Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Eur Urol ; 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39003201

ABSTRACT

We previously reported that tumors harboring any one of four gene mutations (ATM, RB1, FANCC, or ERCC2) were likely to respond to neoadjuvant cisplatin-based chemotherapy (NAC), resulting in cancer-free surgical specimens at the time of cystectomy (pT0). Here, we report our validation of this finding. Using the CARIS 592 Gene Panel (Caris Life Sciences, Phoenix, AZ, USA), we analyzed 105 pre-NAC tumor specimens from a large multicenter trial (S1314) of either neoadjuvant gemcitabine and cisplatin (GC), or dose-dense methotrexate, vinblastine, Adriamycin, and cisplatin (DDMVAC). We found that a mutation in any one of these four genes predicted for pT0 at surgery (odds ratio = 5.36; 95% confidence interval [CI] 2.05, 14.02; two-sided p = 0.0006). The biomarker was better at predicting the presence of disease (negative predictive value for pT0 86%; 95% CI 73%, 94%) than the absence of disease (positive predictive value for pT0 48%; 95% CI 35%, 62%). There was no evidence of an interaction between the treatment arm (DDMVAC vs GC) and the genetic variant in terms of pT0. When combined with clinical assessment, these findings help inform patient selection for bladder preservation after cisplatin-based chemotherapy. PATIENT SUMMARY: A common standard of care for patients with muscle-invasive bladder cancer is neoadjuvant chemotherapy (NAC) followed by cystectomy to achieve cure. We previously discovered that specific DNA mutations in tumor samples collected at initial biopsy (transurethral resection of a bladder tumor) were predictive of a complete response to NAC. In other words, patients with these mutations were more likely to have a bladder found to be cancer free after surgery. In this study, we analyzed a larger set of tumor samples from a national clinical trial of chemotherapy followed by cystectomy to validate these earlier findings. We conclude that this biomarker test, when combined with careful clinical assessment, can be used to allocate patients to careful bladder surveillance instead of surgery. This hypothesis has been tested in the RETAIN trial presented previously (NCT02710734).

2.
Eur Urol ; 69(5): 855-62, 2016 05.
Article in English | MEDLINE | ID: mdl-26343003

ABSTRACT

BACKGROUND: Gene expression profiling (GEP) suggests there are three subtypes of muscle-invasive urothelial cancer (UC): basal, which has the worst prognosis; p53-like; and luminal. We hypothesized that GEP of transurethral resection (TUR) and cystectomy specimens would predict subtypes that could benefit from chemotherapy. OBJECTIVE: To explore clinical outcomes for patients treated with dose-dense (DD) methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) and bevacizumab (B) and the impact of UC subtype. DESIGN, SETTING, AND PARTICIPANTS: Sixty patients enrolled in a neoadjuvant trial of four cycles of DDMVAC + B between 2007 and 2010. TUR and cystectomy specimens for GEP were available from 38 and 23 patients, respectively, and from an additional confirmation cohort of 49 patients treated with perioperative MVAC. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Relationships with outcomes were analyzed using multivariable Cox regression and log-rank tests. RESULTS AND LIMITATIONS: Chemotherapy was active, with pT0N0 and ≤pT1N0 downstaging rates of 38% and 53%, respectively, and 5-yr overall survival (OS) of 63%. Bevacizumab had no appreciable impact on outcomes. Basal tumors had improved survival compared to luminal and p53-like tumors (5-yr OS 91%, 73%, and 36%, log-rank p=0.015), with similar findings on multivariate analysis. Bone metastases within 2 yr were exclusively associated with the p53-like subtype (p53-like 100%, luminal 0%, basal 0%; p ≤ 0.001). Tumors enriched with the p53-like subtype at cystectomy suggested chemoresistance for this subtype. A separate cohort treated with perioperative MVAC confirmed the UC subtype survival benefit (5-yr OS 77% for basal, 56% for luminal, and 56% for p53-like; p=0.021). Limitations include the small number of pretreatment specimens with sufficient tissue for GEP. CONCLUSION: GEP was predictive of clinical UC outcomes. The basal subtype was associated with better survival, and the p53-like subtype was associated with bone metastases and chemoresistant disease. PATIENT SUMMARY: We can no longer think of urothelial cancer as a single disease. Gene expression profiling identifies subtypes of urothelial cancer that differ in their natural history and sensitivity to chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bone Neoplasms/secondary , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/genetics , Transcriptome , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Adult , Aged , Bevacizumab/administration & dosage , Carcinoma, Transitional Cell/classification , Carcinoma, Transitional Cell/secondary , Chemotherapy, Adjuvant , Cisplatin/administration & dosage , Cystectomy , Doxorubicin/administration & dosage , Female , Gene Expression Profiling , Humans , Male , Methotrexate/administration & dosage , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Prognosis , Proportional Hazards Models , Survival Rate , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Urinary Bladder Neoplasms/classification , Urinary Bladder Neoplasms/pathology , Vinblastine/administration & dosage
3.
PLoS One ; 10(5): e0128011, 2015.
Article in English | MEDLINE | ID: mdl-26017803

ABSTRACT

Areca nut (AN) is a popular carcinogen used by about 0.6-1.2 billion people worldwide. Although AN contains apoptosis-inducing ingredients, we previously demonstrated that both AN extract (ANE) and its 30-100 kDa fraction (ANE 30-100K) predominantly induce autophagic cell death in both normal and malignant cells. In this study, we further explored the action mechanism of ANE 30-100K-induced autophagy (AIA) in Jurkat T lymphocytes and carcinoma cell lines including OECM-1 (mouth), CE81T/VGH (esophagus), SCC25 (tongue), and SCC-15 (tongue). The results showed that chemical- and small hairpin RNA (shRNA)-mediated inhibition of AMP-activated protein kinase (AMPK) resulted in the attenuation of AIA in Jurkat T but not in OECM-1 cells. Knockdown of Atg5 and Beclin 1 expressions ameliorated AIA in OECM-1/CE81T/VGH/Jurkat T and OECM-1/SCC25/SCC-15, respectively. Furthermore, ANE 30-100K could activate caspase-3 after inhibition of Beclin 1 expression in OECM-1/SCC25/SCC15 cells. Meanwhile, AMPK was demonstrated to be the upstream activator of the extracellular-regulated kinase (ERK) in Jurkat T cells, and inhibition of MEK attenuated AIA in Jurkat T/OECM-1/CE81T/VGH cells. Finally, we also found that multiple myeloma RPMI8226, lymphoma U937, and SCC15 cells survived from long-term non-cytotoxic ANE 30-100K treatment exhibited stronger resistance against serum deprivation through upregulated autophagy. Collectively, our studies indicate that Beclin-1 and Atg5 but not AMPK are commonly required for AIA, and MEK/ERK pathway is involved in AIA. Meanwhile, it is also suggested that long-term AN usage might increase the resistance of survived tumor cells against serum-limited conditions.


Subject(s)
Areca/chemistry , Autophagy/drug effects , Mouth Neoplasms/drug therapy , Nuts/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Caspase 3/metabolism , Cell Line, Tumor , Humans , Jurkat Cells , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mouth/drug effects , Mouth/metabolism , Mouth Neoplasms/metabolism , U937 Cells , Up-Regulation/drug effects
4.
Cancer Epidemiol Biomarkers Prev ; 23(8): 1569-78, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24859869

ABSTRACT

BACKGROUND: Oral cavity squamous cell carcinoma (OSCC) is one of the most common cancers worldwide, and its incidence is still increasing. Approximately 50% of patients with OSCC die within 5 years after diagnosis, mostly ascribed to the fact that the majority of patients present advanced stages of OSCC at the time of diagnosis. METHODS: To discover salivary biomarkers for ameliorating the detection of OSCC, herein, we developed a multiplexed bead-based platform to simultaneously detect auto-antibodies (auto-Abs) in salivary samples. RESULTS: Compared with healthy individuals, the salivary levels of anti-p53, anti-survivin, anti-Hsp60, and anti-RPLP0 were significantly elevated in patients with OSCC. Noteworthily, the elevated levels of anti-p53, anti-survivin, and anti-Hsp60 were already observed in individuals with oral potentially malignant disorder. Moreover, the salivary levels of anti-p53, anti-survivin, anti-Hsp60, anti-RPLP0, and anti-CK8 were significantly elevated in patients with early-stage OSCC compared with those in healthy individuals. Most importantly, the use of a combined panel of salivary anti-p53, anti-survivin, anti-Hsp60, and anti-RPLP0 largely improves the detection of OSCC. CONCLUSION: Collectively, our results reveal that the salivary auto-Abs are effective OSCC biomarkers and the four-auto-Ab panel provides a novel and practicable approach for OSCC screening. IMPACT: This study provides the first evidence for the potential clinical application of salivary auto-Abs in OSCC diagnosis.


Subject(s)
Antibodies, Neoplasm , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/diagnosis , Mouth Neoplasms/diagnosis , Saliva/chemistry , Adult , Aged , Aged, 80 and over , Autoantibodies , Carcinoma, Squamous Cell/immunology , Female , Humans , Immunoassay , Male , Middle Aged , Mouth Neoplasms/immunology , Saliva/immunology , Sensitivity and Specificity
5.
Cancer Cell ; 25(2): 152-65, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24525232

ABSTRACT

Muscle-invasive bladder cancers (MIBCs) are biologically heterogeneous and have widely variable clinical outcomes and responses to conventional chemotherapy. We discovered three molecular subtypes of MIBC that resembled established molecular subtypes of breast cancer. Basal MIBCs shared biomarkers with basal breast cancers and were characterized by p63 activation, squamous differentiation, and more aggressive disease at presentation. Luminal MIBCs contained features of active PPARγ and estrogen receptor transcription and were enriched with activating FGFR3 mutations and potential FGFR inhibitor sensitivity. p53-like MIBCs were consistently resistant to neoadjuvant methotrexate, vinblastine, doxorubicin and cisplatin chemotherapy, and all chemoresistant tumors adopted a p53-like phenotype after therapy. Our observations have important implications for prognostication, the future clinical development of targeted agents, and disease management with conventional chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Basal Cell/pathology , Drug Resistance, Neoplasm/genetics , Muscle Neoplasms/pathology , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Urinary Bladder Neoplasms/pathology , Aged , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma, Basal Cell/drug therapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Cell Proliferation , Cisplatin/administration & dosage , Clinical Trials, Phase II as Topic , Cohort Studies , Doxorubicin/administration & dosage , Female , Gene Expression Profiling , Humans , Male , Methotrexate/administration & dosage , MicroRNAs/genetics , Muscle Neoplasms/classification , Muscle Neoplasms/drug therapy , Neoadjuvant Therapy , Neoplasm Invasiveness , Neoplasm Staging , PPAR gamma/genetics , PPAR gamma/metabolism , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Urinary Bladder Neoplasms/classification , Urinary Bladder Neoplasms/drug therapy , Vinblastine/administration & dosage
6.
J Urol ; 190(4): 1404-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23500642

ABSTRACT

PURPOSE: KU7 is a popular urothelial carcinoma cell line that was isolated from the bladder of a patient at Keio University in 1980. It has subsequently been widely used in laboratories around the world. We describe how routine cell line authentication revealed that KU7 was cross contaminated almost 30 years ago with HeLa, a cervical carcinoma cell line. MATERIALS AND METHODS: Presumed KU7 clones dating from 1984 to 1999 were provided by M.D. Anderson Cancer Center, Vancouver Prostate Centre, Kyoto University, Tokyo Medical University and Keio University. HeLa was obtained from ATCC. Genomic DNA was isolated and short tandem repeat analysis was performed at the M.D. Anderson Cancer Center Characterized Cell Line Core Facility, Johns Hopkins University Fragment Analysis Facility and RIKEN BioResource Center, Ibaraki, Japan. Comparative genomic hybridization was performed on a platform (Agilent Technologies, Santa Clara, California) at Vancouver Prostate Centre. RESULTS: The short tandem repeat profile of all KU7 clones was an exact match with that of HeLa. Comparative genomic hybridization of all samples revealed an abundance of shared chromosomal aberrations. Slight differences in some genomic areas were explained by genomic drift in different KU7 clones separated by many years. CONCLUSIONS: Our analysis identified that cross contamination of KU7 with HeLa occurred before 1984 at the source institution. All KU7 clones in the urological literature should be considered HeLa and experimental results should be viewed in this light. Our results emphasize the need to authenticate cell lines in oncological research.


Subject(s)
DNA Contamination , HeLa Cells , Comparative Genomic Hybridization , Gene Expression Profiling , Humans , Time Factors , Urinary Bladder Neoplasms/pathology
7.
J Biol Chem ; 288(5): 3275-88, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23239884

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a physiological process that plays important roles in tumor metastasis, "stemness," and drug resistance. EMT is typically characterized by the loss of the epithelial marker E-cadherin and increased expression of EMT-associated transcriptional repressors, including ZEB1 and ZEB2. The miR-200 family and miR-205 prevent EMT through suppression of ZEB1/2. p53 has been implicated in the regulation of miR-200c, but the mechanisms controlling miR-205 expression remain elusive. Here we report that the p53 family member and p63 isoform, ΔNp63α, promotes miR-205 transcription and controls EMT in human bladder cancer cells. ΔNp63α, E-cadherin and miR-205 were coexpressed in a panel of bladder cancer cell lines (n = 28) and a cohort of primary bladder tumors (n = 98). Stable knockdown of ΔNp63α in the "epithelial" bladder cancer cell line UM-UC6 decreased the expression of miR-205 and induced the expression of ZEB1/2, effects that were reversed by expression of exogenous miR-205. Conversely, overexpression of ΔNp63α in the "mesenchymal" bladder cancer cell line UM-UC3 induced miR-205 and suppressed ZEB1/2. ΔNp63α knockdown reduced the expression of the primary and mature forms of miR-205 and the miR-205 "host" gene (miR-205HG) and decreased binding of RNA Pol II to the miR-205HG promoter, inhibiting miR-205HG transcription. Finally, high miR-205 expression was associated with adverse clinical outcomes in bladder cancer patients. Together, our data demonstrate that ΔNp63α-mediated expression of miR-205 contributes to the regulation of EMT in bladder cancer cells and identify miR-205 as a molecular marker of the lethal subset of human bladder cancers.


Subject(s)
Epithelial-Mesenchymal Transition , MicroRNAs/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Base Sequence , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Kaplan-Meier Estimate , MicroRNAs/genetics , Molecular Sequence Data , Protein Binding/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Transcription Factors/genetics , Treatment Outcome , Tumor Suppressor Proteins/genetics , Urothelium/metabolism , Urothelium/pathology , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
8.
Biochem Pharmacol ; 84(8): 1036-44, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22846602

ABSTRACT

Quercetin is a major flavonoid in a wide range of fruits and vegetables. Consumption of quercetin may contribute to a reduction in risk of cardiovascular disease (CVD). Following ingestion, flavonoids are metabolized rapidly by methylation or glucuronidation, which can alter their biological activity. Certain dietary flavonoids have been shown to upregulate the expression of adenosine monophosphate-activated protein kinase (AMPK). AMPK is a conserved key enzyme in cellular energy homeostasis that affects fatty acid oxidation. The aim of the present study was to investigate the effects of supraphysiological concentrations of quercetin and its methyl and glucuronide metabolites (3'-O-methyl-quercetin and quercetin-3-O-glucuronide) on activation of AMPK and eNOS in human aortic endothelial cells (HAECs) and endothelial function in isolated aortic rings from C57BL mice. We found that 5 and 10 µM quercetin and its metabolites, and pretreatment of arteries with quercetin and its metabolites can protect vessels against hypochlorous acid-induced endothelial dysfunction in isolated arteries (P < 0.05). Inhibition of AMPK blocked these protective effects. We also found that 5 and 10 µM quercetin and its metabolites can induce activation of AMPK and eNOS in human aortic endothelial cells, and lead to an increase in the concentrations of S-nitrosothiols and nitrite in cell culture media (P < 0.05). These results provide further support for the cardioprotective effects of certain dietary flavonoids. They suggest that beneficial effects of quercetin on endothelial cell functions are in part mediated via AMPK pathway.


Subject(s)
Adenylate Kinase/metabolism , Blood Vessels/drug effects , Nitric Oxide Synthase Type III/biosynthesis , Quercetin/pharmacology , Animals , Blood Vessels/enzymology , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Phosphorylation
9.
Cancer Biol Ther ; 13(7): 477-86, 2012 May.
Article in English | MEDLINE | ID: mdl-22361733

ABSTRACT

Antimitotics such as taxanes are being considered as alternatives to conventional cisplatin-based chemotherapy in patients with bladder cancer, but the molecular determinants of sensitivity or resistance to these agents in bladder cancer cells have not been defined. Here we examined the cytotoxic effects of a novel antimitotic, the Eg5 inhibitor AZD4877, in a molecularly diverse panel of human bladder cancer cell lines. The cells displayed heterogeneous responses to the drug that correlated closely with sensitivity to docetaxel but not with sensitivity to cisplatin. Global gene expression profiling identified p63 as the top gene that was differentially expressed between sensitive and resistant cell lines. Stable knockdown of p63 inhibited cell death induced by either AZD4877 or docetaxel and was associated with decreased proliferation and decreased expression of c-myc. Furthermore, c-myc knockdown also rendered cells resistant to AZD4877 or docetaxel. Together, our results implicate p63 and its downstream target c-myc as determinants of sensitivity to anti-mitotics in bladder cancer cells. Our data also suggest that anti-mitotics and cisplatin target different subsets of bladder cancer cells, a conclusion that may have important implications for the therapy of muscle-invasive bladder cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Kinesins/antagonists & inhibitors , Pyrimidinones/pharmacology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
10.
PLoS One ; 7(1): e30206, 2012.
Article in English | MEDLINE | ID: mdl-22253920

ABSTRACT

BACKGROUND: p63 is a member of the p53 family that has been implicated in maintenance of epithelial stem cell compartments. Previous studies demonstrated that p63 is downregulated in muscle-invasive bladder cancers, but the relationship between p63 expression and survival is not clear. METHODOLOGY/PRINCIPAL FINDINGS: We used real-time PCR to characterize p63 expression and several genes implicated in epithelial-to-mesenchymal transition (EMT) in human bladder cancer cell lines (n = 15) and primary tumors (n = 101). We correlated tumor marker expression with stage, disease-specific (DSS), and overall survival (OS). Expression of E-cadherin and p63 correlated directly with one another and inversely with expression of the mesenchymal markers Zeb-1, Zeb-2, and vimentin. Non-muscle-invasive (Ta and T1) bladder cancers uniformly expressed high levels of E-cadherin and p63 and low levels of the mesenchymal markers. Interestingly, a subset of muscle-invasive (T2-T4) tumors maintained high levels of E-cadherin and p63 expression. As expected, there was a strongly significant correlation between EMT marker expression and muscle invasion (p<0.0001). However, OS was shorter in patients with muscle-invasive tumors that retained p63 (p = 0.007). CONCLUSIONS/SIGNIFICANCE: Our data confirm that molecular markers of EMT are elevated in muscle-invasive bladder cancers, but interestingly, retention of the "epithelial" marker p63 in muscle-invasive tumors is associated with a worse outcome.


Subject(s)
Muscles/pathology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/classification , Urinary Bladder Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Databases, Genetic , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Matrix Metalloproteinase 9/metabolism , Middle Aged , Neoplasm Invasiveness , Prognosis , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/genetics
11.
Clin Cancer Res ; 17(9): 2863-73, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21415218

ABSTRACT

PURPOSE: We investigated the effect of the mTOR inhibitor RAD001 (everolimus) on human bladder cancer (BC) cells in vitro and in vivo. EXPERIMENTAL DESIGN: The effect of RAD001 on the growth of UM-UC-3, UM-UC-6, UM-UC-9, and UM-UC-14 BC cells were assessed by crystal violet and [(3)H]thymidine incorporation assays. Flow cytometric cell-cycle analyses were done to measure the apoptotic cell fraction. Protein synthesis was measured using tritium-labeled leucine incorporation assays. The effects of RAD001 on the mTOR pathway were analyzed by Western blotting. To test the effects of RAD001 in vivo, UM-UC-3, UM-UC-6, and UM-UC-9 cells were subcutaneously implanted into nude mice. Tumor-bearing mice were treated orally with RAD001 or placebo. Tumors were harvested for immunohistochemical analysis. RESULTS: In vitro, RAD001 transiently inhibited BC cell growth in a dose-dependent manner. This effect was augmented by re-treatment of cells after 3 days. UM-UC-14 cells were the most sensitive to RAD001, whereas UM-UC-9 cells were the least sensitive. After re-treatment with RAD001, only sensitive cell lines showed G(1)-phase arrest, with no evidence of apoptosis. RAD001 significantly inhibited the growth of tumors that were subcutaneously implanted in mice. Inhibition of protein synthesis through the S6K and 4EBP1 pathways seems to be the main mechanism for the RAD001-induced growth inhibition. However, inhibition of angiogenesis was the predominant mechanism of the effect of RAD001 on UM-UC-9 cells. CONCLUSIONS: The mTOR inhibitor RAD001 inhibits growth of BC cells in vitro. RAD001 is effective in treating BC tumors in an in vivo nude mouse model despite the heterogeneity of in vitro responses.


Subject(s)
Carcinoma, Transitional Cell/drug therapy , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Transitional Cell/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Everolimus , Female , Humans , Mice , Mice, Nude , Protein Biosynthesis/drug effects , Sirolimus/pharmacology , Sirolimus/therapeutic use , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Cancer Prev Res (Phila) ; 3(6): 776-86, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20501863

ABSTRACT

Transitional cell carcinoma (TCC) of the bladder ranks fourth in incidence of all cancers in the developed world, yet the mechanisms of its origin and progression remain poorly understood. There are also few useful diagnostic or prognostic biomarkers for this disease. We have combined a transgenic mouse model for invasive bladder cancer (UPII-SV40Tag mice) with DNA microarray technology to determine molecular mechanisms involved in early TCC development and to identify new biomarkers for detection, diagnosis, and prognosis of TCC. We have identified genes that are differentially expressed between the bladders of UPII-SV40Tag mice and their age-matched wild-type littermates at 3, 6, 20, and 30 weeks of age. These are ages that correspond to premalignant, carcinoma in situ, and early-stage and later stage invasive TCC, respectively. Our preliminary analysis of the microarray data sets has revealed approximately 1,900 unique genes differentially expressed (> or =3-fold difference at one or more time points) between wild-type and UPII-SV40Tag urothelium during the time course of tumor development. Among these, there were a high proportion of cell cycle regulatory genes and a proliferation signaling genes that are more strongly expressed in the UPII-SV40Tag bladder urothelium. We show that several of the genes upregulated in UPII-SV40Tag urothelium, including RacGAP1, PCNA, and Hmmr, are expressed at high levels in superficial bladder TCC patient samples. These findings provide insight into the earliest events in the development of bladder TCC as well as identify several promising early-stage biomarkers.


Subject(s)
Carcinoma in Situ/genetics , Carcinoma, Transitional Cell/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Neoplasm Proteins/genetics , Urinary Bladder Neoplasms/genetics , Animals , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Disease Models, Animal , Disease Progression , Gene Regulatory Networks , Humans , Hyperplasia , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Neoplasm Proteins/biosynthesis , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Urinary Bladder Diseases/genetics , Urinary Bladder Diseases/metabolism , Urinary Bladder Diseases/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urothelium/metabolism , Urothelium/pathology
13.
J Contam Hydrol ; 100(3-4): 91-100, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18649972

ABSTRACT

In situ chemical oxidation (ISCO) is considered a reliable technology to treat groundwater contaminated with high concentrations of organic contaminants. An ISCO oxidant, persulfate anion (S(2)O(8)(2-)) can be activated by ferrous ion (Fe(2+)) to generate sulfate radicals (E(o)=2.6 V), which are capable of destroying trichloroethylene (TCE). The property of polarity inhibits S(2)O(8)(2-) or sulfate radical (SO(4)(-)) from effectively oxidizing separate phase TCE, a dense non-aqueous phase liquid (DNAPL). Thus the oxidation primarily takes place in the aqueous phase where TCE is dissolved. A bench column study was conducted to demonstrate a conceptual remediation method by flushing either S(2)O(8)(2-) or Fe(2+) through a soil column, where the TCE DNAPL was present, and passing the dissolved mixture through either a Fe(2+) or S(2)O(8)(2-) fluid sparging curtain. Also, the effect of a solubility enhancing chemical, hydroxypropyl-beta-cyclodextrin (HPCD), was tested to evaluate its ability to increase the aqueous TCE concentration. Both flushing arrangements may result in similar TCE degradation efficiencies of 35% to 42% estimated by the ratio of TCE degraded/(TCE degraded+TCE remained in effluent) and degradation byproduct chloride generation rates of 4.9 to 7.6 mg Cl(-) per soil column pore volume. The addition of HPCD did greatly increase the aqueous TCE concentration. However, the TCE degradation efficiency decreased because the TCE degradation was a lower percentage of the relatively greater amount of dissolved TCE by HPCD. This conceptual treatment may serve as a reference for potential on-site application.


Subject(s)
Oxygen/chemistry , Sulfates/chemistry , Trichloroethylene/chemistry , Water Pollutants, Chemical/analysis , Chlorides/chemistry , Equipment Design , Free Radicals , Iron/chemistry , Models, Chemical , Oxygen/metabolism , Time Factors , Trichloroethylene/analysis , Water Purification , beta-Cyclodextrins/chemistry
14.
Urol Oncol ; 26(6): 641-5, 2008.
Article in English | MEDLINE | ID: mdl-18367112

ABSTRACT

OBJECTIVES: Cyclooxygenase 2 (COX-2) is aberrantly expressed in multiple tumor types including bladder cancer and is associated with enhanced growth, resistance to apoptosis, invasion, and angiogenesis. To evaluate the mechanisms through which COX-2 expression alters normal urothelium, we transfected the SV-40 immortalized human urothelial cell line SV-HUC with COX-2. METHODS: SV-HUC cells were stably transfected with a plasmid containing COX-2 under a CMV promoter. Following isolation of monoclonal transfectants, COX-2 expression was determined by Western and Northern analyses. Prostaglandin E2 (PGE2) in the culture supernatant was measured by ELISA. Cell growth was measured by crystal violet assay. Cellular invasion through Matrigel and anchorage-independent growth in 0.4% agarose were assessed. Tumorigenicity was evaluated by subcutaneous injection of cells in nude mice with and without Matrigel. RESULTS: Four of 12 clones stably overexpressing COX-2 at high levels relative to vector-transfected control cells were chosen for further study. Cell growth rates of these 4 clones were higher than vector control cells. PGE(2) production was elevated in 3 of these 4 clones, and PGE2 levels correlated significantly with invasion through Matrigel. COX-2-transfected cells did not form colonies in soft agarose or tumors in nude mice. CONCLUSIONS: Forced COX-2 expression in SV-HUC immortalized urothelial cells contributes to increased PGE2 production and increased invasion through Matrigel. However, it is insufficient to induce malignant transformation.


Subject(s)
Cyclooxygenase 2/physiology , Dinoprostone/biosynthesis , Urinary Bladder Neoplasms/etiology , Urinary Bladder/pathology , Cell Line , Cyclooxygenase 2/genetics , Humans , Neoplasm Invasiveness , Transfection , Urinary Bladder Neoplasms/pathology
15.
Urology ; 71(2): 346-50, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18308117

ABSTRACT

OBJECTIVES: Urothelial differentiation is essential for the maintenance of urinary bladder function. We explored the expression and function of 15-hydroxyprostaglandin dehydrogenase (PGDH) during urothelial differentiation. METHODS: We evaluated expression of PGDH by Northern and Western blotting and immunostaining in human urothelial cultures, cell lines, and tissues. We determined enzymatic function using enzyme-linked immunosorbent assay. Small inhibitory ribonucleic acids were used to inhibit PGDH expression in human bladder cancer cells. RESULTS: We found PGDH messenger ribonucleic acid was increased in an in vitro model of human urothelial differentiation by Northern blotting. Western blotting of human bladder cancer cell lines showed expression in the well-differentiated RT4 cells and no expression in poorly differentiated UC3 cells. Immunostaining showed that PGDH expression increased with differentiation in normal bladder urothelium. The enzyme was functional in the well-differentiated RT4 human bladder cancer cell line. Inhibition of PGDH expression resulted in disruption of E-cadherin expression at cell-cell contacts in well-differentiated RT4 bladder cancer cells. CONCLUSIONS: These studies indicate that PGDH expression is associated with urothelial differentiation, and loss of PGDH expression results in disruption of urothelial differentiation.


Subject(s)
Hydroxyprostaglandin Dehydrogenases/physiology , Urothelium/physiology , Cell Differentiation , Cells, Cultured , Humans , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Urothelium/cytology
16.
Chemosphere ; 70(3): 426-35, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17692892

ABSTRACT

In situ chemical oxidation with persulfate anion (S2O82*) is a viable technique for remediation of groundwater contaminants such as trichloroethylene (TCE). An accelerated reaction using S2O82* to destroy TCE can be achieved via chemical activation with ferrous ion to generate sulfate radicals (SO4*)(E degrees =2.6 V). The column study presented here simulates persulfate oxidation of TCE in porous media (glass beads and a sandy soil). Initial experiments were conducted to investigate persulfate transport in the absence of TCE in the column. The persulfate flushing exhibited a longer residence time and revealed a moderate persulfate interaction with soils. In TCE treatment experiments, the results indicate that the water or persulfate solution would push dissolved TCE from the column. Therefore, the effluent TCE concentration gradually increased to a maximum when about one pore volume was replaced with the flushing solution in the column. The presence of Fe2+ concentration within the column caused a quick drop in effluent TCE concentration and more TCE degradation was observed. When a TCE solution was flushing through the soil column, breakthrough of TCE concentration in the effluent was relatively slow. In contrast, when the soil column was flushed with a mixed solution of persulfate and TCE, persulfate appeared to preferentially oxidize soil oxidizable matter rather than TCE during transport. Hence, persulfate oxidation of soil organics may possibly reduce the interaction between TCE and soil (e.g., adsorption) and facilitate the transport of TCE through soil columns resulting in faster breakthrough.


Subject(s)
Iron/chemistry , Oxidants/chemistry , Sodium Compounds/chemistry , Sulfates/chemistry , Trichloroethylene/chemistry , Water Pollutants, Chemical/chemistry , Glass , Oxidation-Reduction , Porosity , Soil , Waste Management/methods
17.
J Urol ; 175(3 Pt 1): 1133-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16469639

ABSTRACT

PURPOSE: Cell lines have become an essential component for the investigation of cancer. We have developed a panel of cell lines derived from human urothelial cancers and we describe some of their important characteristics. MATERIALS AND METHODS: Ten human urothelial cancer cell lines were characterized by their growth in athymic nude mice, CAR expression and their susceptibility to adenoviral mediated transfer of the green fluorescence protein gene. TP53 mutation status and immunochemical analysis of p53, pRB and p16 were also examined. RESULTS: Five cell lines rapidly produced tumors in athymic nude mice. Two cell lines produced tumors in 1 month, 1 produced them in 3 months and 2 were nontumorigenic. The cell lines varied in CAR expression and in their susceptibility to adenoviral mediated gene transduction. There was no direct correlation between CAR expression and susceptibility to adenoviral mediated gene transduction. Seven cell lines had TP53 mutations, of which 2 had large deletions and did not express p53 protein by immunostaining. All cell lines expressed abnormal pRB by immunochemical analysis (3 had no staining and 7 had homogenously strong staining) and 8 did not express p16 (7 showed homogeneously strong pRB staining). CONCLUSIONS: Our panel of 10 human urothelial cell lines differed in genetic alterations, growth in nude mice, susceptibility to adenoviral mediated gene transduction, and expression of p53, p16 and pRB. The availability of various urothelial cancer cell lines with differing genotypic and phenotypic features will facilitate further research into bladder cancer.


Subject(s)
Adenoviridae , Gene Transfer Techniques , Receptors, Virus/biosynthesis , Urologic Neoplasms/pathology , Urothelium/pathology , Animals , Cell Division , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Humans , Mice , Mice, Nude , Urologic Neoplasms/genetics , Urologic Neoplasms/metabolism , Urologic Neoplasms/virology
18.
Oncol Rep ; 15(2): 471-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16391871

ABSTRACT

Selective COX-2 inhibitors such as celecoxib and NS-398 are being evaluated as chemopreventive and therapeutic agents for bladder and other cancers. We investigated the effects of these nonsteroidal anti-inflammatory agents on a panel of bladder cancer cell lines, and assessed their effects on anchorage-dependent and -independent growth, cell cycle, apoptosis and morphology. The human bladder cancer cell lines UM-UC-1, -3, and -6 were assayed for COX-2 expression by Western analysis using a monoclonal antibody to COX-2. UM-UC-1, -3, and -6 cells were grown in the presence of increasing concentrations of NS-398 and celecoxib, and cell growth was quantitated over 7 days by crystal violet elution. The cell lines were treated with NS-398 and celecoxib for 48 h and analyzed by flow cytometry with propidium iodide staining and Br-dUTP staining for apoptosis. Anchorage-independent growth was assessed using an agarose growth assay. Western analysis demonstrated that COX-2 expression in UM-UC-1, -6, and -3 was high, low, and undetectable, respectively. NS-398 and celecoxib produced dose-dependent growth inhibition of UM-UC-1 and -6. Both NS-398 and celecoxib also inhibited anchorage-dependent and -independent growth of UM-UC-3 in a dose-dependent fashion, despite the low basal expression of COX-2 in this cell line. Cell cycle analyses of UM-UC-1 and -6 revealed a 50% reduction in S-phase in the presence of 100 microM NS-398 whereas a smaller reduction in S-phase was noted in UM-UC-3 cells. Furthermore, treatment with 100 microM celecoxib resulted in significant apoptosis in all three cell lines, which was associated with downregulation of Bcl-2. COX-2 selective inhibitors NS-398 and celecoxib produced dose-dependent growth inhibition of bladder cancer cells associated with a significant reduction in S-phase. Induction of apoptosis in all three cell lines by celecoxib was associated with downregulation of Bcl-2. These changes occur independently of COX-2 expression levels suggesting the presence of a COX-2 independent pathway.


Subject(s)
Apoptosis/drug effects , Carcinoma, Transitional Cell/drug therapy , Cell Proliferation/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Urinary Bladder Neoplasms/drug therapy , Blotting, Western , Celecoxib , Cell Line, Tumor , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Flow Cytometry , Humans , Nitrobenzenes/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazoles/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology
19.
Int J Oncol ; 25(6): 1631-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15547700

ABSTRACT

Chromosomal aneuploidy is associated with invasive bladder cancer and one of the genes implicated in these changes is Aurora-A/STK15/BTAK, that is localized on chromosome 20q13 and encodes a centrosome-associated serine/threonine kinase. To better understand the association between Aurora-A/STK15 expression, tumor aneuploidy and clinical prognosis, we sought to determine whether overexpression of Aurora-A/STK15 in cultured urothelial cells facilitated chromosomal instability. Using immunofluorescence staining, Northern and Western blot analyses, we verified that overexpression of Aurora-A/STK15 in bladder tumor cell lines enhanced chromosomal instability. Additionally, we observed that some bladder tumor cell lines expressed more Aurora-A/STK15 than cultured normal urothelial cells and that Aurora-A/STK15 expression was higher in an immortalized E7 urothelial cell line having 20q amplification than in an E6 line lacking 20q amplification. These results were consistent with our observations of higher mRNA levels in some T3 invasive bladder tumors than in T1 superficial tumors and adjacent normal bladder tissue. Overall our results suggest that overexpression of Aurora-A/STK15 in bladder tumor cells contributes to tumor progression by promoting chromosomal instability leading to aneuploidy.


Subject(s)
Aneuploidy , Gene Expression Regulation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/pharmacology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Aurora Kinase A , Aurora Kinases , Disease Progression , Fluorescent Antibody Technique , Humans , Prognosis , Up-Regulation , Urothelium/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...