in head and neck squamous cell carcinoma by inhibiting an HB‐EGF autocrine loop. Oncogene. 2013;32(40):4766‐4777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Jimenez L, Sharma VP, Condeelis J, et al. MicroRNA‐375 suppresses extracellular matrix degradation and Invadopodial activity in head and neck squamous cell carcinoma. Arch Pathol Lab Med. 2015;139(11):1349‐1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Pehkonen H, von Nandelstadh P, Karhemo PR, et al. Liprin‐alpha1 is a regulator of vimentin intermediate filament network in the cancer cell adhesion machinery. Sci Rep. 2016;6(1):24486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Chen CN, Chen YT, Yang TL. The data of establishing a three‐dimensional culture system for in vitro recapitulation and mechanism exploration of tumor satellite formation during cancer cell transition. Data Brief. 2017;15:545‐561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Chen CN, Chen YT, Yang TL. Application of three‐dimensional collagen scaffolds to recapitulate and monitor the dynamics of epithelial‐mesenchymal transition during tumor satellite formation of head and neck cancer. Biomaterials. 2018;154:134‐146. [DOI] [PubMed] [Google Scholar]
- 54. Akagi K, Li J, Broutian TR, et al. Genome‐wide analysis of HPV integration in human cancers reveals recurrent, focal genomic instability. Genome Res. 2014;24(2):185‐199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Glogauer JE, Sun CX, Bradley G, Magalhaes MA. Neutrophils increase oral squamous cell carcinoma invasion through an invadopodia‐dependent pathway. Cancer Immunol Res. 2015;3(11):1218‐1226. [DOI] [PubMed] [Google Scholar]
- 56. Olthof NC, Huebbers CU, Kolligs J, et al. Viral load, gene expression and mapping of viral integration sites in HPV16‐associated HNSCC cell lines. Int J Cancer. 2015;136(5):E207‐E218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Gary C, Hajek M, Biktasova A, Bellinger G, Yarbrough WG, Issaeva N. Selective antitumor activity of roscovitine in head and neck cancer. Oncotarget. 2016;7(25):38598‐38611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Dok R, Glorieux M, Holacka K, Bamps M, Nuyts S. Dual role for p16 in the metastasis process of HPV positive head and neck cancers. Mol Cancer. 2017;16(1):113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Kannan A, Hertweck KL, Philley JV, Wells RB, Dasgupta S. Genetic mutation and exosome signature of human papilloma virus associated oropharyngeal Cancer. Sci Rep. 2017;7:46102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Thomas RJ, Oleinik N, Panneer Selvam S, et al. HPV/E7 induces chemotherapy‐mediated tumor suppression by ceramide‐dependent mitophagy. EMBO Mol Med. 2017;9(8):1030‐1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Jimenez L, Jayakar SK, Ow TJ, Segall JE. Mechanisms of invasion in head and neck Cancer. Arch Pathol Lab Med. 2015;139(11):1334‐1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Friedl P, Wolf K. Tumour‐cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3(5):362‐374. [DOI] [PubMed] [Google Scholar]
- 63. Friedl P, Locker J, Sahai E, Segall JE. Classifying collective cancer cell invasion. Nat Cell Biol. 2012;14(8):777‐783. [DOI] [PubMed] [Google Scholar]
- 64. Clark AG, Vignjevic DM. Modes of cancer cell invasion and the role of the microenvironment. Curr Opin Cell Biol. 2015;36:13‐22. [DOI] [PubMed] [Google Scholar]
- 65. Hoppe‐Seyler K, Bossler F, Braun JA, Herrmann AL, Hoppe‐Seyler F. The HPV E6/E7 oncogenes: key factors for viral carcinogenesis and therapeutic targets. Trends Microbiol. 2018;26(2):158‐168. [DOI] [PubMed] [Google Scholar]
- 66. Walline HM, Komarck CM, McHugh JB, et al. Genomic integration of high‐risk HPV alters gene expression in oropharyngeal squamous cell carcinoma. Mol Cancer Res. 2016;14(10):941‐952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Weijzen S, Zlobin A, Braid M, Miele L, Kast WM. HPV16 E6 and E7 oncoproteins regulate Notch‐1 expression and cooperate to induce transformation. J Cell Physiol. 2003;194(3):356‐362. [DOI] [PubMed] [Google Scholar]
- 68. Accardi R, Rubino R, Scalise M, et al. E6 and E7 from human papillomavirus type 16 cooperate to target the PDZ protein Na/H exchange regulatory factor 1. J Virol. 2011;85(16):8208‐8216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Sabeh F, Shimizu‐Hirota R, Weiss SJ. Protease‐dependent versus ‐independent cancer cell invasion programs: Three‐dimensional amoeboid movement revisited. J Cell Biol. 2009;185(1):11‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Hotary K, Li XY, Allen E, Stevens SL, Weiss SJ. A cancer cell metalloprotease triad regulates the basement membrane transmigration program. Genes Dev. 2006;20(19):2673‐2686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Parekh A, Weaver AM. Regulation of cancer invasiveness by the physical extracellular matrix environment. Cell Adh Migr. 2009;3(3):288‐292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Kauppila S, Stenback F, Risteli J, Jukkola A, Risteli L. Aberrant type I and type III collagen gene expression in human breast cancer in vivo. J Pathol. 1998;186(3):262‐268. [DOI] [PubMed] [Google Scholar]
- 73. Kauppila S, Bode MK, Stenback F, Risteli L, Risteli J. Cross‐linked telopeptides of type I and III collagens in malignant ovarian tumours in vivo. Br J Cancer. 1999;81(4):654‐661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Condeelis J, Segall JE. Intravital imaging of cell movement in tumours. Nat Rev Cancer. 2003;3(12):921‐930. [DOI] [PubMed] [Google Scholar]
- 75. Demou ZN, Awad M, McKee T, et al. Lack of telopeptides in fibrillar collagen I promotes the invasion of a metastatic breast tumor cell line. Cancer Res. 2005;65(13):5674‐5682. [DOI] [PubMed] [Google Scholar]
- 76. Bajenoff M, Egen JG, Koo LY, et al. Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes. Immunity. 2006;25(6):989‐1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Nagano S, Perentes JY, Jain RK, Boucher Y. Cancer cell death enhances the penetration and efficacy of oncolytic herpes simplex virus in tumors. Cancer Res. 2008;68(10):3795‐3802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Beadle C, Assanah MC, Monzo P, Vallee R, Rosenfeld SS, Canoll P. The role of myosin II in glioma invasion of the brain. Mol Biol Cell. 2008;19(8):3357‐3368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Glentis A, Oertle P, Mariani P, et al. Cancer‐associated fibroblasts induce metalloprotease‐independent cancer cell invasion of the basement membrane. Nat Commun. 2017;8(1):924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol. 2003;5(8):711‐719. [DOI] [PubMed] [Google Scholar]
- 81. Sanz‐Moreno V, Gadea G, Ahn J, et al. Rac activation and inactivation control plasticity of tumor cell movement. Cell. 2008;135(3):510‐523. [DOI] [PubMed] [Google Scholar]
- 82. Lammermann T, Sixt M. Mechanical modes of ‘amoeboid’ cell migration. Curr Opin Cell Biol. 2009;21(5):636‐644. [DOI] [PubMed] [Google Scholar]
- 83. Rowe RG, Weiss SJ. Breaching the basement membrane: Who, when and how? Trends Cell Biol. 2008;18(11):560‐574. [DOI] [PubMed] [Google Scholar]
- 84. Sodek KL, Brown TJ, Ringuette MJ. Collagen I but not Matrigel matrices provide an MMP‐dependent barrier to ovarian cancer cell penetration. BMC Cancer. 2008;8(1):223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Sedgwick AE, Clancy JW, Olivia Balmert M, D'Souza‐Schorey C. Extracellular microvesicles and invadopodia mediate non‐overlapping modes of tumor cell invasion. Sci Rep. 2015;5(1):14748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Au Yeung CL, Tsang TY, Yau PL, Kwok TT. Human papillomavirus type 16 E6 induces cervical cancer cell migration through the p53/microRNA‐23b/urokinase‐type plasminogen activator pathway. Oncogene. 2011;30(21):2401‐2410. [DOI] [PubMed] [Google Scholar]
- 87. Wang S, Pang T, Gao M, et al. HPV E6 induces eIF4E transcription to promote the proliferation and migration of cervical cancer. FEBS Lett. 2013;587(6):690‐697. [DOI] [PubMed] [Google Scholar]
- 88. Gomez‐Gomez Y, Organista‐Nava J, Gariglio P. Deregulation of the miRNAs expression in cervical cancer: Human papillomavirus implications. Biomed Res Int. 2013;2013:407052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Hu D, Zhou J, Wang F, Shi H, Li Y, Li B. HPV‐16 E6/E7 promotes cell migration and invasion in cervical cancer via regulating cadherin switch in vitro and in vivo. Arch Gynecol Obstet. 2015;292(6):1345‐1354. [DOI] [PubMed] [Google Scholar]
- 90. Ben W, Yang Y, Yuan J, et al. Human papillomavirus 16 E6 modulates the expression of host microRNAs in cervical cancer. Taiwan J Obstet Gynecol. 2015;54(4):364‐370. [DOI] [PubMed] [Google Scholar]
- 91. Jiang L, Liu X, Kolokythas A, et al. Downregulation of the rho GTPase signaling pathway is involved in the microRNA‐138‐mediated inhibition of cell migration and invasion in tongue squamous cell carcinoma. Int J Cancer. 2010;127(3):505‐512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Zhang J, He X, Ma Y, et al. Overexpression of ROCK1 and ROCK2 inhibits human laryngeal squamous cell carcinoma. Int J Clin Exp Pathol. 2015;8(1):244‐251. [PMC free article] [PubMed] [Google Scholar]
- 93. Wang X, Huang Y, Guo R, et al. Clinicopathological significance of ROCK1 and PIK3CA expression in nasopharyngeal carcinoma. Exp Ther Med. 2017;13(3):1064‐1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Dourado MR, de Oliveira CE, Sawazaki‐Calone I, Sundquist E, Coletta RD, Salo T. Clinicopathologic significance of ROCK2 expression in oral squamous cell carcinomas. J Oral Pathol Med. 2018;47(2):121‐127. [DOI] [PubMed] [Google Scholar]
- 95. Clark K, Langeslag M, Figdor CG, van Leeuwen FN. Myosin II and mechanotransduction: A balancing act. Trends Cell Biol. 2007;17(4):178‐186. [DOI] [PubMed] [Google Scholar]
- 96. Huveneers S, Danen EH. Adhesion signaling—crosstalk between integrins, Src and Rho. J Cell Sci. 2009;122(Pt 8):1059‐1069. [DOI] [PubMed] [Google Scholar]
- 97. Marjoram RJ, Lessey EC, Burridge K. Regulation of RhoA activity by adhesion molecules and mechanotransduction. Curr Mol Med. 2014;14(2):199‐208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Nobes CD, Hall A. Rho GTPases control polarity, protrusion, and adhesion during cell movement. J Cell Biol. 1999;144(6):1235‐1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Ridley AJ. Rho GTPases and cell migration. J Cell Sci. 2001;114(Pt 15):2713‐2722. [DOI] [PubMed] [Google Scholar]
- 100. Balaban NQ, Schwarz US, Riveline D, et al. Force and focal adhesion assembly: A close relationship studied using elastic micropatterned substrates. Nat Cell Biol. 2001;3(5):466‐472. [DOI] [PubMed] [Google Scholar]
- 101. Rape AD, Guo WH, Wang YL. The regulation of traction force in relation to cell shape and focal adhesions. Biomaterials. 2011;32(8):2043‐2051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Stricker J, Aratyn‐Schaus Y, Oakes PW, Gardel ML. Spatiotemporal constraints on the force‐dependent growth of focal adhesions. Biophys J. 2011;100(12):2883‐2893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Sakurai‐Yageta M, Recchi C, Le Dez G, et al. The interaction of IQGAP1 with the exocyst complex is required for tumor cell invasion downstream of Cdc42 and RhoA. J Cell Biol. 2008;181(6):985‐998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Roh‐Johnson M, Bravo‐Cordero JJ, Patsialou A, et al. Macrophage contact induces RhoA GTPase signaling to trigger tumor cell intravasation. Oncogene. 2014;33(33):4203‐4212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Hwang YS, Lee J, Zhang X, Lindholm PF. Lysophosphatidic acid activates the RhoA and NF‐kappaB through Akt/IkappaBalpha signaling and promotes prostate cancer invasion and progression by enhancing functional invadopodia formation. Tumour Biol. 2016;37(5):6775‐6785. [DOI] [PubMed] [Google Scholar]
- 106. Ma X, Choudhury SN, Hua X, Dai Z, Li Y. Interaction of the oncogenic miR‐21 microRNA and the p53 tumor suppressor pathway. Carcinogenesis. 2013;34(6):1216‐1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Min BM, Baek JH, Shin KH, Gujuluva CN, Cherrick HM, Park NH. Inactivation of the p53 gene by either mutation or HPV infection is extremely frequent in human oral squamous cell carcinoma cell lines. Eur J Cancer B Oral Oncol. 1994;30B(5):338‐345. [DOI] [PubMed] [Google Scholar]
- 108. Zhou G, Wang J, Zhao M, et al. Gain‐of‐function mutant p53 promotes cell growth and cancer cell metabolism via inhibition of AMPK activation. Mol Cell. 2014;54(6):960‐974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Scott RW, Hooper S, Crighton D, et al. LIM kinases are required for invasive path generation by tumor and tumor‐associated stromal cells. J Cell Biol. 2010;191(1):169‐185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Bravo‐Cordero JJ, Oser M, Chen X, Eddy R, Hodgson L, Condeelis J. A novel spatiotemporal RhoC activation pathway locally regulates cofilin activity at invadopodia. Curr Biol. 2011;21(8):635‐644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Bravo‐Cordero JJ, Hodgson L, Condeelis JS. Spatial regulation of tumor cell protrusions by RhoC. Cell Adh Migr. 2014;8(3):263‐267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Semprucci E, Tocci P, Cianfrocca R, et al. Endothelin a receptor drives invadopodia function and cell motility through the beta‐arrestin/PDZ‐RhoGEF pathway in ovarian carcinoma. Oncogene. 2016;35(26):3432‐3442. [DOI] [PubMed] [Google Scholar]
- 113. Murphy DA, Courtneidge SA. The 'ins' and 'outs' of podosomes and invadopodia: characteristics, formation and function. Nature Reviews. 2011;12(7):413‐426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Beaty BT, Condeelis J. Digging a little deeper: The stages of invadopodium formation and maturation. Eur J Cell Biol. 2014;93(10–12):438‐444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Chernock RD. Morphologic features of conventional squamous cell carcinoma of the oropharynx: ‘Keratinizing’ and ‘nonkeratinizing’ histologic types as the basis for a consistent classification system. Head Neck Pathol. 2012;6(Suppl 1):S41‐S47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Westra WH. The morphologic profile of HPV‐related head and neck squamous carcinoma: implications for diagnosis, prognosis, and clinical management. Head Neck Pathol. 2012;6(Suppl 1):S48‐S54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Lewis JS Jr. Morphologic diversity in human papillomavirus‐related oropharyngeal squamous cell carcinoma: Catch me if you can! Mod Pathol. 2017;30(s1):S44‐S53. [DOI] [PubMed] [Google Scholar]
- 118. Pai SI, Westra WH. Molecular pathology of head and neck cancer: Implications for diagnosis, prognosis, and treatment. Annu Rev Pathol. 2009;4(1):49‐70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Paszek MJ, Zahir N, Johnson KR, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell. 2005;8(3):241‐254. [DOI] [PubMed] [Google Scholar]
- 120. Jaalouk DE, Lammerding J. Mechanotransduction gone awry. Nature Reviews. 2009;10(1):63‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Kai F, Laklai H, Weaver VM. Force matters: Biomechanical regulation of cell invasion and migration in disease. Trends Cell Biol. 2016;26(7):486‐497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Furukawa MK, Kubota A, Hanamura H, Furukawa M. Clinical application of real‐time tissue elastography to head and neck cancer‐evaluation of cervical lymph node metastasis with real‐time tissue elastography. Nihon Jibiinkoka Gakkai Kaiho. 2007;110(7):503‐505. [DOI] [PubMed] [Google Scholar]
- 123. Bhatia KS, Rasalkar DD, Lee YP, et al. Real‐time qualitative ultrasound elastography of miscellaneous non‐nodal neck masses: Applications and limitations. Ultrasound Med Biol. 2010;36(10):1644‐1652. [DOI] [PubMed] [Google Scholar]
- 124. Furukawa MK, Furukawa M. Diagnosis of lymph node metastases of head and neck cancer and evaluation of effects of chemoradiotherapy using ultrasonography. Int J Clin Oncol. 2010;15(1):23‐32. [DOI] [PubMed] [Google Scholar]
- 125. Bhatia K, Tong CS, Cho CC, Yuen EH, Lee J, Ahuja AT. Reliability of shear wave ultrasound elastography for neck lesions identified in routine clinical practice. Ultraschall Med. 2012;33(5):463‐468. [DOI] [PubMed] [Google Scholar]
- 126. Bhatia KS, Lee YY, Yuen EH, Ahuja AT. Ultrasound elastography in the head and neck. Part II. Accuracy for malignancy. Cancer imaging: The official publication of the international Cancer imaging. Society. 2013;13(2):260‐276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Kaluzny J, Kopec T, Szczepanek‐Parulska E, et al. Shear wave elastography: A new noninvasive tool to assess the intensity of fibrosis of irradiated salivary glands in head and neck cancer patients. Biomed Res Int. 2014;2014:157809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Lehn CN, Rapoport A. The desmoplastic lymph node reaction as a prognostic factor of cancer of the tongue and floor of the mouth. Sao Paulo Med J. 1994;112(3):591‐596. [DOI] [PubMed] [Google Scholar]
- 129. Rosenthal E, McCrory A, Talbert M, Young G, Murphy‐Ullrich J, Gladson C. Elevated expression of TGF‐beta1 in head and neck cancer‐associated fibroblasts. Mol Carcinog. 2004;40(2):116‐121. [DOI] [PubMed] [Google Scholar]
- 130. Lentsch EJ, Goudy S, Sosnowski J, Major S, Bumpous JM. Microvessel density in head and neck squamous cell carcinoma primary tumors and its correlation with clinical staging parameters. Laryngoscope. 2006;116(3):397‐400. [DOI] [PubMed] [Google Scholar]
- 131. Kawashiri S, Tanaka A, Noguchi N, et al. Significance of stromal desmoplasia and myofibroblast appearance at the invasive front in squamous cell carcinoma of the oral cavity. Head Neck. 2009;31(10):1346‐1353. [DOI] [PubMed] [Google Scholar]
- 132. Canel M, Secades P, Garzon‐Arango M, et al. Involvement of focal adhesion kinase in cellular invasion of head and neck squamous cell carcinomas via regulation of MMP‐2 expression. Br J Cancer. 2008;98(7):1274‐1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Le QT, Harris J, Magliocco AM, et al. Validation of lysyl oxidase as a prognostic marker for metastasis and survival in head and neck squamous cell carcinoma: Radiation therapy oncology group trial 90‐03. J Clin Oncol. 2009;27(26):4281‐4286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. de Vicente JC, Rodrigo JP, Rodriguez‐Santamarta T, Lequerica‐Fernandez P, Allonca E, Garcia‐Pedrero JM. Cortactin and focal adhesion kinase as predictors of cancer risk in patients with premalignant oral epithelial lesions. Oral Oncol. 2012;48(7):641‐646. [DOI] [PubMed] [Google Scholar]
- 135. Theocharis S, Klijanienko J, Giaginis C, Alexandrou P, Patsouris E, Sastre‐Garau X. FAK and Src expression in mobile tongue squamous cell carcinoma: Associations with clinicopathological parameters and patients survival. J Cancer Res Clin Oncol. 2012;138(8):1369‐1377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Rosado P, Lequerica‐Fernandez P, Pena I, Alonso‐Duran L, de Vicente JC. In oral squamous cell carcinoma, high FAK expression is correlated with low P53 expression. Virchows Arch. 2012;461(2):163‐168. [DOI] [PubMed] [Google Scholar]
- 137. de Vicente JC, Rosado P, Lequerica‐Fernandez P, Allonca E, Villallain L, Hernandez‐Vallejo G. Focal adhesion kinase overexpression: Correlation with lymph node metastasis and shorter survival in oral squamous cell carcinoma. Head Neck. 2013;35(6):826‐830. [DOI] [PubMed] [Google Scholar]
- 138. Chiu YW, Liou LY, Chen PT, et al. Tyrosine 397 phosphorylation is critical for FAK‐promoted Rac1 activation and invasive properties in oral squamous cell carcinoma cells. Lab Invest; a Journal of Technical Methods and Pathology. 2016;96(3):296‐306. [DOI] [PubMed] [Google Scholar]
- 139. Hsin CH, Chou YE, Yang SF, et al. MMP‐11 promoted the oral cancer migration and Fak/Src activation. Oncotarget. 2017;8(20):32783‐32793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Parekh A, Weaver AM. Regulation of invadopodia by mechanical signaling. Exp Cell Res. 2016;343(1):89‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. El‐Mofty SK, Zhang MQ, Davila RM. Histologic identification of human papillomavirus (HPV)‐related squamous cell carcinoma in cervical lymph nodes: a reliable predictor of the site of an occult head and neck primary carcinoma. Head Neck Pathol. 2008;2(3):163‐168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Lopez F, Rodrigo JP, Silver CE, et al. Cervical lymph node metastases from remote primary tumor sites. Head Neck. 2016;38(Suppl 1):E2374‐E2385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Lydiatt WM, Patel SG, O'Sullivan B, et al. Head and neck cancers‐major changes in the American joint committee on cancer eighth edition cancer staging manual. CA Cancer J Clin. 2017;67(2):122‐137. [DOI] [PubMed] [Google Scholar]
- 144. Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363(1):24‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Huang SH, Perez‐Ordonez B, Liu FF, et al. Atypical clinical behavior of p16‐confirmed HPV‐related oropharyngeal squamous cell carcinoma treated with radical radiotherapy. Int J Radiat Oncol Biol Phys. 2012;82(1):276‐283. [DOI] [PubMed] [Google Scholar]
- 146. O'Sullivan B, Huang SH, Siu LL, et al. Deintensification candidate subgroups in human papillomavirus‐related oropharyngeal cancer according to minimal risk of distant metastasis. J Clin Oncol. 2013;31(5):543‐550. [DOI] [PubMed] [Google Scholar]
- 147. Maxwell JH, Grandis JR, Ferris RL. HPV‐associated head and neck Cancer: Unique features of epidemiology and clinical management. Annu Rev Med. 2016;67(1):91‐101. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Fig. S1. Relative expression (fold change) in mRNA levels of E6 and E7 in HPV− SCC‐25 and HPV+ SCC‐47 cells evaluated with quantitative real‐time PCR which was performed in triplicate for each experiment and overall for 3 independent experiments. Expression levels of E6 and E7 in HPV+ SCC‐47 cells were statistically similar to those in the SCC‐25 E6/E7 OE in Figure 2A. All data are presented as box and whisker plots with the black lines indicating the medians, the whiskers representing the 10th and 90th percentiles, and * indicating p < 0.05.
