The cell cycle regulatory genes p16/CDKN2 and RB are frequently deleted in prostate cancers. In this study, we examined the role of alterations in p16 and pRb during growth, senescence, and immortalization in vitro of human prostate epithelial cells (HPECs). HPECs are established from normal prostate tissues and cultured on collagen-coated dishes. Our results show that p16 is reproducibly elevated at senescence in HPECs. HPECs are immortalized using human papilloma virus 16 E6 and/or E7 as molecular tools to inactivate p53 and/or pRb, respectively. Immortalization occurs infrequently in this system and only after a latent period during which additional genetic/epigenetic changes are thought to occur. Notably, all of the E6-immortalized HPEC lines but none of the E7 lines show inactivation of p16/CDKN2 (by deletion, methylation, or mutation) in association with immortalization. In contrast, E7 lines, in which pRb function is abrogated by E7 binding, retain the high levels of p16 observed at senescence. Thus, all lines show either a p16 or pRb inactivation. Analysis of six independent lines from metastatic prostate cancers reveals a similar loss of either p16 or pRb. Comparative genomic hybridization of HPECs shows that gains of chromosomes 5q, 8q, and 20 are nonrandomly associated with bypassing senescence (probability = 0.95). These results suggest that high levels of the cyclin-dependent kinase inhibitor p16 mediate senescence G1 arrest in HPECs and that bypassing this block by a p16/pRb pathway alteration is required for immortalization in vitro and possibly tumorigenesis in vivo. Our results further indicate that inactivation of the p16/pRb pathway alone is not sufficient to immortalize HPECs and that additional genetic alterations are required for this process.

Normal human cells have a finite proliferative potential (usually 50–100 doublings) in vitro, after which cell cycle arrest occurs (1). This process is termed replicative senescence, and in contrast to apoptotic or programmed cell death, it represents a stable, protracted state in which cells remain viable and metabolically active. The process of immortalization requires overcoming or escaping replicative cellular senescence. Bypassing senescence is thought to be important in the development of human cancer for several reasons (2, 3): (a) immortalization increases cell susceptibility to malignant progression by permitting the extensive cell divisions that are necessary for cells to acquire multiple genetic alterations; (b) tumors, both naturally occurring and experimentally induced, often contain cells that are immortal or have an extended replicative life span (4, 5); and (c) genetic regions associated with the immortalization of cell types in vitro are found to be altered in clinical cancers of the same cell type (6). Therefore, an understanding of the genes controlling normal prostate epithelial cell mortality may provide relevant information on the mechanisms underlying abnormal cell growth and tumorigenesis in prostate epithelial cells.

A series of classical observations have shown that replicative life span can be extended by the expression of DNA tumor virus oncoproteins, including SV40 and HPV163 E6 and E7 (3, 7). These tumor viruses are known to inactivate p53 and pRb (8), thus implicating these antiproliferative proteins in the process of overcoming senescence. This initial period of extended life span requires overcoming the M1 (mortality) block. However, overcoming M2, which results in an infinite life span, clearly requires additional genetic events (9, 10). Immortalization occurs infrequently, which indicates that the inactivation of several pathways is required. Alterations in other cell cycle regulatory genes, including p21WAF1 and p16/CDKN2, have been implicated in mediating senescence (11, 12, 13). Notably, all of these genes encode putative tumor suppressor proteins, the functions of which are lost in many human cancers, including prostate cancers (14, 15, 16, 17, 18, 19).

Increases in wild-type p53 induce antiproliferation signals, including the downstream cyclin-dependent kinase p21, that are distinct from apoptosis (20). Levels of p53 have been found to increase at late passage in human fibroblasts, putatively contributing to senescence growth arrest (11, 21). Inhibiting the expression of p53, either by antisense RNA methods (22) or by use of p53 transdominant mutants (23), leads to a delay in senescence. Elevation of p21 levels has also been found in aging fibroblasts (13). However, because levels decrease at senescence, the significance of the role of p21 in senescence growth arrest is unclear. Its role in tumorigenesis is also uncertain: a recent study showed that mice with a homozygous deletion of p21/WAF1 failed to form tumors (24).

pRb and p16 function in a common pathway that appears to be important in the growth arrest of cells after a finite number of population doublings. Phosphorylation regulates pRb function by inducing E2F release and the subsequent expression of E2F-dependent proteins, such as cdc2 and cyclin A. These genes are not expressed in senescent fibroblasts consistent with a block in pRb phosphorylation at senescence (25). Furthermore, pRb is found in a hypophosphorylated state in senescent fibroblasts (26). Overcoming this senescence block is possible by fusion with cells containing the viral oncogene E7, which binds unphosphorylated pRb (26). p16 blocks pRb phosphorylation by binding cdk4 and cdk6 and inhibiting their association with cyclin D (27). This results in a failure of pRb phosphorylation and E2F release and culminates in G1 cell cycle arrest. Several studies have shown that p16 levels increase in human and rodent fibroblasts as cells are passed to terminal senescence (11, 28). However, only one study to date has demonstrated increased p16 at senescence in a human epithelial cell type, namely urothelial cells (12).

p16 and pRb inactivation are generally not found in the same cancer, consistent with the observation that they function within the same pathway (29, 30). In support of this model, the cell cycle arrest induced by the introduction of p16 occurs only in cells that retain functional pRb (31). The importance of p16 as a tumor suppressor is emphasized by its frequent inactivation in different types of human malignancies (32). We recently reported that p16/CDKN2, located at 9p21, was deleted or hypermethylated in >50% of advanced prostate cancers and that this finding was associated with transcriptional inactivation (19). RB loss of heterozygosity is also frequently found in prostate cancers (16, 17), although whether homozygous alterations occur is not known. The loss of pRb expression does correlate with a poor clinical prognosis (33).

In summary, an alteration in the p16/pRb pathway is commonly seen in prostate cancers. Several lines of evidence discussed above suggest that these alterations may play a role in bypassing the tumor suppressor checkpoint of cellular senescence. Here, we test this hypothesis using an HPEC in vitro transformation system that uses HPV16 E6 and E7 to inactivate p53 and pRb, respectively. We report here for the first time with HPECs that p16 levels are reproducibly elevated at senescence and that bypassing senescence in HPV16 E6- or E7- transformed cells is always accompanied by an alteration in the p16/pRb pathway. Furthermore, we identify additional genetic alterations, including +5q, +8q, and +20, that are nonrandomly associated with HPEC immortalization. We conclude that alteration of the p16/pRb pathway is necessary but not sufficient for overcoming senescence in HPECs.

Tissue Acquisition/Prostate Epithelial Cell Culture.

Biopsies of histologically confirmed normal prostate tissue from men (ages 47–65 years) without diagnosed prostate cancer were obtained from cystoprostatectomy specimens or from cadavers. The culture system used is based on the work of Peehl et al. (34, 35) and Reznikoff et al. (36). To establish HPECs, we minced fresh prostate tissues and plated them on 100-mm collagen dishes in F-12+ medium with penicillin/streptomycin (9). F-12+ is a supplemented Ham’s medium (Life Technologies, Inc., Gaithersburg, MD; intermediate Ca2+, 0.3 mm) that also contains 1% FBS. Bovine pituitary extract (25 μg/ml; Life Technologies, Inc.) and cholera toxin (100 ng/ml; Life Technologies, Inc.) were added to this medium. HPECs were passaged routinely when confluent after dispersion with trypsin-EDTA (Life Technologies, Inc.). Prostate cancer cell lines were grown in RPMI with 10% FBS.

Immunohistochemistry.

Immunohistochemistry was performed on early-passage HPECs and HPEC E6- and E7- immortalized cell lines after harvesting and 10% formalin fixation (37). Primary antibodies and dilutions included cytokeratins 5 (1:500; Enzo) and 18 (1:100; Sigma Chemical Co., St. Louis, MO) and prostate-specific antigen (1: 100; Sigma). Biotinylated antimouse or rabbit IgG was used as a secondary antibody (Sigma). Staining was accomplished with the ABC reagent (Vector Laboratories, Burlingame, CA) and the substrate diaminobenzidine. Immunohistochemistry was duplicated on each of three independent HPEC cultures. Controls included tissues with no primary antibody and uncultured normal prostate tissues.

SA β-Galactosidase Staining.

HPECs were transferred to chambered slides (Nunc, Naperville, IL) and grown for 2 days. Cells were then rinsed and fixed in a β-galactosidase stain solution as described (38). Positive β-galactosidase staining, as defined by blue cytoplasmic and nuclear staining, was a marker for senescence and was not present in differentiated or apoptotic cells (38).

Transformation of HPECs with HPV16 E6 and/or E7.

Transformation of HPECs with HPV16 E6 and/or E7 and selection for immortal clones was performed as described for human urothelial cells (9). Briefly, retroviruses carrying either the HPV16 E6 and/or E7 gene(s) (received from Dr. D. Galloway, Seattle, WA) were prepared (39). Subconfluent proliferating HPECs were infected with 103–105 infectious viral units at early-passage (∼5 × 105 cells per 100-mm dish) in 3 ml of 1% FBS-F-12+ containing 4 μg/ml polybrene (Sigma). The virus was then removed after 6 h, and selection with 50 μg/ml G418 (Life Technologies, Inc.) was performed for a minimum of 7 days. Two uninfected control dishes were monitored for senescence. Clones infected with E6, E7, or E6/E7 were checked for expression of E6 and E7 transcripts using RT-PCR as described previously (9). Southern blot analysis was also performed as described previously (9) using a 0.8-kb BamHI-HindIII fragment from p1321 that contains the HPV16 E6/E7 genes. Fifteen μg of DNA were loaded in each lane.

Western Blot Analysis.

Western blots were performed as described previously (12) Briefly, cells were lysed in buffer containing protease inhibitors and resolved (50 μg/lane) on 12.5% SDS-polyacrylamide gels. After transfer to a nylon blot (Immobilon P; Millipore, Bedford, MA), primary antibodies were applied. These included specific antibodies to p53 (AB2; Oncogene Science), phosphorylated and unphosphorylated forms of pRb (14001A; PharMingen), p16 (C-20, Santa Cruz Biotechnology, Santa Cruz, CA), and p21(Oncogene Science). Immunoreactive proteins were visualized using enhanced chemiluminescence. Urothelial cells immortalized with HPV16 E7 (12) were used as positive controls for p16, p53, and p21. Immunoblotting was performed in duplicate.

Sequencing of p16/CDKN2 and Methylation Analysis.

Sample DNA was amplified by PCR using primers spanning exons 1 and 2 of the p16/CDKN2 gene (40, 41). The amplified products were cloned into pCR 2.1-TOPO using the TOPO TA cloning kit from Invitrogen according to manufacturer’s instructions. The cloned fragments were sequenced using an automated DNA sequencer. Methylation analysis was performed as described previously using a 340-bp p16/CDKN2 exon 1 probe (19). Serial concentrations of HPEC E6-15 DNA was used in Southern blotting to confirm deletion of p16/CDKN2.

CGH of HPV16 HPEC Immortal Lines and Statistical Analysis.

Hybridization of differentially labeled immortalized cell lines to metaphase chromosomes from normal peripheral blood was performed exactly as described previously (42). We have described how we define changes in the relative copy number of DNA sequences (i.e., gains and losses) in an earlier manuscript (43). CGH loss and gain data were analyzed using a simple statistical model, as described previously (44). Briefly, the null hypothesis asserts that changes are sporadic and thus present randomly according to a background rate that is constant among chromosome arms. The alternative hypothesis allows that some arms exhibit an elevated rate of change. The model also allows the possibility that changes on the p and q arms are linked. Gain data exhibit a pattern significantly different from one would expect under the null hypothesis, with P = 0.001, whereas loss data are consistent with sporadic change (P = 0.31).

Growth and Senescence of HPECs in Vitro.

The growth of HPEC explants on collagen-coated dishes in F-12+ medium results in cultures of prostate epithelial cells that are epithelial in morphology (i.e., tightly adherent and polygonal) and apparently uncontaminated with other cell types (Fig. 1,A). After four to five passages, HPEC proliferation slows, and cells enter a nonproliferative state. These HPECs adopt a morphological phenotype characteristic of senescence (Fig. 1,B) and acquire SA β-galactosidase activity (Ref. 38; Fig. 1 C).

Characterization of Cultured HPECs.

To assess the phenotype of cultured HPECs using our conditions, we performed immunohistochemical staining for cytokeratin markers of luminal or basal prostate epithelial cells (45, 46) in both uncultured tissue sections and cultured HPECs. Cytokeratin 5, a basal cell marker, and cytokeratin 18, a luminal protein, are both expressed (data not shown). Another marker of differentiated luminal epithelial cells, prostate-specific antigen, is not found at detectable levels in HPECs using immunohistochemistry. It is, however, detectable using RT-PCR (data not shown). These results demonstrate that HPECs coexpress both basal and luminal keratins, thus representing the amplifying or stem cell population that is hypothesized to be the cellular precursor to prostate cancer (46). Additionally, expression of these prostate epithelial markers essentially excludes contamination by cells of stromal origin.

Elevation of p16 but not p21 or p53 at Senescence in Normal HPECs.

To determine whether alterations of pRb, p53, p21, and p16 might play a role in mediating prostate epithelial senescence, we compared protein levels in proliferating (presenescent) and senescent HPECs. In contrast to previous studies in fibroblasts (11), results showed no alterations in p53 or p21 levels at senescence in HPECs. However, HPECs at senescence reproducibly show at least a 10-fold elevation of p16 (Fig. 2), when compared to early-passage proliferating HPECs. We also note a consistently decreased level of phosphorylated pRb in senescent cells consistent with a G1 cell cycle block. pRb is detectable on longer exposure. The antibody used for pRb detection recognizes primarily phosphorylated forms of pRb.

Generation of Isogeneic HPV16 E6 and/or E7 HPEC Lines.

HPV16 E6 selectively abrogates p53, whereas HPV16 E7 binds and inactivates pRb. These viral genes were used as molecular tools to test, in HPECs, the functional significance of losing pRb and p53 in the process of overcoming senescence. Results showed that immortalization of human epithelial cell lines by HPV16 genes occurs in two stages (M1 and M2), as was initially described in mammary epithelial cells by Shay et al.(47). After infection of HPECs at passage 2 (P2) and selection with G418, all HPV16-transformed cultures (E6/E7, E6, and E7) showed an extended life span of two to three additional passages (eight versus five passages) before undergoing senescence. E6- and/or E7-transformed HPECs then entered an extended crisis period lasting 2–3 months in which no net gain in cell numbers was evident. After 2–3 months, small, single colonies of HPV16-transformed HPECs began to proliferate and could eventually be dispersed and passaged. These colonies represent the emergence of immortal clones from the so-called M2 stage, and they occurred at a low frequency of ∼1 × 10−5. E6/E7 immortal colonies typically emerge sooner than E6 or E7 colonies (∼6 weeks versus 12 weeks). Using this approach, we established three sets of HPV16 E6, E7, and E6/E7 HPEC immortal cell lines. Each set of cell lines was generated from a single initial prostate epithelial culture taken from one of three different individuals, thus the lines can be defined as isogeneic in origin.

Phenotypic differences are noted between the E6- and E7-transformed prostate epithelial cell lines. E6 HPEC and E6/E7 HPEC lines are characterized by a pleomorphic cell size, irregular shape, and loose adhesion (Fig. 3,A). In contrast, E7 HPEC cells have flat, tightly adherent cells more typical of normal epithelial morphology (Fig. 3 B). All HPV16-transformed HPEC lines are now at P40 or more (400 doublings), consistent with the immortal phenotype. Cytokeratin staining for 5 and 18 confirmed their epithelial origin.

Independence and Clonality of HPV16 E6 and/or E7 HPECs.

Southern blot analysis for HPV16 insertion was performed on the eight HPV cell lines generated at an early-passage (P18→P24). Lines were not considered immortal until approximately P15. Results show a single integration site in 5 cell lines, as indicated by a single band on Southern blot (Fig. 4,A). Two bands are demonstrated in HPEC lines E6-9, E6/E7-9, and E7–14. Given their apparent clonal origin (see above) and equal band intensity at different passages, these two bands probably represent two integration sites in each of these cell lines. RT-PCR confirms the correct expression of E6 and/or E7 RNA within the E6, E7, or E6/E7 HPEC lines (Fig. 4 B).

p16 or pRb Inactivation in HPV16 Immortal HPECs and in Metastatic Prostate Cancer Lines.

The sufficiency of pRb or p53 inactivation in overcoming senescence during immortalization was evaluated in HPECs. Consistent with a functioning HPV16 E6, Western blot analysis of HPEC E6 and E6/E7 immortal lines (P20) shows that p53 levels are undetectable (Fig. 5). In contrast, HPEC E7 cell lines show slightly elevated p53 levels compared to normal HPECs (Fig. 2). p21 expression was positively regulated by p53, and consistent with this, p21 was strongly expressed in HPEC E7. In contrast, even with prolonged Western exposure times, p21 protein was not detectable in p53 negative HPEC E6 lines. Therefore, p21 levels correlate with p53 expression and p53 function in E7 cell lines was confirmed.

pRb is nondetectable in the E7 cell lines and present at low levels in E6/E7 HPEC when compared to proliferating normal HPECs. The low level of pRb detected in HPEC E6/E7 lines may represent incomplete ubiquitin-induced degradation of pRb (48). pRb was strongly detected in HPEC E6 lines, as expected. These levels of pRb protein were comparable to those seen in normal prostate epithelium. However, in all three HPEC E6 lines, immortalization is accompanied by p16 loss. In contrast, p16 is abundantly expressed in the HPEC E7 and E6/E7 immortal lines (that have lost pRb function). These elevated levels of p16 are comparable to the levels seen in HPECs at senescence (Fig. 2). High levels of p16 do not inhibit proliferation in E7-transformed HPECs because of the inactivation of downstream regulatory protein pRb. Therefore, all HPV16 E6- and/or E7-transformed HPECs showed either p16 or pRb inactivation.

To further test the association between inactivation of pRb expression or p16 in bypassing senescence in HPECs, we evaluated a series of immortal prostate cancer cell lines (Fig. 5). These lines derive from independent biopsies of metastatic prostate cancer. Two cell lines, Du145 and LnCaP, showed no pRb expression. The Du145 prostate cancer cell line contains a known RB mutation (49). In both of these lines, p16 was present and detectable by Western analysis. All other metastatic immortal prostate cancer lines expressed apparently wild-type pRb, but no p16 protein was detectable. These results demonstrate a loss of p16 or pRb in all immortalized prostate cancer cells.

Abnormal p53 expression was detected in the majority of metastatic cell lines, including PC3, TSU-PR1, DuPro, and PPC-1. Inactivating mutations within the coding exons (exons 5–8) have been described previously in PC3 and TSU-PR1 (50). Du145 contains a stabilized mutant p53. The mutant p53 was undetectable in PC3 and TSU-PR1. In LnCaP, p53 is wild type, and low levels were detectable only by immunoprecipitation (50). Therefore, p53 inactivation was frequent but not necessary for overcoming senescence and acquisition of the metastatic phenotype in prostate cancer.

CDKN2/p16 Is Inactivated by Hypermethylation, Deletion, or Mutation in HPEC E6 and Metastatic Prostate Cancer Lines.

To identify mechanisms underlying the loss of p16 expression in immortal HPEC E6 lines and in representative metastatic prostate cancer lines described above, we performed methylation analysis using Southern blot (Fig. 6). Loss of p16 expression in two of three HPEC E6 lines (E6-9 and E6-14) was due to biallelic DNA hypermethylation, as evidenced by the presence of a 6-kb fragment after restriction with the methylation-sensitive enzyme SmaI. Methylation of this region has been previously correlated with loss of transcription (40). The radiolabeled probe used for Southern analysis demonstrates a reproducibly decreased signal (>50%) in cell line HPEC E6-15, indicating a heterozygous deletion of p16/CDKN2. This is consistent with CGH results (Table 1). Sequencing of exons 1 and 2 of p16/CDKN2 was performed in all HPEC E6 lines. E6-15 contains a mutation in exon 1 in codon 33 (GAG→TAG) that generates a stop codon and has been described previously (51). A second mutation was detected downstream in codon 34 (GCG→ACG; Glu→Thr). A complete loss of p16 expression from E6-15 resulted from a deletion of the other allele of CDKN2/p16 (Fig. 6).

An analysis of the p16/CDKN2 locus in prostate cancer cell lines by Southern demonstrated no methylation in LnCaP and Du145 (19), both of which express p16 by Western blot. However, a missense mutation in exon 2 on codon 84 (GAC→TAC; Arg→Leu) is present in Du145. This was identified in all sequencing reactions performed and confirmed an earlier report of mutation in this line (19). It did not alter expression of the protein. Hypermethylation was detected in PC3, TSU-PR1 (data not shown), DuPro, and PPC-1. Therefore, the loss of p16 expression in HPEC E6 and metastatic prostate cancer lines can, in all cases, be explained by hypermethylation, mutation, or deletion.

Additional Genomic Losses and Gains Accompany HPEC Immortalization.

CGH on early-passage HPV16 immortalized HPECs was used to identify genetic regions that were gained or lost in association with immortalization (Table 1). A gain of chromosome 20 was present in seven of eight HPEC-immortalized lines. 8q was gained in four cell lines. Gain of 5q was seen in all HPEC E7. Loss of 8p, a commonly deleted region (∼80%) in clinical prostate cancer (52), was present in one HPEC E6. Notably, CGH is relatively insensitive for the detection of genomic losses smaller than 10 Mb. Further mapping may detect smaller deletions. Nevertheless, these findings suggest that genetic alterations, including +20, +8q, and +5q may contribute to overcoming senescence.

To test this, we analyzed CGH loss and gain data using a simple statistical model as described earlier (5, 44). The null hypothesis asserts that changes are sporadic and thus present randomly according to a background rate that is constant among chromosome arms. The alternative hypothesis allows that some arms exhibit an elevated rate of change. The model allows the possibility that changes on the p and q arms are linked. Gain data exhibited a pattern that was significantly different from that which one would expect under the null hypothesis with P = 0.001, whereas loss data were consistent with sporadic change (P = 0.31). Among gains, 5q, 8q, and 20 were significant in the sense that the probability exceeded 0.95 (in each case) that gain of each arm exhibited an elevated rate. No significant losses were detected in our sample group.

Replicative senescence is a mechanism of tumor suppression and represents a safeguard against the development of neoplasia (2, 4). Therefore, the identification of genes that function in normal replicative senescence in prostate epithelial cells is important for our understanding of tumorigenesis in the prostate. In this study, we report for the first time that replicative senescence in normal prostate epithelial cell cultures is reproducibly associated with an elevation of p16. We also demonstrate for the first time using prostate epithelial cells that bypassing senescence requires an alteration in the p16/pRb pathway. However, this latter alteration is apparently insufficient for bypassing senescence, as nonrandom gains of 5q, 8q, and 20q also accompany immortalization.

Our data showing that p16 is elevated at senescence in prostate epithelial cells supports a model for replicative senescence that has been previously proposed for human fibroblast and urothelial cells (11, 12). p16 specifically binds to and inhibits CDK4 blocking progression of the cell cycle beyond G1. Thus, in this model, elevated levels of p16 play a critical role in senescence G1 growth arrest. The mechanisms underlying the dramatic increase in p16 levels at terminal senescence have not been defined but may result from an accumulation of p16, possibly due to an increased stabilization of p16 mRNA and/or a loss of p16 repression by decreased levels of pRb (53). We find a consistent down-regulation of phosphorylated forms of pRb in senescent human prostate epithelial cells, which may, in turn, contribute to a failure of cell cycle progression. Functional pRb is required for normal G1 growth arrest, as studies using transfected and expressed p16 have shown (31, 54). Our findings, therefore, implicate p16 as important in the G1 cell cycle arrest characteristic of senescence in normal prostate epithelial cells.

Notably, increases in expression of p21 and p53 do not occur during replicative cellular senescence in prostate epithelial cells. This result is similar to findings in cultured human urothelial cells (12) but differs from findings in human and rodent fibroblasts (11, 28). Some studies suggest that p21, which functions to arrest cells in cycle by binding to cyclins D, A, and E, is overexpressed in and is a mediator of replicative senescence in fibroblasts (13). However, other data question the finding that p21 is elevated at terminal senescence in fibroblasts (11). It is also uncertain whether p53 up-regulation is critical to replicative senescence. For example, p53-null fibroblasts from patients with Li-Fraumeni syndrome undergo normal senescence in the absence of expression changes in either p53 or p21 (55). Therefore, p21 and p53 elevation at senescence may represent a characteristic of selected fibroblast cell strains, but it does not appear to be important for senescence G1 cell cycle arrest in human prostate (or urothelial) epithelial cells.

To test our hypothesis that p16 elevation plays a critical role in replicative senescence, we examined the status of p16 and pRb in three isogeneic sets of HPV16 E6 and/or E7 immortal prostate epithelial cells that bypassed senescence. Each cell set arises from an initial single epithelial culture generated from one of three independent normal prostate specimens. Our second important finding is that overcoming the cell cycle block associated with senescence requires an alteration in the p16/pRb pathway. In the cells lines containing E7, pRb function is lost by the binding of E7 oncoprotein to underphosphorylated pRb and by an enhancement of ubiquitin-induced degradation of pRb (48). HPV16 E7 immortal lines show elevated levels of p16, similar to those seen at senescence. This finding supports the observation that the cell cycle arrest imposed by p16 is only apparent in cells that retain functional pRb (31, 54). The high levels of p16 in E7 pRb deficient cells provide further evidence for a feedback regulatory loop involving pRb and p16. We also tested spontaneously immortalized cell lines derived from biopsies of metastatic prostate cancer and demonstrate that pRb expression is lost and p16 is elevated in two of six lines.

An alternate pathway for reentry into the cell cycle from senescence in the presence of wild-type pRb involves a loss of p16 expression. Because pRb is a downstream component of the p16 pathway, loss of p16 function would, in theory, function equivalently to pRb loss. This scenario is found in all HPEC E6-immortalized cell lines that have lost functional p53 and retained pRb. In the E6 cell lines tested, p16 loss is mediated most commonly by DNA hypermethylation. We also find that one line, E6-15, contains an allelic deletion and a mutation on the remaining allele. Therefore, several common mechanisms for inactivating p16 are demonstrated in our experimental model using normal prostate epithelial cells. The requirement for a p16/pRb pathway alteration is also met in the spontaneously immortalized metastatic prostate cell lines. Loss of p16 expression due to hypermethylation of p16/CDKN2, along with wild-type pRb, is found in four of six of these cell lines. Hypermethylation of p16/CDKN2 is a selective mechanism for inactivating p16 expression, and does not appear to alter the expression of the alternative reading frame splice variant p14/ARF in bladder cancer cell lines (56). The finding of methylation inactivation of p16/CDKN2 in prostate cancer and HPEC E6-immortalized lines (six of nine) emphasizes a unique epigenetic feature of the tumorigenic process in prostate cells. Mutations of p16/CDKN2 occur rarely in prostate cancer (19) in contrast to other tumors (57, 58). The Du145 cell line, which contains a pRb mutation, we have found on sequencing to contain a p16/CDKN2 missense mutation (19, 59). However, wild-type p16 is apparently encoded from the second allele in this pRb-negative line.

The results above document that an alteration in the p16/pRb pathway is critical for immortalization of HPEC. However, we have identified a number of additional genetic alterations that are nonrandomly associated with overcoming the senescence block in HPECs. Both HPEC E7- and E6-infected cells undergo a crisis period of low to undetectable proliferation (M2 block) for several months before proliferative clones that give rise to immortal lines are detected. The most significant genetic change, identified in seven of eight immortal lines, is a gain of chromosome 20. Gain of chromosome 20 has been seen in many human cancer types, including bladder, breast, ovarian, and prostate (9, 43, 60). By CGH, centromeric regions of 20 are amplified in almost half of in vivo prostate cancer metastases (15). This amplification is infrequently found in primary prostate tumors. Finally, gain of chromosome 20 has been identified in human urothelial cells transformed in vitro by HPV16 (9). These observations indicate the presence on chromosome 20 of one or several oncogenes. Several candidate genes have been identified, including ZNF217, NABC1, and CAS(61). Other regions of gain in HPEC E6 and E7 lines include 5q and 8q. 8q gains are noted infrequently in primary prostate cancers, but they occur commonly in metastatic and recurrent (80%) prostate cancers (15). c-Myc is located at 8q24, and Myc protein levels are increased in E6 cells by an undefined posttranscriptional mechanism. Recently, it was found that telomerase induction is required for E6 epithelial immortalization, in addition to alterations in the p16/pRb pathway (10). Notably, we have demonstrated telomerase activity in cultured normal prostate epithelium, as well as normal bladder, ureter, and mammary epithelium (62). It has also been noted that Myc protein increases telomerase mRNA through an undefined mechanism (63).

In the current model of prostate carcinogenesis, inactivation of the p16-pRb pathway appears to play an important role in overcoming the cell cycle block imposed by p16 at senescence. RB, on 13q14, is a region of intermediate frequency (∼30%) deletion in prostate cancer samples (16, 64) We have previously demonstrated that p16/CDKN2 alterations occur frequently (∼50%) in prostate cancer (19). Given the present data supporting the mutual exclusion of p16 and pRb alterations within the same cell line, inactivation of this pathway may represent an extremely common alteration in prostate cancer. Although this correlation has not been tested in prostate cancers in vivo, it has been demonstrated in several other tumors (29, 65). Inactivation of the p16/pRb pathway is necessary but not sufficient for immortalization of HPEC. Thus, our in vitro model using E6 and E7 contains genetic gains and losses also seen in in vivo tumors. Therefore, it may be useful in identifying genes altered in prostate cancer and defining pathways of cancer progression via different combinations of genetic and epigenetic alterations.

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

      
1

This work was supported by NIH Grant CA76184 and a Howard Hughes Faculty Development award (to D. F. J.).

            
3

The abbreviations used are: HPV16, human papilloma virus 16; HPEC, human prostate epithelial cell; FBS, fetal bovine serum; RT-PCR, reverse transcriptase-PCR; CGH, comparative genomic hybridization; SA, senescence-associated.

Fig. 1.

Phase contrast microscopy and SA-β-galactosidase staining of proliferating and senescent normal human prostate epithelial cells in culture. A, early-passage HPECs demonstrate tightly juxtaposed cells that grow in large islands (phase microscopy, ×200). B, nonproliferating passage five HPECs demonstrate morphological changes consistent with senescence including flattened, enlarged cells, and multiple nuclei. C, SA-β-galactosidase staining (38) of nonproliferating prostate epithelial cells demonstrates blue nuclear/cytoplasmic staining indicating senescence (×400). Early-passage cultures (data not shown) were negative for SA-β-galactosidase staining.

Fig. 1.

Phase contrast microscopy and SA-β-galactosidase staining of proliferating and senescent normal human prostate epithelial cells in culture. A, early-passage HPECs demonstrate tightly juxtaposed cells that grow in large islands (phase microscopy, ×200). B, nonproliferating passage five HPECs demonstrate morphological changes consistent with senescence including flattened, enlarged cells, and multiple nuclei. C, SA-β-galactosidase staining (38) of nonproliferating prostate epithelial cells demonstrates blue nuclear/cytoplasmic staining indicating senescence (×400). Early-passage cultures (data not shown) were negative for SA-β-galactosidase staining.

Close modal
Fig. 2.

Western analysis of p53, pRb, p16, and p21 in early- and late-passage, senescent normal prostate epithelial cells. Protein expression from three separate proliferating human prostate epithelial cell cultures is shown (Pre-Senescent, Lanes 1–3). These cells were continuously passaged and analyzed at passage 5 after growth arrest (Senescent, Lanes 1–3). Senescence was confirmed by positive SA-β-galactosidase staining (data not shown). p16 is strongly expressed in senescent cells compared to early-passage cells. In contrast, no alterations in expression are found in p53 or p21. Phosphorylated pRb levels are decreased at senescence. HPEC-E7 is used as a positive control because it strongly expresses p53, p21, and p16.

Fig. 2.

Western analysis of p53, pRb, p16, and p21 in early- and late-passage, senescent normal prostate epithelial cells. Protein expression from three separate proliferating human prostate epithelial cell cultures is shown (Pre-Senescent, Lanes 1–3). These cells were continuously passaged and analyzed at passage 5 after growth arrest (Senescent, Lanes 1–3). Senescence was confirmed by positive SA-β-galactosidase staining (data not shown). p16 is strongly expressed in senescent cells compared to early-passage cells. In contrast, no alterations in expression are found in p53 or p21. Phosphorylated pRb levels are decreased at senescence. HPEC-E7 is used as a positive control because it strongly expresses p53, p21, and p16.

Close modal
Fig. 3.

Phase-contrast microscopy of HPV16 E6 and E7 HPEC lines. The morphology of HPV16 immortal cells was recorded at passage 20 in subconfluent dishes (×200 magnification). A, the morphology of E6 human prostate epithelial cell lines demonstrates irregular shaped, loosely packed cells with poor adherence. E6/E7 cell lines have a similar morphology (data not shown) to E6 lines. B, in contrast, E7 lines show flat, adherent cells with morphology similar to early-passage HPECs.

Fig. 3.

Phase-contrast microscopy of HPV16 E6 and E7 HPEC lines. The morphology of HPV16 immortal cells was recorded at passage 20 in subconfluent dishes (×200 magnification). A, the morphology of E6 human prostate epithelial cell lines demonstrates irregular shaped, loosely packed cells with poor adherence. E6/E7 cell lines have a similar morphology (data not shown) to E6 lines. B, in contrast, E7 lines show flat, adherent cells with morphology similar to early-passage HPECs.

Close modal
Fig. 4.

Demonstration of HPV16 E6 and E7 DNA insertion and gene expression. A, Southern analysis for HPV16 insertions. DNA from the eight cell lines was harvested between passages 18 and 25, digested with HindIII, and probed with a radiolabeled 0.8-kb HPV16 fragment. Results show single bands in some lanes, demonstrating single sites for retroviral insertion and single clonal origin. Multiple bands seen in Lanes 1, 3, and 5 (from left to right) represent multiple viral insertion sites. B, RT-PCR for HPV16 E6 or E7 gene expression in human prostate epithelial cell lines. The HPEC cell lines immortalized with HPV16 E6, E7, or E6/E7 were assessed at passage 18–25 for expression of E6 and E7. Expression of the E6 (347 and 194 bp) and E7 gene products (165 bp) in immortalized HPEC cell sets (HPEC 9, 14, and 15) is shown. Each set is derived from separate single prostate epithelial tissue specimen.

Fig. 4.

Demonstration of HPV16 E6 and E7 DNA insertion and gene expression. A, Southern analysis for HPV16 insertions. DNA from the eight cell lines was harvested between passages 18 and 25, digested with HindIII, and probed with a radiolabeled 0.8-kb HPV16 fragment. Results show single bands in some lanes, demonstrating single sites for retroviral insertion and single clonal origin. Multiple bands seen in Lanes 1, 3, and 5 (from left to right) represent multiple viral insertion sites. B, RT-PCR for HPV16 E6 or E7 gene expression in human prostate epithelial cell lines. The HPEC cell lines immortalized with HPV16 E6, E7, or E6/E7 were assessed at passage 18–25 for expression of E6 and E7. Expression of the E6 (347 and 194 bp) and E7 gene products (165 bp) in immortalized HPEC cell sets (HPEC 9, 14, and 15) is shown. Each set is derived from separate single prostate epithelial tissue specimen.

Close modal
Fig. 5.

Western analysis of p53, pRb, p16, and p21 in HPV16 E6-, E7-, or E6/E7-immortalized prostate epithelial cells and metastatic prostate cancer cell lines. Lanes 1–8 (from left to right), immortalized prostate epithelial cell lines generated by retroviral transfection of HPV16 E7 and/or E6 that inactivate pRb and p53, respectively. p16 expression is uniformly lost in HPV16 E6 cell lines in contrast to E7 cell lines. p16 is not detectable by increasing the length of exposure or protein levels. Immortalized prostate cancer cell lines (Lanes 9–14) from metastatic prostate cancers demonstrate an inverse correlation between p16 and pRb expression. p53 is wild type in cancer cell line LNCaP and shows detectable levels upon longer exposure.

Fig. 5.

Western analysis of p53, pRb, p16, and p21 in HPV16 E6-, E7-, or E6/E7-immortalized prostate epithelial cells and metastatic prostate cancer cell lines. Lanes 1–8 (from left to right), immortalized prostate epithelial cell lines generated by retroviral transfection of HPV16 E7 and/or E6 that inactivate pRb and p53, respectively. p16 expression is uniformly lost in HPV16 E6 cell lines in contrast to E7 cell lines. p16 is not detectable by increasing the length of exposure or protein levels. Immortalized prostate cancer cell lines (Lanes 9–14) from metastatic prostate cancers demonstrate an inverse correlation between p16 and pRb expression. p53 is wild type in cancer cell line LNCaP and shows detectable levels upon longer exposure.

Close modal
Fig. 6.

Southern blot of p16/CDKN2 in HPV16-immortalized prostate epithelial cells and metastatic prostate cancer cell lines. The methylation-sensitive enzyme SmaI and flanking enzyme HindIII demonstrate hypermethylation (6.0 kb) in HPEC E6-9, HPEC E6-14, DuPro, and PPC-1 cell lines. Digestion fragments of 2.1 and 3.9 kb are shown in unmethylated cell lines HPEC E6-15 and E7-9. HPEC E6-15 contains a heterozygous deletion, as evidenced by the reproducible decrease in signal intensity. The other allele of HPEC E6-15 is unmethylated but mutated (see text).

Fig. 6.

Southern blot of p16/CDKN2 in HPV16-immortalized prostate epithelial cells and metastatic prostate cancer cell lines. The methylation-sensitive enzyme SmaI and flanking enzyme HindIII demonstrate hypermethylation (6.0 kb) in HPEC E6-9, HPEC E6-14, DuPro, and PPC-1 cell lines. Digestion fragments of 2.1 and 3.9 kb are shown in unmethylated cell lines HPEC E6-15 and E7-9. HPEC E6-15 contains a heterozygous deletion, as evidenced by the reproducible decrease in signal intensity. The other allele of HPEC E6-15 is unmethylated but mutated (see text).

Close modal
Table 1

Genetic alteration in HPV16 E-6, E-7, and E6/E7-immortalized HPECs by CGH

Cell linePassageaLossesGainsb
HPEC E7-9 26  4, 5, 8, 9, 14, 20c 
HPEC E7-14 21 10p 5q, 8, 11, 20 
HPEC E7-15 16  5, 7, 20 
HPEC E6-9 25  5, 8q22–qter, 16, 20, Y 
HPEC E6-14 14 22,Y 8q21.1–qter, 15q21–qter, 20 
HPEC E6-15 16 8p, 9p 9q31-qter 
HPEC E6/E7-9 24  7, 20,c Yp 
HPEC E6/E7-14 17 9, 20c 
Cell linePassageaLossesGainsb
HPEC E7-9 26  4, 5, 8, 9, 14, 20c 
HPEC E7-14 21 10p 5q, 8, 11, 20 
HPEC E7-15 16  5, 7, 20 
HPEC E6-9 25  5, 8q22–qter, 16, 20, Y 
HPEC E6-14 14 22,Y 8q21.1–qter, 15q21–qter, 20 
HPEC E6-15 16 8p, 9p 9q31-qter 
HPEC E6/E7-9 24  7, 20,c Yp 
HPEC E6/E7-14 17 9, 20c 
a

Denotes total passage number since the initial culture was generated. Immortalized cell lines generally arose between passages 8 and 12; therefore, these cultures represent early passages.

b

Gains at 5q, 8q, and 20 are significant. The probability exceeds 0.95 (in each case) that that region exhibits an elevated rate of gain (44).

c

In these cases, gains of chromosome 20 occurred to a greater degree than in other lines, suggesting possible amplification.

Our special thanks to Robert Huffman (Department of Surgery) and Dr. David Uehling (Division of Urology) of the University of Wisconsin for their support.

1
Hayflick L. The limited in vitro lifespan of human diploid cell strains.
Exp. Cell Res.
,
25
:
585
-621,  
1965
.
2
Newbold R. F., Overell R. W., Connell J. R. Induction of immortality is an early event in malignant transformation of mammalian cells by carcinogens.
Nature (Lond.)
,
299
:
633
-635,  
1982
.
3
Smith J. R., Pereira-Smith O. M. Replicative senescence: implications for in vivo aging and tumor suppression.
Science (Washington DC)
,
273
:
63
-67,  
1996
.
4
Dimri G. P., Campisi J. Molecular and cell biology of replicative senescence.
Cold Spring Harbor Symp. Quant. Biol.
,
59
:
67
-73,  
1994
.
5
Yeager T. R., DeVries S., Jarrard D. F., Kao C., Nakada S. Y., Moon T. D., Bruskewitz R., Stadler W. M., Meisner L. F., Gilchrist K. W., Newton M. A., Waldman F. M., Reznikoff C. A. Overcoming cellular senescence in human cancer pathogenesis.
Genes Dev.
,
12
:
163
-174,  
1998
.
6
Reznikoff C. A., Belair C. D., Yeager T. R., Savelieva E., Blelloch R. H., Puthenveettil J. A., Cuthill S. A Molecular genetic model of human bladder cancer pathogenesis.
Semin. Oncol.
,
23
:
571
-584,  
1996
.
7
Shay J. W., Pereira-Smith O. M., Wright W. E. A role for both RB and p53 in the regulation of human cellular senescence.
Exp. Cell Res.
,
196
:
33
-39,  
1991
.
8
Kao C., Huang J., Wu S. Q., Hauser P., Reznikoff C. A. Role of SV40 T antigen binding to pRB and p53 in multistep transformation in vitro of human uroepithelial cells.
Carcinogenesis (Lond.)
,
14
:
2297
-2302,  
1993
.
9
Reznikoff C. A., Belair C., Savelieva E., Zhai Y., Pfeifer K., Yeager T., Thompson K. J., DeVries S., Bindley C., Newton M. A. Long-term genome stability and minimal genotypic and phenotypic alterations in HPV16 E7-, but not E6-, immortalized human uroepithelial cells.
Genes Dev.
,
8
:
2227
-2240,  
1994
.
10
Kiyono T., Foster S. A., Koop J. L., McDougall J. K., Galloway D. A., Klingelhutz A. J. Both Rb/p16INK4A inactivation and telomerase activity are required to immortalize human epithelial cells.
Nature (Lond.)
,
396
:
84
-88,  
1998
.
11
Alcorta D. A., Xiong Y., Phelps D., Hannon G., Beach D., Barrett J. C. Involvement of the cyclin-dependent kinase inhibitor p16 (INK4a) in replicative senescence of normal human fibroblasts.
Proc. Natl. Acad. Sci. USA
,
93
:
13742
-13747,  
1996
.
12
Reznikoff C. A., Yeager T. R., Belair C. D., Savelieva E., Puthenveettil J. A., Stadler W. M. Elevated p16 at senescence and loss of p16 at immortalization in human papillomavirus 16 E6, but not E7, transformed human uroepithelial cells.
Cancer Res.
,
56
:
2886
-2890,  
1996
.
13
Tahara H., Sato E., Noda A., Ide T. Increase in expression level of p21/cip1/waf1 with increasing division age in both normal and SV40-transformed human fibroblasts.
Oncogene
,
10
:
835
-840,  
1995
.
14
Brooks J. D., Bova G. S., Ewing C. M., Piantadosi S., Carter B. S., Robinson J. C., Epstein J. I., Isaacs W. B. An uncertain role for p53 gene alterations in human prostate cancers.
Cancer Res.
,
56
:
3814
-3822,  
1996
.
15
Cher M. L., Bova G. S., Moore D. H., Small E. J., Carroll P. R., Pin S. S., Epstein J. I., Isaacs W. B., Jensen R. H. Genetic alterations in untreated metastases and androgen-independent prostate cancer detected by comparative genomic hybridization and allelotyping.
Cancer Res.
,
56
:
3091
-3102,  
1996
.
16
Brooks J. D., Bova G. S., Isaacs W. B. Allelic loss of the retinoblastoma gene in primary human prostatic adenocarcinomas.
Prostate
,
26
:
35
-39,  
1995
.
17
Cunningham J. M., Shan A. L., Wick M. J., McDonnell S. K., Schaid D. J., Tester D. J., Qian J. Q., Takahashi S., Jenkins R. B., Bostwick D. G., Thibodeau S. N. Allelic imbalance and microsatellite instability in prostatic adenocarcinoma.
Cancer Res.
,
56
:
4475
-4482,  
1996
.
18
Aprikian A. G., Sarkis A. S., Fair W. R., Zhang Z. F., Fuks Z., Cordon-Cardo C. Immunohistochemical determination of p53 protein nuclear accumulation in prostatic adenocarcinoma.
J. Urol.
,
151
:
1276
-1280,  
1994
.
19
Jarrard D. F., Bova G. S., Ewing C. M., Pin S. S., Nguyen S. H., Herman J. G., Cairns P., Sidransky D., Baylin S. B., Isaacs W. B. Deletional, mutational and methylation analyses of CDKN2 in primary and metastatic prostate cancer.
Genes Chromosomes Cancer
,
19
:
90
-96,  
1997
.
20
Levine A. J. p53, the cellular gatekeeper for growth and division.
Cell
,
88
:
323
-331,  
1997
.
21
Atadja P., Wong H., Garkavtsev I., Veillette C., Riabowol K. Increased activity of p53 in senescing fibroblasts.
Proc. Natl. Acad. Sci. USA
,
92
:
8348
-8352,  
1995
.
22
Hara E., Tsurui H., Shinozaki A., Nakada S., Oda K. Cooperative effect of antisense-Rb and antisense-p53 oligomers on the extension of life span in human diploid fibroblasts, TIG-1.
Biochem. Biophys. Res. Commun.
,
179
:
528
-534,  
1991
.
23
Bond J. A., Blaydes J. P., Rowson J., Haughton M. F., Smith J. R., Wynford-Thomas D., Wyllie F. S. Mutant p53 rescues human diploid cells from senescence without inhibiting the induction of SKI1/WAF1.
Cancer Res.
,
55
:
2404
-2409,  
1995
.
24
Brugarolas J., Chandrasekaran C., Gordon J. I., Beach D., Jacks T., Hannon G. J. Radiation-induced cell cycle arrest compromised by p21 deficiency.
Nature (Lond.)
,
377
:
552
-557,  
1995
.
25
Cristofalo V. J., Pignolo R. J., Cianciarulo F. L., DiPaolo B. R., Rotenberg M. O. Changes in gene expression during senescence in culture.
Exp. Gerontol.
,
27
:
429
-432,  
1992
.
26
Stein G. H., Beeson M., Gordon L. Failure to phosphorylate the retinoblastoma gene product in senescent human fibroblasts.
Science (Washington DC)
,
249
:
666
-669,  
1990
.
27
Serrano M., Hannon G. J., Beach D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4.
Nature (Lond.)
,
366
:
704
-707,  
1993
.
28
Hara E., Smith R., Parry D., Tahara H., Stone S., Peters G. Regulation of p16CDKN2 expression and its implications for cell immortalization and senescence.
Mol. Cell. Biol.
,
16
:
859
-867,  
1996
.
29
Ueki K., Ono Y., Henson J. W., Efird J. T., von Deimling A., Louis D. N. CDKN2/p16 or RB alterations occur in the majority of glioblastomas and are inversely correlated.
Cancer Res.
,
56
:
150
-153,  
1996
.
30
Otterson G. A., Kratzke R. A., Coxon A., Kim Y. W., Kaye F. J. Absence of p16INK4 protein is restricted to the subset of lung cancer lines that retains wildtype RB.
Oncogene
,
9
:
3375
-3378,  
1994
.
31
Lukas J., Parry D., Aagaard L., Mann D. J., Bartkova J., Strauss M., Peters G., Bartek J. Retinoblastoma-protein-dependent cell-cycle inhibition by the tumour suppressor p16.
Nature (Lond.)
,
375
:
503
-506,  
1995
.
32
Ruas M., Peters G. The p16/CDKN2 tumor suppressor gene and its relatives.
Biochim. Biophys. Acta
,
1378
:
F115
-F117,  
1998
.
33
Theodorescu D., Broder S. R., Boyd J. C., Mills S. E., Frierson H. F., Jr. p53, bcl-2 and retinoblastoma proteins as long-term prognostic markers in localized carcinoma of the prostate.
J. Urol.
,
158
:
131
-137,  
1997
.
34
Peehl D. M., Stamey T. A. Serum-free growth of adult human prostatic epithelial cells.
In Vitro Cell. Dev. Biol.
,
22
:
82
-90,  
1986
.
35
Peehl D. M., Wong S. T., Stamey T. A. Clonal growth characteristics of adult human prostatic epithelial cells.
In Vitro Cell. Dev. Biol.
,
24
:
530
-536,  
1988
.
36
Reznikoff C. A., Johnson M. D., Norback D. H., Bryan G. T. Growth and characterization of normal human urothelium in vitro.
In Vitro
,
19
:
326
-343,  
1983
.
37
Umbas R., Schalken J. A., Aalders T. W., Carter B. S., Karthaus H. F., Schaafsma H. E., Debruyne F. M., Isaacs W. B. Expression of the cellular adhesion molecule E-cadherin is reduced or absent in high-grade prostate cancer.
Cancer Res.
,
52
:
5104
-5109,  
1992
.
38
Dimri G. P., Lee X., Basile G., Acosta M., Scott G., Roskelley C., Medrano E. E., Linskens M., Rubelj I., Pereira-Smith O. A biomarker that identifies senescent human cells in culture and in aging skin in vivo.
Proc. Natl. Acad. Sci. USA
,
92
:
9363
-9367,  
1995
.
39
Halbert C. L., Demers G. W., Galloway D. A. The E7 gene of human papillomavirus type 16 is sufficient for immortalization of human epithelial cells.
J. Virol.
,
65
:
473
-478,  
1991
.
40
Merlo A., Herman J. G., Mao L., Lee D. J., Gabrielson E., Burger P. C., Baylin S. B., Sidransky D. 5′ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers.
Nat. Med.
,
1
:
686
-692,  
1995
.
41
Kamb A., Gruis N. A., Weaver-Feldhaus J., Liu Q., Harshman K., Tavtigian S. V., Stockert E., Day R. S., III, Johnson B. E., Skolnick M. H. A cell cycle regulator potentially involved in genesis of many tumor types.
Science (Washington DC)
,
264
:
436
-440,  
1994
.
42
Kallioniemi A., Kallioniemi O. P., Piper J., Tanner M., Stokke T., Chen L., Smith H. S., Pinkel D., Gray J. W., Waldman F. M. Detection and mapping of amplified DNA sequences in breast cancer by comparative genomic hybridization.
Proc. Natl. Acad. Sci. USA
,
91
:
2156
-2160,  
1994
.
43
Hovey R. M., Chu L., Balazs M., DeVries S., Moore D., Sauter G., Carroll P. R., Waldman F. M. Genetic alterations in primary bladder cancers and their metastases.
Cancer Res.
,
58
:
3555
-3560,  
1998
.
44
Newton M. A., Gould M. N., Reznikoff C. A., Haag J. D. On the statistical analysis of allelic-loss data.
Stat. Med.
,
17
:
1425
-1445,  
1998
.
45
Bonkhoff H., Stein U., Remberger K. Multidirectional differentiation in the normal, hyperplastic, and neoplastic human prostate: simultaneous demonstration of cell-specific epithelial markers.
Hum. Pathol.
,
25
:
42
-46,  
1994
.
46
Verhagen A. P., Ramaekers F. C., Aalders T. W., Schaafsma H. E., Debruyne F. M., Schalken J. A. Colocalization of basal and luminal cell-type cytokeratins in human prostate cancer.
Cancer Res.
,
52
:
6182
-6187,  
1992
.
47
Shay J. W., Wright W. E., Brasiskyte D., Van der Haegen B. A. E6 of human papillomavirus type 16 can overcome the M1 stage of immortalization in human mammary epithelial cells but not in human fibroblasts.
Oncogene
,
8
:
1407
-1413,  
1993
.
48
Vousden K. H. Regulation of the cell cycle by viral oncoproteins.
Semin. Cancer Biol.
,
6
:
109
-116,  
1995
.
49
Bookstein R., Rio P., Madreperla S. A., Hong F., Allred C., Grizzle W. E., Lee W. H. Promoter deletion and loss of retinoblastoma gene expression in human prostate carcinoma.
Proc. Natl. Acad. Sci. USA
,
87
:
7762
-7766,  
1990
.
50
Isaacs W. B., Carter B. S., Ewing C. M. Wild-type p53 suppresses growth of human prostate cancer cells containing mutant p53 alleles.
Cancer Res.
,
51
:
4716
-4720,  
1991
.
51
Cairns P., Mao L., Merlo A., Lee D. J., Schwab D., Eby Y., Tokino K., van der Riet P., Blaugrund J. E., Sidransky D. Rates of p16 (MTS1) mutations in primary tumors with 9p loss.
Science (Washington DC)
,
265
:
415
-417,  
1994
.
52
Vocke C. D., Pozzatti R. O., Bostwick D. G., Florence C. D., Jennings S. B., Strup S. E., Duray P. H., Liotta L. A., Emmert-Buck M. R., Linehan W. M. Analysis of 99 microdissected prostate carcinomas reveals a high frequency of allelic loss on chromosome 8p12–21.
Cancer Res.
,
56
:
2411
-2416,  
1996
.
53
Boring C. C., Squires T. S., Tong T., Montgomery S. Cancer statistics, 1994.
CA Cancer J. Clin.
,
44
:
7
-26,  
1994
.
54
Okamoto A., Demetrick D. J., Spillare E. A., Hagiwara K., Hussain S. P., Bennett W. P., Forrester K., Gerwin B., Serrano M., Beach D. H. Mutations and altered expression of p16INK4 in human cancer.
Proc. Natl. Acad. Sci. USA
,
91
:
11045
-11049,  
1994
.
55
Medcalf A. S., Klein-Szanto A. J., Cristofalo V. J. Expression of p21 is not required for senescence of human fibroblasts.
Cancer Res.
,
56
:
4582
-4585,  
1996
.
56
Gonzalgo M. L., Hayashida T., Bender C. M., Pao M. M., Tsai Y. C., Gonzales F. A., Nguyen H. D., Nguyen T. T., Jones P. A. The role of DNA methylation in expression of the p19/p16 locus in human bladder cancer cell lines.
Cancer Res.
,
58
:
1245
-1252,  
1998
.
57
Caldas C., Hahn S. A., da Costa L. T., Redston M. S., Schutte M., Seymour A. B., Weinstein C. L., Hruban R. H., Yeo C. J., Kern S. E. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma.
Nat. Genet.
,
8
:
27
-32,  
1994
.
58
Hussussian C. J., Struewing J. P., Goldstein A. M., Higgins P. A., Ally D. S., Sheahan M. D., Clark W. H., Jr., Tucker M. A., Dracopoli N. C. Germline p16 mutations in familial melanoma.
Nat. Genet.
,
8
:
15
-21,  
1994
.
59
Liu Q., Neuhausen S., McClure M., Frye C., Weaver-Feldhaus J., Gruis N. A., Eddington K., Allalunis-Turner M. J., Skolnick M. H., Fujimura F. K., Kamb A. CDKN2 (MTS1) tumor suppressor gene mutations in human tumor cell lines.
Oncogene
,
11
:
2455
-2461,  
1995
.
60
Nishizaki T., DeVries S., Chew K., Goodson W. H., III, Ljung B. M., Thor A., Waldman F. M. Genetic alterations in primary breast cancers and their metastases: direct comparison using modified comparative genomic hybridization.
Genes Chromosomes Cancer
,
19
:
267
-272,  
1997
.
61
Collins C., Rommens J. M., Kowbel D., Godfrey T., Tanner M., Hwang S. I., Polikoff D., Nonet G., Cochran J., Myambo K., Jay K. E., Froula J., Cloutier T., Kuo W. L., Yaswen P., Dairkee S., Giovanola J., Hutchinson G. B., Isola J., Kallioniemi O. P., Palazzolo M., Martin C., Ericsson C., Pinkel D., Gray J. W. Positional cloning of ZNF217 and NABC1: genes amplified at 20q13.2 and overexpressed in breast carcinoma.
Proc. Natl. Acad. Sci. USA
,
95
:
8703
-8708,  
1998
.
62
Belair C. D., Yeager T. R., Lopez P. M., Reznikoff C. A. Telomerase activity: a biomarker of cell proliferation, not malignant transformation.
Proc. Natl. Acad. Sci. USA
,
94
:
13677
-13682,  
1997
.
63
Wang J., Xie L. Y., Allan S., Beach D., Hannon G. J. Myc activates telomerase.
Genes Dev.
,
12
:
1769
-1774,  
1998
.
64
Ittmann M. M., Wieczorek R. Alterations of the retinoblastoma gene in clinically localized, stage B prostate adenocarcinomas.
Hum. Pathol.
,
27
:
28
-34,  
1996
.
65
Parry D., Bates S., Mann D. J., Peters G. Lack of cyclin D-Cdk complexes in Rb-negative cells correlates with high levels of p16INK4/MTS1 tumour suppressor gene product.
EMBO J.
,
14
:
503
-511,  
1995
.