Purpose: Osteopontin is a secreted cytokine that binds to the cell surface CD44v6 receptor. We studied osteopontin and CD44v6 expression in laryngeal squamous cell carcinomas and correlated osteopontin expression levels with clinicopathologic tumor features.

Experimental Design: We used immunohistochemistry, immunoblotting, and reverse transcription-PCR to study osteopontin expression in 58 laryngeal squamous cell carcinomas. Cultured squamous carcinoma cells were treated with exogenous osteopontin or with RNA interference to knockdown osteopontin expression.

Results: Osteopontin expression was higher in all the invasive carcinomas than in patient-matched normal mucosa. Its expression levels were significantly correlated with tumor stage and grade and with the presence of lymph node and distant metastases. Osteopontin positivity was negatively correlated with overall survival (P = 0.03). Osteopontin expression was paralleled by intense cell surface reactivity for CD44v6. Treatment of squamous carcinoma cells with recombinant osteopontin sharply increased proliferation and Matrigel invasion in comparison with the untreated cells parallel to activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/mitogen-activated protein kinase signaling cascade. Osteopontin knockdown by RNA interference, anti-CD44 antibodies, and mitogen-activated protein kinase/extracellular signal-regulated kinase kinase inhibition prevented these effects.

Conclusions: These results identify osteopontin as a marker and a potential therapeutic target in cases of aggressive laryngeal squamous cell carcinomas.

Head and neck squamous cell carcinoma (HNSCC) is the sixth most frequent cancer. Laryngeal squamous cell carcinoma (LSCC) is the most common HNSCC (13). Notwithstanding primary prevention, screening, surgical treatment, and radiotherapy, the long-term survival rate of LSCC patients has remained substantially unchanged in the last two decades (4, 5). Stage and histologic grade are prognostic factors in LSCC but do not always distinguish between high-risk and low-risk patients (15). Various biological prognostic markers have been identified for LSCC and other HNSCC types: mutation in the p53 tumor suppressor gene (6), amplification of cyclin D1 (7, 8), overexpression of the epidermal growth factor receptor (9) and vascular endothelial growth factor (10), and reduced expression of the CIP/KIP cell cycle inhibitory proteins (11). However, little is known about the molecular mechanisms that govern the establishment and maintenance of the LSCC neoplastic phenotype.

Osteopontin, also known as secreted phosphoprotein 1, is a highly acidic calcium-binding glycosylated phosphoprotein. It is a cytokine (early T lymphocyte antigen-1 or interleukin-28) that regulates T helper cell-1 function (12, 13). In addition, osteopontin binds to the cell surface receptors αv- or β1-containing integrins and CD44 (14, 15), thereby supporting chemotaxis, attachment, and migration of many epithelial cell types (16, 17). CD44 is expressed as a standard receptor (CD44s) and in multiple splice isoforms (CD44v), whose expression is altered during tumor growth and progression. Expression of the v6 variant exon of CD44 is necessary for osteopontin binding (18). CD44 splice variants are thought to be correlated with invasive growth and metastasis in many tumor types (18). Osteopontin is overexpressed in many human tumors (e.g., colon, breast, liver, prostate, gastric, ovarian, and lung carcinomas; refs. 1922).

We have explored osteopontin expression in LSCC. Our results indicate that osteopontin is a promising molecular marker for LSCC risk assessment. At cellular level, osteopontin binding to CD44v6 promoted SCC cell growth and invasion.

Tumors. Archival tumor samples from 58 patients (57 males and 1 female) with laryngeal cancer were retrieved from the files of the Department of Biomorphological and Functional Sciences, Pathology Section, University of Naples “Federico II,” after having obtained informed consent. Clinicopathologic data were recorded (Table 1). Patients underwent surgery at the Institute of Otolaryngology, University of Naples “Federico II,” between 1997 and 2001. The study was approved by the institutional review board committee. The patients' age ranged between 43 and 83 years, with a mean of 64.7 years. Patients underwent otolaryngologic examination, fiberoptic study, and radiological evaluation and were treated by surgery alone, except for patients with locally advanced tumors (T4), who received postsurgical radiation therapy. The patients underwent a median of 60 months' follow-up, which consisted of clinical and radiological evaluation at 3-month intervals for the first year and at 6-month intervals thereafter. After surgical resection, tissues were fixed in 10% neutral buffered formalin and embedded in paraffin blocks. Sections (4 μm thick) were stained with H&E. Histologic grading and tumor-node-metastasis classification were done according to the recommendations of the International Union Against Cancer (23, 24). The pathologic analysis was done in a blinded manner with respect to the patients' clinical data. For five patients, a 10-μm-thick section was processed for dissection. Paraffin was removed by treatment with xylene for 3 hours at room temperature followed by tissue rehydration through multiple graded ethanol solutions and distilled water. The cancerous region was identified microscopically; normal and tumor tissues were dissected with a sterile 30-gauge hypodermic needle. The collected samples (∼120,000 cells) were placed into 1.5-mL microcentrifuge tubes and processed for RNA extraction.

Table 1.

Clinicopathologic features of studied laryngeal cancer patients

CharacteristicsTotal (%)
No. subjects  
    Male 57 (98) 
    Female 1 (2) 
Disease site  
    Glottis-hypoglottis 43 (74) 
    Supraglottis 15 (26) 
Histologic differentiation  
    G1 19 (33) 
    G2 21 (36) 
    G3 18 (31) 
Stage  
    T1 9 (16) 
    T2 19 (33) 
    T3/T4 30 (52) 
Lymph nodes  
    N+ 15 (26) 
    No 25 (43) 
    Nx 18 (31) 
Metastasis  
    Mo 27 (46) 
    Bone 4 (7) 
    Lung 8 (14) 
    Brain 1 (2) 
    Mx 18 (31) 
CharacteristicsTotal (%)
No. subjects  
    Male 57 (98) 
    Female 1 (2) 
Disease site  
    Glottis-hypoglottis 43 (74) 
    Supraglottis 15 (26) 
Histologic differentiation  
    G1 19 (33) 
    G2 21 (36) 
    G3 18 (31) 
Stage  
    T1 9 (16) 
    T2 19 (33) 
    T3/T4 30 (52) 
Lymph nodes  
    N+ 15 (26) 
    No 25 (43) 
    Nx 18 (31) 
Metastasis  
    Mo 27 (46) 
    Bone 4 (7) 
    Lung 8 (14) 
    Brain 1 (2) 
    Mx 18 (31) 

Immunohistochemistry. Serial tumor sections (4 μm thick) were mounted on poly-l-lysine-coated glass slides. After antigen retrieval, the slides were incubated with anti-osteopontin (5 μg/mL; 10A16, Assay Designs, Ann Arbor, MI) or anti-CD44 (1:100; NCL-CD44v6, clone VFF-7, Novocastra Laboratories Ltd., Newcastle upon Tyne, United Kingdom) monoclonal antibodies (mAb). The sections were incubated (overnight at 4°C) with the primary antibody, biotinylated anti-IgG, and the premixed avidin-biotin complex (Vectastain ABC kits, Vector Laboratories, Burlingame, CA). The immune reaction was revealed with 0.06 mmol/L 3,3′-diaminobenzidine (DAKO, Carpinteria, CA) and 2 mmol/L H2O2. The slides were counterstained with hematoxylin. Anti-osteopontin antibody was preincubated with a 5-fold molar excess of osteopontin peptide to ascertain the specificity of the reaction. Control slides, stained with preimmune serum, were included as an additional negative control. The results of the immunohistochemical staining were evaluated separately by two investigators in a blinded manner. The percentage of positive tumor cells was determined by examining at least five representative microscope areas at a ×400 magnification. For osteopontin, samples were assigned to one of the four following categories: 0, absence of positive cells; +, <10% of positive cells; ++, 10% to 50% of positive cells; and +++, >50% of positive cells. Immunohistochemical staining for CD44v6 was expressed as “basal” or “full thickness” based on the epithelium layers stained.

Cell lines. Human normal epidermal keratinocytes (HNEK; neonatal) were cultured in keratinocyte growth medium according to the recommendation of the manufacturer (Cambrex, East Rutherford, NJ). HN and BHY cell lines were derived from a human oral cavity SCC (25), CAL27 and CAL33 cell lines were from human tongue SCC (26), and Hep2 cells were from a LSCC (27); KB are epidermoid cancer cells (28). Tumor cells were maintained in DMEM supplemented with 10% fetal bovine serum, 2 mmol/L l-glutamine, and 100 units/mL penicillin-streptomycin (Life Technologies, Paisley, PA). For cell treatments, recombinant mouse osteopontin protein was from R&D Systems (Minneapolis, MN). U0126 was from Calbiochem (San Diego, CA). Blocking CD44 mAbs were purified from the KM81 hybridoma cell line (TIB-241, American Type Culture Collection, Manassas, VA; ref. 29). For blocking experiments, cells were preincubated for 30 minutes with the KM81 mAb (10 μg/mL) at 37°C, 5% CO2.

RNA extraction and reverse transcription-PCR. Tissue samples were snap frozen in liquid nitrogen and stored at −80°C before RNA extraction. Tissues were homogenized using a Mixer Mill Homogenizer (Qiagen, Crawley, West Sussex, United Kingdom). Total RNA from the indicated cell cultures and from frozen tissue samples was prepared using the RNeasy Mini kit (Qiagen) according to the manufacturer's instructions. Only tissue samples containing >70% neoplastic cells were used. Total RNA (2.5 μg) was retrotranscribed into cDNA by using the GeneAmp RNA PCR Core kit (Applied Biosystems, Foster City, CA). PCR amplification was done using 2.5 μL of the reverse transcription product in a reaction volume of 25 μL. To exclude DNA contamination, each PCR reaction was also done on untranscribed RNA. The levels of the housekeeping β-actin transcript served as a control of equal RNA loading. Primers were designed with the Primer 3 program (http://www-genome.wi.mit.edu/cgi-bin/primer/primer3_www.cgi) and synthesized by MWG Biotech (Ebersberg, Germany). Primer sequences were osteopontin forward 5′-AGGAGGAGGCAGAGCACA-3′, osteopontin reverse 5′-CTGGTATGGCACAGGTGATG-3′, CD44 (exon 2) forward 5′-GCTTTCAATAGCACCTTGCC-3′, CD44 (exon3) reverse 5′-GTTGTTTGCTGCACAGATGG-3′, CD44 (exon v6) reverse 5′-GTTGCCAAACCACTGTTCCT-3′, β-actin forward 5′-TGCGTGACATTAAGGAGAAG-3′, and β-actin reverse 5′-GCTCGTAGCTCTTCTCCA-3′. Reverse transcription-PCR (RT-PCR) products were loaded on a 2% agarose gel and stained with 0.5 μg/mL ethidium bromide, and the corresponding image was saved with the Typhoon 8600 laser scanning system (Amersham Pharmacia Biotech, Buckinghamshire, United Kingdom). The density and width of each band were quantified using the ImageQuant 5.0 (Amersham Pharmacia Biotech).

Protein studies. Immunoblotting experiments were done according to standard procedures. For tissue protein extraction, samples were snap frozen and homogenized in lysis buffer by using the Mixer Mill apparatus (Qiagen). Protein concentration was estimated with a modified Bradford assay (Bio-Rad, Hercules, CA). Antigens were revealed by an enhanced chemiluminescence detection kit (Amersham Pharmacia Biotech). Anti-osteopontin goat polyclonal antibody (K20) and rabbit polyclonal anti-CD44 (H300) were from Santa Cruz Biotechnology (Santa Cruz, CA). Monoclonal anti-α-tubulin was from Sigma-Aldrich (St. Louis, MO). Anti-phosphorylated mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK) 1/2 (Ser217/Ser221), anti-phosphorylated p44/42 mitogen-activated protein kinase (Thr202/Tyr204; ERK), and anti-p44/42 mitogen-activated protein kinase were from Cell Signaling Technology, Inc. (Beverly, MA).

Flow cytometric analysis. Subconfluent cells (1 × 106) were detached from culture dishes. After saturation with 1 μg human IgG per 105 cells, cells were incubated for 20 minutes on ice with antibodies specific for human CD44v6 (R&D Systems) or isotype control antibody. After incubation, unreacted antibody was removed. Cells were then incubated (30 minutes, 4°C) with 100 μL fluorescein-conjugated goat anti-mouse IgG/M (Jackson ImmunoResearch, West Grove, PA) and analyzed on a FACSCalibur cytofluorimeter using the CellQuest software (Becton Dickinson, San Jose, CA). Analyses were done in triplicate. In each analysis, a total of 104 events were calculated.

Chemoinvasion. The cell suspension (1 × 105 cells per well) was added to the upper chamber of Transwell cell culture chambers on a prehydrated polycarbonate membrane filter of 8-μm pore size (Costar, Cambridge, MA) coated with 35 μg Matrigel (Collaborative Research, Inc., Bedford, MA). The lower chamber was filled with complete medium, and when required, purified recombinant osteopontin was added at the concentration of 100 ng/mL. To inhibit Matrigel invasion, cells were preincubated with 10 μg/mL CD44-blocking antibodies (KM81) or with 10 μmol/L U0126. After 24-hour incubation at 37°C, nonmigrating cells on the upper side of the filter were wiped off and migrating cells on the reverse side of the filter were stained with 0.1% crystal violet in 20% methanol for 15 minutes and photographed. The stained cells were lysed in 10% acetic acid. Triplicate samples were analyzed at 570 nm with an ELISA reader (model 550 microplate reader, Bio-Rad). The results were expressed as percentage of migrating cells.

Bromodeoxyuridine incorporation. Cells were seeded on glass coverslips and bromodeoxyuridine (BrdUrd) was added to the cell culture medium at a final concentration of 100 μg/mL (BrdUrd Labeling and Detection kit, Boehringer Mannheim, Mannheim, Germany). After 1-hour incubation, cells were fixed with 70% ethanol/50 mmol/L glycine (pH 2.0). Coverslips were incubated with anti-BrdUrd mouse mAb and with a FITC-conjugated anti-mouse antibody. All coverslips were counterstained in PBS containing Hoechst 33258 (final concentration, 1 μg/mL; Sigma-Aldrich), rinsed in water and mounted in Moviol on glass slides. The fluorescent signal was visualized with an epifluorescent microscope (Axioskop 2, Zeiss) interfaced with the image analyzer software Axiovision (Zeiss, Gottingen, Germany).

RNA silencing. Small inhibitor duplex RNAs targeting human osteopontin were designed with a small interfering RNA (siRNA) selection program available online at http://jura.wi.mit.edu/siRNAext/ and were chemically synthesized by PROLIGO (Boulder, CO). Sense strand for siRNA targeting was (osteopontin siRNA) 5′-AAGCAGCUUUACAACAAAUACCC-3′. As a control, a nonspecific siRNA duplex containing the same nucleotides but in irregular sequence (scrambled) was used. The day before transfection, 1 × 105 cells were plated in 35-mm dishes in DMEM supplemented with 10% fetal bovine serum and without antibiotics. Transfection was done using 360 pmol siRNA and 18 μL Oligofectamine reagent (Invitrogen, Groningen, the Netherlands) following the manufacturer's instruction. Cells were kept in 2.5% serum and BrdUrd incorporation was measured 48 hours after transfection.

Statistical analysis. Statistical evaluation of the data was done with a two-tailed Student's t test when simple comparison between two groups was required; χ2 test was used to establish the statistical significance of distributions. Nonparametric Spearman's correlation coefficient method was used to assess the statistical significance of the correlation between clinicopathologic characteristics of tumor and osteopontin expression. Survival curves of the patients were calculated using the Kaplan-Meier method and analysis was done by the log-rank test. Differences were significant at P < 0.05. Statistical analysis was done using the JMP software program version 5.1.1 (SAS Institute, Inc., Austin, TX).

Immunohistochemical detection of osteopontin up-regulation in laryngeal squamous cell carcinomas. Fifty-eight larynx carcinomas at different grades of malignancy and the corresponding normal tissues (Table 1) were tested for osteopontin expression by immunohistochemistry with an anti-osteopontin-specific mAb. Representative immunohistochemical stainings are shown in Fig. 1 and the entire data set is reported in Table 2. Osteopontin was virtually undetectable (<2.0% of cells) in normal tissues (n = 58; data not shown). Hyperplastic epithelia (n = 20) were also constantly negative for osteopontin staining (Fig. 1E; data not shown). In contrast, 93% (54 of 58) of the tumor samples were osteopontin positive (Fig. 1A and C). The signal was confined to tumor cells. The specificity of signal was shown by competition with a molar excess of osteopontin-blocking peptide (Fig. 1B, D, and F). Overall, 37% of T2 tumors and 73% of T3/T4 tumors showed intense (+++) osteopontin immunostaining. Moreover, osteopontin immunostaining was intense (+++) in 21%, 52%, and 78% of G1, G2, and G3 tumors, respectively. Finally, 60% of tumors with lymph node metastases and 92% of tumors with distant metastases had intense osteopontin staining. Accordingly, metastatic tissues were intensely osteopontin positive (data not shown). Thus, osteopontin reactivity was correlated with tumor stage (P < 0.0001) and grade (P < 0.0001) and with the presence of lymph node (P < 0.0046) and distant (P < 0.0001) metastases (Table 3). Importantly, the 5-year survival rate for LSCC patients was negatively correlated with intense osteopontin staining. Five-year survival was 71.40% for osteopontin-positive cases (++) and 51.70% for osteopontin-positive cases (+++) as shown by the Kaplan-Meier survival curves reported in Fig. 2 (P = 0.03, two-sided log-rank test).

Fig. 1.

Immunohistochemical detection of osteopontin in laryngeal carcinomas. A, an infiltrating LSCC (G2, N+, M0) showing intense (+++) osteopontin staining (×106). B, the same sample as in (A): no signal was detected after preincubation with a molar excess of osteopontin-blocking peptide. C, an infiltrating LSCC (G3, N0, M0) showing (++) osteopontin immunoreactivity (×106;). D, the same sample as in (C) after preincubation with a molar excess of osteopontin-blocking peptide. E, hyperplastic epithelium (without dysplasia) negative for osteopontin staining (×150). F, the same sample as in (E) after preincubation with osteopontin-blocking peptide.

Fig. 1.

Immunohistochemical detection of osteopontin in laryngeal carcinomas. A, an infiltrating LSCC (G2, N+, M0) showing intense (+++) osteopontin staining (×106). B, the same sample as in (A): no signal was detected after preincubation with a molar excess of osteopontin-blocking peptide. C, an infiltrating LSCC (G3, N0, M0) showing (++) osteopontin immunoreactivity (×106;). D, the same sample as in (C) after preincubation with a molar excess of osteopontin-blocking peptide. E, hyperplastic epithelium (without dysplasia) negative for osteopontin staining (×150). F, the same sample as in (E) after preincubation with osteopontin-blocking peptide.

Close modal
Table 2.

Osteopontin positivity in laryngeal carcinomas

Characteristics (no. samples)Osteopontin positivity*
Disease site  
    Glottis-hypoglottis (43) 22/43 (+++) 
 16/43 (++) 
 3/43 (+) 
 2/43 (0) 
    Supraglottis (15) 7/15 (+++) 
 5/15 (++) 
 1/15 (+) 
 2/15 (0) 
Stage  
    T1 (9) 3/9 (++) 
 2/9 (+) 
 4/9 (0) 
    T2 (19) 7/19 (+++) 
 10/19 (++) 
 2/19 (+) 
    T3/T4 (30)  
 22/30 (+++) 
 8/30 (++) 
Grade  
    G1 (19) 4/19 (+++) 
 8/19 (++) 
 3/19 (+) 
 4/19 (0) 
    G2 (21) 11/21 (+++) 
 9/21 (++) 
 1/21 (+) 
    G3 (18) 14/18 (+++) 
 4/18 (++) 
Lymph node metastases  
    N+ (15) 9/15 (+++) 
 6/15 (++) 
    No (25) 6/25 (+++) 
 9/25 (++) 
 8/25 (+) 
 2/25 (0) 
    Nx (18)  
Distant metastases  
    M+ (13) 12/13 (+++) 
 1/13 (++) 
    Mo (27) 7/27 (+++) 
 12/27 (++) 
 4/27 (+) 
 4/27 (0) 
    Mx (18)  
Characteristics (no. samples)Osteopontin positivity*
Disease site  
    Glottis-hypoglottis (43) 22/43 (+++) 
 16/43 (++) 
 3/43 (+) 
 2/43 (0) 
    Supraglottis (15) 7/15 (+++) 
 5/15 (++) 
 1/15 (+) 
 2/15 (0) 
Stage  
    T1 (9) 3/9 (++) 
 2/9 (+) 
 4/9 (0) 
    T2 (19) 7/19 (+++) 
 10/19 (++) 
 2/19 (+) 
    T3/T4 (30)  
 22/30 (+++) 
 8/30 (++) 
Grade  
    G1 (19) 4/19 (+++) 
 8/19 (++) 
 3/19 (+) 
 4/19 (0) 
    G2 (21) 11/21 (+++) 
 9/21 (++) 
 1/21 (+) 
    G3 (18) 14/18 (+++) 
 4/18 (++) 
Lymph node metastases  
    N+ (15) 9/15 (+++) 
 6/15 (++) 
    No (25) 6/25 (+++) 
 9/25 (++) 
 8/25 (+) 
 2/25 (0) 
    Nx (18)  
Distant metastases  
    M+ (13) 12/13 (+++) 
 1/13 (++) 
    Mo (27) 7/27 (+++) 
 12/27 (++) 
 4/27 (+) 
 4/27 (0) 
    Mx (18)  
*

Osteopontin expression was assessed by immunohistochemistry and scored as follows: 0, absence of positive cells; +, <10% positive cells; ++, 10% to 50% positive cells; and +++, 50% to 100% positive cells.

Table 3.

Correlation of osteopontin expression and clinicopathologic characteristics of laryngeal carcinomas

Osteopontin positivityrsP
Tumor stage 0.6230 <0.0001 
Grade 0.5169 <0.0001 
Node 0.4391 <0.0046 
Distant metastases 0.7821 <0.0001 
Osteopontin positivityrsP
Tumor stage 0.6230 <0.0001 
Grade 0.5169 <0.0001 
Node 0.4391 <0.0046 
Distant metastases 0.7821 <0.0001 

NOTE: Correlation between osteopontin expression and tumor stage, grade, node, and distant metastases analyzed by the Spearman rank correlation test: correlation coefficient (rs) and Ps are shown (Ps < 0.05 were considered significant).

Fig. 2.

Osteopontin staining negatively correlates with LSCC patient survival. Kaplan-Meier survival plots for LSCC patients grouped by the level of expression of osteopontin (OPN). LSCC tumoral samples were stratified in three categories [0/+ (n = 8), ++ (n = 21), and +++ (n = 29)] based on intensity of osteopontin immunostaining. P was determined by a two-sided log-rank test. For patients whose tumors had osteopontin (++) immunostain, the 2- and 5-year overall survival rates were 76.19% [95% confidence interval (95% CI), 56.53-95.87%] and 71.40% (95% CI, 48.53-94.27%), respectively. For patients whose tumors had osteopontin (+++) immunostain, the 2- and 5-year overall survival rates were 58.60% (95% CI, 35.85-81.35%) and 51.70% (95% CI, 26.41-76.99%), respectively. All patients whose tumors had osteopontin (0/+) stain were still alive at the end of the study.

Fig. 2.

Osteopontin staining negatively correlates with LSCC patient survival. Kaplan-Meier survival plots for LSCC patients grouped by the level of expression of osteopontin (OPN). LSCC tumoral samples were stratified in three categories [0/+ (n = 8), ++ (n = 21), and +++ (n = 29)] based on intensity of osteopontin immunostaining. P was determined by a two-sided log-rank test. For patients whose tumors had osteopontin (++) immunostain, the 2- and 5-year overall survival rates were 76.19% [95% confidence interval (95% CI), 56.53-95.87%] and 71.40% (95% CI, 48.53-94.27%), respectively. For patients whose tumors had osteopontin (+++) immunostain, the 2- and 5-year overall survival rates were 58.60% (95% CI, 35.85-81.35%) and 51.70% (95% CI, 26.41-76.99%), respectively. All patients whose tumors had osteopontin (0/+) stain were still alive at the end of the study.

Close modal

The interaction of osteopontin with the CD44 cell surface receptor has been implicated in many signal transduction pathways. CD44 pre-mRNA is encoded by 20 exons. The constant 5′-terminal five exons encode the NH2-terminal extracellular portion of the CD44 protein, whereas the constant 3′-terminal five exons encode the transmembrane and the short cytosolic tail of the protein. An additional 10 exons (variants v1-v10) are alternatively spliced and encode the extracellular membrane-proximal stem structure (18). Cancer cells often overexpress CD44 variants that include a variable number of “v” exons. The v6 exon has been reported to be important for efficient osteopontin binding (18). Thus, we sought to verify whether CD44v6 molecules were expressed in LSCC. LSCC samples (n = 58) were constantly CD44v6 positive at immunohistochemistry (representative samples are shown in Fig. 3A-C). In tumors, CD44v6-positive cells showed full thickness staining, whereas only basal cells were CD44v6 positive in normal stratified epithelium (data not shown).

Fig. 3.

Expression of CD44v6 in LSCC. A, an infiltrating LSCC (G2, N0, M0) strongly immunoreactive for CD44v6 (×150). B, full thickness CD44v6 positivity in a G2, N+, M0 LSCC sample (×150). C, full thickness CD44v6 positivity in a G2, N0, M0 LSCC sample (×400). D, LSCC: absence of signal in the presence of preimmune serum (×400).

Fig. 3.

Expression of CD44v6 in LSCC. A, an infiltrating LSCC (G2, N0, M0) strongly immunoreactive for CD44v6 (×150). B, full thickness CD44v6 positivity in a G2, N+, M0 LSCC sample (×150). C, full thickness CD44v6 positivity in a G2, N0, M0 LSCC sample (×400). D, LSCC: absence of signal in the presence of preimmune serum (×400).

Close modal

Osteopontin and CD44 up-regulation in laryngeal squamous cell carcinomas at protein and mRNA level. Protein lysates were harvested from selected high-stage/grade snap-frozen LSCC samples (T3, G3; n = 10) and from the corresponding adjacent normal mucosa from the same patients, and osteopontin protein levels were examined by immunoblotting. As shown in Fig. 4A, the osteopontin protein (Mr ∼60,000) was abundantly expressed in all carcinomas but was barely detectable in matched normal tissues (Fig. 4A). To determine whether up-regulation occurred at transcriptional level, we subjected the same LSCC samples (n = 10) to RT-PCR. Osteopontin mRNA was abundantly overexpressed at mRNA level in tumors with respect to adjacent normal mucosa from the same patients (Fig. 4B; data not shown). Phosphorimaging analysis of band intensity indicated that osteopontin mRNA was 15 ± 3–fold higher in LSCC samples than in the normal tissue counterpart. To validate the results, we analyzed osteopontin mRNA levels in purified dissected tumor cells from five representative high-stage/grade LSCC samples and corresponding normal cells by RT-PCR; the representative samples shown in Fig. 4C showed that osteopontin mRNA accumulation was restricted to tumor cells.

Fig. 4.

Osteopontin up-regulation in LSCC samples at the protein and mRNA levels. A, levels of osteopontin protein were evaluated by immunoblot in LSCC and in adjacent normal epithelium: T, tumoral sample; N, normal epithelium. Anti-α-tubulin were used for normalization. Representative of three independent experiments. B, semiquantitative RT-PCR (25 cycles) was done to detect osteopontin mRNA levels in the indicated LSCC samples and in adjacent normal epithelium. β-Actin mRNA detection was used for normalization. Band intensity was calculated by phosphorimaging. Representative of three independent experiments. C, RT-PCR (28 cycles) was done on purified cells from two representative samples of LSCC (sample 1: G3, N+, M0; sample 2: G2, N0, M0) and from the corresponding normal cells after microscope-guided manual dissection.

Fig. 4.

Osteopontin up-regulation in LSCC samples at the protein and mRNA levels. A, levels of osteopontin protein were evaluated by immunoblot in LSCC and in adjacent normal epithelium: T, tumoral sample; N, normal epithelium. Anti-α-tubulin were used for normalization. Representative of three independent experiments. B, semiquantitative RT-PCR (25 cycles) was done to detect osteopontin mRNA levels in the indicated LSCC samples and in adjacent normal epithelium. β-Actin mRNA detection was used for normalization. Band intensity was calculated by phosphorimaging. Representative of three independent experiments. C, RT-PCR (28 cycles) was done on purified cells from two representative samples of LSCC (sample 1: G3, N+, M0; sample 2: G2, N0, M0) and from the corresponding normal cells after microscope-guided manual dissection.

Close modal

We next examined CD44 expression in LSCC (T3, G3) samples (n = 10) using immunoblotting. An intense broad band of a relative molecular mass of 90 kDa was detected in tumors (Fig. 5A; data not shown). In contrast, CD44 was weakly expressed (<10 ± 3–fold compared with LSCC samples) in normal tissue counterparts (Fig. 5A). Then, we used different combinations of amplimers in RT-PCR experiments with RNA extracted from LSCC (T3, G3) samples (n = 10). LSCC overexpressed CD44 mRNA species containing the v6 or both v3 and v6 variant exons (Fig. 5B; data not shown).

Fig. 5.

CD44 up-regulation in LSCC samples. A, equal amounts of proteins (100 μg) from LSCC tumor samples and adjacent normal epithelium underwent Western blotting with an anti-CD44 antibody. Anti-α-tubulin was used for normalization. Representative of three independent experiments. B, semiquantitative RT-PCR (25 cycles) was used to detect mRNA levels of CD44 variants in LSCC samples. Band intensity was calculated by phosphorimaging. Amplimers mapping on exons 2 and 3 were used to detect all CD44 mRNA species, whereas the exon 2-v6 primer pair was used to detect variant mRNA species containing exon v6. In LSCC, the latter primer pair amplified two major products of 734 and 876 bp containing, along with standard exons 2 to 5, exon v6 or both exon v3 and v6 as proven by subsequent Southern hybridization with exon-specific probes (data not shown). β-Actin mRNA detection was used for normalization. Representative of three independent experiments.

Fig. 5.

CD44 up-regulation in LSCC samples. A, equal amounts of proteins (100 μg) from LSCC tumor samples and adjacent normal epithelium underwent Western blotting with an anti-CD44 antibody. Anti-α-tubulin was used for normalization. Representative of three independent experiments. B, semiquantitative RT-PCR (25 cycles) was used to detect mRNA levels of CD44 variants in LSCC samples. Band intensity was calculated by phosphorimaging. Amplimers mapping on exons 2 and 3 were used to detect all CD44 mRNA species, whereas the exon 2-v6 primer pair was used to detect variant mRNA species containing exon v6. In LSCC, the latter primer pair amplified two major products of 734 and 876 bp containing, along with standard exons 2 to 5, exon v6 or both exon v3 and v6 as proven by subsequent Southern hybridization with exon-specific probes (data not shown). β-Actin mRNA detection was used for normalization. Representative of three independent experiments.

Close modal

Osteopontin and CD44v6 overexpression in squamous cell carcinoma cell lines. We evaluated osteopontin expression in six cultured human SCC lines. A primary culture of HNEK was used as a control. Osteopontin protein and mRNA expression was >7-fold higher in SCC than in normal cells (Fig. 6A and B). Moreover, all the SCC cell lines featured high levels (>10-fold with respect to HNEK) of standard and v6-containing CD44 species (Fig. 6C and D). All the cell lines featured abundant cell surface CD44v6 expression by flow cytometry (Fig. 6E; data not shown).

Fig. 6.

Osteopontin and CD44 up-regulation in cultured SCC cells. A, osteopontin protein levels were evaluated by immunoblot in the indicated cell lines. Anti-α-tubulin was used for normalization. B, semiquantitative RT-PCR (25 cycles) was used to detect osteopontin mRNA levels in the indicated cell lines. β-Actin mRNA detection was used for normalization. Band intensity was calculated by phosphorimaging. C, CD44 protein levels were determined by immunoblot. D, semiquantitative RT-PCR (25 cycles) was done (see Fig. 5 legend) to detect CD44 mRNA levels in the indicated cell lines. E, flow cytometric analysis of cell surface expression of CD44v6 in the indicated cell lines. Black histogram, negative control antibody.

Fig. 6.

Osteopontin and CD44 up-regulation in cultured SCC cells. A, osteopontin protein levels were evaluated by immunoblot in the indicated cell lines. Anti-α-tubulin was used for normalization. B, semiquantitative RT-PCR (25 cycles) was used to detect osteopontin mRNA levels in the indicated cell lines. β-Actin mRNA detection was used for normalization. Band intensity was calculated by phosphorimaging. C, CD44 protein levels were determined by immunoblot. D, semiquantitative RT-PCR (25 cycles) was done (see Fig. 5 legend) to detect CD44 mRNA levels in the indicated cell lines. E, flow cytometric analysis of cell surface expression of CD44v6 in the indicated cell lines. Black histogram, negative control antibody.

Close modal

Osteopontin activates intracellular signaling, growth, and invasiveness of squamous cell carcinoma cells. Various SCC cell lines, washed in serum-free medium, were stimulated with exogenous recombinant osteopontin and harvested at different time points. Protein lysates were probed with phosphorylated MEK and phosphorylated ERK (ERK1/2) antibodies. MEK and ERK were readily activated in osteopontin-stimulated cells, peaking at 5 to 15 minutes (Fig. 7A), but not in normal HNEK cells (data not shown). We then examined the ability of SCC cells to synthesize DNA in basal conditions and in the presence of exogenous recombinant osteopontin. Osteopontin stimulated DNA synthesis in SCC cells, washed in serum-free medium, but not in normal HNEK cells (P = 0.01, two tailed Student's t test). Thus, we asked whether osteopontin expression was required for the growth of SCC cells. BrdUrd incorporation was obtained in triplicate after BHY and CAL27 cell transfection with osteopontin or scrambled siRNA. The transient silencing of osteopontin (Fig. 7C, inset) significantly inhibited the growth of BHY cells, whereas the negative control siRNA had virtually no effect (Fig. 7C; data not shown). To determine whether the CD44 receptor mediated these events, we treated BHY, after washing in serum-free medium, with exogenous osteopontin after CD44 blockade with specific antibodies or chemical ERK blockade by the U0126 MEK inhibitor. Activation of MEK and ERK was virtually abrogated by pretreatment with U0126 (Fig. 7A). Stimulation of cell proliferation by osteopontin was obstructed by both anti-CD44 and U0126 (Fig. 7C). We next examined the ability of SCC cells to invade Matrigel in basal conditions and in the presence of exogenous osteopontin. Treatment with osteopontin induced a strong migratory response of tumor cells but not of normal HNEK cells (P = 0.04, χ2 test; Fig. 7D). Treatment with CD44-blocking antibodies or U0126 sharply inhibited these effects (P = 0.03, two tailed Student's t test; Fig. 7E).

Fig. 7.

Osteopontin-mediated signaling, growth, and Matrigel invasion in SCC cells. A, total cell lysates were prepared at various time points after stimulation of SCC cells, washed in serum-free medium, with recombinant osteopontin (100 ng/mL). Immunoblots were probed with the indicated phosphospecific antibodies. Anti-ERK was used for normalization. B, after starvation, the indicated cell lines were treated (48 hours) or not with exogenous recombinant osteopontin (100 ng/mL). Cells were exposed to BrdUrd for 1 hour, and cells were fixed and processed for immunofluorescence. Average results of three independent experiments in which at least 400 cells were counted; bars, 95% CI. C, BrdUrd incorporation was evaluated in BHY cells, washed in serum-free medium, in response to osteopontin (100 ng/mL) with and without U0126 (10 μmol/L) or KM81 CD44-blocking mAb (10 μg/mL). NT, not treated cells. Moreover, osteopontin knockdown was obtained by transient transfection with by siRNA (inset). Mock-transfected cells and cells transfected with scrambled siRNA served as a control. BrdUrd was evaluated in transfected cells. All experiments were carried out in triplicate. Bars, 95% CI. D, Matrigel invasion of the various SCC cells in response to normal culture medium or exogenous recombinant osteopontin. The percentage of migrating cells was quantified with an ELISA reader. Top, average results of three independent experiments; bars, 95% CI. Bottom, representative micrographs. E, cells were preincubated with U0126 (10 μmol/L) or CD44-blocking antibody (10 μg/mL) and Matrigel invasion was analyzed as described in (D).

Fig. 7.

Osteopontin-mediated signaling, growth, and Matrigel invasion in SCC cells. A, total cell lysates were prepared at various time points after stimulation of SCC cells, washed in serum-free medium, with recombinant osteopontin (100 ng/mL). Immunoblots were probed with the indicated phosphospecific antibodies. Anti-ERK was used for normalization. B, after starvation, the indicated cell lines were treated (48 hours) or not with exogenous recombinant osteopontin (100 ng/mL). Cells were exposed to BrdUrd for 1 hour, and cells were fixed and processed for immunofluorescence. Average results of three independent experiments in which at least 400 cells were counted; bars, 95% CI. C, BrdUrd incorporation was evaluated in BHY cells, washed in serum-free medium, in response to osteopontin (100 ng/mL) with and without U0126 (10 μmol/L) or KM81 CD44-blocking mAb (10 μg/mL). NT, not treated cells. Moreover, osteopontin knockdown was obtained by transient transfection with by siRNA (inset). Mock-transfected cells and cells transfected with scrambled siRNA served as a control. BrdUrd was evaluated in transfected cells. All experiments were carried out in triplicate. Bars, 95% CI. D, Matrigel invasion of the various SCC cells in response to normal culture medium or exogenous recombinant osteopontin. The percentage of migrating cells was quantified with an ELISA reader. Top, average results of three independent experiments; bars, 95% CI. Bottom, representative micrographs. E, cells were preincubated with U0126 (10 μmol/L) or CD44-blocking antibody (10 μg/mL) and Matrigel invasion was analyzed as described in (D).

Close modal

Here, we show that osteopontin expression was closely correlated with advanced stage, high grade, metastatic disease, and poor survival of LSCC. This is in accordance with our observation that osteopontin affected the signaling and the mitogenic and motile phenotypes of carcinoma cells. Thus, although larger, prospective studies are needed to elucidate the relevance of osteopontin status versus other prognostic factors, our data suggest that osteopontin expression could be exploited as a predictor of outcome in LSCC patients. Interestingly, osteopontin plasma levels have been associated with treatment outcome and survival of HNSCC patients (30).

After its identification as a protein secreted by neoplastic cells (31), osteopontin has been detected in several human tumor types (i.e., gliomas and lung, prostate, gastric, esophageal, and ovarian carcinomas; refs. 16, 17, 19, 20). Osteopontin is also a major determinant of breast (21) and liver (22) cancer metastatization. Numerous growth factors (32), the RAS (33) and SRC (34) oncogenes, and the tumor suppressor p53 (35) regulate the expression of osteopontin. Many of these oncogenic proteins are involved in the pathogenesis of HNSCC (1) and thus might be responsible for osteopontin deregulation in such a tumor type. Osteopontin is able to engage several receptors, including integrins and CD44 variants, and thus may stimulate diverse signaling pathways and influence cellular events that, in turn, favor tumorigenesis and metastasis (16, 17). In particular, osteopontin binds CD44 proteins that contain v6-encoded sequences (14, 15), and osteopontin-CD44v6 binding has been implicated in carcinogenesis (1618). In agreement with a report of CD44 (the standard CD44 variant in that case) expression in HNSCC (36), we show that osteopontin up-regulation is paralleled by intense expression of CD44 (in particular, CD44v6) in LSCC tissue samples as well as in a panel of SCC cell lines. Thus, CD44v6 is a candidate receptor for osteopontin in LSCC cells. Accordingly, CD44 blockade obstructed osteopontin-mediated cellular effects. CD44 activates a wealth of signaling proteins, among which ERK (37), RAC (38), and RHO (39), as well as secretion of cytokines (40), angiogenic factors, and metalloproteinases (41). This could explain the effects exerted by the osteopontin-CD44v6 axis. On the other hand, osteopontin induces CD44v6 overexpression (42). Of note, our experiments show that osteopontin-CD44v6 binding mediates the effects occurring in SCC cells, but they do not exclude that interactions between osteopontin and integrins (1618) and between CD44, hyaluronan (1618), and other membrane receptors, such as members of the MET and ERBB family (4345), are involved in the osteopontin-CD44 axis as well.

In conclusion, the results of this study suggest that patients affected by LSCC that express high levels of osteopontin protein may be more prone to a poor outcome than patients with low osteopontin-expressing LSCC. Thus, therapies targeted at the molecular mechanism (e.g., ligands that antagonize the interaction of osteopontin with CD44 or antibodies directed against osteopontin and CD44) may prove useful in LSCC patients.

Grant support: Associazione Italiana per la Ricerca sul Cancro, Progetto Strategico Oncologia of the Consiglio Nazionale delle Ricerche/Ministero dell'Istruzione, dell'Università e della Ricerca, Italian Ministero della Salute, Centro Regionale di Competenza Genomic for Applied Research, and Naples Oncogenomic Center (M. Santoro) and BioGeM scarl fellowships (MDC and VG).

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.

Note: D. Testa and S. Staibano contributed equally to the work.

We thank L. Vitiello and L. Racioppi for fluorescence-activated cell sorting analysis and Jean Gilder for text editing.

1
Mao L, Hong WK, Papadimitrakopoulou VA. Focus on head and neck cancer.
Cancer Cell
2004
;
5
:
311
–6.
2
Licitra L, Bernier J, Grandi C, et al. Cancer of the larynx.
Crit Rev Oncol Hematol
2003
;
l47
:
65
–80.
3
Vokes EE, Weichselbaum RR, Lippman SM, Hong WK. Head and neck cancer.
N Engl J Med
1993
;
328
:
184
–94.
4
Hoffman HT, Karnell LH, Funk GF, Robinson RA, Menck HR. The National Cancer Data Base report on cancer of the head and neck.
Arch Otolaryngol Head Neck Surg
1998
;
124
:
951
–62.
5
Tabor MP, Brakenhoff RH, Ruijter-Schippers HJ, et al. Multiple head and neck tumours frequently originate from a single preneoplastic lesion.
Am J Pathol
2002
;
161
:
1051
–60.
6
Shin DM, Lee JS, Lippman SM, et al. p53 expressions: predicting recurrence and second primary tumors in head and neck squamous cell carcinoma.
J Natl Cancer Inst
1996
;
88
:
519
–29.
7
Bellacosa A, Almadori G, Cavallo S, et al. Cyclin D1 gene amplification in human laryngeal squamous cell carcinomas: prognostic significance and clinical implications.
Clin Cancer Res
1996
;
2
:
175
–80.
8
Pignataro L, Pruneri G, Carboni N, et al. Clinical relevance of cyclin D1 protein overexpression in laryngeal squamous cell carcinoma.
J Clin Oncol
1998
;
9
:
3069
–77.
9
Rubin Grandis J, Melhem MF, Gooding WE, et al. Levels of TGF-α and EGFR protein in head and neck squamous cell carcinoma and patient survival.
J Natl Cancer Inst
1998
;
90
:
824
–32.
10
Smith BD, Smith GL, Carter D, Sasaki CT, Haffty BG. Prognostic significance of vascular endothelial growth factor protein levels in oral and oropharyngeal squamous cell carcinoma.
J Clin Oncol
2000
;
18
:
2046
–52.
11
Pruneri G, Pignataro L, Carboni N, et al. Clinical relevance of expression of the CIP/KIP cell-cycle inhibitors p21 and p27 in laryngeal cancer.
J Clin Oncol
1999
;
17
:
3150
–9.
12
Ashkar S, Weber GF, Panoutsakopoulou V, et al. Eta-1 (osteopontin): an early component of type-1 (cell-mediated) immunity.
Science
2000
;
287
:
860
–4.
13
Chabas D, Baranzini SE, Mitchell D, et al. The influence of the proinflammatory cytokine, osteopontin on autoimmune demyelinating disease.
Science
2001
;
294
:
1731
–5.
14
Weber GF, Ashkar S, Glimcher MJ, Cantor H. Receptor-ligand interaction between CD44 and osteopontin (Eta-1).
Science
1996
;
271
:
509
–12.
15
Katagiri YU, Sleeman J, Fujii H, et al. CD44 variants but not CD44s cooperate with β1-containing integrins to permit cells to bind to osteopontin independently of arginine-glycine-aspartic acid, thereby stimulating cell motility and chemotaxis.
Cancer Res
1999
;
59
:
219
–26.
16
Rittling SR, Chambers AF. Role of osteopontin in tumour progression.
Br J Cancer
2004
;
90
:
1877
–81.
17
Weber GF. The metastasis gene osteopontin: a candidate target for cancer therapy.
Biochim Biophys Acta
2001
;
1552
:
61
–85.
18
Ponta H, Sherman L, Herrlich PA. CD44: from adhesion molecules to signalling regulators.
Nat Rev Mol Cell Biol
2003
;
4
:
33
–45.
19
Agrawal D, Chen T, Irby R, et al. Osteopontin identified as lead marker of colon cancer progression, using pooled sample expression profiling.
J Natl Cancer Inst
2002
;
94
:
513
–21.
20
Schorge JO, Drake RD, Lee H, et al. Osteopontin as an adjunct to CA125 in detecting recurrent ovarian cancer.
Clin Cancer Res
2004
;
10
:
3474
–8.
21
Kang Y, Siegel PM, Shu W, et al. A multigenic program mediating breast cancer metastasis to bone.
Cancer Cell
2003
;
3
:
537
–49.
22
Ye QH, Qin LX, Forgues M, et al. Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning.
Nat Med
2003
;
9
:
416
–23.
23
Hermanek P, Sobin LH. Larynx. In: Hermanek P, Sobin LH, editors. TNM classification of malignant tumors. Berlin (Germany): Springer-Verlag; 1987. p. 25–8.
24
Rosai J, Carcangiu ML, DeLellis RA. Atlas of tumor pathology—tumors of the larynx, 3rd series. Washington: Armed Force Institute of Pathology; 1992.
25
Kawamata H, Nakashiro K, Uchida D, Harada K, Yoshida H, Sato M. Possible contribution of active MMP2 to lymph-node metastasis and secreted cathepsin L to bone invasion of newly established human oral-squamous-cancer cell lines.
Int J Cancer
1997
;
70
:
120
–7.
26
Gioanni J, Fischel JL, Lambert JC, et al. Two new human tumor cell lines derived from squamous cell carcinomas of the tongue: establishment, characterization and response to cytotoxic treatment.
Eur J Cancer Clin Oncol
1988
;
24
:
1445
–55.
27
Moore AE, Sabachewsky L, Toolan HW. Culture characteristics of four permanent lines of human cancer cells.
Cancer Res
1955
;
15
:
598
–602.
28
Eagle H. Propagation in a fluid medium of a human epidermoid carcinoma, strain KB.
Proc Soc Exp Biol Med
1955
;
89
:
362
–4.
29
Weiss JM, Renkl AC, Maier CS, et al. Osteopontin is involved in the initiation of cutaneous contact hypersensitivity by inducing Langerhans and dendritic cell migration to lymph nodes.
J Exp Med
2001
;
194
:
1219
–29.
30
Le QT, Sutphin PD, Raychaudhuri S, et al. Identification of osteopontin as a prognostic marker for head and neck squamous cell carcinomas.
Clin Cancer Res
2003
;
9
:
31
–2.
31
Senger DR, Wirth DF, Hytnes RO. Transformed mammalian cells secrete specific proteins and phosphoproteins.
Cell
1979
;
16
:
885
–93.
32
Medico E, Gentile A, Lo Celso C, et al. Osteopontin is an autocrine mediator of hepatocyte growth factor-induced invasive growth.
Cancer Res
2001
;
61
:
5861
–8.
33
Wu Y, Denhardt DT, Rittling SR. Osteopontin is required for full expression of the transformed phenotype by the ras oncogene.
Br J Cancer
2000
;
83
:
156
–63.
34
Chackalaparampil I, Peri A, Nemir M, et al. Cells in vivo and in vitro from osteopetrotic mice homozygous for c-src disruption show suppression of synthesis of osteopontin, a multifunctional extracellular matrix protein.
Oncogene
1996
;
12
:
1457
–67.
35
Morimoto I, Sasaki Y, Ishida S, Imai K, Tokino T. Identification of the osteopontin gene as a direct target of TP53.
Genes Chromosomes Cancer
2002
;
3
:
270
–8.
36
Liu M, Lawson G, Delos M, et al. Prognostic value of cell proliferation markers, tumour suppressor proteins and cell adhesion molecules in primary squamous cell carcinoma of the larynx and hypopharynx.
Eur Arch Otorhinolaryngol
2003
;
260
:
28
–34.
37
Bourguignon LY, Gilad E, Rothman K, Peyollier K. Hyaluronan-CD44 interaction with IQGAP1 promotes Cdc42 and ERK signaling leading to actin binding, Elk-1/estrogen receptor transcriptional activation and ovarian cancer progression.
J Biol Chem
2005
;
280
:
11961
–72.
38
Teramoto H, Castellone MD, Malek RL, et al. Autocrine activation of an osteopontin-CD44-Rac pathway enhances invasion and transformation by H-RasV12.
Oncogene
2005
;
24
:
489
–501.
39
Bourguignon LY, Singleton PA, Zhu H, Diedrich F. Hyaluronan-mediated CD44 interaction with RhoGEF and Rho kinase promotes Grb2-associated binder-1 phosphorylation and phosphatidylinositol 3-kinase signaling leading to cytokine (macrophage-colony stimulating factor) production and breast tumor progression.
J Biol Chem
2003
;
278
:
29420
–34.
40
Murphy JF, Lennon F, Steele C, Kelleher D, Fitzgerald D, Long A. Engagement of CD44 modulates cyclooxygenase induction, VEGF generation, and cell proliferation in human vascular endothelial cells.
FASEB J
2005
;
19
:
446
–8.
41
Zhang Y, Thant AA, Machida K, et al. Hyaluronan-CD44s signaling regulates matrix metalloproteinase-2 secretion in a human lung carcinoma cell line QG90.
Cancer Res
2002
;
62
:
3962
–5.
42
Gao C, Guo H, Downey L, Marroquin C, Wei J, Kuo PC. Osteopontin-dependent CD44v6 expression and cell adhesion in HepG2 cells.
Carcinogenesis
2003
;
24
:
1871
–8.
43
Orian-Rousseau V, Chen L, Sleeman JP, Herrlich P, Ponta H. CD44 is required for two consecutive steps in HGF/c-Met signaling.
Genes Dev
2002
;
16
:
3074
–86.
44
Ghatak S, Misra S, Toole BP. Hyaluronan constitutively regulates ErbB2 phosphorylation and signaling complex formation in carcinoma cells.
J Biol Chem
2005
;
280
:
8875
–83.
45
Tuck AB, Hota C, Wilson SM, Chambers AF. Osteopontin-induced migration of human mammary epithelial cells involves activation of EGF receptor and multiple signal transduction pathways.
Oncogene
2003
;
22
:
1198
–205.