Purpose: Invasion is the critical step in progression of a precancerous lesion to squamous cell carcinoma of the head and neck (HNSCC). Invasion is regulated by multiple proinflammatory mediators. Tristetraprolin (TTP) is an mRNA-degrading protein that regulates multiple proinflammatory mediators. TTP may serve as an excellent treatment target. Rap1 is a ras-like oncoprotein that induces critical signaling pathways. In this study, the role of rap1 in TTP-mediated invasion was investigated.

Experimental Design: Using complementary approaches, we modulated TTP and altered expression of interleukin (IL)-6 and matrix metalloproteinase (MMP) 2/9, which were quantified by ELISA and zymogram. Invasion was evaluated in vitro using the oral-cancer-equivalent (OCE) three-dimensional model and in vivo in the chick chorioallantoic membrane (CAM). The role of rap1 and p38 were established using knockdown strategies.

Results: Downregulation of TTP significantly increased invasion via secretion of MMP9/2 and IL-6. In the novel OCE and CAM invasion models of HNSCC, cells with downregulated TTP destroyed the basement membrane to invade the underlying connective tissue. Rap1 induces p38 mitogen-activated protein kinase (p38)-mediated inactivation of TTP. Inactive TTP enhances transcript stability via binding to the 3′-untranslated region (UTR). High IL-6 and MMP9 are prognostic for poor clinical outcomes in patients with HNSCC.

Conclusions: Targeting the rap1-p38-TTP cascade is an attractive novel treatment strategy in HNSCC to concurrently suppress multiple mediators of invasion. Clin Cancer Res; 19(5); 1169–79. ©2012 AACR.

Translational Relevance

Invasion, a critical step in tumor progression, is induced by concurrent upregulation of multiple cytokines and proinflammatory mediators. This may explain why targeted therapy against a single mediator in squamous cell carcinoma of the head and neck (HNSCC) is only marginally successful and underscores the importance of identifying a common regulatory mechanism. Using novel invasion models that recapitulate the oral mucosa, we show that loss or phosphorylation of tristetraprolin (TTP) promotes invasion via matrix metalloproteinase (MMP) 9, MMP2, and interleukin (IL)-6. Moreover, high IL-6 and/or MMP9 are prognostic for poor outcomes in patients with HNSCC. Rap1B, a critical signaling molecule in HNSCC, induces phosphorylation of TTP via p38. These data suggest that targeting TTP expression or phosphorylation may be a novel therapeutic strategy to inhibit invasion and improve patient survival.

Novel treatment strategies are required for squamous cell carcinoma of the head and neck (HNSCC) because current regimens have only marginally improved survival in 4 decades. HNSCC is the sixth most common cancer globally, with approximately 600,000 new cases a year (1). At approximately 50%, the 5-year survival rate is poorer than breast cancer or melanoma (2). The dismal survival rate is attributed to tumor invasion and metastasis at the time of diagnosis (1). Destruction of the basement membrane and invasion of tumor cells into the underlying tissue are required for the progression of a precancerous lesion to HNSCC. The basement membrane separates the oral epithelium, the tissue of origin of HNSCC, from the underlying connective tissue. Tumor-derived proinflammatory mediators such as matrix metalloproteinase (MMP) 9, MMP2, and interleukin (IL)-6 promote invasion (3, 4), an essential phenotype of cancer progression (5). MMP9 and MMP2 are gelatinases that degrade type IV collagen in the basement membrane (6). The simultaneous upregulation of these cytokines and proinflammatory mediators, each correlated with poor prognosis, may explain why targeted therapy against a single growth factor or its receptor had limited success (7).

Multiple cytokines and proinflammatory mediators promote invasion, underscoring the importance of identifying and targeting a common regulatory mechanism. Proinflammatory mediators are modulated during transcription, posttranscriptionally, and posttranslationally. RNA-binding proteins (RNA-BP) that impact the stability of transcripts have a significant role in tumor progression. RNA-BPs regulate mRNA posttranscriptionally by binding the adenylate-uridylate–rich elements (ARE) in the 3′ untranslated region (3′-UTR) of mRNA, thereby inducing decay or stabilization of the transcript (8, 9). Tristetraprolin [(TTP), also known as TISS11, ZFP36, and Nup475] is an RNA-BP that presents transcripts to the decay machinery (9, 10). Because RNA-BPs regulate multiple proinflammatory mediators that promote invasion and tumor progression, RNA-BPs may represent potential treatment targets.

We recently showed that TTP expression is inversely correlated with invasion in HNSCC (11). In macrophages, TTP is inactivated by phosphorylation (12). It is unclear whether these mechanisms are conserved in cancer. In the current study, we investigate the mechanism by which TTP mediates invasion of HNSCC. We show that suppression or p-38–mediated phosphorylation of TTP promotes invasion due to increased secretion of IL-6, MMP9, and MMP2.

The basement membrane, the first and most robust structural barrier to invasion, separates the epithelium from the underlying connective tissue (13). A fundamental challenge of invasion studies is the lack of models for human HNSCC that recapitulate the complexity of the basement membrane and underlying connective tissue. Therefore, we developed novel in vitro and in vivo models of invasion of human HNSCC.

In this study, we investigated the mechanism by which TTP mediates invasion of HNSCC. Using novel models of invasion of human HNSCC, we show that suppression of TTP leads to an invasive phenotype in vitro and in vivo due to increased secretion of IL-6, MMP9, and MMP2. In addition, we show that rap1 induces p38 mitogen-activated protein kinase (MAPK)-mediated inactivation of TTP, which enhances transcript stability of IL-6, MMP9, and MMP2 via the 3′-UTR. TTP and its upstream regulators may represent novel therapeutic targets to suppress multiple proinflammatory mediators simultaneously with the potential to improve patient survival in HNSCC.

Tissue microarray

A tissue microarray (TMA), described previously (14, 15), from patients with stage III/IV HNSCC treated with chemotherapy and radiation or surgery and radiation after induction chemotherapy (16), was used for these studies. The TMA was scored for IL-6 (Supplementary Fig. S1) and MMP9 based on no, low, medium, and high staining intensity interpreted as described (11, 14). Representative scoring intensities for IL-6 are shown in Supplementary Fig. S1. Similar reference scoring intensities for MMP9 were previously published (14). Clinical outcomes of the patients analyzed were time to indication of surgery at primary site and time to recurrence, second primary, or death from disease. Marker values were dichotomized into low staining and high staining. Associations were evaluated with Cox proportional hazards models that related time-to-event outcomes to marker levels and other covariates. Models with each marker alone, each marker plus clinical stage, and interaction models with pairs of markers and their interactions were explored. Kaplan–Meier survival curves represent the significant findings of an interaction term between 2 markers in the Cox interaction model. P values represent the results of a Wald χ2 test of the interaction.

Cell culture

HNSCC cell lines from the University of Michigan (Ann Arbor, MI) were validated (genotyped) from frozen stock cultures and maintained via continuous passages. Normal human oral keratinocytes were from frozen stocks purchased from ScienCell Research Laboratories and were cultured as described (14, 17). Individual siRNAs were used to knockdown TTP (J-01789-13), IL-6 (J-007993-07), and rap1B (J-010364-06 and -07; Dharmacon). For MMP9, MMP2 (14), and p38 knockdown, ON-TARGETplus siRNA SMART-pools and control NonTargeting SMART-pools were used. Transfections were conducted as described (11). For stable knockdown of TTP, HNSCC cells were transduced with short hairpin RNA [shRNA (shTTP and shscramble control)] in lentiviral particles containing a GFP (Open Biosystems) and were selected as described (11).

Western blot analysis

Whole-cell lysates were immunoblotted with phospho-p38 (p-p38), p38, rap1B, actin, and MMP2 (Cell Signaling), MMP9, TTP, and phosphoserine (Abcam) IL-6 (R&D) primary and horseradish peroxidase (HRP)-conjugated secondary antibodies (Jackson Laboratories). For immunoprecipitation of TTP (IP-TTP) detection, HRP-conjugated Rbt-TrueBlot anti-immunoglobulin G (IgG; eBioscience) secondary antibody was used.

Organotypic oral cancer equivalent

AlloDerm (LifeCell) is an acellular dermal matrix used to generate normal oral mucosa with human oral keratinocytes (18). We took advantage of this tissue matrix to develop a three-dimensional (3D) model of invasion of human HNSCC. AlloDerm sections were rehydrated in PBS. The rehydrated tissue with the epidermal side up was coated with human type IV collagen (50 μg/mL; Sigma-Aldrich) overnight at 4°C. UM-SCC-1-shTTP or -shControl cells were trypsinized and seeded on the tissue matrix. Cells proliferated for 2 days on tissue submerged in Dulbecco's modified Eagle's medium (DMEM)/FBS. This oral cancer equivalent (OCE) was raised to the air/liquid interface for another 2 days to allow cellular stratification. The harvested, formalin-fixed tissue was divided into 3 parallel segments at 2 to 3 mm intervals before paraffin embedding. Tissue sections were stained with hematoxylin and eosin. Invasive islands and the total number of invasive cells were quantified in 3 separate sections from each OCE, each section with all 3 segments of tissue.

Chick chorioallantoic membrane

The chick chorioallantoic membrane (CAM) consists of surface chorionic epithelium separated by connective tissue from the endodermal allantoic membrane (19, 20). The chorionic epithelium is separated from the underlying connective tissue by an intact epithelial-derived basement membrane that contains type IV collagen (19). UM-SCC-1-shTTP or UM-SCC-1-shControl cells were seeded on the CAM. Day 11 chick embryos were used, as described (19, 21). After 2 days, the CAM was harvested, frozen, sectioned, and stained with hematoxylin and eosin or immunostained for collagen IV and diamidino-2-phenylindole (DAPI; Invitrogen) to highlight the basement membrane and nuclei, respectively (19). Images for GFP, collagen IV, and DAPI were merged in 50% ratios with DP Manager. Invasive islands were quantified in multiple fields and graphed after normalization to control.

ELISA and gelatin zymography

Conditioned media was collected as described (11) and concentrated in Centrifugal Filter Units (Millipore). Gelatin enzyme activity for MMP9 and MMP2 was evaluated as described (14). IL-6 was measured by ELISA (R&D).

cDNA synthesis and qRT-PCR

QIAzol (Qiagen) was used to isolate total RNA, and cDNA was synthesized (11). Quantitative real-time PCR was conducted with SYBR Green Master mix on an Applied Biosystems 7600HT Real-Time PCR machine. Forward and reverse primers are listed in Supplementary Table S1.

mRNA stability

A total of 10 nmol/L of actinomycin D (Sigma) was used to halt transcription for 0 to 3 hours. RNA was collected and processed (11). Cycle threshold values for IL-6, MMP9, and MMP2 were normalized to glyceraldehyde-3-phosphate dehydrogenase and expressed as a percentage of time 0.

3′UTR luciferase assay

UM-SCC-1 and -81B cells were transfected with siRNA targeting TTP (siTTP) or nontarget control siRNA (NT). A promoter-driven firefly luciferase reporter construct containing the 3′UTR of IL-6, MMP9, or MMP2 (SwitchGear) and a Renilla reporter construct (normalization control) were cotransfected with Lipofectomine 2000 (Invitrogen). Luciferase and Renilla were quantified in cell extracts using the DualGlo Reporter Luciferase System (Promega) on LMaxx384 (Molecular Devices). Firefly luciferase activity was normalized to Renilla activity and expressed as relative luminescence units.

Immunoprecipitation of TTP

IP-TTP was conducted with TTP antibody (Santa Cruz) crosslinked to Amino-Link Plus Coupling Resin Beads (Pierce) in the presence of protease (Roche) and phosphatase (Sigma) inhibitors.

Data analysis

Statistical analysis was conducted using a Student t test. A P value of < 0.05 was considered to be significant.

IL-6 and MMP9 are predictive of poor outcomes in HNSCC

The interaction between IL-6 and MMP9 was explored in terms of negative sequelae for HNSCC including short time to recurrence, second primary tumor, or surgery or death from disease (Fig. 1). Patients with low IL-6 and high MMP9 or with high IL-6 and low MMP9 had the poorest outcomes (P = 0.020) followed by patients with both high IL-6 and high MMP9 (P = 0.020). Patients with low IL-6 and low MMP9 had the best outcome with respect to tumor recurrence, second primary tumor, surgery or death from disease. These findings suggest that IL-6 and MMP9, individually or together, have a significant role in HNSCC progression.

Figure 1.

IL-6 and MMP9 are predictive of poor outcome in HNSCC. Immunohistochemistry was conducted on tissue sections of a human HNSCC TMA. Interactions with IL-6 and MMP9 were determined with a COX interaction model. Patient groups are indicated by color lines. Low MMP9- and low IL-6–staining intensity (red, n = 14); low MMP9 and high IL-6 (yellow, n = 8); high MMP9 and low IL-6 (blue, n = 6); high MMP9 and high IL-6 (green, n = 7). Subjects who did not experience the events were censored (P = 0.020). DFD, death from disease.

Figure 1.

IL-6 and MMP9 are predictive of poor outcome in HNSCC. Immunohistochemistry was conducted on tissue sections of a human HNSCC TMA. Interactions with IL-6 and MMP9 were determined with a COX interaction model. Patient groups are indicated by color lines. Low MMP9- and low IL-6–staining intensity (red, n = 14); low MMP9 and high IL-6 (yellow, n = 8); high MMP9 and low IL-6 (blue, n = 6); high MMP9 and high IL-6 (green, n = 7). Subjects who did not experience the events were censored (P = 0.020). DFD, death from disease.

Close modal

Downregulation of TTP promotes invasion through the basement membrane

Invasion, an essential step in tumor progression, is required for transformation of precancerous lesions to HNSCC. In initial in vitro and in vivo studies, we investigated whether TTP modulates invasion. Transient suppression of TTP was optimized with 4 individual siRNAs and the siRNA that had the best sustained knockdown was used for subsequent studies (Supplementary Fig. S2). When TTP was suppressed in UM-SCC-1, a HNSCC cell line, invasion was significantly increased compared with nontarget controls, as well as migration (Fig. 2A, left and right, respectively; *, P < 0.01). TTP suppression was verified by immunoblot analysis (Fig. 2B).

Figure 2.

Downregulation of TTP promotes invasion. A, UM-SCC-1 cells transfected with NT or siTTP were analyzed for invasion and migration. B, whole-cell lysates were immunoblotted with TTP and actin. Signal intensity was quantified as arbitrary densitometric unit (DU) by Image J software, normalized to actin, and presented as the percentage of NT. C, whole-cell lysates from UM-SCC-1 cells stably transduced with shControl (shC) and shTTP were immunoblotted with TTP and actin, as a loading control. Signal intensity (DU) was quantified, normalized to actin, and presented as the percentage of shC. D, left, hematoxylin and eosin staining of UM-SCC-1-shC and -shTTP cells grown as an OCE. Black arrows depict invasive islands, which are more visible at the higher magnification (right). Middle, quantification of total number of invasive islands. Right, quantification of total number of invaded cells, normalized to total cell number. Data are representative of 2 independent experiments with 2 replicates in each experiment. E, left, UM-SCC-1shC and -shTTP CAM sections were stained with hematoxylin and eosin. Images were taken under bright-field and fluorescence (GFP) at × 20 and merged to visualize cells invading past the basement membrane (basement membrane, white arrows). Right, quantification of invasive islands. Results are representative of 3 independent experiments. F, CAM sections were immunostained for collagen IV to identify the basement membrane (yellow arrows). Images were taken at × 20 under fluorescence and merged (green-tumor cells, red-Col IV, blue-DAPI). White arrows show invading tumor cells (also Supplementary Fig. S3). *, P < 0.01.

Figure 2.

Downregulation of TTP promotes invasion. A, UM-SCC-1 cells transfected with NT or siTTP were analyzed for invasion and migration. B, whole-cell lysates were immunoblotted with TTP and actin. Signal intensity was quantified as arbitrary densitometric unit (DU) by Image J software, normalized to actin, and presented as the percentage of NT. C, whole-cell lysates from UM-SCC-1 cells stably transduced with shControl (shC) and shTTP were immunoblotted with TTP and actin, as a loading control. Signal intensity (DU) was quantified, normalized to actin, and presented as the percentage of shC. D, left, hematoxylin and eosin staining of UM-SCC-1-shC and -shTTP cells grown as an OCE. Black arrows depict invasive islands, which are more visible at the higher magnification (right). Middle, quantification of total number of invasive islands. Right, quantification of total number of invaded cells, normalized to total cell number. Data are representative of 2 independent experiments with 2 replicates in each experiment. E, left, UM-SCC-1shC and -shTTP CAM sections were stained with hematoxylin and eosin. Images were taken under bright-field and fluorescence (GFP) at × 20 and merged to visualize cells invading past the basement membrane (basement membrane, white arrows). Right, quantification of invasive islands. Results are representative of 3 independent experiments. F, CAM sections were immunostained for collagen IV to identify the basement membrane (yellow arrows). Images were taken at × 20 under fluorescence and merged (green-tumor cells, red-Col IV, blue-DAPI). White arrows show invading tumor cells (also Supplementary Fig. S3). *, P < 0.01.

Close modal

To recapitulate the basement membrane of the oral epithelium and the complexity of human connective tissue, we developed a 3D OCE model of invasion to evaluate whether TTP suppression promotes invasion. To facilitate sustained knockdown of TTP for the OCE model, UM-SCC-1 cells were stably transduced with control shRNA (UM-SCC-1-shC or shC) or shRNA targeting TTP (UM-SCC-1-shTTP or shTTP). Knockdown was verified by immunoblot analysis (Fig. 2C). In the OCE model, loss of TTP significantly increased invasion compared with corresponding control cells (Fig. 2D, left; arrowheads show tumor islands completely detached from surface epithelium in shTTP). There were significantly more invasive tumor islands (Fig. 2D middle) as well as invasive cells (Fig. 2D, right) in OCE-shTTP compared with control OCE-shC (*, P < 0.01).

The oral mucosa consists of surface epithelium separated from the underlying connective tissue by basement membrane. HNSCC occurs when transformed surface oral epithelial cells invade the connective tissue. The effect of TTP on invasion was verified in the CAM in vivo model. Similar to the oral mucosa, the CAM consists of surface epithelium separated from the underlying connective tissue by an intact epithelial-derived basement membrane (19, 22). The human HNSCC cells are placed on top of the surface epithelium and must destroy the basement membrane to invade the connective tissue, thereby simulating human HNSCC. Tumors with UM-SCC-1-shTTP showed destruction of the basement membrane and more GFP-tagged tumor cells invading the connective tissue compared with corresponding control tumors (Fig. 2E, left, arrows on merged shTTP). There were significantly more invasive islands per field in shTTP compared with control (Fig. 2E, right; *, P < 0.01). In tumors with shTTP, the basement membrane was completely disrupted (Fig. 2F, bottom; and Supplementary Fig. S3, type IV collagen staining; white arrows highlight invasive tumor cells), whereas in control tumors, the basement membrane seemed intact (Fig. 2F, top, yellow arrows show intact basement membrane), supporting that low TTP in HNSCC promotes invasion.

Downregulation of TTP promotes invasion via upregulation of IL-6, MMP9, and MMP2

Previously, we showed that TTP inhibits IL-6, but its effect on gelatinases (MMP2, MMP9), which promote invasion in HNSCC, is unknown (11, 14). The impact of TTP expression on MMP9 and MMP2 secretion was evaluated by gelatin zymography in conditioned medium from HNSCC cells transfected with NT and siTTP (Fig. 3A). In UM-SCC-1 cells, suppression of TTP increased MMP9 secretion by 89% and MMP2 secretion by 42% compared with cells transfected with NT (Fig 3A middle). TTP knockdown was verified (Fig. 3A right). Similar findings were observed in UM-SCC-81B, another independent HNSCC cell line (Supplementary Fig. S4A).

Figure 3.

Downregulation of TTP promotes invasion via IL-6, MMP9, and MMP2. A, UM-SCC-1 cells were transiently transfected with NT or siTTP. Gelatin zymography (left) was used to quantify MMP9/2 in conditioned medium and expressed as the percentage of corresponding control (middle). TTP knockdown was verified (right). Data are representative of 3 independent experiments. B, whole-cell lysates from UM-SCC-1-shTTP cells transiently transfected with siIL-6, siMMP9, siMMP2, or NT were immunoblotted with IL-6, MMP9, or MMP2 and actin antibodies. Signal intensity (DU) was quantified, normalized to actin, and expressed as the percentage of control. C, invasion of UM-SCC-1-shTTP cells transiently transfected with siIL-6, siMMP9, siMMP2, or NT was quantified and expressed as the percentage of NT. Data are representative of 2 independent experiments with 3 replicates in each experiment. *, P < 0.01.

Figure 3.

Downregulation of TTP promotes invasion via IL-6, MMP9, and MMP2. A, UM-SCC-1 cells were transiently transfected with NT or siTTP. Gelatin zymography (left) was used to quantify MMP9/2 in conditioned medium and expressed as the percentage of corresponding control (middle). TTP knockdown was verified (right). Data are representative of 3 independent experiments. B, whole-cell lysates from UM-SCC-1-shTTP cells transiently transfected with siIL-6, siMMP9, siMMP2, or NT were immunoblotted with IL-6, MMP9, or MMP2 and actin antibodies. Signal intensity (DU) was quantified, normalized to actin, and expressed as the percentage of control. C, invasion of UM-SCC-1-shTTP cells transiently transfected with siIL-6, siMMP9, siMMP2, or NT was quantified and expressed as the percentage of NT. Data are representative of 2 independent experiments with 3 replicates in each experiment. *, P < 0.01.

Close modal

To determine whether downregulation of TTP induces invasion via overexpression of IL-6, MMP9, and MMP2, we investigated whether knockdown of these proinflammatory mediators suppresses the shTTP-induced invasion. UM-SCC-1-shTTP cells were transfected with siRNA targeting IL-6, MMP9, or MMP2. Knockdown of IL-6, MMP9, and MMP2 was verified by immunoblot analysis (Fig. 3B). Invasion was evaluated in a FluoroBlok invasion assay (Fig 3C). UM-SCC-1-shTTP cells exhibit an invasive phenotype (Fig. 3C, NT), which was suppressed when IL-6, MMP2, or MMP9 were downregulated with siRNA (Fig. 3C). Suppression of IL-6, MMP9, and MMP2, all significantly decreased invasion (*, P < 0.01). No differences in migration were observed. Similar results were observed in UM-SCC-81B (Supplementary Fig. S4B and S4C). Taken together, these results suggest that low TTP in HNSCC favors secretion of IL-6, MMP9, and MMP2 to promote invasion.

TTP regulates the stability of transcripts for IL-6, MMP2, and MMP9 via the 3′-UTR

TTP binds AREs in the 3′-UTR of mRNAs to promote degradation (9, 23). AREs are present in multiple cytokines and proinflammatory factors including MMP2 and MMP9 (24). However, TTP-mediated regulation of MMPs via the 3′-UTR has not been investigated. In initial studies, the effect of downregulation of TTP on transcripts for IL-6, MMP9, and MMP2 was investigated. The steady state mRNA levels of IL-6, MMP9, and MMP2 were significantly increased in UM-SCC-1-siTTP cells compared withcontrol cells (Fig. 4A, *, P < 0.01). Similar results were observed in UM-SCC-81B (Supplementary Fig. S5A).

Figure 4.

TTP regulates the stability of transcripts of IL-6, MMP9, and MMP2 via the 3′-UTR. A, left three panels, IL-6, MMP9, and MMP2 mRNA levels were quantified by qRT-PCR from UM-SCC-1 cells transfected with NT and siTTP. Data are representative of 2 independent experiments with 3 replicates in each experiment. Right, whole-cell lysates were immunoblotted with TTP and actin. Signal intensity (DU) was quantified, normalized to actin, and expressed as the percentage of NT. B, left 3, UM-SCC-1shC or shTTP cells were treated with actinomycin D followed by RNA isolation and quantification via qRT-PCR. Right, whole-cell lysates were immunoblotted with TTP and actin. Signal intensity (DU) was quantified, normalized to actin, and expressed as percent of shC. C, left 3, UM-SCC-1 cells transfected with NT or siTTP were cotransfected with a luciferase reporter construct containing the 3′-UTR of IL-6, MMP9, or MMP2 and a Renilla construct for normalization. Data are representative of 3 independent experiments with 3 replicates in each experiment. Right, whole-cell lysates were immunoblotted with TTP and actin antibodies. Signal intensity (DU) was quantified, normalized to actin, and expressed as the percentage of NT. *, P < 0.01.

Figure 4.

TTP regulates the stability of transcripts of IL-6, MMP9, and MMP2 via the 3′-UTR. A, left three panels, IL-6, MMP9, and MMP2 mRNA levels were quantified by qRT-PCR from UM-SCC-1 cells transfected with NT and siTTP. Data are representative of 2 independent experiments with 3 replicates in each experiment. Right, whole-cell lysates were immunoblotted with TTP and actin. Signal intensity (DU) was quantified, normalized to actin, and expressed as the percentage of NT. B, left 3, UM-SCC-1shC or shTTP cells were treated with actinomycin D followed by RNA isolation and quantification via qRT-PCR. Right, whole-cell lysates were immunoblotted with TTP and actin. Signal intensity (DU) was quantified, normalized to actin, and expressed as percent of shC. C, left 3, UM-SCC-1 cells transfected with NT or siTTP were cotransfected with a luciferase reporter construct containing the 3′-UTR of IL-6, MMP9, or MMP2 and a Renilla construct for normalization. Data are representative of 3 independent experiments with 3 replicates in each experiment. Right, whole-cell lysates were immunoblotted with TTP and actin antibodies. Signal intensity (DU) was quantified, normalized to actin, and expressed as the percentage of NT. *, P < 0.01.

Close modal

To determine whether TTP destabilizes transcripts of IL-6, MMP9, and MMP2, UM-SCC-1-shTTP or UM-SCC-1-shC cells were treated with actinomycin D, which inhibits transcription. Loss of TTP enhanced stability of IL-6, MMP9, and MMP2 transcripts compared with control cells (Fig. 4B, left three). IL-6 mRNA stability was shown previously in an independent experiment ref. (11; *, P < 0.01). Downregulation of TTP was verified (Fig. 4B, right). This data implies that TTP promotes mRNA degradation of IL-6, MMP9, and MMP2.

To verify that loss of TTP stabilizes the transcripts for IL-6, MMP9, and MMP2 via the 3′-UTR, UM-SCC-1-siTTP, and control were cotransfected with a firefly luciferase reporter construct containing the 3′-UTR of IL-6 (schematic in Fig. 4C), MMP9 and MMP2, and a Renilla construct. Suppression of TTP significantly increased the relative luminescence of IL-6, MMP9, and MMP2 after normalization to Renilla compared with control cells (Fig. 4C, left three; *, P < 0.01). TTP knockdown was verified (Fig. 4C, right). Similar results were observed in UM-SCC-81B (Supplementary Fig. S5B). Taken together, these studies show that TTP promotes degradation of mRNA for IL-6, MMP9, and MMP2 via the 3′-UTR.

Rap1B activates p38 in HNSCC

We previously showed that rap1B is a critical signaling molecule in HNSCC and induces extracellular signal-regulated kinase (ERK)/MAPK activation (14, 17, 25, 26). To determine whether rap1B mediates p38 activation in HNSCC, UM-SCC-1 cells were induced with IL-1β (1 ng/mL). An increase in active, GTP-bound rap1B was observed before p38 phosphorylation, suggesting that rap1B is upstream of p38 (Fig. 5A). Similar results were observed in UM-SCC-81B (Supplementary Fig. S6A).

Figure 5.

Rap1B induces p38-mediated phosphorylation of TTP, which promotes secretion of proinflammatory mediators. A, whole-cell lysates from UM-SCC-1 cells treated with IL-1β were immunoblotted with pp38, p38, rap1B, and actin. Active GTP-bound rap1B (rap1BGTP) was retrieved by a ralGDS pull-down assay. p38 was used for normalization of p-p38 and rap1B was used for normalization of rap1BGTP. Signal intensity (DU) was quantified, normalized to its corresponding loading control, and expressed as the percentage of time zero. Data are representative of 3 independent experiments. B, cells were transfected with 2 siRNAs targeting rap1B (si6 and si7) or NT and stimulated with IL-1β or PBS (control) for 10 minutes. Whole-cell lysates were immunoblotted with p-p38, p38, and rap1B. Signal intensity (DU) was quantified, normalized to p38, and expressed as the percentage of stimulated NT. Experiments were carried out in triplicate. C, whole-cell lysates from HNSCC and human oral keratinocytes (HOK) were immunoblotted for p-p38, p38, and actin. Signal intensity (DU) was quantified, normalized to p38 then actin, and expressed as the percentage of HOK. D, TTP was immunoprecipitated from UM-SCC-1, -11A, and -81B, and were immunoblotted with phosphoserine or TTP. Signal intensity (DU) was quantified, normalized to input, and expressed as the percentage of UM-SCC-1. E, UM-SCC-1 cells were serum starved and treated with 10 μmol/L of SB203580. Clarified cell lysates were immunoprecipitated with TTP antibody, and blotted with anti-TTP and anti-phosphoserine antibodies. F, cells transfected with sip38 or NT were incubated with PBS (control) or IL-1β for 10 minutes. Top 2 panels, immunoprecipitation was conducted as above. Signal intensity (DU) was quantified, normalized to input, and expressed as the percentage of unstimulated NT. Bottom 3 panels, whole cell lysates from NT and sip38 were also immunoblotted with p-p38, p38, and actin. Signal intensity (DU) was quantified, normalized to p38 then actin, and expressed as the percentage of unstimulated NT. Three independent experiments were carried out. G, cells were transfected with sip38 or NT and cell lysates were immunoblotted with p38 and actin antibodies. Signal intensity (DU) was quantified, normalized, and expressed as the percentage of NT. IL-6 secretion was determined with 2 replicates in each of 3 independent experiments. *, P < 0.01.

Figure 5.

Rap1B induces p38-mediated phosphorylation of TTP, which promotes secretion of proinflammatory mediators. A, whole-cell lysates from UM-SCC-1 cells treated with IL-1β were immunoblotted with pp38, p38, rap1B, and actin. Active GTP-bound rap1B (rap1BGTP) was retrieved by a ralGDS pull-down assay. p38 was used for normalization of p-p38 and rap1B was used for normalization of rap1BGTP. Signal intensity (DU) was quantified, normalized to its corresponding loading control, and expressed as the percentage of time zero. Data are representative of 3 independent experiments. B, cells were transfected with 2 siRNAs targeting rap1B (si6 and si7) or NT and stimulated with IL-1β or PBS (control) for 10 minutes. Whole-cell lysates were immunoblotted with p-p38, p38, and rap1B. Signal intensity (DU) was quantified, normalized to p38, and expressed as the percentage of stimulated NT. Experiments were carried out in triplicate. C, whole-cell lysates from HNSCC and human oral keratinocytes (HOK) were immunoblotted for p-p38, p38, and actin. Signal intensity (DU) was quantified, normalized to p38 then actin, and expressed as the percentage of HOK. D, TTP was immunoprecipitated from UM-SCC-1, -11A, and -81B, and were immunoblotted with phosphoserine or TTP. Signal intensity (DU) was quantified, normalized to input, and expressed as the percentage of UM-SCC-1. E, UM-SCC-1 cells were serum starved and treated with 10 μmol/L of SB203580. Clarified cell lysates were immunoprecipitated with TTP antibody, and blotted with anti-TTP and anti-phosphoserine antibodies. F, cells transfected with sip38 or NT were incubated with PBS (control) or IL-1β for 10 minutes. Top 2 panels, immunoprecipitation was conducted as above. Signal intensity (DU) was quantified, normalized to input, and expressed as the percentage of unstimulated NT. Bottom 3 panels, whole cell lysates from NT and sip38 were also immunoblotted with p-p38, p38, and actin. Signal intensity (DU) was quantified, normalized to p38 then actin, and expressed as the percentage of unstimulated NT. Three independent experiments were carried out. G, cells were transfected with sip38 or NT and cell lysates were immunoblotted with p38 and actin antibodies. Signal intensity (DU) was quantified, normalized, and expressed as the percentage of NT. IL-6 secretion was determined with 2 replicates in each of 3 independent experiments. *, P < 0.01.

Close modal

To verify that rap1B activates p38 in HNSCC, 2 siRNAs targeting rap1B were used. Downregulation of rap1B inhibited IL-1β–induced p38 phosphorylation by 40% in UM-SCC-1 cells compared with corresponding stimulated cells transfected with NT (Fig. 5B). Similar findings were observed in UM-SCC-81B (Supplementary Fig. S6B). Thus, rap1B induces p38 in HNSCC.

p38-mediated phosphorylation of TTP promotes secretion of proinflammatory mediators

In macrophages, p38 inactivates TTP via MK2-mediated phosphorylation at 2 serine sites (12, 27). However, the role of p38 in TTP phosphorylation in other cells, including cancer cells, is unknown. Moreover, signaling mechanisms vary between cell types, even in different types of cancer cells. Initial studies focused on constitutive p38 activity in HNSCC. Active, p-p38 was evaluated in whole-cell lysates from a panel of 6 HNSCC cell lines (Fig. 5C). All 6 cell lines exhibited an increase of more than 200% in p-p38 expression compared with normal human oral keratinocytes after normalization to total p38 and actin. Subsequently, TTP phosphorylation was evaluated in 3 HNSCC cell lines that express variable levels of TTP. Because a phospho-TTP antibody was not commercially available, TTP was immunoprecipitated with TTP antibody and immunoblotted with phosphoserine antibody. Input and IgG controls are shown for each cell lines. After normalization to TTP eluted after immunoprecipitation, UM-SCC-1 had the lowest expression of phospho-TTP of the 3 HNSCC cell lines (Fig. 5D).

To verify that TTP phosphorylation is mediated by p38, we used the chemical inhibitor SB203580, which inhibits the catalytic activity of p38. SB203580 binds to the ATP-binding pocket of p38, thereby inhibiting downstream affector molecules without altering phosphorylation of p38 itself (28). UM-SCC-1 cells were serum starved for 4 hours followed by 2 hours preincubation with 10 μmol/L of SB203580 before stimulation with IL-1β. TTP was immunoprecipitated from cell lysates and blotted with phosphoserine and TTP antibodies to detect phospho-TTP and as a loading control, respectively. IL-1β–induced phosphorylation of TTP (Fig. 5E, lane 4) was significantly inhibited with SB203580 (Fig 5E, lane 6).

In a complementary approach, we inhibited TTP phosphorylation by siRNA-mediated knockdown of p38 and evaluated phospho-TTP (29). UM-SCC-1 cells were transfected with nontarget or sip38 before stimulation with IL-1β followed by immunoprecipitation of TTP. IL-1β–induced phosphorylation of TTP was abrogated by knockdown of p38 (Fig. 5F, top, lanes 2, and 4). Knockdown of p38 was verified by immunoblot analysis (Fig. 5F, bottom, lanes 3, and 4). Similar results were observed in UM-SCC-81B (Supplementary Fig. S6). Taken together, our findings with a chemical inhibitor of p38 and with siRNA show that p38 mediates IL-1β–induced phosphorylation of TTP.

To determine whether phosphorylation of TTP promotes secretion of proinflammatory mediators, p38 was suppressed by siRNA. Knockdown of p38 reduced IL-6 secretion by 45% in UM-SCC-1 (Fig. 5G) and by 75% in UM-SCC-81B, (Supplementary Fig. S6D) compared with control cells (P < 0.01). Together these findings show that p38-mediated phosphorylation of TTP promotes secretion of proinflammatory mediators.

Poor survival is correlated with secretion of proinflammatory mediators (11, 14). Targeted therapy against individual mediators was only marginally successful in treating HNSCC (7), likely due to the concurrent upregulation of multiple mediators with overlapping functions in tumor progression. Targeting each factor may improve prognosis but is impractical, emphasizing the importance of identifying a common regulatory mechanism for multiple mediators. The complementary approaches presented here, conclusively show that downregulation or inactivation of TTP in HNSCC promotes invasion via secretion of multiple proinflammatory mediators. We show that IL-1β stimulates rap1B, which in turn induces p38-mediated phosphorylation of TTP to promote secretion of multiple proinflammatory mediators. Downregulation or p38-mediated inactivation of TTP promotes invasion via stabilization of mRNAs of MMP9/2 and IL-6. Active TTP promotes transcript degradation via the 3′UTR. Secretion of MMP9/2 and IL-6 promote invasion and progression of HNSCC. This rap1B-p38–induced signaling cascade represents a potential therapeutic target for HNSCC because p38 regulates multiple mediators of invasion. Moreover, p38 inhibitors are orally bioavailable and are already in clinical trials for inflammatory diseases (30).

TMA findings in this study, based on a small number of patients, show that either high IL-6 or high MMP9 are correlated with poor outcomes, suggesting that IL-6 and MMP-9 may also be regulated by a mechanism independent of TTP. In fact, expression of proinflammatory cytokines is impacted by transcriptional regulation of genes as well as RNA stability (24). The variation in gene transcription with each HNSCC and redundance in function between proinflammatory mediators underscores the relevance of targeting a common regulatory mechanism of transcript stability, to effectively inhibit adverse effects.

Other RNA-binding proteins that bind transcripts of proinflammatory mediators have been identified but are mostly uncharacterized in HNSCC. HuR (human antigen–related protein), which is overexpressed in HNSCC, increases stability of COX2 and VEGF transcripts (24, 31). ARE/poly(U)-binding and degradation factor (AUF-1) stabilizes granulocyte-macrophage colony-stimulating factor (32), whereas CUG binding protein 1, destabilizes transcripts of TNF-α (33), but neither has been investigated in HNSCC.

MMPs have a critical role in remodeling the extracellular matrix. While remodeling is important for wound healing, excessive MMP production is associated with invasion (34). Invasion is an essential event in tumor progression. We previously showed that MMP9/2 promote invasion and are prognostic of poor outcomes in HNSCC (14). Although we and others showed that the loss of TTP promotes invasion in cancer (11, 35) and MMP9/2 have AREs in the 3′UTR, the role of MMP2 and MMP9 in TTP-mediated invasion has not been investigated. Knockdown of TTP promotes invasion of HNSCC, which was significantly suppressed when IL-6, MMP2, or MMP9 was downregulated with siRNA. Concurrent knockdown of IL-6 and MMP9 lead to such severe apoptosis that the remaining cells were insufficient to conduct invasion assays (not shown). These findings are consistent with an overlap in function between multiple proinvasive mediators. In support of the possibility that loss of TTP promotes invasion via multiple proteins, loss of TTP enhances MMP1, urokinase plasminogen activator, and IL-8 in other cancers (35, 36), all of which promote tumor progression.

While suppression of TTP leads to a greater increase in MMP9 than MMP2 secretion (Figs. 3A and S4A), the smaller increase in MMP2 has a significant effect on invasion given that downregulation of MMP2 had a similar impact on invasion as knockdown of MMP9.

Although p38 mediates TTP phosphorylation in macrophages and endothelial cells (37, 38), this mechanism has not been investigated in cancer, which is important given that each cell type has a unique signaling repertoire. Our data from 2 different HNSCC cell lines using biochemical and siRNA approaches show that p38 phosphorylates and inactivates TTP, and increases secretion of proinflammatory mediators.

Rap1, a critical signaling mediator in HNSCC, facilitates adhesion, proliferation, survival, migration, and invasion in HNSCC (17, 39). Two isoforms exist in HNSCC, rap1A and rap1B. Although active, GTP-bound rap1B has a critical role in ERK/MAPK activation in HNSCC (39), its role in modulating p38 has not been investigated in any cancer. Our findings show a novel mechanism of p38-mediated TTP regulation in cancer via rap1B (Fig. 6). Although we established that IL-1β rapidly activates rap1B in HNSCC, this does not eliminate the possibility that other secreted proteins activate p38 via rap1B in HNSCC.

Figure 6.

Proposed model for role of TTP in invasion. TTP, downregulated or functionally inactivated by rap1B and p38, promotes tumor invasion and progression via mRNA stabilization and increased secretion of IL-6, MMP9, and MMP2.

Figure 6.

Proposed model for role of TTP in invasion. TTP, downregulated or functionally inactivated by rap1B and p38, promotes tumor invasion and progression via mRNA stabilization and increased secretion of IL-6, MMP9, and MMP2.

Close modal

Together, the novel OCE and CAM models of invasion of human HNSCC supported the in vitro findings that loss of TTP facilitates invasion. In precancerous oral lesions (epithelial dysplasia or carcinoma-in-situ), epithelial changes are restricted to the surface epithelium, that is, above the basement membrane (40). Destruction of the basement membrane and invasion of tumor cells into the underlying connective tissue are required for progression of a precancerous lesion to HNSCC. Thus, the basement membrane is the first structural barrier to invasion (13), but current models for human HNSCC do not recapitulate this structure. Two dimensional invasion assays lack the basement membrane and the complexity of the connective tissue. Alternative 3D assays that investigate invasion on a collagen matrix lack basement membrane and connective tissue. The novel in vitro OCE 3D invasion model described here, recapitulates invasion through a basement membrane barrier into human connective tissue. This model was based on the organotypic oral mucosa model that is used for tissue regeneration of normal oral mucosa (18).

Given the structural complexity of the basement membrane and the cellular and structural complexity of the connective tissue itself (13), we developed an in vivo model for invasion of human HNSCC using the chick embryo CAM. Mouse models of human HNSCC are inadequate for invasion because tumor cells are injected directly into the connective tissue thereby bypassing the basement membrane of the surface epithelium. The CAM has been used for breast, brain, and ovarian cancers (19, 22, 41) but not HNSCC. The CAM consists of surface chorionic epithelium separated from the underlying connective tissue by an intact epithelial-derived basement membrane that contains type IV collagen (13, 19). Tissues underlying the CAM contain types I and III collagen and blood vessels (42). Human HNSCC cells are seeded on top of the chorionic epithelium. To invade the underlying connective tissue, the HNSCC cells must destroy the basement membrane thereby recapitulating the process that occurs in human HNSCC. Invasion is easily visualized and quantified. Thus, the CAM model of invasion simulates the basement membrane, the complexity of the connective tissue, and the microscopic features that simulate invasion of human HNSCC. Studies in the OCE and CAM models highlight the role of TTP-mediated invasion in HNSCC.

A recent study further supported the role of low TTP in cancer progression; low TTP mRNA level is a negative prognostic indicator in breast cancer (43). Overexpression of TTP decreases tumor growth and VEGF secretion in colon cancer (44) and decreases invasion in breast cancer (35). Moreover, when treating patients with deficient TTP, a synonymous polymorphism in TTP genes predicted failure to respond to Herceptin/Trastuzumab in patients with HER2-positive breast cancer (45).

Taken together, our findings provide insight into a master regulatory mechanism that promotes invasion in HNSCC. To establish this mechanism, we developed in vitro and in vivo models for HNSCC that recapitulate the complexity of invasion in human oral mucosa. By elucidating TTP-mediated invasion, we uncovered a possible explanation for failure of HNSCC to respond favorably to inhibitors targeting a single proinflammatory mediator. The studies presented here suggest that targeting the p38-TTP cascade is an attractive treatment strategy in HNSCC.

No potential conflicts of interest were disclosed.

Conception and design: E.A. Van Tubergen, M. Liu, N.J. D'Silva

Development of methodology: E.A. Van Tubergen, M. Liu, S. Feinberg, N.J. D'Silva

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): E.A. Van Tubergen, R. Banerjee, M. Liu, G.T. Wolf, T.E. Carey, M. Prince, N.J. D'Silva

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): E.A. Van Tubergen, R. Banerjee, M. Liu, R.J.V. Broek, E. Light, G.T. Wolf, T.E. Carey, K.L. Kirkwood, N.J. D'Silva

Writing, review, and/or revision of the manuscript: E.A. Van Tubergen, R. Banerjee, R.J.V. Broek, E. Light, S. Kuo, S. Feinberg, G.T. Wolf, T.E. Carey, C.R. Bradford, M. Prince, F.P. Worden, K.L. Kirkwood, N.J. D'Silva

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): E.A. Van Tubergen, R. Banerjee, S. Kuo, S. Feinberg, A.L. Willis, T.E. Carey, K.L. Kirkwood

Study supervision: R. Banerjee, N.J. D'Silva

The authors thank Dr. Stephen Weiss' laboratory for help with the CAM.

This work was supported by National Institute of Dental and Craniofacial Research grants DE018512, DE019513, and DE017977 (to N.J. D'Silva) and DE021305 (to E.A. Van Tubergen) and University of Michigan Head and Neck SPORE grant (P50 CA97248).

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

1.
Leemans
CR
,
Braakhuis
BJ
,
Brakenhoff
RH
. 
The molecular biology of head and neck cancer
.
Nat Rev Cancer
2011
;
11
:
9
22
.
2.
Jemal
A
,
Siegel
R
,
Xu
J
,
Ward
E
. 
Cancer statistics, 2010
.
CA Cancer J Clin
2010
;
60
:
277
300
.
3.
Badache
A
,
Hynes
NE
. 
Interleukin 6 inhibits proliferation and, in cooperation with an epidermal growth factor receptor autocrine loop, increases migration of T47D breast cancer cells
.
Cancer Res
2001
;
61
:
383
91
.
4.
Kanazawa
T
,
Nishino
H
,
Hasegawa
M
,
Ohta
Y
,
Iino
Y
,
Ichimura
K
, et al
Interleukin-6 directly influences proliferation and invasion potential of head and neck cancer cells
.
Eur Arch Otorhinolaryngol
2007
;
264
:
815
21
.
5.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
6.
Rundhaug
JE
. 
Matrix metalloproteinases, angiogenesis, and cancer: commentary re: A. C. Lockhart et al., Reduction of wound angiogenesis in patients treated with BMS-275291, a broad spectrum matrix metalloproteinase inhibitor
.
Clin Cancer Res
2003
;
9
:
551
4
.
7.
Sharafinski
ME
,
Ferris
RL
,
Ferrone
S
,
Grandis
JR
. 
Epidermal growth factor receptor targeted therapy of squamous cell carcinoma of the head and neck
.
Head Neck
2010
;
32
:
1412
21
.
8.
Sanduja
S
,
Blanco
FF
,
Dixon
DA
. 
The roles of TTP and BRF proteins in regulated mRNA decay
.
Wiley Interdiscip Rev RNA
2011
;
2
:
42
57
.
9.
Carballo
E
,
Lai
WS
,
Blackshear
PJ
. 
Evidence that tristetraprolin is a physiological regulator of granulocyte-macrophage colony-stimulating factor messenger RNA deadenylation and stability
.
Blood
2000
;
95
:
1891
9
.
10.
Taylor
GA
,
Lai
WS
,
Oakey
RJ
,
Seldin
MF
,
Shows
TB
,
Eddy
RL
 Jr
, et al
The human TTP protein: sequence, alignment with related proteins, and chromosomal localization of the mouse and human genes
.
Nucleic Acids Res
1991
;
19
:
3454
.
11.
Van Tubergen
E
,
Vander
Broek R
,
Lee
J
,
Wolf
G
,
Carey
T
,
Bradford
C
, et al
Tristetraprolin regulates interleukin-6, which is correlated with tumor progression in patients with head and neck squamous cell carcinoma
.
Cancer
2011
;
117
:
2677
89
.
12.
Stoecklin
G
,
Stubbs
T
,
Kedersha
N
,
Wax
S
,
Rigby
WF
,
Blackwell
TK
, et al
MK2-induced tristetraprolin:14-3-3 complexes prevent stress granule association and ARE-mRNA decay
.
EMBO J
2004
;
23
:
1313
24
.
13.
Rowe
RG
,
Weiss
SJ
. 
Breaching the basement membrane: who, when and how?
Trends Cell Biol
2008
;
18
:
560
74
.
14.
Mitra
RS
,
Goto
M
,
Lee
JS
,
Maldonado
D
,
Taylor
JM
,
Pan
Q
, et al
Rap1GAP promotes invasion via induction of matrix metalloproteinase 9 secretion, which is associated with poor survival in low N-stage squamous cell carcinoma
.
Cancer Res
2008
;
68
:
3959
69
.
15.
Kumar
B
,
Cordell
KG
,
Lee
JS
,
Worden
FP
,
Prince
ME
,
Tran
HH
, et al
EGFR, p16, HPV Titer, Bcl-xL and p53, sex, and smoking as indicators of response to therapy and survival in oropharyngeal cancer
.
J Clin Oncol
2008
;
26
:
3128
37
.
16.
Worden
FP
,
Kumar
B
,
Lee
JS
,
Wolf
GT
,
Cordell
KG
,
Taylor
JM
, et al
Chemoselection as a strategy for organ preservation in advanced oropharynx cancer: response and survival positively associated with HPV16 copy number
.
J Clin Oncol
2008
;
26
:
3138
46
.
17.
Mitra
RS
,
Zhang
Z
,
Henson
BS
,
Kurnit
DM
,
Carey
TE
,
D'Silva
NJ
. 
Rap1A and rap1B ras-family proteins are prominently expressed in the nucleus of squamous carcinomas: nuclear translocation of GTP-bound active form
.
Oncogene
2003
;
22
:
6243
56
.
18.
Izumi
K
,
Terashi
H
,
Marcelo
CL
,
Feinberg
SE
. 
Development and characterization of a tissue-engineered human oral mucosa equivalent produced in a serum-free culture system
.
J Dent Res
2000
;
79
:
798
805
.
19.
Ota
I
,
Li
XY
,
Hu
Y
,
Weiss
SJ
. 
Induction of a MT1-MMP and MT2-MMP-dependent basement membrane transmigration program in cancer cells by Snail1
.
Proc Natl Acad Sci U S A
2009
;
106
:
20318
23
.
20.
Matschke
K
,
Da Silva-Azevedo
L
,
Hlushchuk
R
,
Djonov
V
,
Baum
O
. 
Annexins as cell-type-specific markers in the developing chicken chorionallantoic membrane
.
Cell Tissue Res
2006
;
323
:
395
404
.
21.
Sabeh
F
,
Ota
I
,
Holmbeck
K
,
Birkedal-Hansen
H
,
Soloway
P
,
Balbin
M
, et al
Tumor cell traffic through the extracellular matrix is controlled by the membrane-anchored collagenase MT1-MMP
.
J Cell Biol
2004
;
167
:
769
81
.
22.
Hagedorn
M
,
Javerzat
S
,
Gilges
D
,
Meyre
A
,
de Lafarge
B
,
Eichmann
A
, et al
Accessing key steps of human tumor progression in vivo by using an avian embryo model
.
Proc Natl Acad Sci U S A
2005
;
102
:
1643
8
.
23.
Zhao
W
,
Liu
M
,
D'Silva
NJ
,
Kirkwood
KL
. 
Tristetraprolin regulates interleukin-6 expression through p38 MAPK-dependent affinity changes with mRNA 3′ untranslated region
.
J Interferon Cytokine Res
2011
;
31
:
629
37
.
24.
Palanisamy
V
,
Jakymiw
A
,
Van Tubergen
EA
,
D'Silva
NJ
,
Kirkwood
KL
. 
Control of cytokine mRNA expression by RNA-binding proteins and microRNAs
.
J Dent Res
2012
;
91
:
651
8
.
25.
Banerjee
R
,
Henson
BS
,
Russo
N
,
Tsodikov
A
,
D'Silva
NJ
. 
Rap1 mediates galanin receptor 2-induced proliferation and survival in squamous cell carcinoma
.
Cell Signal
2011
;
23
:
1110
8
.
26.
Banerjee
R
,
Mani
RS
,
Russo
N
,
Scanlon
CS
,
Tsodikov
A
,
Jing
X
, et al
The tumor suppressor gene rap1GAP is silenced by miR-101-mediated EZH2 overexpression in invasive squamous cell carcinoma
.
Oncogene
2011
;
30
:
4339
49
.
27.
Clement
SL
,
Scheckel
C
,
Stoecklin
G
,
Lykke-Andersen
J
. 
Phosphorylation of tristetraprolin by MK2 impairs AU-rich element mRNA decay by preventing deadenylase recruitment
.
Mol Cell Biol
2011
;
31
:
256
66
.
28.
Kumar
S
,
Jiang
MS
,
Adams
JL
,
Lee
JC
. 
Pyridinylimidazole compound SB 203580 inhibits the activity but not the activation of p38 mitogen-activated protein kinase
.
Biochem Biophys Res Commun
1999
;
263
:
825
31
.
29.
Lee
JC
,
Laydon
JT
,
McDonnell
PC
,
Gallagher
TF
,
Kumar
S
,
Green
D
, et al
A protein kinase involved in the regulation of inflammatory cytokine biosynthesis
.
Nature
1994
;
372
:
739
46
.
30.
Anand
P
,
Shenoy
R
,
Palmer
JE
,
Baines
AJ
,
Lai
RY
,
Robertson
J
, et al
Clinical trial of the p38 MAP kinase inhibitor dilmapimod in neuropathic pain following nerve injury
.
Eur J Pain
2011
;
15
:
1040
8
.
31.
Kakuguchi
W
,
Kitamura
T
,
Kuroshima
T
,
Ishikawa
M
,
Kitagawa
Y
,
Totsuka
Y
, et al
HuR knockdown changes the oncogenic potential of oral cancer cells
.
Mol Cancer Res
2010
;
8
:
520
8
.
32.
Xu
N
,
Chen
CY
,
Shyu
AB
. 
Versatile role for hnRNP D isoforms in the differential regulation of cytoplasmic mRNA turnover
.
Mol Cell Biol
2001
;
21
:
6960
71
.
33.
Zhang
L
,
Lee
JE
,
Wilusz
J
,
Wilusz
CJ
. 
The RNA-binding protein CUGBP1 regulates stability of tumor necrosis factor mRNA in muscle cells: implications for myotonic dystrophy
.
J Biol Chem
2008
;
283
:
22457
63
.
34.
Coussens
LM
,
Fingleton
B
,
Matrisian
LM
. 
Matrix metalloproteinase inhibitors and cancer: trials and tribulations
.
Science
2002
;
295
:
2387
92
.
35.
Al-Souhibani
N
,
Al-Ahmadi
W
,
Hesketh
JE
,
Blackshear
PJ
,
Khabar
KS
. 
The RNA-binding zinc-finger protein tristetraprolin regulates AU-rich mRNAs involved in breast cancer-related processes
.
Oncogene
2010
;
29
:
4205
15
.
36.
Suswam
E
,
Li
Y
,
Zhang
X
,
Gillespie
GY
,
Li
X
,
Shacka
JJ
, et al
Tristetraprolin down-regulates interleukin-8 and vascular endothelial growth factor in malignant glioma cells
.
Cancer Res
2008
;
68
:
674
82
.
37.
Carballo
E
,
Blackshear
PJ
. 
Roles of tumor necrosis factor-alpha receptor subtypes in the pathogenesis of the tristetraprolin-deficiency syndrome
.
Blood
2001
;
98
:
2389
95
.
38.
Al Ghouleh
I
,
Magder
S
. 
NADPH oxidase-derived superoxide destabilizes LPS-induced interleukin-8 mRNA via p38, Erk1/2 MAPK and the destabilizing factor tristetraprolin1
.
Shock
2012
;
37
:
433
40
.
39.
Zhang
Z
,
Mitra
RS
,
Henson
BS
,
Datta
NS
,
McCauley
LK
,
Kumar
P
, et al
Rap1GAP inhibits tumor growth in oropharyngeal squamous cell carcinoma
.
Am J Pathol
2006
;
168
:
585
96
.
40.
Scanlon
CS
,
Van Tubergen
EA
,
Inglehart
RC
,
D'Silva
NJ
. 
Biomarkers of epithelial-mesenchymal transition in squamous cell carcinoma
.
J Dent Res
2013
;
92
:
114
21
41.
Lokman
NA
,
Elder
AS
,
Ricciardelli
C
,
Oehler
MK
. 
Chick chorioallantoic membrane (CAM) assay as an in vivo model to study the effect of newly identified molecules on ovarian cancer invasion and metastasis
.
Int J Mol Sci
2012
;
13
:
9959
70
.
42.
Rowe
SL
,
Stegemann
JP
. 
Microstructure and mechanics of collagen-fibrin matrices polymerized using ancrod snake venom enzyme
.
J Biomech Eng
2009
;
131
:
061012
.
43.
Brennan
SE
,
Kuwano
Y
,
Alkharouf
N
,
Blackshear
PJ
,
Gorospe
M
,
Wilson
GM
. 
The mRNA-destabilizing protein tristetraprolin is suppressed in many cancers, altering tumorigenic phenotypes and patient prognosis
.
Cancer Res
2009
;
69
:
5168
76
.
44.
Lee
HH
,
Son
YJ
,
Lee
WH
,
Park
YW
,
Chae
SW
,
Cho
WJ
, et al
Tristetraprolin regulates expression of VEGF and tumorigenesis in human colon cancer
.
Int J Cancer
2010
;
126
:
1817
27
.
45.
Griseri
P
,
Bourcier
C
,
Hieblot
C
,
Essafi-Benkhadir
K
,
Chamorey
E
,
Touriol
C
, et al
A synonymous polymorphism of the Tristetraprolin (TTP) gene, an AU-rich mRNA-binding protein, affects translation efficiency and response to Herceptin treatment in breast cancer patients
.
Hum Mol Genet
2011
;
20
:
4556
68
.

Supplementary data