Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 11(1)2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30583471

ABSTRACT

Glioblastoma multiform (GBM) tumors are very heterogeneous, organized in a hierarchical pattern, including cancer stem cells (CSC), and are responsible for development, maintenance, and cancer relapse. Therefore, it is relevant to establish new GBM cell lines with CSC characteristics to develop new treatments. A new human GBM cell line, named R2J, was established from the cerebro-spinal fluid (CSF) of a patient affected by GBM with leptomeningeal metastasis. R2J cells exhibits an abnormal karyotype and form self-renewable spheres in a serum-free medium. Original tumor, R2J, cultured in monolayer (2D) and in spheres showed a persistence expression of CD44, CD56 (except in monolayer), EGFR, Ki67, Nestin, and vimentin. The R2J cell line is tumorigenic and possesses CSC properties. We tested in vitro the anticancer effects of sodium selenite (SS) compared to temozolomide TMZ. SS was absorbed by R2J cells, was cytotoxic, induced an oxidative stress, and arrested cell growth in G2M before inducing both necrosis and apoptosis via caspase-3. SS also modified dimethyl-histone-3-lysine-9 (H3K9m2) levels and decreased histone deacetylase (HDAC) activity, suggesting anti-invasiveness potential. This study highlights the value of this new GBM cell line for preclinical modeling of clinically relevant, patient specific GBM and opens a therapeutic window to test SS to target resistant and recurrent GBM.

2.
Oncotarget ; 9(53): 30066-30078, 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-30046388

ABSTRACT

Potent inhibitors of PI3K (GDC-0941) and Src (Saracatinib) exhibit as individual agents, excellent oral anticancer activity in preclinical models and have entered phase II clinical trials in various cancers. We found that PI3K and Src kinases are dysregulated in clear cell renal carcinomas (ccRCCs), an aggressive disease without effective targeted therapies. In this study we addressed this challenge by testing GDC-0941 and Saracatinib as either single agents or in combination in ccRCC cell lines, as well as in mouse and PDX models. Our findings demonstrate that combined inhibition of PI3K and Src impedes cell growth and invasion and induces cell death of renal carcinoma cells providing preclinical evidence for a pairwise combination of these anticancer drugs as a rational strategy to improve renal cancer treatment.

3.
J Trace Elem Med Biol ; 44: 161-176, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28965572

ABSTRACT

Glioblastoma (GBM) is the most common type of primary tumor of the central nervous system with a poor prognosis, needing the development of new therapeutic drugs. Few studies focused on sodium selenite (SS) effects in cancer cells cultured as multicellular tumor spheroids (MCTS or 3D) closer to in vivo tumor. We investigated SS anticancer effects in three human GBM cell lines cultured in 3D: LN229, U87 (O(6)-methyguanine-DNA-methyltransferase (MGMT) negative) and T98G (MGMT positive). SS absorption was evaluated and the cytotoxicity of SS and temozolomide (TMZ), the standard drug used against GBM, were compared. SS impacts on proliferation, cell death, and invasiveness were evaluated as well as epigenetic modifications by focusing on histone deacetylase (HDAC) activity and dimethyl-histone-3-lysine-9 methylation (H3K9m2), after 24h to 72h SS exposition. SS was absorbed by spheroids and was more cytotoxic than TMZ (i.e., for LN229, the IC50 was 38 fold-more elevated for TMZ than SS, at 72h). SS induced a cell cycle arrest in the S phase and apoptosis via caspase-3. SS decreased carbonic anhydrase-9 (CA9) expression, invasion on a Matrigel matrix and modulated E- and N-Cadherin transcript expressions. SS decreased HDAC activity and modulated H3K9m2 levels. 3D model provides a relevant strategy to screen new drugs and SS is a promising drug against GBM that should now be tested in GBM animal models.


Subject(s)
Glioblastoma/drug therapy , Glioblastoma/pathology , Sodium Selenite/therapeutic use , Spheroids, Cellular/pathology , Antigens, Neoplasm/metabolism , Apoptosis/drug effects , Cadherins/metabolism , Carbonic Anhydrase IX/metabolism , Caspase 3/metabolism , Cell Aggregation/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Humans , Necrosis , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium Selenite/pharmacology , Spheroids, Cellular/drug effects
4.
Anticancer Agents Med Chem ; 16(4): 490-500, 2016.
Article in English | MEDLINE | ID: mdl-26286659

ABSTRACT

AIMS: Selenium (Se) is an essential trace element for human health which also has antitumor properties. Little is known about its effects on brain tumor cells (BTC). The aim of this study was to investigate the anticancer effects of sodium selenite (SS) including histone deacetylase (HDAC) activity in three human glioblastoma (GBM) cell lines (LN229, T98G and U87). MATERIALS & METHODS: LN229, T98G and U87 GBM cell lines were treated with variable doses of SS for time varying from 24 to 72h. HDAC activity, cell proliferation, toxicity, cell death process, caspase-3 and MMP2 activities and Se absorption were evaluated. RESULTS: SS modulated all the parameters tested in a dose- and time-dependent manner. We found that SS decreased HDAC activity, blocked cell proliferation and cell cycle at the G2 phase, triggered an apoptotic cell death process caspase-3-dependent and reduced MMP2 activities. All these effects were performed whereas SS was weakly absorbed (<2%). CONCLUSIONS: SS decreasing HDAC activity exhibited interesting antitumor properties in GBM cells which may be taken into account in the novel strategies for achieving tumor growth inhibition and cytotoxicity. Epigenetic modifications induced by SS should be evaluated in further studies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Glioblastoma/enzymology , Glioblastoma/pathology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Sodium Selenite/pharmacology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioblastoma/drug therapy , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Metallomics ; 6(9): 1683-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24994457

ABSTRACT

Selenium (Se) is an essential trace element with a narrow safety zone and unclear effects on skin photoageing. The aim of this work was to investigate the photoageing properties of sodium selenite or selenomethionine (SeMet) after a long term (6 days) Se supplementation in normal human skin fibroblasts (NHSF) subjected to ultraviolet-A (UVA) irradiation inducing 30% cell death. The uptake, toxicity and antioxidant effects of sodium selenite and SeMet were compared to better understand their photoageing properties. SeMet uptake was better than sodium selenite and their uptake by fibroblasts was not via an actively transport process. Sodium selenite induced a higher toxicity than SeMet. At 5 µM, sodium selenite inhibited cell proliferation associated with a blockage in the G2 phase and induced DNA fragmentation leading to caspase-3-dependent apoptosis cell death. At low doses (<1 µM), SeMet and sodium selenite induced glutathione peroxidase-1 (GPX1) activity and selenoproteinW1 (SEPW1) transcript expression but metalloproteinase (MMP)-1 was only induced by sodium selenite. SeMet and sodium selenite did not protect NHSFs from UVA-induced cell death. However, SeMet decreased malondialdehyde (MDA) and protected NHSFs from UVA-induced MMP1 and MMP3. We then observed a large difference in terms of photoprotection according to selenium forms. SeMet may be a potential agent for the prevention and treatment of skin photoageing.


Subject(s)
Cytoprotection/radiation effects , Fibroblasts/drug effects , Fibroblasts/radiation effects , Selenium/pharmacology , Selenium/toxicity , Skin/cytology , Ultraviolet Rays , Antioxidants/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cytoprotection/drug effects , Fibroblasts/cytology , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Selenium/metabolism , Selenomethionine/pharmacology , Selenomethionine/toxicity , Sodium Selenite/pharmacology , Sodium Selenite/toxicity , Spectrophotometry, Atomic
6.
Anticancer Agents Med Chem ; 14(6): 862-71, 2014.
Article in English | MEDLINE | ID: mdl-24568163

ABSTRACT

The present work describes the anticancer activity of a new indolylcoumarin named COUFIN and more specifically, its efficiency against clear cell renal carcinoma (CCRC). COUFIN inhibited microtubule formation and bound on tubulin to or near the colchicine site. In vitro, COUFIN showed potent anticancer activity on renal carcinoma cells (RCC) both in monolayer (2D culture) (IC50 of 88 ± 8 nM) and multicellular tumor spheroid (3D culture) (IC50 of 180 ± 20 nM). The compound blocked cell cycle transition at G2/M phase, induced a subsequent apoptotic process but did not modulate clonal growth of CFU-GM. On the other hand, the coumarin derivative decreased the activity of P-gp and BCRP but was not substrate for these ABC pumps. In vivo, the indolylcoumarin increased the survival rate after 3 weeks of treatment. Based on the present study, COUFIN was identified as a bifunctional molecule able to inhibit renal carcinoma cells proliferation without being effluxed by ABC proteins. Thus COUFIN could be a promising chemotherapeutic agent for treating tumor cells over-expressing efflux pumps and tumor cells irrigated by vessels lined with endothelial cells responsible of poor distribution of conventional anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Coumarins/pharmacology , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hematopoietic System , Humans , Indoles/chemical synthesis , Indoles/chemistry , Mice , Mice, Inbred C57BL , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Toxicol In Vitro ; 27(8): 2305-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24134853

ABSTRACT

Chalcones are naturally occurring compounds with diverse pharmacological activities. Chalcones derive from the common structure: 1,3-diphenylpropenone. The present study aims to better understand the mechanistic pathways triggering chalcones anticancer effects and providing evidences that minor structural difference could lead to important difference in mechanistic effect. We selected two recently investigated chalcones (A and B) and investigated them on glioblastoma cell lines. It was found that chalcone A induced an apoptotic process (type I PCD), via the activation of caspase-3, -8 and -9. Chalcone A also increased CDK1/cyclin B ratios and decreased the mitochondrial transmembrane potential (ΔΨm). Chalcone B induced an autophagic cell death process (type II PCD), ROS-related but independent of both caspases and protein synthesis. Both chalcones increased Bax/Bcl2 ratios and decreased Ki67 and CD71 antigen expressions. The present investigation reveals that despite the close structure of chalcones A and B, significant differences in mechanism of effect were found.


Subject(s)
Antineoplastic Agents/pharmacology , Chalcones/pharmacology , Catalase/genetics , Cell Cycle/drug effects , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Glutathione Peroxidase/genetics , Humans , Malondialdehyde/metabolism , Mitotic Index , Reactive Oxygen Species/metabolism , Glutathione Peroxidase GPX1
8.
Biol Trace Elem Res ; 154(2): 288-98, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23771685

ABSTRACT

The beneficial effect of selenium (Se) on cancer is known to depend on the chemical form, the dose and the duration of the supplementation. The aim of this work was to explore long term antagonist (antioxidant versus toxic) effects of an inorganic (sodium selenite, Na2SeO3) and an organic (seleno-L-methionine, SeMet) forms in human immortalized keratinocytes HaCaT cells. HaCaT cells were supplemented with Na2SeO3 or SeMet at micromolar concentrations for 144 h, followed or not by UVA radiation. Se absorption, effects of UVA radiation, cell morphology, antioxidant profile, cell cycle processing, DNA fragmentation, cell death triggered and caspase-3 activity were determined. At non-toxic doses (10 µM SeMet and 1 µM Na2SeO3), SeMet was better absorbed than Na2SeO3. The protection of HaCaT from UVA-induced cell death was observed only with SeMet despite both forms increased glutathione peroxidase-1 (GPX1) activities and selenoprotein-1 (SEPW1) transcript expression. After UVA irradiation, malondialdehyde (MDA) and SH groups were not modulated whatever Se chemical form. At toxic doses (100 µM SeMet and 5 µM Na2SeO3), Na2SeO3 and SeMet inhibited cell proliferation associated with S-G2 blockage and DNA fragmentation leading to apoptosis caspase-3 dependant. SeMet only led to hydrogen peroxide production and to a decrease in mitochondrial transmembrane potential. Our study of the effects of selenium on HaCaT cells reaffirm the necessity to take into account the chemical form in experimental and intervention studies.


Subject(s)
G2 Phase Cell Cycle Checkpoints/drug effects , Keratinocytes/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Selenomethionine , Sodium Selenite , Trace Elements , Caspase 3/metabolism , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Transformed , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/radiation effects , Humans , Hydrogen Peroxide/metabolism , Keratinocytes/pathology , Malondialdehyde/metabolism , S Phase Cell Cycle Checkpoints/radiation effects , Selenium/adverse effects , Selenium/pharmacology , Selenomethionine/adverse effects , Selenomethionine/pharmacology , Sodium Selenite/adverse effects , Sodium Selenite/pharmacology , Trace Elements/adverse effects , Trace Elements/pharmacology , Ultraviolet Rays/adverse effects
9.
Cancer Genomics Proteomics ; 7(2): 87-92, 2010.
Article in English | MEDLINE | ID: mdl-20335523

ABSTRACT

BACKGROUND: In a previous investigation, we showed that the janus kinase (JNK) inhibitor SP600125 induced several phenotypic and genomic changes in leukemia cells. However, the molecular mechanisms that sustain these changes remain unknown. The purpose of the present study was to examine gene expression changes in THP-1 leukemia cells treated with SP600125. MATERIALS AND METHODS: Gene expression levels were investigated using Affymetrix hybridization technology and quantitative reverse transcriptase polymerase chain reaction. RESULTS: Affymetrix technology showed that the expression of 1,038 genes with a biological process description well known in gene ontology was modulated. Fifteen genes were related to kinases or phosphatases, 20 genes were involved in the cell cycle regulation, and 23 genes were involved in apoptosis. A network of 15 correlated genes was obtained showing a primordial role for the myelocytomatosis viral oncogene homolog (MYC). CONCLUSION: These findings show that SP600125 exhibits cytostatic and cytolytic activities through MYC gene modulation.


Subject(s)
Anthracenes/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oligonucleotide Array Sequence Analysis/methods , Cell Line, Tumor , Cluster Analysis , Down-Regulation/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Genes, Neoplasm/genetics , Humans , Reproducibility of Results , Up-Regulation/drug effects
10.
BMC Cancer ; 9: 242, 2009 Jul 20.
Article in English | MEDLINE | ID: mdl-19619277

ABSTRACT

BACKGROUND: Over the past decades, in spite of intensive search, no significant increase in the survival of patients with glioblastoma has been obtained. The role of the blood-brain barrier (BBB) and especially the activity of efflux pumps belonging to the ATP Binding Cassette (ABC) family may, in part, explain this defect. METHODS: The in-vitro activities of JAI-51 on cell proliferation were assessed by various experimental approaches in four human and a murine glioblastoma cell lines. Using drug exclusion assays and flow-cytometry, potential inhibitory effects of JAI-51 on P-gp and BCRP were evaluated in sensitive or resistant cell lines. JAI-51 activity on in-vitro microtubule polymerization was assessed by tubulin polymerization assay and direct binding measurements by analytical ultracentrifugation. Finally, a model of C57BL/6 mice bearing subcutaneous GL26 glioblastoma xenografts was used to assess the activity of the title compound in vivo. An HPLC method was designed to detect JAI-51 in the brain and other target organs of the treated animals, as well as in the tumours. RESULTS: In the four human and the murine glioblastoma cell lines tested, 10 muM JAI-51 inhibited proliferation and blocked cells in the M phase of the cell cycle, via its activity as a microtubule depolymerising agent. This ligand binds to tubulin with an association constant of 2 x 105 M-1, overlapping the colchicine binding site. JAI-51 also inhibited the activity of P-gp and BCRP, without being a substrate of these efflux pumps. These in vitro studies were reinforced by our in vivo investigations of C57BL/6 mice bearing GL26 glioblastoma xenografts, in which JAI-51 induced a delay in tumour onset and a tumour growth inhibition, following intraperitoneal administration of 96 mg/kg once a week. In accordance with these results, JAI-51 was detected by HPLC in the tumours of the treated animals. Moreover, JAI-51 was detected in the brain, showing that the molecule is also able to cross the BBB. CONCLUSION: These in vitro and in vivo data suggest that JAI-51 could be a good candidate for a new treatment of tumours of the CNS. Further investigations are in progress to associate the title compound chemotherapy to radiotherapy in a rat model.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain Neoplasms/metabolism , Chalcone/analogs & derivatives , Chalcones/pharmacology , Glioblastoma/metabolism , Microtubules/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Blood-Brain Barrier , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcr/antagonists & inhibitors , Rats
11.
Exp Hematol ; 36(1): 28-36, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17949889

ABSTRACT

OBJECTIVE: The roles of phosphatidylinositol 3 (PI3K) and mitogen-activated protein kinases (MAPK) have been widely studied in terms of the differentiation process induced by several drugs (phorbol ester, vitamin D-3, retinoic acid, etc.), but their exact functions in leukemic cells' phenotype and their potential therapeutic role remain incompletely clarified. MATERIALS AND METHODS: In order to investigate this query, leukemia cells were cultured in presence of kinase inhibitors (KIs). Proliferation, apoptosis, and differentiation were analyzed at the cellular and molecular levels, using flow cytometry and reverse transcriptase quantitative polymerase chain reaction. RESULTS: SB203580, a P38 MAPK inhibitor, had no effect on cell proliferation, whereas LY294002, a PI3K inhibitor, and PD098059, a selective inhibitor of mitogen-activated extracellular regulated kinase (MEK) phosphorylation, arrested cells in G(0)/G(1). However, LY294002 and PD098059 acted using different mechanisms: LY294002 decreased the expression of phosphorylated S6RP, whereas PD098059 increased P21/waf1 antigen expression. SP600125, an inhibitor of N-terminal c-jun kinases, arrested cells in G(2) and induced an endoreplicative process. SP600125 increased p21 at both the mRNA and protein levels. G(2) blockage is dependent on the PI3K pathway and the endoreplicative process is dependent on the PI3K and extracellular regulated kinase (ERK) pathways and mRNA synthesis. On the other hand, PD098059 potentiated the apoptotic process induced by either SP600125 or LY294002. Modulation of the expression of CD11, CD15, CD18, and CD54 was cell-dependent. CONCLUSION: Our results suggest that KIs modulate proliferation of leukemia cells and that the MEK/ERK inhibitor, PD098059, in combination with either SP600125 or LY294002, could have clinical value.


Subject(s)
Leukemia/enzymology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Anthracenes/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Chromones/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Flavonoids/pharmacology , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells/drug effects , HL-60 Cells/enzymology , Humans , Imidazoles/pharmacology , Leukemia/drug therapy , Leukemia/pathology , Monocytes/drug effects , Monocytes/enzymology , Morpholines/pharmacology , Pyridines/pharmacology , U937 Cells/drug effects , U937 Cells/enzymology
12.
Bioorg Med Chem Lett ; 17(1): 208-13, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17049235

ABSTRACT

2-Arylidenedihydroindole-3-ones were assayed for their antiproliferative and apoptotic abilities as potential drug candidates to treat bladder tumor. These compounds were tested on cell lines obtained from bladder tumors of various stages [superficial (pTa and pT1) vs. invasive (pT2)]. The most active compound (3c) inhibited the proliferation, induced apoptosis, and decreased the expression of p-Stat5 and p-Pyk2 in DAG-1 and RT112 lines in which the FGFR3 is either mutated or overexpressed. Knowing that FGFR3 is involved in cell proliferation, differentiation, and migration through cell signaling pathways including p-Stat5 way via p-Pyk2, let us assume that compound 3c may probably act through FGFR3 pathway.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma/enzymology , Indoles/chemistry , Indoles/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Urinary Bladder Neoplasms/enzymology , Antineoplastic Agents/chemical synthesis , Apoptosis , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Focal Adhesion Kinase 2/metabolism , Humans , Indoles/chemical synthesis , Neoplasm Staging , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Urinary Bladder Neoplasms/pathology
13.
Leuk Res ; 30(11): 1407-16, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16757024

ABSTRACT

UM384 cells, derived from the human myeloid leukemia U937 cell line, fail to differentiate in response to 12-O-tetradecanoylphorbol-13-acetate (TPA). Using cDNA microarray and real-time quantitative PCR (RT-QPCR) approaches, we observed a difference in the response to TPA treatment: all the genes from U937 cells were continuously modulated from 2 to 24h. In UM384 cells, 60% of the genes were transiently modulated at 2h, then returned to control levels at 24h. Moreover, HuR, an AU-rich element-binding protein (ARE-BP), was differentially located in the two cell lines. Therefore, a defect of mRNA stabilization could be responsible for the resistance of UM384 cells to TPA-induced differentiation, suggesting a possible role for the post-transcriptional regulation in the leukemogenesis.


Subject(s)
Cell Differentiation/drug effects , Drug Resistance, Neoplasm , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , RNA Stability , RNA, Messenger/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Profiling , HL-60 Cells , Humans , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/genetics , Leukemia, Myeloid/genetics , Naphthalenes/pharmacology , Oligonucleotide Array Sequence Analysis/methods , Protein Kinase C/drug effects , Protein Kinase C/genetics , Protein Kinase C beta , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Structure-Activity Relationship , Transcription, Genetic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
14.
Eur Urol ; 48(5): 846-51, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16046050

ABSTRACT

OBJECTIVE: Investigating whether extracellular factors are possible actors in tumoral progression in bladder carcinoma. METHODS: RT112/G2 bladder tumour cells were grown in presence of TGFbeta and analysed by immunological and cDNA microarray techniques. RESULTS: TGFbeta inhibited cell proliferation, reduced TNFalpha- and IFNgamma-induced apoptosis by decreasing TNFalpha-RI and IFNgamma-R antigen expression. It also inhibited cleaved caspase 8 and 9 expression, decreased E-cadherin, and increased BclxL and cyclooxygenase-2 expression. The cDNA microarray approach showed that TGFbeta up-regulated the expression of genes with defined roles in tumoral progression sometimes associated with poor outcome in bladder cancer. CONCLUSION: These results suggest that a part of the bladder tumoral progression process may be related to the action of exogenous TGFbeta confirming the possible role for the microenvironment.


Subject(s)
Transforming Growth Factor beta/metabolism , Urinary Bladder Neoplasms/metabolism , Apoptosis/physiology , Cadherins/genetics , Cadherins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Interferon-gamma/metabolism , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transforming Growth Factor beta/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Interferon gamma Receptor
15.
Cancer Genomics Proteomics ; 1(5-6): 455-464, 2004.
Article in English | MEDLINE | ID: mdl-31394635

ABSTRACT

BACKGROUND: TNFα and IFNγ, two main cytokines secreted in the urine of bladder cancer patients after Bacillus Calmette Guerin immunotherapy (BCG therapy), exert various responses ranging from growth arrest, apoptosis, phenotypic changes and differentiation. MATERIALS AND METHODS: To identify their transcriptional and translational targets, the highly sensitive bladder cancer cell line (RT112) was treated for 24 hours with increasing doses of IFNγ or TNFα and analyzed for cellular and molecular changes using a cDNA microarray technique (Transcriptome) containing 800 genes. RESULTS: High doses (>10 ng/ml) induced an apoptotic cell death, whereas low doses (<5 ng/ml) induced a survival program. TNFα-inducible genes, IFNγ-inducible genes and genes modulated by TNFα and IFNγ together were identified. All were related to the tumor progression program including cell proliferation, apoptosis/survival, angiogenesis and metastatic processes. CONCLUSION: These results suggest that the transcriptomic approach could be a good methodology to determine the molecular mechanisms involved in bladder tumor progression processes in relation to a low response to BCG treatment. However, mRNA and protein expression did not always correlate, suggesting that translational regulation is a vital process in bladder tumor progression.

16.
Clin Cancer Res ; 9(12): 4562-9, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14555531

ABSTRACT

PURPOSE: IFN-gamma is detected in the urine of bladder cancer patients after intravesical bacillus Calmette Guerin instillation. Because it acts in the anticancer process, we studied its cellular and molecular mechanisms of action on human bladder cancer cell lines. RESULTS: IFN-gamma (>5 ng.ml(-1))(>400 IU.ml(-1)) inhibited the growth of bladder cancer cell lines and modified the expression of the tumor-associated markers tissue-type plasminogen activators, Plasminogen activator inhibitor-2, urokinase plasminogen activator receptor, colony-stimulating factor 1, intercellular adhesion molecule 1, and class II MHC. Interestingly, IFN-gamma-induced apoptosis of the low-grade bladder cancer cell lines (RT4/G1 and RT112/G2) related to a cleavage of caspases 1, 8, and 9. This process was inhibited by the phosphatidylinositol 3'-kinase inhibitor (LY294002) and the protein synthesis inhibitor (cycloheximide). Moreover, low doses of IFN-gamma (<5 ng.ml(-1))(<400 IU.ml(-1)) increased the resistance to the cytotoxic effect of tumor necrosis factor alpha in the RT112 cells but not in the RT4 cells. This acquired resistance was associated with morphological changes and with an increase of the cell migration and scattering. CONCLUSIONS: We demonstrated that in the low-grade bladder cancer cell lines, the effect of IFN-gamma was dose dependent: high doses (>5 ng.ml(-1)) induced apoptosis of RT4 and RT112 cells, whereas low doses (<5 ng.ml(-1)) induced a resistance to the cytotoxic effect of tumor necrosis factor alpha and increase the metastatic potential of the RT112 cells. Therefore, we propose that a similar phenomenon could participate to the immunotherapy failure observed during tumor progression of bladder cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Interferon-gamma/pharmacology , Urinary Bladder Neoplasms/pathology , Biomarkers, Tumor/metabolism , Caspases/metabolism , Cell Division/drug effects , Cell Movement/drug effects , Drug Resistance, Neoplasm , Hepatocyte Growth Factor/metabolism , Humans , Signal Transduction , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/secondary
17.
Urol Res ; 30(5): 301-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12389118

ABSTRACT

The expression of plasminogen- and colony-stimulating factor-1-associated markers was first investigated in seven bladder carcinoma cell lines and in 15 primary bladder tumors using RT-PCR (mRNAs), zymography (protein activity), ELISA and immunocytochemistry analysis (ICC) (protein levels). The mRNAs expression, the activity and the levels of the secreted proteins were not informative. Only urokinase plasminogen activator receptor (uPA-R/CD87) and possibly plasminogen activator inhibitor type-2 (PAI2) antigen expression at the cellular levels seem to be useful markers. uPA-R antigen expression correlated with the secretion of hepatocyte growth factor (HGF) ( P=0.016) and the motility of the bladder tumor cells ( P=0.014), two markers associated with a poor prognosis in bladder carcinoma. To validate our technique and confirm these preliminary results, uPA-R and PAI2 antigen expression was determined in the imprints from 129 resected bladder carcinoma fragments. uPA-R correlated with the grade ( P=0.002), tumor invasion ( P=0.003) and the ploidy ( P=0.05) of the bladder carcinomas and with the low overall survival ( P=0.045) of the patients. PAI2 correlated only with the stage ( P=0.02) and low overall survival ( P=0.038). We conclude that in bladder carcinomas, studying the transcripts of PAs, PAIs, CSF-1 and its receptor, as well as measuring their concentration or activity in culture supernatants was of no clinical interest in terms of diagnostic or prognostic value. Only the ICC of uPA-R, which correlated with the major histopathological parameters of tumors and the low overall survival, proved to be a diagnostic and prognostic marker.


Subject(s)
Carcinoma/diagnosis , Carcinoma/metabolism , Plasminogen Activator Inhibitor 2/metabolism , Receptors, Cell Surface/metabolism , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma/pathology , Female , Hepatocyte Growth Factor/metabolism , Humans , Immunohistochemistry , Macrophage Colony-Stimulating Factor/metabolism , Male , Middle Aged , Neoplasm Invasiveness/pathology , Plasminogen/metabolism , Prognosis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...