Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667323

ABSTRACT

Bladder cancer aggressiveness is correlated with abnormal N-cadherin transmembrane glycoprotein expression. This protein is cleaved by the metalloprotease ADAM10 and the γ-secretase complex releasing a pro-angiogenic N-terminal fragment (NTF) and a proliferation-activating soluble C-terminal fragment (CTF2). Tetraspanin 15 (Tspan15) is identified as an ADAM10-interacting protein to induce selective N-cadherin cleavage. We first demonstrated, in invasive T24 bladder cancer cells, that N-cadherin was cleaved by ADAM10 generating NTF in the extracellular environment and leaving a membrane-anchored CTF1 fragment and that Tspan15 is required for ADAM10 to induce the selective N-cadherin cleavage. Targeting N-cadherin function in cancer is relevant to preventing tumor progression and metastases. For antitumor molecules to inhibit N-cadherin function, they should be complete and not cleaved. We first showed that the GW501516, an agonist of the nuclear receptor PPARß/δ, decreased Tspan15 and prevented N-cadherin cleavage thus decreasing NTF. Interestingly, the drug did not modify ADAM10 expression, which was important because it could limit side effects since ADAM10 cleaves numerous substrates. By targeting Tspan15 to block ADAM10 activity on N-cadherin, GW501516 could prevent NTF pro-tumoral effects and be a promising molecule to treat bladder cancer. More interestingly, it could optimize the effects of the N-cadherin antagonists those such as ADH-1 that target the N-cadherin ectodomain.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Antigens, CD , Cadherins , Dipeptides , Hydroxamic Acids , Membrane Proteins , Tetraspanins , Urinary Bladder Neoplasms , Humans , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Cadherins/metabolism , Cell Line, Tumor , Membrane Proteins/metabolism , Neoplasm Invasiveness , Proteolysis/drug effects , Tetraspanins/metabolism , Tetraspanins/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics
2.
Mol Cell Biochem ; 471(1-2): 113-127, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32519230

ABSTRACT

N-cadherin is a transmembrane glycoprotein expressed by mesenchymal origin cells and is located at the adherens junctions. It regulates also cell motility and contributes to cell signaling. In previous studies, we identified that its anomalous expression in bladder carcinoma was a tumor progression marker. A pharmacological approach to inhibit N-cadherin expression or to block its function could be relevant to prevent disease progression and metastasis development. The morphological exploration of T24 invasive bladder cancer cells by atomic force microscopy (AFM) revealed a spindle-like shape with fibrous structures. By engaging force spectroscopy with AFM tip functionalized with anti-E or anti-N-cadherin antibodies, results showed that T24 cells expressed only N-cadherin as also demonstrated by Western blotting and confocal microscopy. For the first time, we demonstrated by RTqPCR and Western blotting analyses that the peroxisome proliferator-activated receptor ß/δ (PPARß/δ) agonist GW501516 significantly decreased N-cadherin expression in T24 cells. Moreover, high non-cytotoxic doses of GW501516 inhibited confluent T24 cell wound healing closure. By using AFM, a more sensitive nanoanalytical method, we showed that the treatment modified the cellular morphology and diminished N-cadherin cell surface coverage through the decreasing of these adhesion molecule-mediated interaction forces. We observed a greater decrease of N-cadherin upon GW501516 exposure with AFM than that detected with molecular biology techniques. AFM was a complementary tool to biochemical techniques to perform measurements on living cells at the nanometer resolution level. Taken together, our data suggest that GW501516 could be an interesting therapeutic strategy to avoid bladder cancer cell spreading through N-cadherin decrease.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Epithelial-Mesenchymal Transition , Microscopy, Atomic Force/methods , PPAR delta/agonists , PPAR-beta/agonists , Thiazoles/pharmacology , Urinary Bladder Neoplasms/metabolism , Antigens, CD/ultrastructure , Cadherins/ultrastructure , Cell Line, Tumor , Cell Movement , Humans , Signal Transduction , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/ultrastructure
3.
BMC Cancer ; 18(1): 1239, 2018 Dec 10.
Article in English | MEDLINE | ID: mdl-30526555

ABSTRACT

BACKGROUND: Non-muscle invasive bladder cancers (NMIBC: pTa, pT1) are characterised by a high risk of recurrence and/or progression. Identification of prognostic markers is needed to improve both diagnosis and management of the disease. The aim of this study was to analyse the expression of A-FABP (adipocyte-fatty acid binding protein) and to evaluate its prognostic value in bladder cancer with a long term clinical follow-up. METHODS: A-FABP expression was investigated by immunohistochemistry in 236 tumours (114 pTa, 61 pT1, 61 pT2-4). Immunostaining was classified as negative (absent or weak immunostaining and moderate or strong staining on ≤10% of cells) or positive (moderate or strong staining on > 10% of cells). Event-free survival (EFS) and overall survival (OS) were determined with a 87.3 months median follow-up in the overall cohort. Recurrence-free survival (RFS) and progression-free survival (PFS) were established in NMIBC. RESULTS: Loss of A-FABP was associated with higher mean age, high stage/grade, and the presence of metastatic lymph nodes. It was correlated with shorter median EFS (17.5 vs 62.5 months; p = 0.001) and mean OS (76.7 vs 154.2 months; p = 0.009) and with higher risk of progression in the pTa/pT1 subgroup (HR, 0.36; 95% CI, 0.13-0.96; p = 0.041) and importantly in the pTa tumours (HR, 0.34; 95% CI, 0.10-0.97; p = 0.045). CONCLUSION: These results demonstrated that loss of A-FABP expression following a long follow-up was predictive of pTa and pTa/pT1 progression. Immunohistochemistry on diagnostic biopsy is easy to use and could be of value to help clinicians to propose appropriate treatment for these tumours.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Transitional Cell/pathology , Fatty Acid-Binding Proteins/biosynthesis , Urinary Bladder Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/mortality , Disease Progression , Down-Regulation , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Progression-Free Survival , Retrospective Studies , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality
4.
Oncotarget ; 8(64): 107744-107762, 2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29296202

ABSTRACT

Known activators of the Peroxisome Proliferator-Activated Receptor γ (PPARγ), thiazolidinediones (TZD) induce apoptosis in a variety of cancer cells through dependent and/or independent mechanisms of the receptor. We tested a panel of TZD (Rosiglitazone, Pioglitazone, Ciglitazone) to shed light on their potential therapeutic effects on three cervical cancer cell lines (HeLa, Ca Ski, C-33 A). In these cells, only ciglitazone triggered apoptosis through PPARγ-independent mechanisms and in particular via both extrinsic and intrinsic pathways in Ca Ski cells containing Human PapillomaVirus (HPV) type 16. It also inhibits cervical cancer xenograft development in nude mice. Ciglitazone kills cervical cancer cells by activating death receptor signalling pathway, caspase cascade and BH3 interacting-domain death agonist (Bid) cleavage through the up-regulation of Death Receptor 4 (DR4)/DR5 and soluble and membrane-bound TNF related apoptosis inducing ligand (TRAIL). Importantly, the drug let TRAIL-resistant Ca Ski cells to respond to TRAIL through the downregulation of cellular FLICE-Like Inhibitory Protein (c-FLIP) level. For the first time, we revealed that ciglitazone is able to decrease E6 viral oncoprotein expression known to block TRAIL pathway and this was associated with cell death. Our results highlight the capacity of ciglitazone to restore TRAIL sensitivity and to prevent E6 blocking action to induce apoptosis in cervical cancer cells.

5.
Tumour Biol ; 37(11): 14789-14802, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27638828

ABSTRACT

GW501516 is a selective and high-affinity synthetic agonist of peroxisome proliferator-activated receptor ß/δ (PPARß/δ). This molecule promoted the inhibition of proliferation and apoptosis in few cancer cell lines, but its anticancer action has never been investigated in bladder tumor cells. Thus, this study was undertaken to determine whether GW501516 had antiproliferative and/or apoptotic effects on RT4 and T24 urothelial cancer cells and to explore the molecular mechanisms involved. Our results indicated that, in RT4 cells (derived from a low-grade papillary tumor), GW501516 did not induce cell death. On the other hand, in T24 cells (derived from an undifferentiated high-grade carcinoma), this PPARß/δ agonist induced cytotoxic effects including cell morphological changes, a decrease of cell viability, a G2/M cell cycle arrest, and the cell death as evidenced by the increase of the sub-G1 cell population. Furthermore, GW501516 triggered T24 cell apoptosis in a caspase-dependent manner including both extrinsic and intrinsic apoptotic pathways through Bid cleavage. In addition, the drug led to an increase of the Bax/Bcl-2 ratio, a mitochondrial dysfunction associated with the dissipation of ΔΨm, and the release of cytochrome c from the mitochondria to the cytosol. GW501516 induced also ROS generation which was not responsible for T24 cell death since NAC did not rescue cells upon PPARß/δ agonist exposure. For the first time, our data highlight the capacity of GW501516 to induce apoptosis in invasive bladder cancer cells. This molecule could be relevant as a therapeutic drug for high-grade urothelial cancers.


Subject(s)
Apoptosis/drug effects , PPAR delta/agonists , PPAR-beta/agonists , Thiazoles/pharmacology , Urinary Bladder Neoplasms/pathology , Blotting, Western , Cell Proliferation/drug effects , Flow Cytometry , Humans , Neoplasm Invasiveness , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism
6.
Cell Signal ; 26(2): 433-43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24172859

ABSTRACT

Peroxisome Proliferator-Activated Receptor-ß (PPARß) is a ligand-inducible transcription factor activated by both natural (fatty acids and derivatives) and high affinity synthetic agonists. It is thought to play a role in angiogenesis development and Vascular Endothelial Growth Factor (VEGF) regulation but its contribution remains unclear. Until now, the PPARß agonism effect on VEGF expression in cervical cancer cells was unknown. This led to our interest in assessing the effect of PPARß activation on the regulation of different VEGF isoforms mRNA expression and the impact of E6 viral oncoprotein and its target p53 on this regulation in cervical cancer cells. Here, we showed that the PPARß agonist L-165041 induces VEGF(121), VEGF(165) and VEGF(189) expression in HPV (Human Papillomavirus) positive HeLa cells but not in HPV negative cells. The underlying mechanisms did involve neither E6 oncoprotein nor p53. We highlighted a novel mode of PPARß ligand action including a post-transcriptional regulation of VEGF mRNA expression through the p38 MAPK signaling pathway and the activation of the mRNA-stabilizing factor HuR. But most importantly, we clearly demonstrated that L-165041 acts independently of PPARß since its effect was not reversed by a chemical inhibition with a specific antagonist and the siRNA-mediated knockdown of the nuclear receptor. As VEGF is crucial for cancer development, the impact of PPARß ligands on VEGF production is of high importance. Thus, the molecular mechanism of their action has to be elucidated and as a result, PPARß agonists currently in clinical trials should be carefully monitored.


Subject(s)
Human papillomavirus 18/metabolism , PPAR-beta/agonists , Phenoxyacetates/pharmacology , RNA Stability/drug effects , RNA, Messenger/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , DNA-Binding Proteins , ELAV Proteins/antagonists & inhibitors , ELAV Proteins/genetics , ELAV Proteins/metabolism , HeLa Cells , Humans , Oncogene Proteins, Viral/antagonists & inhibitors , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , PPAR-beta/genetics , PPAR-beta/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
PLoS One ; 6(12): e28354, 2011.
Article in English | MEDLINE | ID: mdl-22174792

ABSTRACT

BACKGROUND: Ciglitazone belongs to the thiazolidinediones class of antidiabetic drug family and is a high-affinity ligand for the Peroxisome Proliferator-Activated Receptor γ (PPARγ). Apart from its antidiabetic activity, this molecule shows antineoplastic effectiveness in numerous cancer cell lines. METHODOLOGY/PRINCIPAL FINDINGS: Using RT4 (derived from a well differentiated grade I papillary tumor) and T24 (derived from an undifferentiated grade III carcinoma) bladder cancer cells, we investigated the potential of ciglitazone to induce apoptotic cell death and characterized the molecular mechanisms involved. In RT4 cells, the drug induced G2/M cell cycle arrest characterized by an overexpression of p53, p21(waf1/CIP1) and p27(Kip1) in concomitance with a decrease of cyclin B1. On the contrary, in T24 cells, it triggered apoptosis via extrinsic and intrinsic pathways. Cell cycle arrest and induction of apoptosis occurred at high concentrations through PPARγ activation-independent pathways. We show that in vivo treatment of nude mice by ciglitazone inhibits high grade bladder cancer xenograft development. We identified a novel mechanism by which ciglitazone kills cancer cells. Ciglitazone up-regulated soluble and membrane-bound TRAIL and let TRAIL-resistant T24 cells to respond to TRAIL through caspase activation, death receptor signalling pathway and Bid cleavage. We provided evidence that TRAIL-induced apoptosis is partially driven by ciglitazone-mediated down-regulation of c-FLIP and survivin protein levels through a proteasome-dependent degradation mechanism. CONCLUSIONS/SIGNIFICANCE: Therefore, ciglitazone could be clinically relevant as chemopreventive or therapeutic agent for the treatment of TRAIL-refractory high grade urothelial cancers.


Subject(s)
Apoptosis/drug effects , Hypoglycemic Agents/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Thiazolidinediones/pharmacology , Up-Regulation/drug effects , Urinary Bladder Neoplasms/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BH3 Interacting Domain Death Agonist Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Enzyme Activation/drug effects , G2 Phase/drug effects , Humans , Hypoglycemic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mitosis/drug effects , Neoplasm Grading , PPAR gamma/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Survivin , Thiazolidinediones/therapeutic use , Transcriptional Activation/drug effects , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/enzymology , Xenograft Model Antitumor Assays
8.
Int J Cancer ; 127(8): 1769-84, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20099277

ABSTRACT

Thiazolidinediones, including rosiglitazone and troglitazone, are insulin-sensitizing drugs and high-affinity ligands for the peroxisome proliferator-activated receptor gamma (PPARgamma). Apart from their antidiabetic activity, these molecules possess antitumor properties. We investigated their potential apoptotic effects on RT4 (derived from a well-differentiated Grade I papillary tumor) and T24 (derived from an undifferentiated Grade III carcinoma) bladder cancer cells. Rosiglitazone induced G2/M or G0/G1 phase cell cycle arrest in RT4 and T24 cells, respectively. Only troglitazone triggered apoptosis via extrinsic and intrinsic pathways in both cell lines. Interestingly, rosiglitazone amplified TRAIL-induced apoptosis in TRAIL-sensitive RT4 cells or let TRAIL-resistant T24 cells to respond to TRAIL. Thiazolidinediones acted through PPARgamma activation-independent mechanisms. The underlying mechanisms involved for the first time in cancer cells the upregulation of soluble and/or membrane-bound TRAIL. This was associated with increased cell surface death receptor 5 expression and c-FLIP and survivin downregulation, mediated in part through proteasome-dependent degradation in troglitazone-promoted cell death. Therefore, the combination of rosiglitazone and TRAIL could be clinically relevant as chemopreventive or therapeutic agents for the treatment of TRAIL-resistant high-grade urothelial cancers.


Subject(s)
Apoptosis/drug effects , PPAR gamma/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Thiazolidinediones/pharmacology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Survivin , Transcriptional Activation , Tumor Cells, Cultured , Urinary Bladder Neoplasms/drug therapy
9.
Urol Oncol ; 28(2): 180-8, 2010.
Article in English | MEDLINE | ID: mdl-19070520

ABSTRACT

OBJECTIVES: A particular interest in epithelial-mesenchymal transition (EMT), which takes place during embryonic development, provided potential mechanisms involved in the progression of many epithelial tumors, including bladder cancer (BC). The phospho-Akt signaling pathway is supposed to be involved in invasion and progression of human tumors, including BC. Moreover, it has been demonstrated in bladder cancer cell lines that N-cadherin or phospho-epithelial growth factor receptor (EGFR) expression are correlated to tumor progression. Our objectives were to evaluate the potential phospho-Akt pathway involvement in N-cadherin and/or phospho-EGFR positive BC cell lines and to evaluate the prognostic value of E- and N-cadherin expression in patients undergoing cystectomy for invasive BC. MATERIALS AND METHODS: We screened a panel of invasive and noninvasive BC cell lines for E- and N-cadherin, phospho-EGFR, and phospho-Akt expression using the Western blot technique (WB). The potential role of N-cadherin in invasion was assessed by Matrigel assays with and without the N-cadherin blocking monoclonal antibody GC-4. Then we used the Affymetrix microarray technique to evaluate the prognostic value of E- and N-cadherin expression in 30 patients undergoing a cystectomy for invasive BC. RESULTS: N-cadherin and phospho-EGFR expression are associated with Akt activation and with invasive behavior modulation. Even if Akt activation is sufficient in promoting invasion, its inactivation by LY294002 (PI-3 kinase inhibitor) is less efficient on invasion than inhibition of N-cadherin and phospho-EGFR by GC-4 (monoclonal antibody) and gefitinib (anti-tyrosine kinase), respectively. N-cadherin and phospho-EGFR inhibition decreased phospho-Akt activation but also caused restoration and reinforcing of E-cadherin expression, respectively, while phospho-Akt inhibition did not have any impact on E-cadherin expression. In a group of high-risk bladder tumors (T(1)G(3)), N- and E-cadherin expression could be considered as a prognostic marker. In a group of patients with invasive BC (pT(2)-T(4)) undergoing cystectomy, we showed a shorter overall survival when BC expressed N-cadherin (P = 0.0064) and when E-cadherin expression was down-regulated (P = 0.00165). The N (positive) /E (negative) profile has the worst prognosis (P = 0.00153). CONCLUSIONS: We confirmed the partial responsibility of p-Akt activation in invasion of some BC cell lines expressing N-cadherin or p-EGFR and also the potential role of N-cadherin and p-EGFR as target in cancer therapy. N/E- cadherin expression profile has a significant prognostic value in invasive BC.


Subject(s)
Cadherins/metabolism , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Urinary Bladder Neoplasms/metabolism , Biomarkers, Tumor/analysis , Blotting, Western , Cadherins/genetics , Cell Line, Tumor , ErbB Receptors/genetics , Gene Expression , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Neoplasm Invasiveness/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Prognosis , Proto-Oncogene Proteins c-akt/genetics , Reverse Transcriptase Polymerase Chain Reaction , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
10.
Oncol Rep ; 21(6): 1495-504, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19424629

ABSTRACT

The present investigation was conducted first to determine whether correlation exists between VEGF-A and -B mRNA levels and clinicopathological parameters and to assess their prognostic value in bladder cancer, then to clarify the expression level and biological significance of VEGF-A isoforms. Total RNA was isolated from 37 specimens of bladder cancer. Northern blot analysis revealed that VEGF-B mRNA was not expressed either in normal urothelium or in bladder cancer and detected three VEGF-A transcripts of 5.2, 4.5 and 1.7 kb in length, respectively. The VEGF-A transcript levels were greater in cancer tissues than in normal urothelium. They were significantly higher in pT2-T4 than in pTa and pT1 urothelial tumors and thus, were correlated to the pathologic stage. Contrary to the 4.5 kb transcript, elevated expression of the 5.2 and 1.7 kb transcripts was correlated with the histologic grade and the presence of carcinoma in situ. Patients with higher VEGF-A mRNA levels had a significantly shorter survival without progression compared to those with lower levels. Three VEGF-A splice variants were detected by southern blotting namely, VEGF121, 165 and 189. The expression intensity of each isoform was evaluated by quantitative real-time RT-PCR in 20 new fresh frozen recent tumors. VEGF121 and VEGF165 were expressed at the similar level. On the contrary, they were significantly more expressed than VEGF189 (p<0.05). The three isoforms were higher expressed in pT2 bladder cancers than in pTa tumors (p<0.05). There was only a significant correlation between the increased expression level of VEGF121 and 165 and the histological grade of the lesion (p<0.05). To conclude, VEGF-A mRNA level is a potential prognostic indicator of progression in bladder cancer as well as the expression level of the different VEGF-A splice variants.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma in Situ/genetics , Carcinoma, Transitional Cell/genetics , Urinary Bladder Neoplasms/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor B/genetics , Alternative Splicing , Blotting, Northern , Blotting, Southern , Carcinoma in Situ/mortality , Carcinoma in Situ/pathology , Carcinoma in Situ/therapy , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/therapy , Cell Line, Tumor , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Neoplasm Invasiveness , Neoplasm Staging , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Treatment Outcome , Up-Regulation , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy
11.
Int J Cancer ; 124(8): 1820-8, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19115207

ABSTRACT

Superficial pT1 bladder tumors are characterized by a high risk of recurrence and progression in grade and stage. Few studies provided evidence that loss of adipocyte-fatty acid binding protein (A-FABP) expression was associated with bladder cancer progression. A-FABP is a lipid binding protein playing a role in intracellular lipid transport and metabolism, as well as in signal transduction. We reported from bladder tumors that decrease of A-FABP transcript level significantly correlated to tumor stage and to histologic grade (p < 0.05). Namely, in poor prognosis high grade pT1 tumors there was a loss of A-FABP expression compared to good prognosis tumors suggesting that re-expression of A-FABP could be a therapeutic approach in early stage bladder cancer to prevent disease progression. We demonstrated for the first time that this marker is upregulated by Peroxisome Proliferator-Activated Receptor (PPAR) alpha, beta and gamma in T24 cells (derived from an undifferentiated grade III carcinoma) and only by PPARbeta in RT4 cells (derived from a well differentiated grade I papillary tumor). This effect occurred through a PPAR-dependent transcriptional mechanism without modifying mRNA stability and interestingly required de novo protein synthesis. Data as a whole suggest a prognostic significance of A-FABP in bladder cancer outcome and the potential utility of overexpression of this protein by PPAR agonists open up new perspectives in the treatment of bladder cancer. (c) 2008 Wiley-Liss, Inc.


Subject(s)
Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Peroxisome Proliferator-Activated Receptors/metabolism , Urinary Bladder Neoplasms/metabolism , Urothelium/metabolism , Adult , Aged , Aged, 80 and over , Disease Progression , Fatty Acid-Binding Proteins/metabolism , Humans , Middle Aged , Signal Transduction
12.
Exp Cell Res ; 313(15): 3239-50, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17678892

ABSTRACT

Altered angiogenic response is associated with high-grade cervical dysplasia and with invasive squamous carcinoma of the cervix. Vascular Endothelial Growth Factor (VEGF) is one of the most potent inducers of angiogenesis and is up-regulated in carcinoma of the cervix. Infection by high-risk human papillomavirus and persistent expression of viral oncogene E6 are etiologically linked to the development of cervical cancer. E6 is able to immortalize cells and induce malignant transformation by inactivating p53. In cervical cancer, regulation of VEGF expression is poorly described. Thus, we investigated whether E6 oncoprotein could regulate VEGF expression in HPV18-positive cervical cancer-derived HeLa cells harboring a wild-type p53. The alternative splicing of vegf mRNA renders three major isoforms of 121, 165 and 189 amino-acids in humans. We have designed isoform specific real time QRT-PCR assays to quantitate vegf transcripts and VEGF121 was the predominant isoform. Silencing HPV18 E6 mRNA with specific siRNA reduced VEGF121 expression by at least 50% whereas silencing of p53 did not alter its expression. Treatment with cycloheximide did not inhibit E6-induced VEGF121 expression. Collectively, these results suggest that HPV18 E6 oncoprotein contributes to tumor angiogenesis by inducing VEGF transcription from the promoter in a p53-independent manner.


Subject(s)
DNA-Binding Proteins/physiology , Human papillomavirus 18/metabolism , Oncogene Proteins, Viral/physiology , Tumor Suppressor Protein p53/physiology , Vascular Endothelial Growth Factor A/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/virology , Alternative Splicing , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vascular Endothelial Growth Factor A/genetics
13.
Clin Cancer Res ; 12(9): 2780-7, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16675571

ABSTRACT

PURPOSE: Loss of intercellular adhesion and increased cell motility promote tumor cell invasion and spreading. In bladder cancer, loss or reduced E-cadherin expression has been associated with poor survival, and aberrant expression of N-cadherin has been associated with the invasive phenotype of bladder carcinoma cells. The purpose of this study was to investigate whether N-cadherin expression was associated with the bladder tumor progression. EXPERIMENTAL DESIGN: E-cadherin and N-cadherin expression was evaluated by immunohistochemistry in 101 tumors (pT1 and pT2-T3) and by reverse transcription-PCR analysis and immunohistochemistry in 28 other fresh frozen tumors (pT(a), pT1, and pT2-T3). RESULTS: N-cadherin expression was absent in normal urothelium, appeared in stage pT1, and increased in pT2-pT3 tumors. In most cases, increased N-cadherin expression in invasive tumors was associated with loss of E-cadherin expression. Progression-free survival and multivariate analyses revealed that N-cadherin expression is an independent prognostic marker for pT1 tumor progression. Analysis of the 28 frozen tumors by immunohistochemistry and reverse transcription-PCR showed a good correlation between protein and gene expression in pT1 and pT2-T3 tumors. Interestingly, in pT(a) tumors, N-cadherin was not immunodetected, whereas mRNA was present in 50% of cases. CONCLUSION: Regulatory defects in the N-cadherin promoter, abnormalities at the translational, or protein processing levels could explain the discrepancies between protein and mRNA expression. Most importantly, this study identified N-cadherin as a novel prognostic marker of progression in superficial urothelial tumors. Clearly, N-cadherin acts in an invasive mode in bladder cancer, but whether it has a primary role in urothelial neoplastic progression has yet to be investigated.


Subject(s)
Cadherins/genetics , Urinary Bladder Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Staging , Polymerase Chain Reaction , Prognosis , Reproducibility of Results , Urinary Bladder Neoplasms/surgery
14.
Am J Clin Pathol ; 125(1): 119-26, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16483000

ABSTRACT

Loss of intercellular adhesion facilitates tumor invasion. To clarify the relation between altered expression of cell adhesion molecules and progression of T1 superficial bladder tumors, 101 cases (71 T1 tumors, 30 T2/T3 tumors) were examined immunohistochemically for E-cadherin and alpha-, beta-, and gamma-catenins. A highly significant correlation was observed between the decreased expression of all molecules and increased TNM stage (P < .001). Univariate analysis, performed in cases of T1 tumors, revealed association of abnormal E-cadherin with beta-catenin diminution. Survival curves were established with the Kaplan-Meier method and analyzed according to clinical and histopathologic parameters using the log-rank test. Cox multivariate analysis revealed only gamma-catenin as an independent predictor of progression-free survival in patients with stage T1 bladder urothelial tumors. The characterization of T1 tumors that will progress could lead to the identification of patients who might benefit from surgery to avoid vesical muscle invasion and, consequently, metastasis.


Subject(s)
Biomarkers, Tumor/analysis , Cadherins/biosynthesis , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Disease Progression , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , alpha Catenin/biosynthesis , beta Catenin/biosynthesis , gamma Catenin/biosynthesis , Aged , Female , Humans , Male , Muscle Neoplasms/metabolism , Muscle Neoplasms/secondary , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survival Analysis , Urothelium/metabolism , gamma Catenin/analysis
15.
J Biol Chem ; 277(26): 23534-43, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-11980898

ABSTRACT

The growth of any solid tumor depends on angiogenesis. Vascular endothelial growth factor (VEGF) plays a prominent role in vesical tumor angiogenesis regulation. Previous studies have shown that the peroxisome proliferator-activated receptor gamma (PPARgamma) was involved in the angiogenesis process. Here, we report for the first time that in two different human bladder cancer cell lines, RT4 (derived from grade I tumor) and T24 (derived from grade III tumor), VEGF (mRNA and protein) is differentially up-regulated by the three PPAR isotypes. Its expression is increased by PPARalpha, beta, and gamma in RT4 cells and only by PPARbeta in T24 cells via a transcriptional activation of the VEGF promoter through an indirect mechanism. This effect is potentiated by an RXR (retinoid-X-receptor), selective retinoid LG10068 providing support for a PPAR agonist-specific action on VEGF expression. While investigating the downstream signaling pathways involved in PPAR agonist-mediated up-regulation of VEGF, we found that only the MEK inhibitor PD98059 reduced PPAR ligand-induced expression of VEGF. These data contribute to a better understanding of the mechanisms by which PPARs regulate VEGF expression. They may lead to a new therapeutic approach to human bladder cancer in which excessive angiogenesis is a negative prognostic factor.


Subject(s)
Endothelial Growth Factors/genetics , Gene Expression Regulation, Neoplastic , Lymphokines/genetics , Neoplasm Proteins , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins , Urinary Bladder Neoplasms/metabolism , Carrier Proteins/physiology , Culture Media, Conditioned , Endothelial Growth Factors/analysis , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Humans , Lymphokines/analysis , Mitogen-Activated Protein Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...