Purpose: Cluster-of-differentiation antigen 9 (CD9) protein, a member of the tetraspanin family, has been implicated in carcinogenesis of various human tumors. Although decreased expression of the CD82 tetraspanin protein, a close CD9 relative, is associated with prostate cancer progression, CD9 expression has not been analyzed in this malignancy.

Experimental Design: CD9 expression in human prostatic adenocarcinoma was analyzed by immunohistochemistry on 167 primary tumors and 88 lymph node or bone metastases. CD9 cDNA was sequenced from two human prostate cancer cell lines, prostatic adenocarcinoma, high-grade prostatic intraepithelial neoplasia (PIN), and normal prostatic tissues.

Results: Although CD9 was detected in the epithelium of normal prostatic tissues, reduced or loss of CD9 expression within neoplastic cells was observed in 24% of 107 clinically localized primary adenocarcinomas, 85% of 60 clinically advanced primary adenocarcinomas, 85% of 65 lymph node metastases, and 65% of 23 bone metastases. Difference in CD9 expression between clinically localized and advanced diseases was highly significant (P < 1 × 10−7). Whereas there was no alteration of CD9 cDNA in normal tissues, all PC-3–derived cell lines, one PIN, and four prostatic adenocarcinomas harbored deletions in their CD9 cDNAs. Recurring CD9 point mutations were also found in PC-3M-LN4 cells, one PIN, and seven prostatic adenocarcinomas.

Conclusions: CD9 expression is significantly reduced and even lost during prostate cancer progression. Moreover, deletions and mutations of the CD9 mRNA may be associated with loss of protein expression observed in tumor cells. Our data suggest that CD9 inactivation may play an important role in prostate cancer progression.

In North America, adenocarcinoma of prostate is the most frequently diagnosed cancer and the second leading cause of cancer death in men (1). Contrasting with these alarming figures, the mechanisms implicated in the progression of prostatic adenocarcinoma are not completely understood. In addition, because the natural history of prostatic adenocarcinoma is highly variable and unpredictable, more accurate markers to predict tumor progression are urgently needed for optimal therapy.

Using microcell-mediated chromosome transfer, we reported previously that the transfer of a fragment of a normal chromosome 12 (from human foreskin fibroblasts) was sufficient to suppress tumorigenicity of prostate carcinoma DU145 cell–derived hybrids (2). The fact that this suppression was lost on subsequent removal of the introduced human chromosome 12 fragment from these hybrids (2) argues strongly for the presence of one or more tumor suppressor gene(s) and/or other genes whose inactivation plays a role in prostate tumorigenesis. Human chromosome 12 had already been implicated in prostate cancer progression. Indeed, a small region of the long arm of this chromosome was sufficient to inhibit prostate cancer metastatic colonization of the Dunning AT6.1 rat prostate cancer cell line (3). Moreover, loss of heterozygosity (4) as well as reduced expression of specific genes present in a 2-Mb region of chromosome 12p12-13 has been associated with advanced prostate adenocarcinoma (5). Using irradiation, we further fragmented chromosome 12, and radiation hybrids exhibiting either tumorigenic or nontumorigenic phenotypes, depending on the portion of chromosome 12 retained, were generated. Using differential display reverse transcription-PCR, cDNA fragments whose expression varied between tumorigenic and nontumorigenic hybrids were identified. One of these cDNA fragments was derived from the cluster-of-differentiation antigen 9 (CD9) gene, located on human chromosome 12p13.3 (6, 7).

CD9, also referred to as MRP-1 (for motility related protein 1, not to be confused with the multidrug resistance protein 1), is a 25- to 27-kDa cell membrane glycoprotein (810) and a member of tetraspanins (11). Tetraspanins have been identified as a superfamily of transmembrane proteins containing over 32 members (12) expressed in many cell types. Members of this protein family associate with each other, with adhesion molecules, and other membrane proteins to form multimolecular complexes (11). It is believed that through these associations, tetraspanins can form a “tetraspanin web” (13) and thus serve as “molecular facilitators” or “organizers” of multimolecular complexes on the cell surface (11, 14, 15). The tetraspanin web, as the name suggests, provides a dynamic network for molecular interactions. Accordingly, tetraspanins are implicated in a variety of normal and pathologic processes, such as tissue differentiation (16), egg-sperm fusion (17, 18), virus-induced syncytium formation (19), and tumor-cell metastasis (11). Tetraspanins have also been involved in cell adhesion and motility (20). Interestingly, transfection of CD9 reduces metastasis in vivo and this effect has been related to the suppression of tumor cell growth and motility (21).

In cancer, decreased expression of CD9 protein has been implicated in progression of breast (22), non–small cell lung (23), and colon cancers (24). Moreover, decreased CD9 expression correlated with poor prognosis in several human malignancies, such as colon (25), lung (26), breast (22, 27), and ovarian carcinomas (28).

Based on our findings in tumorigenic and nontumorigenic human prostate cancer cells, our objective was to determine the role played by the CD9 protein in prostate cancer progression. Using immunohistochemistry, CD9 expression was characterized in 167 primary tumors (107 localized and 60 advanced), 65 lymph node metastases, and 23 bone metastases. Our results show that CD9 expression is greatly reduced and even lost during prostate cancer progression. To understand the mechanisms responsible for the loss of CD9 protein expression, CD9 cDNA was amplified, cloned, and sequenced from prostate cancer cell line DU145-N19 and its derived nontumorigenic hybrid DBM9-7 and from PC-3 and its derivative cell lines. CD9 cDNA was also cloned from prostatic adenocarcinoma, prostatic intraepithelial neoplasia (PIN), and normal prostate tissues obtained from 11 patients. Our results indicated that cDNAs derived from four prostatic adenocarcinoma cell lines, one PIN, and four prostatic adenocarcinoma specimens harbored one of three repeatedly occurring deletions affecting the CD9 coding region. Moreover, one prostatic adenocarcinoma cell line, one PIN, and seven prostatic adenocarcinoma specimens contained at least one and up to three of four point mutations, including a stop codon.

Cell lines and culture conditions. DU145-N19 is a tumorigenic, G418-resistant derivative of DU145. DBM9-7 is a nontumorigenic DU145-N19–derived microcell hybrid containing an extra fragment [del(12)(q13)] of a normal human chromosome 12 (2). DU145-N19 was grown in MEM supplemented with 10% fetal bovine serum (Canadian Life Technologies, Burlington, Ontario, Canada) and 800 μg/mL G418 (Roche Diagnostics, Laval, Quebec, Canada). DBM9-7 was grown in the same medium supplemented with 400 μg/mL hygromycin B (Roche Diagnostics) to ensure the retention of the tagged chromosome 12 (2). Both cell lines were grown in the absence of other antibiotics and were free of Mycoplasma as determined by Hoechst 33342 staining.

PC-3 was obtained from the American Type Culture Collection (Manassas, VA), whereas its derivatives, PC-3M, PC-3M-Pro4, and PC-3M-LN4 (29), were kindly provided by Dr. I.J. Fidler (University of Texas M. D. Anderson Cancer Center, Houston, TX); they were cultured in MEM supplemented with 10% fetal bovine serum.

Cloning and analysis of candidate cDNAs. We have used differential display (differential display reverse transcription-PCR; ref. 30) to isolate cDNA fragments expressed at higher level in DBM9-7 (the nontumorigenic hybrid) compared with DU145-N19 (the tumorigenic parental cell line). The expression of candidate fragments was assessed by Northern blot hybridization on membranes containing DU145-N19 and DBM9-7 polyA+ RNA. Interesting candidates were cloned and sequenced.

Western blotting. Protein samples (30 μg) extracted from two benign prostatic hyperplasia specimens, two localized, and three advanced human prostatic adenocarcinomas were separated on 13% SDS-PAGE and transferred to nitrocellulose membrane (0.45 μm). CD9 protein expression was detected on a 2-h incubation with an antihuman CD9 monoclonal mouse antibody (NCL-CD9; 1:500; Novocastra Laboratories/Vector Laboratories, Burlington, Ontario, Canada); the signal was amplified with a 1:2,000 dilution of a horseradish peroxidase–coupled goat anti-mouse antibody and revealed using enhanced chemiluminescence Western blotting protocol (GE Healthcare, Baie d'Urfe, Quebec, Canada) as described previously (31).

Patients and tissue materials. Patients provided inform consent and were investigated and treated at the McGill University Health Center. One hundred and seven patients had clinically localized disease and were treated with radical prostatectomy. Of these, 42 patients had received 3 months of preoperative combined androgen blockade hormone therapy. Formaldehyde-fixed and paraffin-embedded whole-mount sections of prostatectomy specimens were used for immunohistochemical analysis. Twenty-seven patients were at pathologic stage T1N0M0, 32 at stage T2N0M0, 47 at stage T3N0M0, and 1 at stage T2N1M0. The average age was 62 years (range, 44-76) at diagnosis, and the average follow-up interval was 62 months (range, 1.6-102).

Sixty patients had clinically advanced disease; in these cases, needle biopsy or transurethral resection of prostate material was used for immunohistochemical study. These patients were palliatively treated with bilateral orchiectomy or medical castration. Eleven patients were at stage T3N0M0, 6 were at stage T2-3N1-3M0, and 43 were at stage T2-3N1-3M1. The average age was 71 years (range, 47-87) at diagnosis and the average follow-up interval was 39 months (range, 1-112). Furthermore, 23 bone and 65 lymph node specimens with metastatic involvement were available for immunohistochemical analysis; however, no clinical data were available.

Immunohistochemical technique. All prostatic tissues were fixed in 10% buffered formaldehyde and paraffin embedded. Four-micrometer-thick sections were obtained and deparaffinated in xylene. Tissues were rehydrated in decreasing graded ethanol and TBS. CD9 antigen was retrieved by heating at high pressure in a pressure cooker with 1 mmol/L EDTA (pH 8.0) for 10 to 15 min. Endogenous peroxidase was blocked by 1.5% of hydrogen peroxide in 50% methanol. An immunohistochemistry kit, HistostainTM-SP kit (Zymed, South San Francisco, CA), was used to detect CD9 signals (32). Tissues were first incubated with nonimmune goat serum for 25 min to minimize nonspecific binding and then incubated with monoclonal antihuman CD9 mouse antibody (NCL-CD9; dilution of 1:500) for 1 h. The CD9 antibody recognizes an epitope located in the extracellular region of the protein. The signal was detected with a goat anti-mouse IgG for 30 min and streptavidin-peroxidase conjugate for 15 min. The staining was revealed with the 3,3′-diaminobenzidine substrate (Sigma-Aldrich Canada Ltd., Oakville, Ontario, Canada). Tissues were counterstained with hematoxylin for 20 s and dehydrated in increasing graded ethanol and xylene. Staining intensity for CD9 expression was assessed under standard microscope.

Scoring of immunohistochemical staining. Two independent reviewers (J-C.W. and L.R.B.) analyzed all tissue sections, without knowledge of patient's clinical data, and a consensus was achieved about the final score. The effectiveness and specificity of the antibody (NCL-CD9) for immunohistochemical analysis was verified on a mammary fibroadenoma. As shown previously (22), cytoplasmic expression of CD9 protein was observed on epithelial cells of breast tissue. Ten whole-mount sections from 10 cases of prostatic adenocarcinoma were randomly selected and used to standardize CD9 scoring. The intensity of immunoreactivity (cytoplasmic staining often with apical and/or membranous enhancement) was defined as negative (score 0) versus positive (on a scale of 3; score 1 for weak staining, score 2 for moderate staining, and score 3 for strong staining). The percentage of tumor cells with immunoreactivity for CD9 was defined as ±5% approximation (e.g., 25-30%). The range of intensity within positive tumor cells was also marked. For instance, if one third of cells were stained weakly, whereas two thirds of cells were stained moderately, the range would be 1 to 2; therefore, a predominant intensity staining would be 2. Gleason pattern(s) present on the representative slide was also analyzed at the same time. The cutoff standard for CD9 immunohistochemical staining was adapted from previous analysis (27). In brief, when the total number of tumor cells with immunoreactivity (scores 1-3) for CD9 protein is >50% in a given specimen, the sample was classified as CD9 positive. When there were <50% of tumor cells with immunoreactivity, the sample was classified as reduced.

Statistical analysis. The χ2 test and the Student's t test were done using Excel software (Microsoft Corp., Redmond, WA). Statistical significance was defined as P value <0.01.

Analysis of CD9 cDNA.CD9 cDNA was synthesized using Expand Reverse transcriptase according to the manufacturer's instructions (Roche Diagnostics) from total RNA isolated from 5 prostatic carcinoma cell lines, 10 prostatic carcinoma tissue specimens, 2 PIN tissue specimens, and 6 normal prostate tissue specimens (grossly dissected from frozen slides). CD9 cDNA was PCR amplified using high-fidelity Pfu DNA polymerase (Stratagene, La Jolla, CA). The primer sequences were derived from CD9 cDNA sequence, where position 1 was assigned to the first nucleotide of the coding sequence (33). Forward CD9 primer (5′-ATGCCGGTCAAAGGAGGCAC-3′) starts at nucleotide 1 and reverse CD9 primer (5′-GACCATCTCGCGGTTCCTGC-3′) starts at nucleotide 684. PCR products were purified and cloned using PCR-Script Amp cloning kit (Stratagene).

We also directly sequenced some PCR products (without cloning) using a similar set of primers (forward primer at position 3-22 and reverse primer at position 658-677). For the cloned CD9 cDNAs, sequencing was done using ABI Prism DNA analyzer 3700 (Applied Biosystems, Foster City, CA) with T7 and T3 primers. CD9 cDNA sequence analysis was done using Blast 2 nucleotide alignment.

CD9 mRNA is overexpressed in nontumorigenic hybrids. Differential display reverse transcription-PCR analysis identified 21 cDNA fragments, for which expression levels were higher in the nontumorigenic DBM9-7 cell line when compared with the parental tumorigenic DU145-N19 cell line (data not shown). Northern blot analysis confirmed that one of these candidates hybridized to a 1.4-kb mRNA, which was expressed in DBM9-7 at 3-fold higher levels compared with DU145-N19. Sequencing of this fragment revealed that it was derived from exon 8 of the CD9 gene. Because loss or decreased expression of the CD9 protein had already been associated with poor prognosis in breast and lung cancers (23, 26, 27), we pursued the characterization of CD9 protein expression and the analysis of its transcript in human prostate cancer.

Expression of CD9 in prostatic adenocarcinoma. Using Western immunoblotting, the 27-kDa CD9 protein was detected in two benign prostatic hyperplasia and two localized prostatic adenocarcinoma specimens. However, the CD9 protein was absent in two of three advanced prostatic adenocarcinoma specimens (Fig. 1). Using immunohistochemistry, CD9 expression was analyzed in 255 specimens (167 prostate specimens with primary adenocarcinoma, 65 lymph node specimens with metastasis, and 23 bone specimens with metastasis). Most sections from the 167 prostate specimens contained normal and/or hyperplastic epithelium in the background, thus providing an internal control for CD9 detection.

Fig. 1.

Analysis of CD9 protein expression in prostate diseases. Protein extracts from different tissue samples were analyzed by Western blotting. Arrow, the presence of the 27-kDa CD9 protein. BPH, benign prostatic hyperplasia; PCa, prostate cancer.

Fig. 1.

Analysis of CD9 protein expression in prostate diseases. Protein extracts from different tissue samples were analyzed by Western blotting. Arrow, the presence of the 27-kDa CD9 protein. BPH, benign prostatic hyperplasia; PCa, prostate cancer.

Close modal

In nonneoplastic glands of all prostate specimens, CD9 was predominately expressed as diffuse cytoplasmic staining within secretory (luminal) cells, including focal apical enhancement (Fig. 2A). CD9 immunoreactivity was also observed in a few scattered basal cells (Fig. 2A). In specimens of patients who had preoperative combined androgen blockade, CD9 was predominantly expressed as diffuse cytoplasmic staining within the hyperplastic basal cell constituent of nonneoplastic glands. Intraluminal prostatic secretions had a strong and uniform immunoreactivity for CD9 (Fig. 2A). In agreement with the CD9 distribution in myelin of the central and peripheral nervous systems (34), prostatic peripheral nerves were strongly stained and served as an additional internal positive control for whole-mount sections. CD9 staining was not seen in the stroma.

Fig. 2.

Expression of CD9 protein in normal prostate and prostatic adenocarcinoma. A, normal prostatic glands show varying degrees of cytoplasmic staining for CD9, including a strongly immunoreactive gland in the center. Strong immunostaining is also observed within luminal secretions. Minute adenocarcinomatous components (Gleason pattern 3) are present in the upper and lower right corners. B, adenocarcinoma, Gleason score 8/10 (pattern 4 + 4), showing varied cytoplasmic staining for CD9 (intensity range, 1-3). C, adenocarcinoma, Gleason score 6/10 (pattern 3 + 3), negative (score 0) for CD9 staining. Note the presence of a nerve bundle positive for CD9 staining in the right upper corner. D, altered (i.e., with treatment-induced cytoarchitectural changes) adenocarcinoma following total androgen blockade, negative (score 0) for CD9 staining. E, lymph node metastasis (arrow), negative (score 0) for CD9 staining. F, bone metastasis, negative (score 0) for CD9 staining. Original magnifications, ×100 (A, B, E, and F) and ×400 (C and D).

Fig. 2.

Expression of CD9 protein in normal prostate and prostatic adenocarcinoma. A, normal prostatic glands show varying degrees of cytoplasmic staining for CD9, including a strongly immunoreactive gland in the center. Strong immunostaining is also observed within luminal secretions. Minute adenocarcinomatous components (Gleason pattern 3) are present in the upper and lower right corners. B, adenocarcinoma, Gleason score 8/10 (pattern 4 + 4), showing varied cytoplasmic staining for CD9 (intensity range, 1-3). C, adenocarcinoma, Gleason score 6/10 (pattern 3 + 3), negative (score 0) for CD9 staining. Note the presence of a nerve bundle positive for CD9 staining in the right upper corner. D, altered (i.e., with treatment-induced cytoarchitectural changes) adenocarcinoma following total androgen blockade, negative (score 0) for CD9 staining. E, lymph node metastasis (arrow), negative (score 0) for CD9 staining. F, bone metastasis, negative (score 0) for CD9 staining. Original magnifications, ×100 (A, B, E, and F) and ×400 (C and D).

Close modal

In the adenocarcinomatous component, CD9 expression was characterized by diffuse cytoplasmic staining that was homogeneous (within one scale, 0-3) in the majority (88%) of specimen. In localized prostate cancers, 29% of the cases revealed uniform intensity without any changes, whereas differences in staining intensity (such as scales 1-2 or 2-3) occurred in 51% of the cases. The remaining 20% of the cases harbored more staining variation, ranging from scale 1 to 3 (Fig. 2B). There was no sample that crossed the staining cutoff (scales 0-1); thus, there was no discrepancy for the classification of CD9 status for any given specimen. A very similar staining spectrum was observed for bone specimen with metastases, in which 83% of the cases showed homogeneous staining. In these specimens, 43% had completely lost CD9 staining (scale 0), whereas an additional 22% had a weak staining (scale 1). Similarly, a fairly uniform staining was seen in 98% of advanced prostate cancers, of which 57% had no CD9 staining (scale 0), whereas 27% had weak staining (scale 1). Uniform staining was also detected in 95% of lymph node metastases, with an impressive 64% harboring no CD9 staining (scale 0) and 21% having weak CD9 staining (scale 1).

Eighty-two percent of the localized prostate cancers without hormone pretreatment (Table 1) still preserved CD9 expression with >50% of cells having positive immunoreactivity (Fig. 2B). Thus, only 18% of cases had reduced or negative CD9 expression (Fig. 2C). This percentage was not significantly different (P = 0.08) from the one (33%) obtained in cases with hormone pretreatment (Fig. 2D; Table 1). However, compared with localized prostate cancers, advanced prostate cancers had very significant differences in CD9 expression (85% with reduced or lost CD9 expression), independent of hormone pretreatment or not (P < 1 × 10−7 and P < 1 × 10−12, respectively; Table 1). This finding was also true for lymph node (Fig. 2E) and bone (Fig. 2F) metastases, in which 85% and 65%, respectively, had reduced or lost CD9 expression. There was no statistical difference between advanced prostate cancers, lymph node metastases, and bone metastases (Table 1).

Table 1.

Comparison of CD9 expression status between different groups of cases with prostatic adenocarcinoma

Case groupNo. patientsCD9 expression, n (%)
P*
PositiveReduced
Localized primary PCa     
    Without hormone pretreatment 65 53 (82) 12 (18) 0.08 
    With hormone pretreatment 42 28 (67) 14 (33)  
For primary PCa without hormone pretreatment     
    Localized primary PCa 65 53 (82) 12 (18) <0.000001 
    Advanced primary PCa 60 9 (15) 51 (85)  
    Localized primary PCa 65 53 (82) 12 (18) <0.000001 
    Lymph node metastases 65 10 (15) 55 (85)  
    Localized primary PCa 65 53 (82) 12 (18) <0.0001 
    Bone metastases 23 8 (35) 15 (65)  
For primary PCa with hormone pretreatment     
    Localized primary PCa 42 28 (67) 14 (33) <0.000001 
    Advanced primary PCa 60 9 (15) 51 (85)  
    Localized primary PCa 42 28 (67) 14 (33) <0.000001 
    Lymph node metastases 65 10 (15) 55 (85)  
    Localized primary PCa 42 28 (67) 14 (33) <0.05 
    Bone metastases 23 8 (35) 15 (65)  
Case groupNo. patientsCD9 expression, n (%)
P*
PositiveReduced
Localized primary PCa     
    Without hormone pretreatment 65 53 (82) 12 (18) 0.08 
    With hormone pretreatment 42 28 (67) 14 (33)  
For primary PCa without hormone pretreatment     
    Localized primary PCa 65 53 (82) 12 (18) <0.000001 
    Advanced primary PCa 60 9 (15) 51 (85)  
    Localized primary PCa 65 53 (82) 12 (18) <0.000001 
    Lymph node metastases 65 10 (15) 55 (85)  
    Localized primary PCa 65 53 (82) 12 (18) <0.0001 
    Bone metastases 23 8 (35) 15 (65)  
For primary PCa with hormone pretreatment     
    Localized primary PCa 42 28 (67) 14 (33) <0.000001 
    Advanced primary PCa 60 9 (15) 51 (85)  
    Localized primary PCa 42 28 (67) 14 (33) <0.000001 
    Lymph node metastases 65 10 (15) 55 (85)  
    Localized primary PCa 42 28 (67) 14 (33) <0.05 
    Bone metastases 23 8 (35) 15 (65)  

Abbreviation: PCa, prostate cancer.

*

χ2 test.

There is no significantly statistical difference between advanced PCa and lymph node metastases, between advanced PCa and bone metastases, and between lymph node and bone metastases (P = 0.95, 0.05, and 0.05, respectively).

Inverse correlation of clinicopathologic variables with CD9 expression. In the 167 primary tumors with available clinicopathologic data, CD9 expression was inversely correlated with prostate-specific antigen level at diagnosis (P = 0.0058), tissue differentiation (by Gleason score; P = 0.0027), pathologic stage (P < 1 × 10−8), and metastasis status (P < 1 × 10−8; Table 2). Prostate-specific antigen level in CD9-reduced cases was much higher than in positive cases (mean, 64.4 and 16.0 ng/mL, respectively; P = 0.0058). As histologic differentiation was lost, more CD9-reduced cases were observed (8% for well-differentiated tumors, 40% for moderately differentiated tumors, and 55% for poorly differentiated tumors; P = 0.0027). As the clinical stage was more advanced, more CD9-reduced and/or negative cases were observed (25%, 36%, and 82% for stage T2, T3, and N+ or M+, respectively; P < 1 × 10−8). In addition, 82% (41 of 50) of prostate specimens obtained from patients with metastatic disease had reduced or lost CD9 expression compared with only 31% (36 of 117) for patients without metastasis (P < 1 × 10−8). However, when analyzed with Kaplan-Meier curve using WinSTAT from R.FINCH software, decreased or loss of CD9 immunostaining did not predict biochemical recurrence (as defined by a prostate-specific antigen value >0.1; P = 0.31) in our population of 107 patients, with a median follow-up of 62.7 months.

Table 2.

Inverse correlation between CD9 expression and clinicopathologic variables in 167 patients with primary prostatic adenocarcinoma

CharacteristicsCD9 expression
P
Positive, 90 (54%)Reduced, 77 (46%)
PSA (mean ± SD; ng/mL) 16.0 ± 29.4 64.4 ± 157.1 0.0058 
Histologic differentiation    
    Well (Gleason scores 4-5) 12 (92%) 1 (8%)  
    Moderately (Gleason scores 6-7) 36 (60%) 24 (40%)  
    Poorly (Gleason scores 8-10) 42 (45%) 52 (55%) 0.0027 
Pathologic stage    
    T2N0M0 44 (75%) 15 (25%)  
    T3N0M0 37 (64%) 21 (36%)  
    N+ or M+ 9 (18%) 41 (82%) <0.000001 
Metastatic status    
    No metastasis 81 (69%) 36 (31%)  
    With metastasis 9 (18%) 41 (82%) <0.000001 
CharacteristicsCD9 expression
P
Positive, 90 (54%)Reduced, 77 (46%)
PSA (mean ± SD; ng/mL) 16.0 ± 29.4 64.4 ± 157.1 0.0058 
Histologic differentiation    
    Well (Gleason scores 4-5) 12 (92%) 1 (8%)  
    Moderately (Gleason scores 6-7) 36 (60%) 24 (40%)  
    Poorly (Gleason scores 8-10) 42 (45%) 52 (55%) 0.0027 
Pathologic stage    
    T2N0M0 44 (75%) 15 (25%)  
    T3N0M0 37 (64%) 21 (36%)  
    N+ or M+ 9 (18%) 41 (82%) <0.000001 
Metastatic status    
    No metastasis 81 (69%) 36 (31%)  
    With metastasis 9 (18%) 41 (82%) <0.000001 

Abbreviation: PSA, prostate-specific antigen.

Mutations of CD9 cDNA in prostatic adenocarcinoma. To further understand the mechanisms implicated in the loss of CD9 protein expression during prostatic adenocarcinoma progression, we PCR amplified, cloned (up to 12 clones per amplification reaction), and sequenced its mRNA coding region. No alteration of the CD9 mRNA transcript was detected in 36 clones isolated from six normal prostate specimens. Although wild-type CD9 cDNA clones were obtained in all samples analyzed, clones derived from four PC-3–derived prostatic carcinoma cell lines, four specimens of prostatic adenocarcinoma, and one specimen of PIN also harbored one of three major deletions (Table 3). These deletions removed nucleotides 115 to 487, 190 to 585, and 120 to 619 of the 684 bp CD9 coding sequence. Thus, from the 228 amino acid CD9 protein, amino acids 39 to 163, 64 to 195, and 40 to 207 were eliminated by these deletions. These deletions affected the large extracellular and intracellular domains of the protein. The presence of the PC-3M-LN4 deletion (del 64-195) was confirmed on direct sequencing of the mRNA amplification product (without cloning). These deletions were not detected in genomic DNA derived from some of these samples, arguing for the existence of transcriptional CD9 mRNA modifications. Another deletion was detected in the DU145 cell line, whereas an in-frame insertion was present in mRNA derived from PC-3M-Pro4 (Table 3).

Table 3.

CD9 mRNA modifications observed in human prostatic carcinoma cell lines and tissues

Type of mutations (amino acid position)PCa cell lines or tissuesPredicted consequences and affected CD9 regions
Deletion (39-163) PC-3, PC-3M-Pro4, and in one PCa patient Truncated protein (deleted from extracellular domain 1 to extracellular domain 2) 
Deletion (64-195) PC-3M-LN4 and in three PCa patients Truncated protein (deleted from transmembrane domain 2 to extracellular domain 2) 
Deletion (40-207) PC-3M and in one patient with PIN Truncated protein (deleted from extracellular domain 1 to transmembrane domain 4) 
Deletion (23-59; alternative splicing) DU145-N19 Truncated protein (deleted transmembrane domain 1) and new Stop codon in transmembrane domain 2) 
Insertion of 81 bp (between 22 and 23; alternative splicing) PC-3M-Pro4 Insertion (in frame) of 27 amino acids; abnormal protein with an extra domain 
Thr142Ala PC-3M-LN4 and in four PCa patients Missense in extracellular domain 2 
His147Arg Three PCa patients Missense in extracellular domain 2 
Thr175Ala Two PCa patients Missense in extracellular domain 2 
Gln83Stop Two PCa patients and one PIN Nonsense (Stop) in cytoplasmic domain 2; truncated protein 
Type of mutations (amino acid position)PCa cell lines or tissuesPredicted consequences and affected CD9 regions
Deletion (39-163) PC-3, PC-3M-Pro4, and in one PCa patient Truncated protein (deleted from extracellular domain 1 to extracellular domain 2) 
Deletion (64-195) PC-3M-LN4 and in three PCa patients Truncated protein (deleted from transmembrane domain 2 to extracellular domain 2) 
Deletion (40-207) PC-3M and in one patient with PIN Truncated protein (deleted from extracellular domain 1 to transmembrane domain 4) 
Deletion (23-59; alternative splicing) DU145-N19 Truncated protein (deleted transmembrane domain 1) and new Stop codon in transmembrane domain 2) 
Insertion of 81 bp (between 22 and 23; alternative splicing) PC-3M-Pro4 Insertion (in frame) of 27 amino acids; abnormal protein with an extra domain 
Thr142Ala PC-3M-LN4 and in four PCa patients Missense in extracellular domain 2 
His147Arg Three PCa patients Missense in extracellular domain 2 
Thr175Ala Two PCa patients Missense in extracellular domain 2 
Gln83Stop Two PCa patients and one PIN Nonsense (Stop) in cytoplasmic domain 2; truncated protein 

Furthermore, common missense point mutations were observed in one prostatic carcinoma cell line (PC-3M-LN4), one specimen of PIN, and seven specimens of prostatic adenocarcinoma (Table 3). Some specimens were harboring more than one missense mutation. Interestingly, CD9 protein expression was not detected in most of these cases (except in one specimen of prostatic adenocarcinoma). A bp substitution resulting in a new stop codon, located in the second cytoplasmic domain (amino acid 83), was also present in one PIN and in two prostate cancer patients where we did not detect the CD9 protein (Table 3). Overall, clones harboring CD9 mRNA modifications (either deletions or point mutations) represented 10% to 25% of the clones analyzed. Figure 3 summarizes the CD9 mRNA alterations and their predicted effect on the protein.

Fig. 3.

Schematic representation of deletions and mutations of CD9 coding region in prostatic adenocarcinoma. CD9 domains are the following: N-Cyt, NH2-terminal cytoplasmic tail; TM1-4, transmembrane domains 1 to 4; EC1, small extracellular domain; IC, intracellular domain; EC2, large extracellular domain; C-Cyt, COOH-terminal cytoplasmic tail. Dotted line, deletions; double line, the 27–amino acid insertion. Amino acid changes affected by point mutations. (See also Table 3).

Fig. 3.

Schematic representation of deletions and mutations of CD9 coding region in prostatic adenocarcinoma. CD9 domains are the following: N-Cyt, NH2-terminal cytoplasmic tail; TM1-4, transmembrane domains 1 to 4; EC1, small extracellular domain; IC, intracellular domain; EC2, large extracellular domain; C-Cyt, COOH-terminal cytoplasmic tail. Dotted line, deletions; double line, the 27–amino acid insertion. Amino acid changes affected by point mutations. (See also Table 3).

Close modal

The present study indicates that, like in other human malignancies, loss of CD9 protein expression is associated with prostatic adenocarcinoma progression and metastasis. Indeed, our data showed that CD9 is well expressed in nonmetastatic disease but less expressed or absent in metastatic prostate cancer. In fact, the majority (76%) of our localized prostate cancers, 99% of which are nonmetastatic, were CD9 positive. In contrast, only 15% of our advanced prostate cancers, 82% of which are metastatic, still retained CD9 expression. The prostate-specific antigen level at diagnosis is significantly higher (P < 1 × 10−5) in advanced than localized prostate cancers. Advanced prostate cancers are also less differentiated (P < 0.001) and more advanced in pathologic stage (P < 10−26). Therefore, the inverse correlation of CD9 with clinical pathologic findings is consistent with a putative role for CD9 at some critical point in the metastatic cascade.

There was no significant difference in CD9 expression when cases were stratified according to whether patients had received combined androgen blockade therapy or not (Table 1). CD9 protein was highly expressed in hyperplastic basal cells of nonneoplastic constituents of the prostate from patients with combined androgen blockade therapy. In the adenocarcinomatous component, no significant correlation between CD9 expression and the response rate to combined androgen blockade therapy (35) was detected. Our results contrast with the small induction (1.8-fold) of CD9 mRNA seen in LNCaP prostate cancer cells following 8 h of incubation with the synthetic androgen R1181 (36); the same authors also reported that prostate tissues from one patient treated with androgen ablation therapy did not show any CD9 staining in its luminal cells. As reported in Table 1, we also observed such reduced or absent CD9 staining in primary prostate tumors of patients undergoing combined androgen blockade. However, such treatment did not result in statistically significant difference in CD9 staining (P = 0.08) because some untreated patients with localized primary prostate cancer also had reduced CD9 expression. Although we cannot rule out the possibility that CD9 could be regulated by androgens, our results strongly suggest that such regulation is unlikely to influence disease progression.

The divergent distribution of CD9 expression in localized and advanced prostate cancer hinders the disease-free survival rate and prognosis analysis. There were 47 of 107 cases with cancer relapse in the localized prostate cancers. However, we could not find a statistical correlation with CD9 expression because 25 (53%) of them were CD9 positive. It should also be noted that many patients were lost on follow-up, as shown by the median follow-up of 62.7 months, although all prostatectomies were done before 1996. Conversely, the situation was similar in advanced prostate cancers because most cases (51 of 60) showed a significant reduction or loss of CD9 expression. Because they represent two totally different entities of the disease, we could not combine and analyze these two databases together. Indeed, only 43% of our patients with localized cancers relapsed compared with 100% of those with advanced cancers. It is also true for the mortality rate after 5 years of follow-up because only one patient (1%) died of localized prostate cancer, whereas 42% of patients died from advanced prostate cancer. Although reduced expression of CD9 protein has been associated with cancer progression in different tumor types, this is the first report implicating CD9 mRNA alterations in CD9 protein inactivation. Although wild-type CD9 mRNA was present in all samples analyzed, four deletions, one insertion, and four base substitutions were also detected in tumorigenic and precancerous (PIN) samples. No such alterations were detected in normal samples. The deletion (amino acids 23-59) found in DU145 cells and the 81 bp insertion seen in PC-3M-Pro4 cells are likely the result of alternative splicing. The DU145 deletion completely eliminates exon 2. Consequently, the new encoded protein will have a frameshift producing a stop codon (UGA), thus generating a truncated protein. For the PC-3M-Pro4 insertion, both splicing donor (CT) and acceptor (AG) sites in CD9 genomic DNA are localized at each end of the insertion. Moreover, when analyzed for their strength using a splice-site model accounting for pairwise dependencies (37), the new splicing sites bordering the 81 bp insertion have a maximum enthropy of 7.19 for 3′ splicing site and 6.64 for 5′ splicing site. These values are as good as others found on the boundary of other CD9 exons (maximum enthropy of 4.87 for 3′ splicing site of exon 7 or 6.66 for 3′ splicing site of exon 2), arguing that this region is likely to be inserted.

Three of four deletions were simultaneously found in CD9 cDNAs derived from primary tumor samples and in mRNA of highly tumorigenic and poorly differentiated prostatic carcinoma cell lines of the PC-3 lineage (Table 3). One deletion (amino acids 64-195) was found in four different samples, arguing against random PCR amplification errors. These three deleted regions could not have been generated by conventional alternative splicing due to absence of donor and acceptor splicing sites at the deletion boundaries. However, we noticed that nucleotides present at the beginning of the deleted region were the same as the ones present when the RNA sequence resumed. These repeats varied from three nucleotides (TCA) in deletion (120-619) to eight nucleotides (TCTTCAGACA) in deletion (115-487). The presence of such repeats could be implicated in the generation of the deleted mRNA molecules we have identified and could represent a novel mechanism for RNA alterations. These three deletions affect the large extracellular and intracellular domains of the protein. Such deletions are thus likely to interfere with CD9 functions because these domains are required for CD9 interaction with other proteins (11, 14, 15). Interestingly, all four prostatic adenocarcinomas containing these deletions did not express CD9 protein as shown by immunohistochemical analysis.

The large extracellular domain of CD9, and more precisely amino acids Gly158, Val159, and Thr175, are the major residues responsible for enhancing the binding of diphtheria toxin to its receptor (38, 39). It is worthy to note that one of these residues was substituted (Thr175Ala; A to G) in two different prostatic adenocarcinomas that had lost CD9 expression. In these two cases, the same major deletion (amino acids 64-195) was also detected in their derived cDNAs. An obvious effect of these mutations (deletions and point mutations), if these mRNAs are translated, will be to generate altered and/or inactivated CD9 protein. A similar situation was found in another CD9-negative tumor sample where a stop codon (Gln83Stop), located in the cytoplasmic domain, and a deletion (amino acids 64-195) were detected. The mechanisms responsible for the generation of the deleted mRNAs are still unknown; however, base substitutions in RNA molecules have been reported in mammalian cells as a result of RNA editing (40). Although RNA editing has not yet been reported in prostate cancer cells, all bp substitutions detected in CD9 mRNA are compatible with such mechanism.

The presence of CD9 mRNA substitutions in prostatic adenocarcinomas suggests that the function of the CD9 protein may be hindered by such amino acid substitutions. Thus, CD9-positive staining seen in a high proportion (82%) of localized prostate cancer (and perhaps also in bone metastases, 35% being positive) should be interpreted with caution. It is indeed possible that in cases where CD9 base substitution occurs, the encoded protein, although still detectable with an antibody, is inactive and unable to adequately interact with other proteins. This could also explain why CD9 expression is not a good predictor of recurrence; indeed, such inactivation of CD9 will still be seen as CD9-positive staining in patients with recurrence. Alternatively, the fact that the epitope recognized by the CD9 antibody used for our immunohistochemical staining is located in the large extracellular loop of the protein will preclude CD9 detection if this domain is deleted in the mutated protein. This could explain why we could not detect CD9 in cases where we identified the three major deletions. In these cases, a truncated protein, undetectable with our antibody, could still be present inside the cells.

In conclusion, we have shown that CD9 protein expression is inversely correlated with prostatic adenocarcinoma progression and metastasis. As tumors become less differentiated and more advanced in stage, CD9 expression is greatly reduced and even lost. Moreover, although never detected in normal prostate samples, mRNA modifications could play a role in regulating CD9 function during prostate cancer progression. Combined with previous reports on CD9 expression in other tissues, these findings strongly suggest that CD9 would act as a tumor suppressor protein.

Grant support: The Cancer Research Society, Inc., with a special contribution from the Bernard Richard Funds, the Canadian Institute for Health Research, and Shire BioChem, Inc.

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

We thank France Landry for her technical assistance, Hassan Behlouli for his help in statistical analysis, Than-Vinh Dam for his assistance in image processing and the preparation of frozen prostate slides, Laurent Guy for the establishment of a localized prostate cancer (whole-mount slides) bank, Eltaher A. Omar for the establishment of an advanced prostate cancer bank, Hazem Ismael for his assistance in updating the clinical information of the prostate tissue bank, and Val Zvereff for helpful discussions and revision of the manuscript.

1
Jemal A, Murray T, Ward E, et al. Cancer statistics, 2005.
CA Cancer J Clin
2005
;
55
:
10
–30.
2
Berube NG, Speevak MD, Chevrette M. Suppression of tumorigenicity of human prostate cancer cells by introduction of human chromosome del(12)(q13).
Cancer Res
1994
;
54
:
3077
–81.
3
Jaeger EB, Chekmareva MA, Tennant TR, et al. Inhibition of prostate cancer metastatic colonization by approximately 4.2 Mb of human chromosome 12.
Int J Cancer
2004
;
108
:
15
–22.
4
Kibel AS, Faith DA, Bova GS, Isaacs WB. Loss of heterozygosity at 12P12-13 in primary and metastatic prostate adenocarcinoma.
J Urol
2000
;
164
:
192
–6.
5
Kibel AS, Huagen J, Guo C, et al. Expression mapping at 12p12-13 in advanced prostate carcinoma.
Int J Cancer
2004
;
109
:
668
–72.
6
Boucheix C, Nguyen VC, Perrot JY, et al. Assignment to chromosome-12 of the gene coding for the human cell surface antigen CD9(P24) using the monoclonal antibody Alb6.
Annales de Genetique
1985
;
28
:
19
–24.
7
Benoit P, Gross MS, Frachet P, et al. Assignment of the human CD9 gene to chromosome-12 (region-p13) by use of human specific DNA probes.
Hum Genet
1991
;
86
:
268
–72.
8
Boucheix C, Benoit P, Frachet P, et al. Molecular cloning of the CD9 antigen—a new family of cell surface proteins.
J Biol Chem
1991
;
266
:
117
–22.
9
Lanza F, Wolf D, Fox CF, et al. cDNA cloning and expression of platelet P24/CD9—evidence for a new family of multiple membrane-spanning proteins.
J Biol Chem
1991
;
266
:
10638
–45.
10
Miyake M, Koyama M, Seno M, Ikeyama S. Identification of the motility related protein (MRP-1), recognized by monoclonal antibody M31-15, which inhibits cell motility.
J Exp Med
1991
;
174
:
1347
–54.
11
Boucheix C, Rubinstein E. Tetraspanins.
Cell Mol Life Sci
2001
;
58
:
1189
–205.
12
Boucheix C, Silvie O, Rubinstein E, Mazier D. A tiny thread towards a tetraspanin function.
Pathologie Biologie
2004
;
52
:
55
–7.
13
Rubinstein E, LeNaour F, Lagaudriere-Gesbert C, Billard M, Conjeaud H, Boucheix C. CD9, CD63, CD81, and CD82 are components of a surface tetraspan network connected to HLA-DR and VLA integrins.
Eur J Immunol
1996
;
26
:
2657
–65.
14
Maecker HT, Todd SC, Levy S. The tetraspanin superfamily: molecular facilitators.
FASEB J
1997
;
11
:
428
–42.
15
Hemler ME. Specific tetraspanin functions.
J Cell Biol
2001
;
155
:
1103
–7.
16
Boismenu R, Rhein M, Fischer WH, Havran WL. A role for CD81 in early T cell development.
Science
1996
;
271
:
198
–200.
17
LeNaour F, Rubinstein E, Jasmin C, Prenant M, Boucheix C. Knock-out mice for the tetraspan CD9 molecule have a fertilization defect.
Med Sci (Paris)
2000
;
16
:
287
–8.
18
Miyado K, Yamada G, Yamada S, et al. Requirement of CD9 on the egg plasma membrane for fertilization.
Science
2000
;
287
:
321
–4.
19
Fukudome K, Furuse M, Imai T, et al. Identification of membrane antigen C33 recognized by monoclonal antibodies inhibitory to human T-cell leukemia virus type-1 (Htlv-1) induced syncytium formation—altered glycosylation of C33 antigen in Htlv-1 positive T-cells.
J Virol
1992
;
66
:
1394
–401.
20
Yanez-Mo M, Tejedor R, Rousselle P, Sanchez-Madrid F. Tetraspanins in intercellular adhesion of polarized epithelial cells: spatial and functional relationship to integrins and cadherins.
J Cell Sci
2001
;
114
:
577
–87.
21
Ikeyama S, Koyama M, Yamaoko M, Sasada R, Miyake M. Suppression of cell motility and metastasis by transfection with human motility-related protein (MRP-1/CD9) DNA.
J Exp Med
1993
;
177
:
1231
–7.
22
Miyake M, Nakano K, Ieki Y, et al. Motility related protein-1 (MRP-1/CD9) expression—inverse correlation with metastases in breast cancer.
Cancer Res
1995
;
55
:
4127
–31.
23
Higashiyama M, Taki T, Ieki Y, et al. Reduced motility related protein-1 (MRP-1/CD9) gene expression as a factor of poor prognosis in non-small-cell lung cancer.
Cancer Res
1995
;
55
:
6040
–4.
24
Cajot JF, Sordat I, Silvestre T, Sordat B. Differential display cloning identifies motility-related protein (MRP1/CD9) as highly expressed in primary compared to metastatic human colon carcinoma cells.
Cancer Res
1997
;
57
:
2593
–7.
25
Hashida H, Takabayashi A, Tokuhara T, et al. Integrin α3 expression as a prognostic factor in colon cancer: association with MRP-1/CD9 and KAI1/CD82.
Int J Cancer
2002
;
97
:
518
–25.
26
Higashiyama M, Doi O, Kodama K, et al. Immunohistochemically detected expression of motility-related protein-1 (MRP-1/CD9) in lung adenocarcinoma and its relation to prognosis.
Int J Cancer
1997
;
74
:
205
–11.
27
Miyake M, Nakano K, Itoi S, Koh T, Taki T. Motility-related protein-1 (MRP-1/CD9) reduction as a factor of poor prognosis in breast cancer.
Cancer Res
1996
;
56
:
1244
–9.
28
Houle CD, Ding XY, Foley JF, Afshari CA, Barrett JC, Davis BJ. Loss of expression and altered localization of KAI1 and CD9 protein are associated with epithelial ovarian cancer progression.
Gynecol Oncol
2002
;
86
:
69
–78.
29
Pettaway CA, Pathak S, Greene G, et al. Selection of highly metastatic variants of different human prostatic carcinomas using orthotopic implantation in nude mice.
Clin Cancer Res
1996
;
2
:
1627
–36.
30
Liang P, Pardee AB. Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction.
Science
1992
;
257
:
967
–71.
31
Zvieriev V, Wang JC, Chevrette M. Over-expression of CD9 does not affect in vivo tumorigenic or metastatic properties of human prostate cancer cells.
Biochem Biophys Res Commun
2005
;
337
:
498
–504.
32
Elias JM, Margiotta M, Gaborc D. Sensitivity and detection efficiency of the peroxidase antiperoxidase (PAP), avidin biotin peroxidase complex (ABC), and peroxidase labeled avidin biotin (LAB) methods.
Am J Clin Pathol
1989
;
92
:
62
–7.
33
Rubinstein E, Benoit P, Billard M, et al. Organization of the human CD9 gene.
Genomics
1993
;
16
:
132
–8.
34
Nakamura Y, Iwamoto R, Mekada E. Expression and distribution of CD9 in myelin of the central and peripheral nervous systems.
Am J Pathol
1996
;
149
:
575
–83.
35
Vaillancourt L, Tetu B, Fradet Y, et al. Effect of neoadjuvant endocrine therapy (combined androgen blockade) on normal prostate and prostatic carcinoma—a randomized study.
Am J Surg Pathol
1996
;
20
:
86
–93.
36
Chuan Y, Pang ST, Bergh A, Norstedt G, Pousette A. Androgens induce CD-9 in human prostate tissue.
Int J Androl
2005
;
28
:
291
–6.
37
Yeo G, Burge CB. Maximum entropy modeling of short sequence motifs with applications to RNA splicing signals.
J Comput Biol
2004
;
11
:
377
–94.
38
Nakamura K, Mitamura T, Takahashi T, Kobayashi T, Mekada E. Importance of the major extracellular domain of CD9 and the epidermal growth factor (EGF)-like domain of heparin-binding EGF-like growth factor for up-regulation of binding and activity.
J Biol Chem
2000
;
275
:
18284
–90.
39
Hasuwa H, Shishido Y, Yamazaki A, Kobayashi T, Yu XC, Mekada E. CD9 amino acids critical for upregulation of diphtheria toxin binding.
Biochem Biophys Res Commun
2001
;
289
:
782
–90.
40
Gott JM. Expanding genome capacity via RNA editing.
C R Biol
2003
;
326
:
901
–8.