Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Carcinogenesis ; 22(1): 147-54, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11159753

ABSTRACT

The global genomic repair of DNA adducts was examined in human papillary transitional cell carcinoma (TCC) cell lines after exposure to N:-hydroxy-4-acetylaminobiphenyl (N-OH-AABP), the proximate carcinogenic metabolite of the human bladder carcinogen 4-aminobiphenyl (ABP). (32)P-post-labeling analysis of TCC cultures exposed to N-OH-AABP revealed a major adduct, identified as the 3',5'-bisphosphate derivative of N-(deoxyguanosin-8-yl)-4-aminobiphenyl (dG-C8-ABP). The amount of adduct formation in TCC10 was dependent upon the dose and the duration of exposure and ranged between 1 and 5 adducts/10(7) nucleotides. To test if p53 regulates repair of the dG-C8-ABP adduct in genomic DNA, an isogeneic set of cell lines was obtained by infection of the TCC10 cultures with a retroviral construct expressing a trans-dominant mutant of p53, namely a Val-->Ala mutation at codon 143. The TDM143-TCC10 line expressing the mutant form of p53 was selected. The rate of repair of dG-C8-ABP was compared between TCC10 and TDM143-TCC10 cultures after treatment with 15 microM N-OH-AABP. The rate of disappearance of the adduct was monitored over a period of time after chemical treatment. (32)P-post-labeling analysis of dG-C8-ABP in parental TCC10 showed its rapid removal, the majority of adducts disappearing within 48 h. In contrast to TCC10, TDM143-TCC10 was relatively slower in removal of dG-C8-ABP. After 24 h DNA repair TDM143-TCC10 showed an approximately 3-fold greater amount of dG-C8-ABP compared with TCC10. These results imply that p53 plays a role in the repair of ABP adducts and that in p53 null cells the unrepaired DNA damage could cause accumulation of mutations, which might contribute to increased genomic instability and neoplastic progression.


Subject(s)
Aminobiphenyl Compounds/metabolism , Carcinoma, Transitional Cell/genetics , DNA Adducts/genetics , DNA Repair/genetics , Deoxyguanosine/metabolism , Genes, p53/genetics , Urinary Bladder Neoplasms/genetics , Aminobiphenyl Compounds/pharmacokinetics , Aminobiphenyl Compounds/toxicity , Carcinogens/metabolism , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Carcinoma, Transitional Cell/metabolism , DNA Adducts/biosynthesis , DNA Damage/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/pharmacokinetics , Deoxyguanosine/toxicity , Genotype , Humans , Mutagenesis/genetics , Mutation , Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Urinary Bladder Neoplasms/metabolism
2.
Cancer Res ; 60(14): 3862-71, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919661

ABSTRACT

Inactivation of both the pRb (pRb-cyclin D1/cyclin-dependent kinase 4/6-p16) and p53 (p53-p21(WAF1)-p14(ARF)) pathways is thought to be essential for immortalization in vitro and malignant transformation in vivo. We identified different combinations of pRb and p53 pathway alterations in 12 invasive transitional cell carcinomas (TCCs) and addressed the functional significance of the different combinations observed. Results showed four combinations of alterations including -pRb/-p53 (ie., pRb inactivated in the pRb pathway and p53 inactivated in the p53 pathway; four TCCs), -p16/-p53 (four TCCs), -p16/-p21(WAF1) (one TCC), and -p16/ -p14(ARF) (two TCCs). These groups include two new combinations (ie., -p16/-p53 and -p16/-p21(WAF1)) not reported previously for TCCs. An alteration in the key components of the p53 pathway was not detected in one invasive TCC that had inactivated p16. Note that all four TCCs with inactivated pRb had mutant p53; thus, the combinations of -pRb/ -p21(WAF1) and -pRb/-p14(ARF) were not observed. Only two of eight TCCs with altered p16 had concomitant p14(ARF) loss, demonstrating that simultaneous inactivation of these two 9p21INK4a tumor suppressor genes is not obligatory. To determine the biological phenotypes of TCCs with different combinations of pRb and p53 pathway alterations, their downstream responses to gamma radiation were studied in vitro. As expected, none of eight TCCs with mutant p53 responded to gamma radiation by elevation of p53, p21(WAF1), or mdm2 or by cell cycle arrest. Only two of four TCCs with wild-type p53 and wild-type pRb (the combination of -p16/-p14(ARF)) showed normal downstream responses to gamma radiation and underwent cell cycle arrest. Two TCCs with wild-type pRb and wild-type p53 (the combination of -pl6/-p21(WAF1) and one TCC with -p16) failed to show cell cycle arrest in response to radiation. This was attributed to the absence of p21(WAF1) in one TCC. In summary, these data support a model of invasive bladder cancer pathogenesis in which both the pRb and p53 pathways are usually inactivated and the biology of the tumor is impacted by the mechanism of their inactivations.


Subject(s)
Genes, p53/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Urinary Bladder Neoplasms/genetics , Blotting, Northern , Blotting, Southern , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Carrier Proteins/metabolism , Chromosomes, Human, Pair 9/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Neoplasm Invasiveness/genetics , Phenotype , Proteins/metabolism , Sequence Analysis, DNA , Tumor Suppressor Protein p14ARF , Urinary Bladder Neoplasms/metabolism
3.
Adv Exp Med Biol ; 462: 83-91, 1999.
Article in English | MEDLINE | ID: mdl-10599415

ABSTRACT

PURPOSE: Normal human uroepithelial cells (HUCs) proliferate rapidly in culture during early passage and then spontaneously undergo replicative senescence. We previously reported that the cyclin D1-CDK4/6 inhibitor, p16INK4a, is elevated at senescence in HUCs. Hence, we proposed that p16INK4a may play a critical role in mediating senescence in this cell type. In the current study, we further characterized the senescent state in HUCs. We also tested the possible roles of changes in other cell cycle proteins, including p53, p21WAF1, pRb, and cyclin D1 in HUC senescence. METHODS: Normal HUCs cultured from explants of ureteral mucosa were used for these studies. Senescence associated-beta-galactosidase activity (SA-beta-gal) was used to identify cells in senescence. Flow cytometric analysis was used to determine changes in cell cycle distribution at senescence. Response of cells to serum stimulation was determined by Northern analysis of c-fos. Western analysis was used to assess changes in p53, p21WAF, p16INK4a, cyclin D1 and plasminogen activator inhibitor-1 (PAI-1) levels at senescence. RESULTS: beta-gal-positive HUCs were blocked at G1/S in senescence and failed to show c-fos induction in response to serum stimulation. As previously reported, senescent HUCs also showed elevated p16INK4a. However, unlike human fibroblasts, neither p53 nor p21WAF1 elevation accompanied HUCs senescence. PAI-1 levels were also not elevated in HUC senescence. CONCLUSION: These findings support a model in which elevation of p16INK4a, but not p53 or p21WAF1 plays a critical role in HUC replicative senescence. These findings elucidate the tumor suppressor mechanism of p16INK4a and the frequent loss of either p16INK4a or pRb in invasive human bladder tumors.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Senescence/physiology , Urothelium/cytology , Cell Division , Cells, Cultured , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Inhibitors/metabolism , Humans , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Urothelium/physiology
4.
Genes Chromosomes Cancer ; 26(4): 304-11, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10534765

ABSTRACT

Gain of 20q has been observed in many cancer types, including bladder cancers. However, the biological significance of low-copy-number 20q gain in human cancer pathogenesis has not yet been defined. We reported that immortalization of human uroepithelial cells (HUC) transformed with human papillomavirus 16 (HPV 16) E7 is associated with single-copy 20q gain (P = 2 x 10(-7)). We also observed 20q13.2 amplification in some cell lines, but only after 20 passages. Thus, we hypothesized that low-copy gain of 20q gene(s) contributes in a dominant way to bypassing HUC senescence. To test this hypothesis, we fused precrisis E7-transformed HUCs (pcE7s) with three independent immortal E7-HUCs that acquired a single-copy 20q gain at immortalization. In one of these lines, a single-copy gain of 20q and a 10p12.1-pter loss were the only cytogenetic alterations. Immortal cell hybrids were obtained with all three crosses. Southern analysis for unique HPV16 insertion sites, as well as fluorescence in situ hybridization (FISH) with whole chromosome 20 painting probes (WCP20) for marker chromosomes in the immortal clones, confirmed the hybrid and independent nature of representative immortal clones. In contrast, when we used the same protocol, no immortal somatic cell hybrids were obtained when HPV16 E6 immortal HUC (E6-HUC) that showed 3p and 9p losses, but no 20q gain, were fused with precrisis E6-transformed HUC (pcE6s). This latter observation is consistent with many results demonstrating that recessive changes are required for cell immortalization. Therefore, the new results reported herein for the first time demonstrate that dominant changes can contribute to bypassing senescence, and that such genes may be located on 20q. Genes Chromosomes Cancer 26:304-311, 1999.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/genetics , Chromosomes, Human, Pair 20/genetics , Genes, Dominant/genetics , Cells, Cultured , Cellular Senescence/genetics , Epithelial Cells , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Ureter , Virus Integration/genetics
5.
Cancer Res ; 59(12): 2957-64, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10383161

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epithelial Cells/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Repressor Proteins , Retinoblastoma Protein/metabolism , Aged , Cellular Senescence , Chromosome Aberrations , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA Methylation , Epithelial Cells/pathology , Gene Deletion , Humans , Male , Middle Aged , Mutation , Neoplasm Metastasis , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Prostate/pathology , Prostatic Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
6.
Stat Med ; 17(13): 1425-45, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9695190

ABSTRACT

This paper concerns the statistical analysis of certain binary data arising in molecular studies of cancer. In allelic-loss experiments, tumour cell genomes are analysed at informative molecular marker loci to identify deleted chromosomal regions. The resulting binary data are used to infer properties of putative suppressor genes, genes involved in normal cell cycling. Various factors can complicate this inference, including background loss of heterozygosity, spatial (that is, within chromosome) dependence of the binary responses, non-informativeness of markers, covariates such as protein levels or tumour histology, heterogeneity of cells within tumours, and measurement error. We focus on the first three factors, discussing methods for statistical inference that separate background loss from significant loss. We outline the extension to other inferences, such as comparison questions and the relationship to covariates. Using characteristic features of tumourigenesis, we present a framework for the stochastic modelling of allelic-loss data, and build models within this framework; in particular, we propose a simple model that has chromosome breaks at locations of a Poisson process, and preferential selection cells with inactivated suppressor genes. We illustrate these methods on allelic-loss data from induced rat mammary tumours and human bladder cancers.


Subject(s)
Data Interpretation, Statistical , Loss of Heterozygosity , Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/genetics , Chromosome Breakage , Chromosome Deletion , Female , Genes, Tumor Suppressor/genetics , Genetic Markers/genetics , Humans , Mammary Neoplasms, Experimental/genetics , Poisson Distribution , Rats , Stochastic Processes , Urinary Bladder Neoplasms/genetics
7.
J Urol ; 159(2): 581-5, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9649296

ABSTRACT

PURPOSE: Bladder cancers are frequently treated with combination chemotherapy that includes methotrexate (MTX). The development of drug resistance is a common problem in treatment of bladder cancers. We tested if the status of p53 and/or pRb affects the development of MTX resistance in bladder epithelial cell lines. MATERIALS AND METHODS: We used two isogeneic sets of cell lines in which we manipulated the status of p53 and/or pRb by transformation with Human Papillomavirus (HPV) E6 and/or E7 or with a transdominant TP53 mutant (TP53(143)). One series of isogeneic origin was derived from normal human uroepithelial cells (HUC), and the other was derived from a human transitional cell carcinoma (TCC). Cell lines with p53 and/or pRb alterations were cultured for six months while increasing the MTX concentration in each line, as resistance developed. RESULTS: Two cell lines with both pRb and p53 alterations, alphaE6/E7-HUC and alphaE7-HUCp53mu, acquired the greatest resistance (750 nM) to MTX. One line with p53 loss, E6-TCC#10, acquired intermediate resistance (500 nM), while two lines, alphaE7-HUC and E7-TCC#10, with altered pRb but wildtype p53, showed low levels of MTX resistance (125 nM and 80 nM, respectively). Two clear mechanisms of MTX resistance were identified. All five MTX resistant cell lines showed altered uptake of MTX. In addition, two of five MTX resistant cell lines, both with altered p53, showed dihydrofolate reductase (DHFR) amplification. CONCLUSIONS: p53 alteration increases the risk for development of drug resistance by both DHFR amplification and altered MTX transport in transformed human bladder epithelial cell lines.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Genes, p53/genetics , Methotrexate/pharmacology , Urothelium/drug effects , Drug Resistance , Genes, Retinoblastoma/genetics , Humans
8.
Genes Dev ; 12(2): 163-74, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9436977

ABSTRACT

Elevation of p16, the CDKN2/p16 tumor suppressor gene (TSG) product, occurs at senescence in normal human uroepithelial cells (HUC). Immortal HUCs and bladder cancer cell lines show either alteration of p16 or pRb, the product of the retinoblastoma (RB) TSG. In addition, many human cancers show p16 or pRb alteration along with other genetic alterations that we associated with immortalization, including +20q and -3p. These observations led us to hypothesize that p16 elevation plays a critical role in senescence cell cycle arrest and that overcoming this block is an important step in tumorigenesis in vivo, as well as immortalization in vitro. Using a novel approach, we tested these hypotheses in the present study by examining p16 and pRb status in primary culture (P0) and after passage in vitro of transitional cell carcinoma (TCC) biopsies that represented both superficial bladder tumors and invasive bladder cancers. We demonstrated that all superficial TCCs showed elevated p16 after limited passage in vitro and then senesced, like normal HUCs. In contrast, all muscle invasive TCCs contained either a p16 or a pRb alteration at P0 and all spontaneously bypassed senescence (P = 0.001). Comparative genomic hybridization (CGH) was used to identify regions of chromosome loss or gain in all TCC samples. The application of a statistical model to the CGH data showed a high probability of elevated alteration rates of +20q11-q12 (0.99) and +8p22-pter (0.94) in the immortal muscle invasive TCCs, and of -9q (0.99) in the superficial TCCs. Three myoinvasive TCCs lost 3p13-p14. In this study, four of six myoinvasive TCCs also showed TP53 mutation that associated well with genome instability (P = 0.001), as previously hypothesized. Notably, TP53 mutation, which has been used as a marker of tumor progression in many human cancers, was less significant in associating with progression in this study (P = 0.04) than was p16 or pRb alteration (P = 0.001). Thus, these data support a new model in which overcoming senescence plays a critical role in human cancer pathogenesis and requires at least two genetic changes that occur in several combinations that can include either p16 or pRb loss and at least one additional alteration, such as +20q11-q12, -3p13-p14, or -8p21-pter.


Subject(s)
Carcinoma, Transitional Cell/metabolism , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Neoplasm Invasiveness/genetics , Retinoblastoma/metabolism , Urinary Bladder Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Southern , Blotting, Western , Carcinoma, Transitional Cell/genetics , Chromosome Aberrations , Chromosome Disorders , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epithelium/metabolism , Female , Humans , Male , Middle Aged , Mutation , Retinoblastoma/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Urinary Bladder Neoplasms/genetics
9.
Genes Chromosomes Cancer ; 21(1): 39-48, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9443040

ABSTRACT

Immortalization and tumorigenic transformation of many human cell types, including human uroepithelial cells (HUCs), are frequently associated with loss of genetic material from the short arm of chromosome 3 (3p). In addition, losses of 3p have been observed in many human cancers including renal cell carcinoma, lung cancer, breast cancer, and bladder cancer. Genetic studies suggest that there are at least two regions on 3p in which tumor suppressor genes might be located, but the precise location of these genes is not known. We studied chromosome 3 losses that were specifically associated with immortalization of five independent human papilloma virus 16 (HPV16) E6- or E7-transformed HUCs. Cytogenetic analysis showed that the smallest common region of deletion was 3p14.1-->14.2. Fluorescence in situ hybridization using a 3p13-->14-specific yeast artificial chromosome (YAC) contig showed the precise localization of the breakpoints to be in 3p13 and 3p14.2, thus defining the smallest common overlap of 3p deletions in HPV16 E6- or E7-immortalized HUCs. These results suggest the presence in this region of genes involved in the control of senescence in vitro and possibly tumorigenesis in vivo.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 3/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Repressor Proteins , Ureter/cytology , Cell Line, Transformed , Cell Transformation, Viral , Chromosomes, Artificial, Yeast , Culture Techniques , Genetic Markers , Humans , Karyotyping , Nucleic Acid Hybridization , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins
10.
Int J Cancer ; 70(4): 423-9, 1997 Feb 07.
Article in English | MEDLINE | ID: mdl-9033650

ABSTRACT

Actin, a highly conserved protein comprising cell stress fibers and other cellular structures, is found in both the cytoplasm and nucleus of cells and responds to both epigenetic signals and altered gene expression occurring during tumorigenesis. We have previously shown that changes in the cytoplasmic F- and G-actin ratios reflect bladder cancer risk. To determine whether nuclear actin is also altered and how nuclear and cytoplasmic actin alterations are interrelated in transformation, an in vitro model of carcinogen-induced transformation consisting of 2 human uroepithelial cell lines immortalized by infection with SV-40 was studied. One line, HUC-PC, is tumorigenic in nude mice after incubation with the carcinogen 4-ABP, the other, HUC-BC, is not. Cytoplasmic and nuclear F- and G-actin were determined by QFIA on individual cells using fluorochrome-labeled phallicidin and DNase, I, respectively. Before exposure to 4-ABP, the PC cells had lower cytoplasmic F-actin content, higher cytoplasmic G-actin content, but similar levels of nuclear G- and F-actin in comparison to the BC cells. After incubation with 4-ABP, F-actin decreased and G-actin increased in both cytoplasm and nuclei of PC cells and cytoplasmic F-actin fibers were lost, but only cytoplasmic actin was altered in the BC cells. Northern blot analysis showed the expression of the beta-actin gene was only approximately 20% lower in 4-ABP-treated PC cells than in untreated controls, indicating the cellular change in actin was attributed to a shift between F- and G-actin proteins rather than to net actin synthesis.


Subject(s)
Actins/metabolism , Cell Transformation, Neoplastic/metabolism , Cytoplasm/metabolism , Nuclear Proteins/metabolism , Actins/genetics , Aminobiphenyl Compounds , Blotting, Northern , Carcinogens , Cell Line, Transformed/drug effects , Cell Transformation, Neoplastic/chemically induced , Dimethyl Sulfoxide , HL-60 Cells , Humans , Nuclear Proteins/genetics , RNA, Messenger/metabolism
11.
Oncogene ; 14(5): 551-60, 1997 Feb 06.
Article in English | MEDLINE | ID: mdl-9053853

ABSTRACT

Breast, bladder, colon, and ovarian carcinomas show frequent low level 20q gain and less frequently high level 20q13.2 amplification, but the significance of these 20q amplifications in transformation has not been defined. Using karyotypic and comparative genomic hybridization (CGH) analyses, chromosome losses and gains were analysed in six newly immortalized human uroepithelial cell (HUC) lines transformed by Human Papillomavirus 16 (HPV16) E7. Results showed clonal chromosomes with 20q11->qter gain in all six lines. CGH revealed a peak of 20q13.2 amplification in two cell lines. FISH with whole chromosome 20 paint showed expanded chromosome regions (ECRs) and double minute chromosomes (DMs) that contained chromosome 20 material in cell lines with 20q13.2 amplification. FISH with probes from the center of the 20q13.2 human breast cancer amplicon showed as many as 24 signals in cells with 20q13.2 amplification. The acquisition of genome instability in these E7-HUCs did not correlate with TP53 mutation, as all E7-HUCs contained only wildtype TP53. These results suggest that low level 20q gain is associated with overcoming cellular senescence in E7 transformed cells (P-value=2 x 10(-7)), but does not confer genome instability, while high level 20q13.2 amplification is associated with chromosome instability. Loss of 10p (P-value = 3 x 10(-5)) was also important in immortalization of E7-transformed HUCs. Thus, these results have profound implications for interpreting the significance of high versus low level 20q gains in human cancers.


Subject(s)
Cell Transformation, Neoplastic , Chromosome Aberrations , Chromosome Disorders , Chromosomes, Human, Pair 20 , Gene Amplification , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Breast Neoplasms/genetics , Cell Line , Chromosome Mapping , Colonic Neoplasms/genetics , Female , Genes, p53 , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Oncogene Proteins, Viral/biosynthesis , Ovarian Neoplasms/genetics , Papillomavirus E7 Proteins , Urinary Bladder Neoplasms/genetics , Urothelium
12.
Proc Natl Acad Sci U S A ; 94(25): 13677-82, 1997 Dec 09.
Article in English | MEDLINE | ID: mdl-9391085

ABSTRACT

Telomerase activity is readily detected in most cancer biopsies, but not in premalignant lesions or in normal tissue samples with a few exceptions that include germ cells and hemopoietic stem cells. Telomerase activity may, therefore, be a useful biomarker for diagnosis of malignancies and a target for inactivation in chemotherapy or gene therapy. These observations have led to the hypothesis that activation of telomerase may be an important step in tumorigenesis. To test this hypothesis, we studied telomerase activity in isogeneic samples of uncultured and cultured specimens of normal human uroepithelial cells (HUCs) and in uncultured and cultured biopsies of superficial and myoinvasive transitional cell carcinoma (TCC) of the bladder. Our results demonstrated that four of four TCC biopsies, representing both superficial and myoinvasive TCCs, were positive for telomerase activity, but all samples of uncultured HUC were telomerase negative. However, when the same normal HUC samples were established as proliferating cultures in vitro, telomerase activity was readily detected but usually at lower levels than in TCCs. Consistent with the above observation of the telomerase activity in HUCs, telomeres did not shorten during the HUC in vitro lifespan. Demonstration of telomerase in proliferating human epithelial cells in vitro was not restricted to HUCs, because it was also present in prostate and mammary cell cultures. Notably, telomerase activity was relatively low or undetectable in nonproliferating HUC cultures. These data do not support a model in which telomerase is inactive in normal cells and activated during tumorigenic transformation. Rather, these data support a model in which the detection of telomerase in TCC biopsies, but not uncultured HUC samples, reflects differences in proliferation between tumor and normal cells in vivo.


Subject(s)
Cell Division/physiology , Cell Transformation, Neoplastic/metabolism , Telomerase/metabolism , Base Sequence , Biomarkers , Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/enzymology , Carcinoma, Transitional Cell/pathology , Cells, Cultured , DNA Primers/genetics , Enzyme Activation , Female , Humans , Male , Models, Biological , Substrate Specificity , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/pathology , Urothelium/cytology , Urothelium/enzymology
13.
Cancer Res ; 56(20): 4649-53, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8840979

ABSTRACT

Chronic infection/inflammation of the urinary tract is a significant risk factor for the development of bladder cancer. The present study examined the hypothesis that hydrogen peroxide (H202) and cytokines released during inflammation are involved in the enhancement of bladder carcinogenesis. Using growth in soft agar and tumorigenicity in athymic nude mice as indices of transformation, we examined the effect of H202 and cytokines on the enhancement of N-methyl-N-nitrosourea (MNU)-initiated transformation of MYP3 cells, an anchorage-dependent nontumorigenic rat bladder epithelial cell line. MYP3 cells pretreated with or without MNU were exposed to H202 (0.001 to 0.1 mM) daily for 1 week in monolayer culture and were then tested for growth in soft agar. A marked increase in colony numbers was observed in the cells that were MNU-initiated and exposed to H202 (P < 0.01). Furthermore, H202 exposure alone at 0.01 mM or 0.1 mM caused colony formation in soft agar. The transformants induced by MNU plus H202 or H202 alone formed high-grade transitional cell carcinomas when injected into nude mice. The growth of these transformants was stimulated by several cytokines (interleukin 1alpha, interleukin 6, and tumor necrosis factor-alpha) better than the parental cells both on a plastic surface and in soft agar. Our results indicate that H202 causes genetic change(s) to induce tumorigenic conversion in urothelial cells and that the transformants are stimulated to grow because of their selective response to several cytokines. We suggest that these mechanisms may be involved in the in vivo carcinogenesis associated with chronic urinary tract infection.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Cytokines/toxicity , Hydrogen Peroxide/toxicity , Urinary Bladder Neoplasms/chemically induced , Animals , Carcinoma, Transitional Cell/chemically induced , Cell Division/drug effects , Cystitis/chemically induced , Cystitis/complications , Male , Methylnitrosourea , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Rats , Tumor Cells, Cultured , Tumor Stem Cell Assay
14.
Semin Oncol ; 23(5): 571-84, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8893868

ABSTRACT

An understanding of the biological significance of the multiple genetic alterations identified in clinical bladder cancers to the stepwise pathogenesis of the disease is evolving. Alterations in p53 and pRb, products of the chromosomes 17p13 TP53 and 13q14 RB tumor suppressor genes, occur in approximately 50% and approximately 33% of bladder cancers respectively, and are associated with later stage, higher grade disease. p53 and pRb alterations are also known to occur in early stage bladder carcinoma in situ where they are thought to represent a poor prognosis for tumor progression. Allelic loss of genes on 9p21 occurs in approximately 50% of bladder cancers, but whether the only critical gene in this region is the CDKN2/p16 cyclin/CDK inhibitor is at present uncertain. Amplification and/or overexpression of the oncogenes epidermal growth factor receptor and erbB2 are associated with later stage disease. Finally, recent findings generated using in vitro transformation systems with human uroepithelial cells provide strong evidence that loss of genes on 3p, which occurs in approximately 20% of bladder cancers, and/or gain of genes on 20q play an important role in blocking HUC cellular senescence. This latter phenotype should represent a critical step in oncogenesis, as cells that do not senesce can survive to accumulate the multiple genetic alterations associated with invasive and metastatic bladder cancers. Further understanding of the biochemical mechanisms underlying these genetic changes will provide the additional information needed to design better strategies for bladder cancer intervention and treatment.


Subject(s)
Urinary Bladder Neoplasms/genetics , Chromosome Aberrations , Genes, p53 , Humans , Models, Genetic , Urinary Bladder Neoplasms/etiology
15.
Oncogene ; 13(6): 1123-31, 1996 Sep 19.
Article in English | MEDLINE | ID: mdl-8808685

ABSTRACT

We compared the ability of E6-, versus E7-, immortalized human uroepithelial cells (HUC) to undergo apoptosis in response to gamma radiation. Two independent HPV16 E6-immortalized cell lines, alphaE6#1 and alphaE6#2, that showed low or undetectable p53 levels, failed to undergo apoptosis in response to 18 Gray (Gy) gamma radiation as determined by DNA fragmentation. In contrast, two independent HPV16 E7-immortalized cell lines, alphaE7#1 and alphaE7#2, both of which showed stabilized wildtype p53, underwent apoptosis in the same experiment. Interestingly, both alphaE7#1 and alphaE7#2 showed constitutively elevated BAX and lowered BCL-2 levels, compared to either alphaE6#1 or alphaE6#2. However, elevated BAX and reduced BCL-2 per se were insufficient to trigger apoptosis, as apoptosis occurred only after exposure to gamma radiation. These results support a model in which HPV16 E7-immortalized cells are primed to undergo apoptosis, given an appropriate trigger. This apoptotic response was not observed in alphaE6/E7#1 cells which, like alphaE6-HUCs, showed low p53 levels, nor in late passage alphaE7#1 with spontaneously mutated TP53. These results suggest that E7 immortalization primes HUC for apoptosis in response to gamma radiation, and that this enhanced apoptotic response is p53 dependent.


Subject(s)
Apoptosis/physiology , Cell Transformation, Viral , Oncogene Proteins, Viral/pharmacology , Papillomaviridae , Repressor Proteins , Urinary Tract/cytology , Urinary Tract/virology , Apoptosis/radiation effects , Cell Cycle/radiation effects , Cell Line, Transformed , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Epithelial Cells , Epithelium/radiation effects , Epithelium/virology , Humans , Mutation , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Urinary Tract/radiation effects , bcl-2-Associated X Protein
16.
Cancer Res ; 56(13): 2886-90, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8674033

ABSTRACT

CDKN2/p16 inhibits the cyclin D/cyclin-dependent kinase complexes that phosphorylate pRb, thus blocking cell cycle progression. We previously reported that p16 levels are low to undetectable in normal human uroepithelial cells (HUCs) and in immortalized uroepithelial cells with functional pRb, whereas p16 levels are markedly elevated in immortal HUCs with altered pRb (T. Yeager et al., Cancer Res., 55: 493-497, 1995). We now report that elevation of p16 levels occurs at senescence in HUCs, including HUCs transformed by human papillomavirus 16 E7 or E6, whose oncoprotein products lead to functional loss of pRb and p53, respectively. We also report that six of six independently immortalized E7 HUCs show high levels of p16 similar to those observed at HUC senescence, whereas p16 is undetectable in five of five immortal E6 HUCs. Four of the five independent E6 HUCs that lost p16 at immortalization showed hemizygous deletion of the 9p21 region. However, no homozygous CDKN2 deletions were detected, and only one CDKN2 mutation was identified. For the first time, these data associate elevated p16 with senescence in human epithelial cells. These data also suggest that a component of immortalization may be abrogation, either by pRb inactivation (as in the E7-transformed HUCs) or by p16 inactivation (as in the E6-transformed HUCs), of a p16-mediated senescence cell cycle block.


Subject(s)
Carrier Proteins/metabolism , Cell Transformation, Viral/physiology , Oncogene Proteins, Viral/physiology , Papillomaviridae/physiology , Repressor Proteins , Carrier Proteins/genetics , Cell Cycle/physiology , Cell Cycle/radiation effects , Cells, Cultured , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Chromosomes, Human, Pair 9 , Cyclin-Dependent Kinase Inhibitor p16 , Gene Deletion , Humans , Mutation , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Retinoblastoma Protein/metabolism , Urinary Tract/cytology
17.
Carcinogenesis ; 17(4): 857-64, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8625501

ABSTRACT

The tumorigenic transformation of certain occupationally significant chemicals, such as N-hydroxy-4-4'-methylenebis[2-chloroaniline] (N-OH-MOCA), N-hydroxy-ortho-toluidine (N-OH-OT), 2-phenyl-1,4-benzoquinone (PBQ) and N-hydroxy-4-aminobiphenyl (N-OH-ABP) were tested in vitro using the well established SV40-immortalized human uroepithelial cell line SV-HUC.PC. SV-HUC cells were exposed in vitro to varying concentrations of N-OH-MOCA, N-OH-OT, N-OH-ABP and PBQ that caused approximately 25% and 75% cytotoxicity. The carcinogen treated cells were propagated in culture for about six weeks and subsequently injected subcutaneously into athymic nude mice. Two of the fourteen different groups of SV-HUC.PC treated with different concentrations of N-OH-MOCA, and one of the three groups exposed to N-OH-ABP, formed carcinomas in athymic nude mice. 32P-postlabeling analyses of DNA isolated from SV-HUC.PC after exposure to N-OH-MOCA revealed one major and one minor adduct. The major adduct has been identified as the N-(deoxyadenosin-3',5'-bisphospho-8-yl)-4-amino-3-chlorob enz yl alcohol (pdAp-ACBA) and the minor adduct as N-(deoxyadenosin-3',5'-bisphospho-8-yl)-4-amino-3-chlorot oluene (pdApACT). Furthermore, SV-HUC.PC cytosols catalyzed the binding of N-OH-MOCA to DNA, in the presence of acetyl-CoA, to yield similar adducts. The same adducts were also formed by chemical interaction of N-OH-MOCA with calf thymus DNA, suggesting that the aryl nitrenium ion may be the ultimate reactive species responsible for DNA binding. The tumorigenic activity of N-OH-MOCA in this highly relevant in vitro transformation model, coupled with the findings that SV-HUC.PC cells formed DNA-adducts in vitro and contained enzyme systems that activated N-OH-MOCA to reactive electrophilic species that bound to DNA, strongly suggest that MOCA could be a human bladder carcinogen. These findings are consistent with the International Agency for Research on Cancer's classification of MOCA as a probable human carcinogen.


Subject(s)
Carcinogens/toxicity , DNA/drug effects , Methylenebis(chloroaniline)/toxicity , Urogenital System/drug effects , Animals , Binding Sites , Carcinogens/metabolism , Cell Line, Transformed , Cell Transformation, Neoplastic , DNA/metabolism , Female , Humans , Methylenebis(chloroaniline)/metabolism , Mice , Mice, Nude , Simian virus 40/physiology , Urinary Bladder Neoplasms/chemically induced , Urogenital System/cytology
18.
Genes Dev ; 8(18): 2227-40, 1994 Sep 15.
Article in English | MEDLINE | ID: mdl-7958891

ABSTRACT

Parameters of genome instability and morphological alterations associated with cell transformation were studied in an isogeneic set of clonal human uroepithelial cell (HUC) lines immortalized by the human papilloma virus 16 (HPV16) E6 and/or E7 gene(s). HPV16 E6 binds p53, leading to rapid degradation of p53, whereas E7 binds and alters pRb and other proteins. We report that two independent E7-immortalized HUC lines showed minimal phenotypic or genotypic alterations, except that both lines contained amplification of 20q DNA sequences and a greater polyploidization at an early passage. The E7-immortalized HUC line resembled normal HUC lines, except that they failed to senesce. In contrast, the E6-immortalized HUC lines were morphologically altered, contained numerous random chromosome aberrations, and showed unstable evolving karyotypes with passage in culture. No amplified DNA sequences were detected in E6-immortalized HUC lines. Instead, clonal losses of chromosome regions (i.e., -3p, -6q, -9p), putatively containing tumor suppressor or senescence genes, accompanied the E6-HUC immortalization event. E6-immortalized HUC lines showed transformed phenotypes similar to E6/E7-HUC lines. The difference in genome stability between E6- and E7-immortalized HUC was highly significant statistically (p-value < 10(-6). Thus, the HPV16 E7 gene led to HUC immortalization by a pathway that blocked cellular senescence, but did not disrupt genome stability. These results implicate p53 loss, but not pRb alteration, in genome destabilization.


Subject(s)
Cell Transformation, Viral/genetics , Papillomaviridae/genetics , Base Sequence , Cell Line , DNA Primers/genetics , DNA, Viral/genetics , Epithelium , Genotype , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Models, Biological , Molecular Sequence Data , Papillomaviridae/classification , Phenotype , Urinary Bladder
19.
Stat Med ; 13(8): 839-58, 1994 Apr 30.
Article in English | MEDLINE | ID: mdl-8047739

ABSTRACT

Cytogenetic analysis reveals alterations of chromosome structure (losses, gains, and rearrangements of genetic material) in bladder cancer cells generated using an in vitro/in vivo transformation system. To predict possible locations of bladder cancer suppressor genes, we performed a robust Bayesian analysis of the chromosome-loss data. We postulated a simple stochastic model to describe chromosome loss during tumour progression. Posterior computations are enabled by a dynamic simulation algorithm. Ordered by decreasing posterior probability of putatively harbouring a suppressor gene, we observe significant losses on chromosomes 3, 18, 13, 10, 11, and y.


Subject(s)
Bayes Theorem , Carcinoma, Transitional Cell/genetics , Cell Transformation, Neoplastic/genetics , Chromosome Deletion , Genes, Tumor Suppressor/genetics , Models, Genetic , Urinary Bladder Neoplasms/genetics , Algorithms , Cell Line, Transformed , Chromosome Mapping/statistics & numerical data , Humans , Karyotyping , Mathematical Computing , Probability
20.
Radiat Res ; 138(1): 86-92, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8146304

ABSTRACT

Recent interest has focused on the identification of molecular genetic mechanisms in multistep neoplastic transformation. In vitro exposure of simian virus 40 (SV40)-immortalized human uroepithelial cells (SV-HUC) that are environmentally relevant to bladder carcinogens has been shown to produce tumorigenic transformation, as assessed by the ability of cells exposed to a carcinogen to form xenograph tumors with heterogeneous cancer phenotypes ranging from very aggressive, invasive high-grade carcinomas to superficial low-grade indolent tumors. In addition, exposure of a low-grade indolent tumor generated in the SV-HUC system, MC-T11, to the same carcinogens results in neoplastic progression as assessed by the production of high-grade aggressive cancers. In the present study, we show neoplastic progression of MC-T11 after in vitro exposure to a single dose of 6 Gy X rays. In addition, we show that the chromosome deletions, including losses of 4q, 11p, 13q and 18, observed in these radiation-induced tumors are similar to those observed in carcinogen-induced tumors, thus supporting the hypothesis that the experimental cell system, not the transforming agent, dictates the genetic losses required for tumorigenic transformation and progression.


Subject(s)
Cell Transformation, Neoplastic/radiation effects , Chromosome Deletion , Chromosomes, Human/radiation effects , Urinary Bladder Neoplasms/pathology , Animals , Cell Division/radiation effects , Cell Line , Cell Line, Transformed , Chromosomes, Human, Pair 11/radiation effects , Chromosomes, Human, Pair 13/radiation effects , Chromosomes, Human, Pair 18/radiation effects , Chromosomes, Human, Pair 4/radiation effects , Epithelium/radiation effects , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Kinetics , Mice , Mice, Nude , Simian virus 40/genetics , Time Factors , Transfection , Transplantation, Heterologous , Urinary Bladder/cytology , Urinary Bladder/radiation effects , Urinary Bladder Neoplasms/genetics , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...