The molecular mechanisms behind the aggressiveness of nasopharyngeal carcinoma (NPC), a highly invasive and metastatic head and neck malignancy, have not been made clear. In this study investigating these mechanisms, guanine nucleotide-binding protein α12 subunit (Gα12) signaling was found by microarray analysis to be increased in primary NPC cells and NPC-derived cell lines. Using small interfering RNA to knock down Gα12 in NPC cells resulted in a reduction in cell migration and invasion as well as a reversal in fibroblastoid morphology. Using microarray analysis, we also found a reduction in expression of key actin dynamics regulators and several epithelial-to-mesenchymal transition–related genes in Gα12-depleted NPC cells. Knocking down one of those genes, IQ motif containing GTPase activating protein 1, reduced the migration and formation of adherens junctions and reversed the fibroblastoid morphology of NPC cells, as knocking down Gα12 was found to do. Immunohistochemical analysis found NPC tumors to have significantly greater levels of Gα12 protein than the normal basal epithelial cells. Quantitative real-time PCR analysis revealed a significant correlation between Gα12 mRNA levels and NPC lymph node metastasis. Together, our findings support a model in which activation of Gα12 signaling promotes tumorigenesis and progression of NPC by modulating actin cytoskeleton reorganization and expression of epithelial-to-mesenchymal transition–related genes. [Cancer Res 2009;69(15):6122–30]

In the western world, nasopharyngeal carcinoma (NPC) is a rare head and neck malignancy; in Southeast Asia, it is highly prevalent. Many factors, including viral, genetic, dietary habit, and environment, have been implicated in the etiology of NPC (1). Undifferentiated NPC, the most common type, is characterized by overwhelming lymphocytic infiltration. The association between it and EBV distinguishes NPC from head and neck squamous cell carcinomas, which are often associated with human papillomavirus (2, 3).

NPC is more metastatic than other head and neck carcinomas. Although tumor invasion and metastasis is thought to involve deregulation of cell motility, the mechanisms underlying this dysregulation remain largely unknown. Several extracellular factors, including cytokines and growth factors, may regulate cell motility by modulating the reorganization of cytoskeleton, and many of these extracellular signals are mediated by G protein–coupled receptors (GPCR). The aberrant expression of GPCR has been associated with tumorigenesis of adrenocortical carcinoma (4), and the ectopic expression of vGPCR (a constitutively active GPCR) has been found to help tumor cells escape immune surveillance (5, 6). Moreover, GPCR signaling pathways may play an important role in tumorigenesis in various carcinomas, including non–small cell lung cancer, breast cancer, prostate cancer, melanoma, gastric cancer, diffused large B-cell lymphoma (7), and head and neck squamous cell carcinoma (8, 9). There is no report, however, of the contribution of GPCR to NPC tumorigenesis.

GPCR signaling is modulated primarily through the activation of heterotrimeric G proteins, which are composed of α and βγ subunits. The Gα subunit has four subfamilies: Gs, Gi, Gq, and G12. To activate Gα12, the receptor needs to be bound to ligands, such as lysophosphatidic acid, sphingosine-1-phosphate, thrombin, or thromboxane A2 (10). The activation of Gα12 triggers RhoA-dependent downstream signaling, regulating actin stress fiber formation, focal adhesion assembly, and phosphorylation of focal adhesion-associated proteins, including FAK, p130 CAS, and paxillin (11, 12). Gα12 mediates divergent physiologic responses, including cytoskeleton organization, cell growth, and activation of mitogen-activated protein kinase and phospholipase D and C, by activating small G protein Rho (13, 14). Furthermore, Gα12 has also been found to regulate cell migration through E-cadherin/β-catenin (14) and tight junction formation through Hsp90/Src signaling (1517).

12 has been found to activate oncogenic transformation and tumor cell invasion (1820), although it is not known whether it is also involved in epithelial-to-mesenchymal transition (EMT). EMT is a pathophysiologic process of tumor cells characterized by loss of epithelial junctional proteins and cell polarity and gain of mesenchymal markers. IQ motif containing GTPase activating protein 1 (IQGAP1), an EMT-related factor, is an important effector of Rac1 and Cdc42 for actin dynamics and cell migration (21, 22). Overexpression of IQGAP1 has been associated with decreases in E-cadherin-based cell-cell adhesion (23) and depletion of IQGAP1 with decreases in cell migration and actin meshwork (24). Together, these findings suggest that the regulation of IQGAP1 expression may be critical to the induction of EMT; therefore, it may be important to investigate the molecular mechanism that regulates IQGAP1 during the process of EMT.

In this study, our microarray studies suggest that the Gα12 signaling might be dysregulated during the tumorigenesis and progression of NPC. Functionally, changes in Gα12 expression in NPC cells influenced many cellular activities associated with NPC cell invasiveness and metastasis. Further microarray studies of NPC cells in which Gα12 was knocked down found changes in expression of genes involved in actin cytoskeleton reorganization and EMT. IQGAP1 was found to be a downstream effector of Gα12 signaling in regulating epithelial morphology. In NPC patients, we found a significant correlation between Gα12 level and NPC tumorigenesis and lymph node metastasis. Together, these studies suggest that Gα12 signaling plays a critical role in the tumorigenesis and metastasis of NPC.

Clinical samples. Nine NPC samples and 32 normal nasopharyngeal samples were used for microarray analysis. Clinical data on patients with NPC are reported in Supplementary Table S1. Informed consent was obtained from all participants.

Cell culture. Primary nasopharyngeal epithelial cells obtained from explant cultures were cultured with defined keratinocyte serum-free medium (Invitrogen). Preconfluent primary nasopharyngeal cells were harvested during passages 3 to 10 for RNA extraction. Two NPC-derived cell lines, CNE1 (25) and NPC-TW06 (26) cells, were cultured in DMEM supplemented with 10% fetal bovine serum.

Immunofluorescence and immunohistochemistry. Normal nasopharyngeal epithelium and NPC cells were fixed and immunostained using mouse anti-human cytokeratin AE1/AE3 (DAKO), mouse anti-CD90/Thy-1 (Dianova), rabbit anti-IQGAP1 (Santa Cruz Biotechnology), mouse anti-paxillin (BD Transduction Laboratories), mouse anti-vimentin (Sigma), and mouse anti-vinculin (Sigma) as primary antibodies. Rhodamine-phalloidin was used to label F-actin. Immunohistochemical staining of tissue sections was done as described previously (27) using anti-Gα12 primary antibody (1:100; sc-409; Santa Cruz Biotechnology). Breast and prostate cancer samples were used as the positive controls for Gα12 staining (18, 19). The primary antibody was omitted in the negative controls. The results of immunohistochemistry were reviewed by two independent observers. Staining intensity was defined as “−,” “+,” “++,” and “+++” scoring (negative or <20%, 21-50%, 51-70%, and >71% of positive cells, respectively).

RNA extraction and cDNA microarray experiments. Total RNA was extracted with the RNeasy kit (Qiagen) and analyzed on an Agilent 2100 Bioanalyzer. A pool of total RNA from 32 normal nasopharyngeal epithelium cell strains served as reference. Labeled cDNA samples were hybridized on a customized microarray containing 46,657 cDNA clones (IMAGE consortium), representing ∼26,000 Unigene clusters. The arrays were scanned on a GenePix 4000B scanner and analyzed with the GenePix software package (Axon Instruments). The microarray data can be found at GEO (accession no. GSE14262).

Cell transfection. For small interfering RNA (siRNA) transfection, CNE1 and NPC-TW06 cells were seeded at a density of 5 × 104 per well in the 24-well culture plate 24 h before transfection with either Gα12- or IQGAP1-siRNA or a nontargeting control siRNA (Dharmacon) at a concentration of 90 nmol/L. For functional assays, cells were collected 1 to 3 days post-transfection. For microarray experiments, RNA was extracted from NPC-TW06 cells 48 h post-transfection. For exogenous protein expression, NPC cells were transiently transfected with one of the following expression plasmids, full-length IQGAP1 (pCMV2-Flag-FL-IQGAP1), a COOH-terminal fragment of IQGAP1 (pCMV2-Flag-IQDN4; ref. 28), wild-type Gα12 (pcDNA3-Gα12-WT), or a constitutively active Gα12 (pcDNA3-Gα12Q231L), using Fugene HD transfection reagents (Roche). The transfection efficiency was measured by cotransfection with pEGFP-C1. The Gα12-WT and Gα12Q231L plasmids were obtained from the Missouri S&T cDNA Resource Center.

Western blot. Equal amounts of protein were separated by SDS-PAGE and transferred to polyvinylidene fluoride membranes. Blots were probed with primary antibodies against Gα12 (Santa Cruz Biotechnology), IQGAP1 (Santa Cruz Biotechnology), LAMB3 (Santa Cruz Biotechnology), E-cadherin (BD Transduction Laboratories), vinculin (Sigma), paxillin (BD Transduction Laboratories), vimentin (Sigma), and glyceraldehyde-3-phosphate dehydrogenase (Lab Frontier) followed by horseradish peroxidase–conjugated secondary antibody and developed with enhanced chemiluminescence detection reagents (Amersham Life Science).

Wound-healing assay. At 48 h post-transfection with Gα12-siRNA, IQGAP1-siRNA, or a nontargeting control siRNA, we scratched confluent cell layers with plastic tips. At 0 and 24 h later, we photographed wound widths under a phase-contrast microscope. All experiments were done in triplicate.

Matrigel invasion assay. Invasion capacity was done in an invasion chamber consisting of cell culture inserts with 8 μm pore-sized PET membranes coated with 80 μg Matrigel (BD Biosciences) according to the manufacturer's instructions. CNE1 and NPC-TW06 cells transfected with Gα12-siRNA or a control siRNA were harvested 48 h post-transfection. Aliquots of resuspended cells were seeded into the top chamber. After 30 h incubation at 37°C, the invaded cells were stained with 1% gentian violet. At least five distinct fields were counted for each duplicate chamber.

Quantitative real-time reverse transcription-PCR. Quantitative real-time reverse transcription-PCR (QRT-PCR) was done in a LightCycler PCR system (Roche) with the FastStart DNA Master SYBR Green I Kit (Roche Applied Science). Refer to Supplementary Table S2 for the primers used. The expression level was corrected relative to the level of MAP4, the expression level of which remained unchanged across all microarray experiments. Relative quantification was calculated using RelQuant software (Roche).

12 signaling activated in NPC tumorigenesis. Primary epithelial populations isolated from nasopharyngeal biopsies were used for microarray analysis. We compared the differential gene expression profiles in 9 primary NPC cell strains and 5 NPC-derived cell lines against a pool of 32 normal primary normal nasopharyngeal epithelium cell strains. By selecting significantly (P ≤ 0.05) enriched gene sets, we identified the gene ontology terms and classified them by k-means clustering to identify the cellular processes commonly altered during tumorigenesis or specifically altered in advanced NPC cells (data not shown). A large number of genes involved in Gα12 signaling were up-regulated in NPC (Fig. 1). These genes included 12, ARHGEF12, RhoA, SLC9A1, ROCK1, PFN1, and JNK. This up-regulation suggests that Gα12 signaling might be activated in the tumorigenesis of NPC.

Figure 1.

Functional genomic analysis of primary NPC cells suggests the possibility that activated Gα12/13 signaling pathway plays a role in NPC tumorigenesis. Diagram of representative genes associated with Gα12/13 signaling pathway. The 1.2- and 1.5-fold differences were used as minimum cutoff points when selecting differentially expressed genes in primary NPC cells and NPC-derived cell lines, respectively. Red boxes, positive fold change; green boxes, negative fold change of expression in the majority of NPC samples compared with the normal controls; white boxes, genes that had no significant fold change.

Figure 1.

Functional genomic analysis of primary NPC cells suggests the possibility that activated Gα12/13 signaling pathway plays a role in NPC tumorigenesis. Diagram of representative genes associated with Gα12/13 signaling pathway. The 1.2- and 1.5-fold differences were used as minimum cutoff points when selecting differentially expressed genes in primary NPC cells and NPC-derived cell lines, respectively. Red boxes, positive fold change; green boxes, negative fold change of expression in the majority of NPC samples compared with the normal controls; white boxes, genes that had no significant fold change.

Close modal

12 depletion inhibited cell migration and invasion and reversed fibroblastic changes in NPC cells. To test whether the increased Gα12 was critical to increased NPC aggressiveness, we used siRNA to knock down endogenous Gα12 and assessed the effects on cell mobility. Compared with the siRNA controls, Gα12 mRNA level in both CNE1 and NPC-TW06 cells were reduced by >80% by Gα12-siRNA at 24 h post-transfection (Fig. 2A). Wound-healing assay showed that rate of wound healing was markedly less in Gα12-depleted NPC cells than in siRNA controls. At 24 h post-scratching, the Gα12-depleted cells had a wound gap that was ∼89% that of open gap measured at 0 h. Mock control cells and control siRNA cells had a gap of 33% and 39%, respectively (Fig. 2B). Matrigel invasion assay was done to evaluate the invasion ability of the NPC cells. The Gα12-siRNA-transfected NPC cells were less invasive than siRNA controls at 30 h after plating (Fig. 2C,, left). The difference between their invasiveness was significant (P < 0.0001; Fig. 2C , right). These results suggest that Gα12 may function to regulate the invasive ability of NPC cells.

Figure 2.

Depletion of Gα12 expression impairment of NPC cell mobility. A, silencing effect of Gα12-siRNA occurred in CNE1 and NPC-TW06 cells. Top, QRT-PCR analysis was done at 24 and 48 h post-transfection. Relative expression levels were normalized to an internal control. Western blot of Gα12 in NPC cells transfected with Gα12-siRNA or control siRNA. Bottom, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a loading control. B, wound-healing assays. Top and bottom, photographed with a ×20 phase-contrast objective at initial wounding and 24 h post-wounding, respectively. Wound gap at 0 h after scratching was regarded as 100% (top, white dashed line). C, Matrigel invasion assays of NPC cells transfected with control siRNA or Gα12-siRNA. Invaded cells were stained with gentian violet at 30 h after plating. Quantitative results of the Matrigel invasion assays (right). Relative percentage of invaded NPC cells was presented as mean ± SE of three experiments. *, P < 0.0001 (paired t test).

Figure 2.

Depletion of Gα12 expression impairment of NPC cell mobility. A, silencing effect of Gα12-siRNA occurred in CNE1 and NPC-TW06 cells. Top, QRT-PCR analysis was done at 24 and 48 h post-transfection. Relative expression levels were normalized to an internal control. Western blot of Gα12 in NPC cells transfected with Gα12-siRNA or control siRNA. Bottom, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a loading control. B, wound-healing assays. Top and bottom, photographed with a ×20 phase-contrast objective at initial wounding and 24 h post-wounding, respectively. Wound gap at 0 h after scratching was regarded as 100% (top, white dashed line). C, Matrigel invasion assays of NPC cells transfected with control siRNA or Gα12-siRNA. Invaded cells were stained with gentian violet at 30 h after plating. Quantitative results of the Matrigel invasion assays (right). Relative percentage of invaded NPC cells was presented as mean ± SE of three experiments. *, P < 0.0001 (paired t test).

Close modal

A spindle-like fibroblastic morphology generally correlates with increased cell mobility in many types of epithelial cancers. To find out whether depleting Gα12 in CNE1 and NPC-TW06 cells could reverse fibroblastic changes in epithelial cell morphology, we examined their morphologies and actin cytoskeletons. At 48 h post-transfection of siRNA, the morphology of Gα12-depleted cells, which were flattened and multipolar and had a larger cell-cell contact area, was different from that of either the mock cells or the cells transfected with a control siRNA, which were small and round bipolar with a spindle-like appearance (Supplementary Fig. S1A). Moreover, the spike-like F-actin protrusion at the bipolar ends of mock control cells or cells transfected with control siRNA became less common or even disappeared in Gα12-depleted cells (Supplementary Fig. S1B, compare inserts in middle and right). Together, these results suggest that Gα12 plays a role in the invasive/metastatic potential of NPC cells.

Reduction of Gα12 down-regulated key actin cytoskeleton regulators. We performed microarray analysis to identify transcripts regulated by the depletion of Gα12 in NPC-TW06 cells. Most differentially regulated genes involved in actin polymerization and stabilization, focal adhesion assembly, and actomyosin-mediated contractility were down-regulated in the Gα12-depleted cells (Fig. 3A).

Figure 3.

Depletion of Gα12 alteration of actin cytoskeletal signaling in NPC cells. A, dysregulated genes (fold change ≥2) involved in actin dynamics. Red and green nodes, increased and decreased fold changes, respectively. The greater the color intensity, the greater the fold change. Gray nodes, no significant changes in expression (fold change ≤2); white nodes, missing data. Different shapes of nodes represent different functional classes of gene products. Edges represent the functional links between them. B, k-means clustering of 95 differentially expressed genes involved in actin reorganization in NPC-TW06 cells transfected with Gα12-siRNA.

Figure 3.

Depletion of Gα12 alteration of actin cytoskeletal signaling in NPC cells. A, dysregulated genes (fold change ≥2) involved in actin dynamics. Red and green nodes, increased and decreased fold changes, respectively. The greater the color intensity, the greater the fold change. Gray nodes, no significant changes in expression (fold change ≤2); white nodes, missing data. Different shapes of nodes represent different functional classes of gene products. Edges represent the functional links between them. B, k-means clustering of 95 differentially expressed genes involved in actin reorganization in NPC-TW06 cells transfected with Gα12-siRNA.

Close modal

To further understand how Gα12 modulated actin reorganization, we extracted 95 differentially expressed genes involved in actin cytoskeleton signaling (Supplementary Table S3) from mock control cells and then compared them with those transcripts in Gα12-depleted cells. Using k-means clustering algorithms, we identified genes that have distinct expression patterns (Fig. 3B). Many of the actin-related differentially expressed genes were found to have opposite expression patterns in cells treated with Gα12-siRNA compared with mock cells (Fig. 3B , bottom). Twenty-nine genes were suppressed and only 7 genes were up-regulated by the reduction of Gα12. To validate our microarray results, we performed QRT-PCR on 10 selected genes, 12, IQGAP1, IRS1, ARHGEF12, ARPC5, v-MYC, WASL, FYN, JAK1, and MAP4, in Gα12-depleted NPC cells and in respective control cells. QRT-PCR results corresponded well to those of microarray analysis (data no shown).

IQGAP1 expression was regulated by Gα12 and depletion of IQGAP1 impaired migration of NPC cells. Because increases in the motility and invasiveness of cancer cells are indicative of EMT and are closely dependent on active remodeling of actin cytoskeleton, we identified 7 EMT-related genes (including IQGAP1, ARHGEF12, JAK1, IRS1, ARPC5, v-MYC, and WASL) from the above-mentioned actin-related differentially expressed genes. The QRT-PCR analysis confirmed that the Gα12-siRNA drastically down-regulated their expressions in NPC-TW06 cells. A known Gα12-regulated gene, RhoA, was used as an informative positive control for the assay (Fig. 4A). In contrast, overexpression of Gα12-WT or Gα12Q231L in NPC-TW06 cells increased the expression of IQGAP1 (Fig. 4B). Furthermore, the immunostaining of IQGAP1 in cells overexpressing Gα12-WT or Gα12Q231L also showed stronger signal intensities in the cell membrane and bipolar regions than those found in control cells (data not shown).

Figure 4.

12 regulates expressions of IQGAP1 and other EMT-related markers and depletion of IQGAP1 impairs migration of NPC cells. A, siRNA-mediated depletion of endogenous Gα12 decreases expression of IQGAP1 and other EMT-associated genes in NPC cells. QRT-PCR analysis was done at 48 h after transfection. Relative expression levels were normalized to a consistently expressed internal control (MAP4). Mean ± SE of three experiments. *, P < 0.05, significantly different from siControl groups. B, overexpression of a Gα12-WT or Gα12Q231L in NPC-TW06 cells increased the IQGAP1 expression levels. mRNA expression was analyzed at 72 h after transfection. QRT-PCR values are expressed as mean ± SE of three experiments. *, P < 0.05. C, Western blot of EMT-related markers in NPC cells transfected with Gα12-siRNA, IQGAP1-siRNA, or expression plasmids of IQGAP1-WT, Gα12-WT, or Gα12Q231L. Glyceraldehyde-3-phosphate dehydrogenase was used as a loading control. D, repression of IQGAP1 by siRNA impaired NPC cell migration as measured by wound-healing assays. Cells were photographed with a ×20 phase-contrast objective at initial wounding and 24 h post-wounding, respectively. Wound gap at 0 h after scratching was considered 100% (top, white line).

Figure 4.

12 regulates expressions of IQGAP1 and other EMT-related markers and depletion of IQGAP1 impairs migration of NPC cells. A, siRNA-mediated depletion of endogenous Gα12 decreases expression of IQGAP1 and other EMT-associated genes in NPC cells. QRT-PCR analysis was done at 48 h after transfection. Relative expression levels were normalized to a consistently expressed internal control (MAP4). Mean ± SE of three experiments. *, P < 0.05, significantly different from siControl groups. B, overexpression of a Gα12-WT or Gα12Q231L in NPC-TW06 cells increased the IQGAP1 expression levels. mRNA expression was analyzed at 72 h after transfection. QRT-PCR values are expressed as mean ± SE of three experiments. *, P < 0.05. C, Western blot of EMT-related markers in NPC cells transfected with Gα12-siRNA, IQGAP1-siRNA, or expression plasmids of IQGAP1-WT, Gα12-WT, or Gα12Q231L. Glyceraldehyde-3-phosphate dehydrogenase was used as a loading control. D, repression of IQGAP1 by siRNA impaired NPC cell migration as measured by wound-healing assays. Cells were photographed with a ×20 phase-contrast objective at initial wounding and 24 h post-wounding, respectively. Wound gap at 0 h after scratching was considered 100% (top, white line).

Close modal

Because depletion of Gα12 resulted in a reduction in mesenchymal-like cell formation and IQGAP1 has been previously implicated in the induction of EMT (22), we wanted to investigate whether depletion of Gα12 in NPC cells would influence the expression of EMT markers. Result of Western blot analysis showed that the EMT markers, including LAMB3, vimentin, and paxillin, were down-regulated by depletion of Gα12 and up-regulated by overexpression of a Gα12-WT or Gα12Q231L in NPC-TW06 cells. However, the epithelial markers, including E-cadherin and vinculin, were not noticeably affected by Gα12-siRNA (Fig. 4C), suggesting that the reduction of mesenchymal-like cell formation does not necessarily up-regulate the expression of epithelial cell markers. To test whether IQGAP1 also contributed to maintenance of EMT in NPC cells, we depleted IQGAP1 by siRNA and found a similar effect to that seen by Gα12-siRNA. The protein levels of LAMB3, vimentin, and paxillin were down-regulated by IQGAP1-siRNA and up-regulated by overexpression of IQGAP1 (Fig. 4C). In addition, the epithelial cell markers, the protein levels of which were found to be unaffected by the Gα12 expression level, were markedly up-regulated by IQGAP1-siRNA (Fig. 4C , right). Together, these results suggest that Gα12 and IQGAP1 may act together to regulate EMT in NPC cells.

To further understand the role of IQGAP1 in the invasive potential of NPC cells, we depleted endogenous IQGAP1 in NPC-TW06 cells. As was observed in Gα12-depleted cells (Fig. 2B), the wound-healing assay clearly showed that the migration ability was markedly decreased by the IQGAP1-siRNA at 24 h post-wounding compared with control cells (Fig. 4D). Collectively, these biochemistry, morphology, and functional assays indicate that IQGAP1 plays a critical role in invasive potential of NPC cells.

IQGAP1 is a key downstream effector of Gα12 for the conversion from epithelial to fibroblastoid morphology in cultured NPC cells. Because both IQGAP1 and vinculin are required for the assembly of adherens junctions, which are important for cell migration (29), we wanted to analyze whether suppression of Gα12 would alter the expressions or subcellular localizations of IQGAP1 and vinculin. The immunocytochemical results showed that Gα12-depleted cells had less IQGAP1 and vinculin immunofluorescence than the control cells. The depletion of IQGAP1 also reduced vinculin immunofluorescence (Fig. 5A,, bottom right). Particularly, we found marked reductions in vinculin in the cell-matrix junctions and IQGAP1 in the cell-cell contacts in cells treated with the siRNA for Gα12 or IQGAP1 (Fig. 5A). In addition, the immunofluorescent signals of vimentin and paxillin (two known mesenchymal markers) in Gα12- or IQGAP1-siRNA-treated cells were found dramatically decreased (Supplementary Fig. S2A). Furthermore, using phase-contrast microscopy, we observed cell shape changes in IQGAP1-depleted cells. Their rounded, bipolar, spindle-shaped, fibroblastoid appearance changed to a more epithelial-like, large, flat, spread-out appearance, findings that were similar to those we observed when knocking down Gα12 with siRNA (Fig. 5B , top). These results suggest that the cell-cell contacts, cell-matrix junctions, and mesenchymal morphology are coregulated by Gα12 and IQGAP1 in NPC cells.

Figure 5.

IQGAP1 is a potential downstream effector of Gα12 in NPC cells. A, both Gα12 and IQGAP1 were required for formation of adherens junctions. Immunofluorescence staining of IQGAP1 and vinculin in NPC-TW06 cells transfected with Gα12-siRNA, IQGAP1-siRNA, or siControl. Arrowheads, sites of specific expression patterns. Magnification, ×1,000. B, ectopic expression of IQGAP1-WT or COOH-terminal IQGAP1 (IQDN4) partially reversed fibroblast-like morphology in cells where Gα12 was knocked down with siRNA. Phase-contrast images. Magnification, ×400.

Figure 5.

IQGAP1 is a potential downstream effector of Gα12 in NPC cells. A, both Gα12 and IQGAP1 were required for formation of adherens junctions. Immunofluorescence staining of IQGAP1 and vinculin in NPC-TW06 cells transfected with Gα12-siRNA, IQGAP1-siRNA, or siControl. Arrowheads, sites of specific expression patterns. Magnification, ×1,000. B, ectopic expression of IQGAP1-WT or COOH-terminal IQGAP1 (IQDN4) partially reversed fibroblast-like morphology in cells where Gα12 was knocked down with siRNA. Phase-contrast images. Magnification, ×400.

Close modal

Because depletion of Gα12 decreased IQGAP1 expression and depletion of IQGAP1 resulted in phenotypes similar to that found in the Gα12-depleted cells, we speculated that IQGAP1 might function as a downstream effector of Gα12 signaling. To test this hypothesis, we depleted Gα12 but simultaneously overexpressed IQGAP1-WT or COOH-terminal IQGAP1 (IQDN4; ref. 28) to determine whether this would reverse the morphologic changes induced by depletion of Gα12. Indeed, overexpression of either IQGAP1 proteins reversed, at least in part, Gα12-siRNA-induced morphologic changes (Fig. 5B , bottom) and expression level of mesenchymal markers (Supplementary Fig. S2B). Together, these results support a model in which IQGAP1 functions downstream of Gα12 in NPC cells.

Immunohistochemical assays of Gα12 expression in NPC tumorigenesis. To validate the role of Gα12 in the pathogenesis of NPC, we conducted immunohistochemical analysis of Gα12 on 50 nasopharyngeal biopsies. Gα12 immunoreactivity was predominantly negative in the normal nasopharyngeal epithelium (12 of 13), 1-positive to 2-positive in dysplasic lesions of varying severity (mild, moderate, and severe), and strongly positive in NPC (Fig. 6A). A significant association was observed between expression of Gα12 and NPC (P < 0.01). Of the 31 NPC biopsies, 58% (18 of 31) were 3-positive, 35.5% (11 of 31) were 2-positive, and 6.5% (2 of 31) were 1-positive for immunoreactivity in tumor masses (Fig. 6B). Further, expression of Gα12 was much higher in NPC than in adjacent basal layer epithelium (Supplementary Fig. S3). To further investigate if Gα12 contributed to the invasive ability of NPC, we used quantitative QRT-PCR assay to compare the transcript levels of Gα12 in NPC biopsy samples taken from patients who had neck lymph node metastasis with those who did not. We found a significant correlation between lymph node metastasis and expression of Gα12 (P < 0.05; Fig. 6C). Using microarray, biochemical, cell biology, and histopathologic approaches, we have identified and characterized the Gα12 signaling in tumorigenesis and metastasis of NPC.

Figure 6.

Expression of Gα12 is associated with NPC progression. A,12 expression in NPC tissues. Immunohistochemical staining of Gα12 protein expression in paraffin-embedded sections of nasopharyngeal tissues. Magnification ×400. B, significance of Gα12 immunohistochemistry (IHC)-positive samples. A staining was defined as positive when >20% of the cells were found to be positive for immunoreactivity: +, weak intensity; ++, moderate intensity; and +++, strong intensity. **, P < 0.01 (χ2 or two-sided Fisher's exact test). C, distribution of Gα12 mRNA expression in NPC biopsies diagnosed with or without neck lymph node (L.N.) metastasis using QRT-PCR assays. Line within the box, median value. *, P < 0.05 (Mann-Whitney U test). Expression values measured by QRT-PCR assay were normalized to an internal control (MAP4).

Figure 6.

Expression of Gα12 is associated with NPC progression. A,12 expression in NPC tissues. Immunohistochemical staining of Gα12 protein expression in paraffin-embedded sections of nasopharyngeal tissues. Magnification ×400. B, significance of Gα12 immunohistochemistry (IHC)-positive samples. A staining was defined as positive when >20% of the cells were found to be positive for immunoreactivity: +, weak intensity; ++, moderate intensity; and +++, strong intensity. **, P < 0.01 (χ2 or two-sided Fisher's exact test). C, distribution of Gα12 mRNA expression in NPC biopsies diagnosed with or without neck lymph node (L.N.) metastasis using QRT-PCR assays. Line within the box, median value. *, P < 0.05 (Mann-Whitney U test). Expression values measured by QRT-PCR assay were normalized to an internal control (MAP4).

Close modal

Although it has been suggested that some EBV-encoded genes, such as latent membrane protein 1 and 2, are important for cell motility and invasiveness of NPC cells (30, 31), the molecular mechanisms underlying the EMT and metastasis of NPC remain largely unknown. In this study, we have uncovered a Gα12-mediated pathway that plays an essential role in the tumorigenesis and progression of NPC. Specifically, we found that siRNA-induced depletion of Gα12 resulted in decreased migration, invasiveness, and reappearance of the enlarged, flat morphology of epithelial cells. Global transcriptional analysis revealed the loss of Gα12 function to be accompanied with a reorganization of the actin cytoskeleton. For example, key actin dynamic regulators involved in Cdc42-dependent and actomyosin-related cell contraction signaling pathways were repressed, supporting the hypothesis that Gα12 signaling may regulate motility and invasiveness through the modulation of actin cytoskeleton reorganization in NPC cells.

This pathway was found to possibly play a role in the regulation of EMT in NPC cells. In tumorigenesis, EMT has been specifically linked to tumor invasiveness and metastasis, making treatment difficult (32). To transform into motile fibroblastoid cells, epithelial cells need to go through changes in cell-cell and cell-matrix adhesion contacts, extracellular matrix synthesis, and actin cytoskeleton reorganization. Because Gα12 was shown by this study to be a key determinant of aggressive behavior and fibroblastoid morphology in NPC cells, we investigated whether knocking down Gα12 would reduce the expression of EMT-related genes. QRT-PCR analysis revealed that IQGAP1 and several other EMT-related genes were down-regulated by the depletion of Gα12 in NPC cells. Because previous genome-wide expression analyses of different malignancies, including breast, lung, gastric, and colon cancers, have implicated IQGAP1 dysfunction in tumorigenesis and tumor progression (3337), we wanted to determine whether depletion of IQGAP1 would contribute to inhibition of cell motility. Similar to what we observed in NPC cells lacking Gα12, depletion of IQGAP1 disrupted adherens junctions and transformed the spindle, fibroblastoid-like cells into flattened and enlarged multipolar ones. We also found a decrease in migration in IQGAP1-depleted cells. Interestingly, although several studies reported different signaling partnerships with Gα12 to be associated with cell-cell adhesion and cell migration (14), none have mentioned the role that IQGAP1 may play in this process. Our epistasis analysis suggested that IQGAP1 may be a potential downstream effector of Gα12 signaling in NPC cells. Having established the link between Gα12 and IQGAP1, we set out to investigate whether Gα12 played an important role in EMT. Both depleted and overexpressed Gα12 clearly altered protein levels or subcellular localization of EMT-related markers, suggesting that the Gα12 signaling is essential for EMT. Our immunochemical studies of 50 nasopharyngeal biopsies confirmed that Gα12 signaling played an important role in tumorigenesis and metastasis of NPC.

IQGAP1 is known to bind several specific cell-cell adhesion-related proteins, including calmodulin, actin, E-cadherin, and Cdc42, to modulate cell-cell adhesion, cell polarity, and cell migration (21). Our data showed that the depletion of either Gα12 or IQGAP1 disrupted cell-cell or cell-matrix junctions in NPC cells, suggesting both proteins might be involved in the same signaling cascade. Future studies may want to focus on how IQGAP1 participates in the Gα12-mediated tumorigenesis of NPC. The role of IQGAP1 in canonical Gα12 signaling in vivo also needs further study. To do this, it will be necessary to measure expression level of IQGAP1 and its correlation with Gα12 during the tumorigenesis and metastasis of NPC in clinical samples. Moreover, because the cross-talk between GPCR and EGFR signaling has been shown to be important for the growth and invasion of head and neck squamous cell carcinoma (8), it will be of interest to investigate whether IQGAP1 is also modulated by another GPCR or receptor of other families in NPC.

A further issue related to the association between Gα12 and NPC pathogenesis is how Gα12 expression is triggered. The role of EBV in NPC pathogenesis may provide clues regarding the connection between the induction of Gα12 signaling and NPC pathogenesis. Nearly all undifferentiated NPC cases harboring EBV are severely infiltrated with lymphocytes. The activated immune cells and adjacent tumor cells can express lysophosphatidic acid to further activate the Gα12-RhoA signaling pathway, contributing to the invasive behavior of cancer cells (38). Moreover, the EBV genome encodes a v-cytokine (vIL-10) and a vGPCR (ORF BILF1; refs. 5, 39), both thought to promote lymphocyte infiltration and chemokine secretion (40). Therefore, EBV might trigger some extracellular stimuli, such as lysophosphatidic acid, to potentiate Gα12 signaling. There is some basis for this hypothesis found in a recent study showing that EBV infection of Hodgkin's lymphoma cells induces autotaxin causing lysophosphatidic acid to be generated (41). On the other hand, although the involvement of Gα12/IQGAP1 in other non-EBV-induced head and neck tumors remains unexplored, it may be involved in other head and neck cancers because the E-cadherin/β-catenin pathway, which is regulated by Gα12 signaling, has been shown to be deregulated in head and neck squamous cell carcinoma (42).

This study is the first to report an association between Gα12-mediated pathway and invasiveness of NPC cells. This association offers new clues for further understanding of the molecular mechanisms and may serve as a basis for the development of targeted molecular cancer therapies.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

Y-M. Jen and S.S. Jiang contributed equally to this work.

Grant support: National Health Research Institutes.

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

We thank National Health Research Institutes Optical Biology Core for microscopy, Dr. Stephen P. Goff for IQGAP1 expression plasmids, Drs. Lu-Hai Wang and Jen-Yang Chen for comments on the article, James Steed and C.Y. Foundation for English editing assistance, and Hsin Yi Hsieh for assistance with the Western blotting.

1
Lo KW, To KF, Huang DP. Focus on nasopharyngeal carcinoma.
Cancer Cell
2004
;
5
:
423
–8.
2
Hobbs CG, Sterne JA, Bailey M, Heyderman RS, Birchall MA, Thomas SJ. Human papillomavirus and head and neck cancer: a systematic review and meta-analysis.
Clin Otolaryngol
2006
;
31
:
259
–66.
3
Termine N, Panzarella V, Falaschini S, et al. HPV in oral squamous cell carcinoma vs head and neck squamous cell carcinoma biopsies: a meta-analysis (1988-2007).
Ann Oncol
2008
;
19
:
1681
–90.
4
Mazzuco TL, Chabre O, Feige JJ, Thomas M. Aberrant GPCR expression is a sufficient genetic event to trigger adrenocortical tumorigenesis.
Mol Cell Endocrinol
2007
;
265–266
:
23
–8.
5
Nicholas J. Human gammaherpesvirus cytokines and chemokine receptors.
J Interferon Cytokine Res
2005
;
25
:
373
–83.
6
Graler MH, Goetzl EJ. Lysophospholipids and their G protein-coupled receptors in inflammation and immunity.
Biochim Biophys Acta
2002
;
1582
:
168
–74.
7
Li S, Huang S, Peng SB. Overexpression of G protein-coupled receptors in cancer cells: involvement in tumor progression.
Int J Oncol
2005
;
27
:
1329
–39.
8
Thomas SM, Bhola NE, Zhang Q, et al. Cross-talk between G protein-coupled receptor and epidermal growth factor receptor signaling pathways contributes to growth and invasion of head and neck squamous cell carcinoma.
Cancer Res
2006
;
66
:
11831
–9.
9
Gschwind A, Prenzel N, Ullrich A. Lysophosphatidic acid-induced squamous cell carcinoma cell proliferation and motility involves epidermal growth factor receptor signal transactivation.
Cancer Res
2002
;
62
:
6329
–36.
10
Neves SR, Ram PT, Iyengar R. G protein pathways.
Science
2002
;
296
:
1636
–9.
11
Sah VP, Seasholtz TM, Sagi SA, Brown JH. The role of Rho in G protein-coupled receptor signal transduction.
Annu Rev Pharmacol Toxicol
2000
;
40
:
459
–89.
12
Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors.
J Cell Physiol
2007
;
213
:
589
–602.
13
Etienne-Manneville S, Hall A. Rho GTPases in cell biology.
Nature
2002
;
420
:
629
–35.
14
Kelly P, Casey PJ, Meigs TE. Biologic functions of the G12 subfamily of heterotrimeric G proteins: growth, migration, and metastasis.
Biochemistry
2007
;
46
:
6677
–87.
15
Meyer TN, Hunt J, Schwesinger C, Denker BM. Gα12 regulates epithelial cell junctions through Src tyrosine kinases.
Am J Physiol Cell Physiol
2003
;
285
:
C1281
–93.
16
Meyer TN, Schwesinger C, Denker BM. Zonula occludens-1 is a scaffolding protein for signaling molecules. Gα(12) directly binds to the Src homology 3 domain and regulates paracellular permeability in epithelial cells.
J Biol Chem
2002
;
277
:
24855
–8.
17
Sabath E, Negoro H, Beaudry S, et al. Gα12 regulates protein interactions within the MDCK cell tight junction and inhibits tight-junction assembly.
J Cell Sci
2008
;
121
:
814
–24.
18
Kelly P, Stemmle LN, Madden JF, Fields TA, Daaka Y, Casey PJ. A role for the G12 family of heterotrimeric G proteins in prostate cancer invasion.
J Biol Chem
2006
;
281
:
26483
–90.
19
Kelly P, Moeller BJ, Juneja J, et al. The G12 family of heterotrimeric G proteins promotes breast cancer invasion and metastasis.
Proc Natl Acad Sci U S A
2006
;
103
:
8173
–8.
20
Kumar RN, Shore SK, Dhanasekaran N. Neoplastic transformation by the gep oncogene, Gα12, involves signaling by STAT3.
Oncogene
2006
;
25
:
899
–906.
21
Brown MD, Sacks DB. IQGAP1 in cellular signaling: bridging the GAP.
Trends Cell Biol
2006
;
16
:
242
–9.
22
Noritake J, Watanabe T, Sato K, Wang S, Kaibuchi K. IQGAP1: a key regulator of adhesion and migration.
J Cell Sci
2005
;
118
:
2085
–92.
23
Kuroda S, Fukata M, Nakagawa M, et al. Role of IQGAP1, a target of the small GTPases Cdc42 and Rac1, in regulation of E-cadherin-mediated cell-cell adhesion.
Science
1998
;
281
:
832
–5.
24
Watanabe T, Wang S, Noritake J, et al. Interaction with IQGAP1 links APC to Rac1, Cdc42, and actin filaments during cell polarization and migration.
Dev Cell
2004
;
7
:
871
–83.
25
Zhang S, Wu Y, Zeng Y, Zech L, Klein G. Cytogenetic studies on an epithelioid cell line derived from nasopharyngeal carcinoma.
Hereditas
1982
;
97
:
23
–8.
26
Lin CT, Chan WY, Chen W, et al. Characterization of seven newly established nasopharyngeal carcinoma cell lines.
Lab Invest
1993
;
68
:
716
–27.
27
Chang JL, Tsao YP, Liu DW, Han CP, Lee WH, Chen SL. The expression of type I growth factor receptors in the squamous neoplastic changes of uterine cervix.
Gynecol Oncol
1999
;
73
:
62
–71.
28
Leung J, Yueh A, Appah FS, Jr., Yuan B, de los Santos K, Goff SP. Interaction of Moloney murine leukemia virus matrix protein with IQGAP.
EMBO J
2006
;
25
:
2155
–66.
29
Le Clainche C, Carlier M-F. Regulation of actin assembly associated with protrusion and adhesion in cell migration.
Physiol Rev
2008
;
88
:
489
–513.
30
Tsai CN, Tsai CL, Tse KP, Chang HY, Chang YS. The Epstein-Barr virus oncogene product, latent membrane protein 1, induces the downregulation of E-cadherin gene expression via activation of DNA methyltransferases.
Proc Natl Acad Sci U S A
2002
;
99
:
10084
–9.
31
Horikawa T, Yang J, Kondo S, et al. Twist and epithelial-mesenchymal transition are induced by the EBV oncoprotein latent membrane protein 1 and are associated with metastatic nasopharyngeal carcinoma.
Cancer Res
2007
;
67
:
1970
–8.
32
Thiery JP, Chopin D. Epithelial cell plasticity in development and tumor progression.
Cancer Metastasis Rev
1999
;
18
:
31
–42.
33
Jadeski L, Mataraza JM, Jeong HW, Li Z, Sacks DB. IQGAP1 stimulates proliferation and enhances tumorigenesis of human breast epithelial cells.
J Biol Chem
2008
;
283
:
1008
–17.
34
Walch A, Seidl S, Hermannstadter C, et al. Combined analysis of Rac1, IQGAP1, Tiam1 and E-cadherin expression in gastric cancer.
Mod Pathol
2008
;
21
:
544
–52.
35
Nakamura H, Fujita K, Nakagawa H, et al. Expression pattern of the scaffold protein IQGAP1 in lung cancer.
Oncol Rep
2005
;
13
:
427
–31.
36
Sugimoto N, Imoto I, Fukuda Y, et al. IQGAP1, a negative regulator of cell-cell adhesion, is upregulated by gene amplification at 15q26 in gastric cancer cell lines HSC39 and 40A.
J Human Genet
2001
;
46
:
21
–5.
37
Nabeshima K, Shimao Y, Inoue T, Koono M. Immunohistochemical analysis of IQGAP1 expression in human colorectal carcinomas: its overexpression in carcinomas and association with invasion fronts.
Cancer Lett
2002
;
176
:
101
–9.
38
Bian D, Mahanivong C, Yu J, et al. The G12/13-RhoA signaling pathway contributes to efficient lysophosphatidic acid-stimulated cell migration.
Oncogene
2005
;
25
:
2234
–44.
39
Paulsen SJ, Rosenkilde MM, Eugen-Olsen J, Kledal TN. Epstein-Barr virus-encoded BILF1 is a constitutively active G protein-coupled receptor.
J Virol
2005
;
79
:
536
–46.
40
Ogino T, Moriai S, Ishida Y, et al. Association of immunoescape mechanisms with Epstein-Barr virus infection in nasopharyngeal carcinoma.
Int J Cancer
2007
;
120
:
2401
–10.
41
Baumforth KR, Flavell JR, Reynolds GM, et al. Induction of autotaxin by the Epstein-Barr virus promotes the growth and survival of Hodgkin lymphoma cells.
Blood
2005
;
106
:
2138
–46.
42
Howell GM, Grandis JR. Molecular mediators of metastasis in head and neck squamous cell carcinoma.
Head Neck
2005
;
27
:
710
–7.

Supplementary data