Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(5)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36900323

ABSTRACT

BACKGROUND: The coagulome, defined as the repertoire of genes that locally regulate coagulation and fibrinolysis, is a key determinant of vascular thromboembolic complications of cancer. In addition to vascular complications, the coagulome may also regulate the tumor microenvironment (TME). Glucocorticoids are key hormones that mediate cellular responses to various stresses and exert anti-inflammatory effects. We addressed the effects of glucocorticoids on the coagulome of human tumors by investigating interactions with Oral Squamous Cell Carcinoma, Lung Adenocarcinoma, and Pancreatic Adenocarcinoma tumor types. METHODS: We analyzed the regulation of three essential coagulome components, i.e., the tissue factor (TF), urokinase-type plasminogen activator (uPA), and plasminogen activator inhibitor-1 (PAI-1) in cancer cell lines exposed to specific agonists of the glucocorticoid receptor (GR) (dexamethasone and hydrocortisone). We used QPCR, immunoblots, small-interfering RNA, Chromatin immunoprecipitation sequencing (ChIPseq) and genomic data from whole tumor and single-cell analyses. RESULTS: Glucocorticoids modulate the coagulome of cancer cells through a combination of indirect and direct transcriptional effects. Dexamethasone directly increased PAI-1 expression in a GR-dependent manner. We confirmed the relevance of these findings in human tumors, where high GR activity/high SERPINE1 expression corresponded to a TME enriched in active fibroblasts and with a high TGF-ß response. CONCLUSION: The transcriptional regulation of the coagulome by glucocorticoids that we report may have vascular consequences and account for some of the effects of glucocorticoids on the TME.

2.
Transl Oncol ; 14(8): 101110, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33951601

ABSTRACT

OBJECTIVES: The immune checkpoint molecule PD-L1 (CD274) is a crucial regulator of the tumor immune response. Its expression has been reported in the therapeutic context in Head and Neck Squamous Cell Carcinoma (HNSCC), but it remains unclear how therapeutically approved molecules regulate PD-L1 expression in HNSCC cells. MATERIALS AND METHODS: Three HNSCC cell lines (BICR6, PE/CA-PJ34 and PE/CA-PJ41) were used to analyze PD-L1 expression by immunoblotting, immunofluorescence and QPCR. Freely-available single cell RNAseq data from HNSCC were also used. RESULTS: 5-Fluorouracil (5-FU) increased the expression of PD-L1 with high efficacy in HNSCC cells. Single cell RNAseq data suggested the specificity of the regulation of PD-L1 in this context. The effect of 5-FU on PD-L1 expression was related to its genotoxic effect and was prevented by extracellular application of thymidine or using a chemical inhibitor of the DNA damage Response kinases ATM/ATR. We found that the effect of 5-FU was additive or synergistic with IFN-γ, the canonical inducer of PD-L1 in epithelial cells. QPCR analysis confirmed this finding and identified JAK-dependent transcriptional activation of PD-L1/CD274 as the underlying mechanism. The induction of PD-L1 by 5-FU was partially prevented by Epidermal Growth Factor Receptor (EGFR) inhibition with cetuximab. CONCLUSION: Our study highlights the specific DNA Damage Response- and JAK- dependent induction of PD-L1 by 5-FU in HNSCC cells. This induction is regulated by the cytokine context and is potentially therapeutically actionable.

3.
Biochem Biophys Res Commun ; 533(4): 1083-1087, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33019976

ABSTRACT

Ferroptosis, a regulated form of cell necrosis was previously reported to be induced upon pharmacological targeting of the cystine transporter SLC7A11 in Head and neck Squamous Cell Carcinoma (HNSCC). Whether tumors arising in a context of chronic infection with Human Papillomavirus (HPV) are sensitive to ferroptosis is unknown. Using RNAseq data (both whole-tumor and single-cell sequencing) we report that HPV positive (HPV+ve) tumors have lower expression levels of SLC7A11 compared to HPV negative (HPV-ve) HNSCC. We examined in vitro the effect of erastin, a specific blocker of SLC7A11, applied on two HNSCC cell lines with stable expression of HPV16 E6 and E7 oncoproteins. We report a decrease in total GSH levels and an increased sensitivity to erastin-induced ferroptosis in E6-E7 cells. Cell sensitivity to ferroptosis was specificaly related to a defect in cystine transport since we found no difference in ferroptosis induced by the direct inhibition of GPX4, and N-Acetyl Cystein abolished the difference between WT and E6-E7-expressing cells. Our findings point to SLC7A11 as an HPV-related biomarker of potential therapeutic relevance in HNSCC. Targeting cystine import to promote ferroptosis might be a promising strategy against HPV+ve HNSCC. (188 words).


Subject(s)
Amino Acid Transport System y+/metabolism , Ferroptosis/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/virology , Human papillomavirus 16/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/virology , Acetylcysteine/metabolism , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/genetics , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Biomarkers/metabolism , Cell Line, Tumor , Cystine/metabolism , Ferroptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glutathione/metabolism , Head and Neck Neoplasms/genetics , Humans , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/virology , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Piperazines/pharmacology , RNA-Seq , Repressor Proteins/metabolism , Single-Cell Analysis , Squamous Cell Carcinoma of Head and Neck/genetics
4.
Biosci Rep ; 39(9)2019 09 30.
Article in English | MEDLINE | ID: mdl-31467175

ABSTRACT

The tumor microenvironment is an important determinant of glioblastoma (GBM) progression and response to treatment. How oncogenic signaling in GBM cells modulates the composition of the tumor microenvironment and its activation is unclear. We aimed to explore the potential local immunoregulatory function of ERK1/2 signaling in GBM. Using proteomic and transcriptomic data (RNA seq) available for GBM tumors from The Cancer Genome Atlas (TCGA), we show that GBM with high levels of phosphorylated ERK1/2 have increased infiltration of tumor-associated macrophages (TAM) with a non-inflammatory M2 polarization. Using three human GBM cell lines in culture, we confirmed the existence of ERK1/2-dependent regulation of the production of the macrophage chemoattractant CCL2/MCP1. In contrast with this positive regulation of TAM recruitment, we found no evidence of a direct effect of ERK1/2 signaling on two other important aspects of TAM regulation by GBM cells: (1) the expression of the immune checkpoint ligands PD-L1 and PD-L2, expressed at high mRNA levels in GBM compared with other solid tumors; (2) the production of the tumor metabolite lactate recently reported to dampen tumor immunity by interacting with the receptor GPR65 present on the surface of TAM. Taken together, our observations suggest that ERK1/2 signaling regulates the recruitment of TAM in the GBM microenvironment. These findings highlight some potentially important particularities of the immune microenvironment in GBM and could provide an explanation for the recent observation that GBM with activated ERK1/2 signaling may respond better to anti-PD1 therapeutics.


Subject(s)
Glioblastoma/immunology , Macrophages/immunology , Proteomics , Transcriptome/genetics , B7-H1 Antigen/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Chemokine CCL2/genetics , Gene Expression Regulation, Neoplastic/immunology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , MAP Kinase Signaling System/genetics , Macrophages/pathology , Programmed Cell Death 1 Ligand 2 Protein/genetics , Receptors, G-Protein-Coupled/genetics , Tumor Microenvironment/immunology
5.
Oncol Rep ; 41(3): 2041-2050, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30569112

ABSTRACT

Sorafenib is the standard of care for the treatment of advanced hepatocellular carcinoma (HCC). However, identifying secreted biomarkers that predict sorafenib efficacy in all HCC patients remains challenging. It was recently reported that sorafenib interferes with protein homeostasis and inhibits global translation in tumour cells. A likely consequence of this inhibition would be the interruption of autocrine loops. The aim of the present study was to investigate the effect of sorafenib on two growth factors implicated in autocrine loops and HCC tumour invasion: amphiregulin (AREG) and vascular endothelial growth factor (VEGF). ELISA, quantitative polymerase chain reaction analysis, western blotting and a cytokine array were performed on HCC cell lines and the prognostic role of these two biomarkers in HCC patients was evaluated. Serum AREG and VEGF levels were assayed by ELISA in 55 patients with advanced HCC treated with sorafenib. It was observed that sorafenib decreased AREG, VEGF and cytokine expression at the transcriptional and post­transcriptional levels. All HCC patients in our cohort had detectable concentrations of AREG and VEGF both at baseline and after sorafenib treatment. The decreased serum levels of AREG and VEGF after 15 days of sorafenib treatment were significantly associated with better overall and progression­free survival. The results of the multivariate analysis demonstrated that a decrease in AREG was an independent prognostic indicator of overall survival (hazard ratio, 0.208; 95% confidence interval, 0.173­0.673; P=0.0003). These results suggest that sorafenib inhibits auto-crine loops and that early decrease in serum AREG or VEGF levels predicts sorafenib efficacy in HCC patients.


Subject(s)
Amphiregulin/blood , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Sorafenib/therapeutic use , Vascular Endothelial Growth Factor A/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/blood , Liver Neoplasms/mortality , Male , Middle Aged , Prognosis , Progression-Free Survival , Time Factors , Treatment Outcome
6.
Oncotarget ; 9(9): 8400-8414, 2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29492203

ABSTRACT

Sorafenib is the first line treatment for advanced hepatocellular carcinoma (HCC). We explored its impact on the proteostasis of cancer cells, i.e. the processes that regulate the synthesis, maturation and turn-over of cellular proteins. We observed that sorafenib inhibits the production of the tumour marker alpha-foetoprotein (AFP) in two different HCC cell lines, an effect that correlated with a radical inhibition of protein biosynthesis. This effect was observed at clinically relevant concentrations of sorafenib and was not related to the effect of sorafenib on the transport of amino acids across the plasma membrane or the induction of the unfolded protein response (UPR). Instead, we observed that sorafenib inhibits translation initiation and the mechanistic target of rapamycin (mTOR) signaling cascade, as shown by the analysis of phosphorylation levels of the protein 4EBP1 (eukaryotic translation initiation factor 4E binding protein 1). We explored the consequences of this inhibition in HCC cells. We observed that overall sorafenib is a weak inducer of the UPR that can paradoxically prevent the UPR induced by tunicamycin. We also found no direct synergistic anticancer effect between sorafenib and various strategies that inhibit the UPR. In agreement with the possibility that translation inhibition might be an adaptive stress response in HCC cells, we noted that it protects cancer cell from ferroptosis, a form of oxidative necrosis. Our findings point to the modulation of protein biosynthesis and mTOR signaling as being important, yet complex determinants of the response of HCC cells to sorafenib.

7.
Anticancer Res ; 37(3): 1049-1057, 2017 03.
Article in English | MEDLINE | ID: mdl-28314264

ABSTRACT

BACKGROUND/AIM: Organ transplant patients treated with the immunosuppressive drug cyclosporine A often present malignant kidney tumors. Cyclosporine A can promote oncogenesis in a cell-intrinsic manner by increasing the production of vascular endothelial growth factor (VEGF). MATERIALS AND METHODS: We explored the impact of cyclosporine A and the role of the unfolded protein response (UPR) on three human renal cell carcinoma (RCC) cell lines under normoxic and hypoxic (1% O2) conditions. RESULTS: Cyclosporine A regulated the expression of VEGF at the post-transcriptional level. Cyclosporine A induced the inositol requiring enzyme-1α (IRE1α) arm of the UPR and stabilized neosynthesized proteins in RCC cells. Toyocamycin, an inhibitor of IRE1α, abolished the clonogenic growth of RCC cells and reduced induction of VEGF by cyclosporine A under hypoxia. CONCLUSION: Our findings highlight the impact of cyclosporine A on the proteostasis of RCC cells, and suggest the potential therapeutic interest of targeting the UPR against tumors arising in the context of organ transplantation.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cyclosporine/chemistry , Gene Expression Regulation, Neoplastic , Immunosuppressive Agents/chemistry , Kidney Neoplasms/metabolism , Unfolded Protein Response , Cell Line, Tumor/drug effects , Endoribonucleases/metabolism , Gene Expression Regulation , Humans , Hypoxia , Oxygen/metabolism , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/metabolism , Puromycin/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Toyocamycin/chemistry , Vascular Endothelial Growth Factor A/metabolism
8.
Mol Cancer ; 15(1): 38, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27184800

ABSTRACT

BACKGROUND: Sorafenib, a kinase inhibitor active against various solid tumours, induces oxidative stress and ferroptosis, a new form of oxidative necrosis, in some cancer cells. Clinically-applicable biomarkers that reflect the impact of sorafenib on the redox metabolism of cancer cells are lacking. METHODS: We used gene expression microarrays, real-time PCR, immunoblot, protein-specific ELISA, and gene reporter constructs encoding the enzyme luciferase to study the response of a panel of cancer cells to sorafenib. Tumour explants prepared from surgical hepatocellular carcinoma (HCC) samples and serum samples obtained from HCC patients receiving sorafenib were also used. RESULTS: We observed that genes of the metallothionein-1 (MT1) family are induced in the HCC cell line Huh7 exposed to sorafenib. Sorafenib increased the expression of MT1G mRNA in a panel of human cancer cells, an effect that was not observed with eight other clinically-approved kinase inhibitors. We identified the minimal region of the MT1G promoter that confers inducibility by sorafenib to a 133 base pair region containing an Anti-oxidant Response Element (ARE) and showed the essential role of the transcription factor NRF2 (Nuclear factor erythroid 2-Related Factor 2). We examined the clinical relevance of our findings by analysing the regulation of MT1G in five tumour explants prepared from surgical HCC samples. Finally, we showed that the protein levels of MT1 increase in the serum of some HCC patients receiving sorafenib, and found an association with reduced overall survival. CONCLUSION: These findings indicate that MT1 constitute a biomarker adapted for exploring the impact of sorafenib on the redox metabolism of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Metallothionein/metabolism , Niacinamide/analogs & derivatives , Oxidation-Reduction/drug effects , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Biomarkers , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cysteine/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Metallothionein/genetics , NF-E2-Related Factor 2/metabolism , Niacinamide/pharmacology , Oxidative Stress , Prognosis , Promoter Regions, Genetic , Sorafenib , Transcription, Genetic
9.
Cancer Lett ; 370(2): 242-9, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26546044

ABSTRACT

Sorafenib is the treatment of reference for advanced hepatocellular carcinoma (HCC). A decrease in the serum levels of Alpha-fetoprotein (AFP) is reported to be the biological parameter that is best associated with disease control by sorafenib. In order to provide a biological rationale for the variations of AFP, we analyzed the various steps of AFP production in human HCC cell lines exposed to sorafenib. Sorafenib dramatically reduced the levels of AFP produced by HCC cells independently of its effect on cell viability. The mRNA levels of AFP decreased upon sorafenib treatment, while the AFP protein remained localized in the Golgi apparatus. Sorafenib activated the Regulated Inositol-Requiring Enzyme-1α (IRE-1α) and the PKR-like ER Kinase (PERK)-dependent arms of the Unfolded Protein Response (UPR). The inhibition of IRE-1α partially restored the mRNA levels of AFP upon treatment with sorafenib. The inhibition of both pathways partially prevented the drop in the production of AFP induced by sorafenib. The findings provide new insights on the regulation of AFP, and identify it as a biomarker suitable for the exploration of HCC cell proteostasis in the context of therapeutic targeting.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Homeostasis , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Proteins/metabolism , Unfolded Protein Response , alpha-Fetoproteins/analysis , Biomarkers , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/physiology , Humans , Liver Neoplasms/metabolism , Niacinamide/pharmacology , Regulatory Factor X Transcription Factors , Sorafenib , Transcription Factors/physiology , alpha-Fetoproteins/biosynthesis
10.
Anticancer Res ; 35(3): 1803-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25750346

ABSTRACT

BACKGROUND/AIM: Sorafenib is the medical reference for treatment of hepatocellular carcinoma (HCC). Multiple forms of cytotoxicity are induced by sorafenib in HCC cells in vitro but it is unclear what extent of apoptosis and necrosis is induced in HCC patients receiving sorafenib. PATIENTS AND METHODS: The M30 and M65 biomarkers, which reflect the release of cytokeratin-18 and its apoptotic cleavage fragments, were measured in patients with HCC (n=36) and matched patients with cirrhosis (n=47). A serum sample was collected from 20 patients with HCC four weeks after the onset of treatment with sorafenib. RESULTS: Basal serum levels of M30 and M65 were increased in patients with HCC compared to those with uncomplicated cirrhosis. No statistically significant increase in the level of M30 or M65 was found in the sera of patients with HCC after sorafenib. CONCLUSION: The findings indicate that sorafenib is not a potent inducer of HCC cell death in most patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Aged , Apoptosis , Biomarkers , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Female , Humans , Keratin-18/blood , Liver Neoplasms/blood , Liver Neoplasms/pathology , Male , Necrosis , Niacinamide/blood , Niacinamide/therapeutic use , Phenylurea Compounds/blood , Sorafenib
11.
Cancer Lett ; 356(2 Pt B): 971-7, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25444922

ABSTRACT

Sorafenib is the treatment of reference for advanced hepatocellular carcinoma (HCC), the most frequent form of primary liver tumour. The loss of function of the retinoblastoma (Rb) protein is an important event during liver carcinogenesis, but it is unclear whether the Rb status modulates the response of HCC cells to sorafenib. Here, we examined this question in HCC cells with reduced levels of Rb achieved through stable RNA interference. We show that HCC cells with reduced levels of Rb exhibit a two- to threefold increase in cell death induction upon exposure to sorafenib compared with controls. Sorafenib treatment of Balb/c nude mice that received tumour xenografts derived from HCC cells with reduced Rb levels resulted in complete tumour regression in 50% of the animals treated, compared with tumour stabilization in mice that received control cells. We show that, upon exposure to sorafenib, the Rb-negative status of HCC cells promotes the occurrence of ferroptosis, a form of oxidative necrosis. The findings highlight the role of Rb in the response of HCC cells to sorafenib and the regulation of ferroptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Necrosis , Niacinamide/analogs & derivatives , Oxidative Stress/drug effects , Phenylurea Compounds/pharmacology , Retinoblastoma Protein/metabolism , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Niacinamide/pharmacology , Reactive Oxygen Species/metabolism , Sorafenib , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Anticancer Res ; 34(11): 6417-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25368241

ABSTRACT

BACKGROUND/AIM: Ferroptosis is a recently identified form of regulated necrosis that can be experimentally induced in cancer cells with the chemical inducer erastin. Recently, we identified sorafenib, an inhibitor of oncogenic kinases, as an inducer of ferroptosis in hepatocellular carcinoma cells. Whether sorafenib is able to exert its ferroptotic activity in cancer cells originating from other tissues is presently unclear. MATERIALS AND METHODS: We compared the levels of ferroptosis induced by sorafenib with those induced by the reference compound erastin in a panel of ten human cell lines originating from various tissues. RESULTS: Sorafenib induced ferroptosis in different cancer cell lines. We found a positive correlation between the ferroptotic potency of sorafenib and erastin. Compared to other kinase inhibitors, sorafenib is the only drug that displays ferroptotic efficacy. CONCLUSION: The findings establish sorafenib as the first clinically-approved anticancer drug that can induce ferroptosis.


Subject(s)
Apoptosis/drug effects , Necrosis , Neoplasms/drug therapy , Neoplasms/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Neoplasms/metabolism , Niacinamide/pharmacology , Sorafenib , Tumor Cells, Cultured
13.
Med Sci (Paris) ; 30(8-9): 779-83, 2014.
Article in French | MEDLINE | ID: mdl-25174755

ABSTRACT

Ferroptosis is a form of cell death that has recently been reported during exposure to erastin, a chemical compound identified in a screen for molecules able to kill cancer cells carrying an active Ras oncogene. In cells exposed to inducers of ferroptosis, a catastrophic alteration of the cellular redox metabolism occurs, resulting in massive lipid peroxidation in the plasma membrane and loss of cell viability. We present our recent observations suggesting that sorafenib, the only medical treatment with proven efficacy against hepatocellular carcinoma, induces ferroptosis, a new anti-oncogenic mode of action of this drug. The discovery of ferroptosis sheds light on the critical adaptations of the redox metabolism in cancer cells. It might also foster the discovery of new biomarkers and innovative approaches for the treatment of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Humans , Lipid Peroxidation/drug effects , Oxidation-Reduction/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Signal Transduction/drug effects
14.
Anticancer Res ; 33(4): 1415-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23564781

ABSTRACT

BACKGROUND/AIM: Sorafenib is currently the only medical treatment with proven efficacy against hepatocellular carcinoma (HCC). HCC cell lines display heterogeneous sensitivity to sorafenib, but little is known about the sensitivity of clinical tumors. We aimed to examine this aspect. MATERIALS AND METHODS: Using experimental tumors generated in nude mice, we set up a technique for short-term culture of HCC fragments. We applied this technique to six human HCC samples obtained from surgical resection. RESULTS: HCC fragments in culture retain their morphology and viability for at least 48 h, permitting an in vitro analysis of the effect of sorafenib on the Extracellular signal-regulated kinase (ERK) cascade. HCC exhibit heterogeneous individual responses, ranging from potent inhibition to paradoxical activation of this oncogenic cascade. CONCLUSION: Our observations highlight the heterogeneous sensitivity of HCC to sorafenib, and point to the potential interest of short-term culture of tumor fragments for personalizing the medical treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Aged , Aged, 80 and over , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Female , Humans , Immunoblotting , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Niacinamide/pharmacology , Phosphorylation/drug effects , Sorafenib , Tumor Cells, Cultured
15.
Int J Cancer ; 133(7): 1732-42, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23505071

ABSTRACT

The multikinase inhibitor sorafenib is currently the treatment of reference for advanced hepatocellular carcinoma (HCC). In our report, we examined the cytotoxic effects of sorafenib on HCC cells. We report that the depletion of the intracellular iron stores achieved by using the iron chelator deferoxamine (DFX) strikingly protects HCC cells from the cytotoxic effects of sorafenib. The protective effect of the depletion of intracellular iron stores could not be explained by an interference with conventional forms of programmed cell death, such as apoptosis or autophagic cell death. We also found that DFX did not prevent sorafenib from reaching its intracellular target kinases. Instead, the depletion of intracellular iron stores prevented sorafenib from inducing oxidative stress in HCC cells. We examined the possibility that sorafenib might exert a cytotoxic effect that resembles ferroptosis, a form of cell death in which iron-dependent oxidative mechanisms play a pivotal role. In agreement with this possibility, we found that pharmacological inhibitors (ferrostatin-1) and genetic procedures (RNA interference against IREB-2) previously reported to modulate ferroptosis, readily block the cytotoxic effects of sorafenib in HCC cells. Collectively, our findings identify ferroptosis as an effective mechanism for the induction of cell death in HCC. Ferroptosis could potentially become a goal for the medical treatment of HCC, thus opening new avenues for the optimization of the use of sorafenib in these tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Deferoxamine/pharmacology , Iron/metabolism , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Survival , Cyclohexylamines/pharmacology , Deferoxamine/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Iron/chemistry , Iron Regulatory Protein 2/genetics , Liver Neoplasms/pathology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Niacinamide/pharmacology , Niacinamide/therapeutic use , Oxidative Stress , Phenylenediamines/pharmacology , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , RNA Interference , Siderophores/chemistry , Sorafenib , raf Kinases/metabolism
16.
Int J Cancer ; 131(12): 2961-9, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22514082

ABSTRACT

Sorafenib is currently the medical treatment of reference for hepatocellular carcinoma (HCC), but it is not known whether sorafenib is equally active in all HCC. Here, our aim was to explore intrinsic differences in the response of HCC cells to sorafenib, to identify potential mechanisms leading to primary resistance to this treatment. We analyzed a panel of six human HCC cell lines and compared the activity of the main oncogenic kinase cascades, their clonogenic potential, proliferation and apoptosis upon exposure to sorafenib. We report that HCC cells present important differences in their response to sorafenib, and that some cell lines are more resistant to the actions of sorafenib than others. We identify the activated epidermal growth factor receptor (EGFR) as a parameter that promotes the resistance of HCC cells to sorafenib. In resistant cells, the efficacy of sorafenib was increased when EGFR was inhibited, as was demonstrated using two chemical inhibitors (erlotinib or gefitinib), a monoclonal antibody directed against EGFR (cetuximab), and RNA interference directed against EGFR. A combination of EGFR inhibitors and sorafenib affords a better control over HCC proliferation, most likely through an improved blockade of the RAF kinases. Our findings therefore confirm the importance of RAF kinases as therapeutic targets in HCC, and identify EGFR as a determinant of the sensitivity of HCC cells to sorafenib. Our findings bear possible implications for the improvement of the efficacy of sorafenib in HCC, and might be useful for the identification of predictive biomarkers in this context.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Carcinoma, Hepatocellular/drug therapy , ErbB Receptors/metabolism , Liver Neoplasms/drug therapy , Pyridines/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Niacinamide/analogs & derivatives , Phenylurea Compounds , RNA Interference , Sorafenib
17.
Biochem Biophys Res Commun ; 417(1): 479-83, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22172950

ABSTRACT

Chronic kidney disease (CKD) has recently emerged as a major risk factor for cardiovascular pathology. CKD patients display accelerated atherosclerotic process, leading to circulatory complications. However, it is currently not clear how uremic conditions accelerate atherosclerosis. Apoptosis is an important homeostatic regulator of vascular smooth cells under pathological conditions. In the present study, we explored the regulation of apoptosis in cells of the vascular wall in the uremic context. We analysed the expression and regulation of the proteins of the BCL2 family that play an essential role in apoptosis. Our results, obtained in mice and primary human smooth muscle cells exposed to two uremic toxins, point to the existence of an alteration in expression and function of one pro-apoptotic member of this family, the protein BAD. We explore the regulation of BAD by uremic toxins and report the sensitization of vascular smooth muscle cells to apoptosis upon BAD induction.


Subject(s)
Kidney Failure, Chronic/metabolism , Muscle, Smooth, Vascular/metabolism , Uremia/metabolism , bcl-Associated Death Protein/biosynthesis , Animals , Apoptosis , Cells, Cultured , Creatine/metabolism , Creatine/toxicity , Humans , Kidney Failure, Chronic/pathology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Urea/metabolism , Urea/toxicity , Uremia/pathology
18.
Nephrol Dial Transplant ; 27(6): 2176-81, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22140126

ABSTRACT

BACKGROUND: Patients with chronic kidney disease (CKD) develop various bone abnormalities characterized by impaired bone remodelling. Recent data suggest that accumulation of the uraemic toxin indoxyl sulphate (IS) may be one of the factors involved in bone abnormalities in CKD patients. Indeed, it was recently reported that IS induces skeletal resistance to parathyroid hormone in cultured osteoblastic cells. However, it is not yet known whether IS also affects osteoclast cells. METHODS: In the present study, we assessed the direct effect of IS at uraemic concentrations and in the presence (to reach the 3 mM concentration) or absence of added inorganic phosphate (Pi) on osteoclast (OCL) differentiation and bone-resorbing activity in two well-established cellular models of monocyte/macrophage (peripheral blood mononuclear cells and the RAW 264.7 cell line). RESULTS: We found that IS inhibits both OCL differentiation and bone-resorbing activity in a dose-dependent manner and that these effects were enhanced in the presence of Pi at 3mM concentration. IS induced a gradual inhibition of JNK, Akt, p38, ERK1/2 phosphorylation and AP-1 DNA-binding activity. The effects of IS on OCL differentiation and AP-1 were prevented by probenecid, a competitive inhibitor of organic anion transporters, suggesting that IS's effects occur subsequently to its intake. CONCLUSION: Our findings strongly suggest that IS not only inhibits osteoblast function but also has an inhibitory effect on OCL function and thus could affect bone remodelling in CKD patients.


Subject(s)
Bone Resorption/drug therapy , Cell Differentiation/drug effects , Indican/pharmacology , Osteoclasts/cytology , Osteoclasts/drug effects , Animals , Cells, Cultured , Collagen Type I/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , MAP Kinase Kinase 4/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Mol Pharmacol ; 79(6): 997-1004, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21393385

ABSTRACT

The proteins of the B-cell lymphoma 2 (Bcl-2) family are important regulators of apoptosis under normal and pathological conditions. Chemical compounds that block the antiapoptotic proteins of this family have been introduced, such as 4-[4-[(4'-Chloro[1,1'-biphenyl]-2-yl)methyl]-1-piperazinyl]-N-[[4-[[(1R)-3-(dimethylamino)-1-[(phenylthio)methyl]propyl]amino]-3-nitrophenyl]sulfonyl]benzamide (ABT-737), a BH3-mimetic that neutralizes Bcl-2 and Bcl-xL. In this study, we used ABT-737 to explore the dynamic regulation of Bcl-2 proteins in living cells of different origins. Using ABT-737 as well as RNA interference or the application of growth factors, we examined the impact of the functional availability of the antiapoptotic proteins Bcl-2 and Bcl-2-extra large (Bcl-xL) on the Bcl-2 network. We report that ABT-737 increases the expression of Bcl-2-associated death promoter (Bad), a proapoptotic partner of the proteins Bcl-2 and Bcl-xL. Our observations indicate that Bad overexpression induced by ABT-737 results from the control of its normally rapid protein turnover, leading to the stabilization of this protein. We demonstrate the relevance of Bad post-translational regulation by Bcl-xL to the physiological setting using RNA interference against Bcl-xL as well as the application of epidermal growth factor, a growth factor that promotes the dissociation of Bad from Bcl-xL. Our results highlight a new facet of the mode of action of the antiapoptotic proteins Bcl-2 and Bcl-xL consisting of the regulation of the stability of the protein Bad. Finally, our results shed light on the mode of action of ABT-737, currently the best characterized inhibitor of the antiapoptotic proteins of the Bcl-2 family, and bear important implications regarding its use as an anticancer drug.


Subject(s)
Biphenyl Compounds/pharmacology , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Blotting, Western , Cell Line , Cytosol/metabolism , Humans , Molecular Mimicry , Piperazines/pharmacology , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/chemistry , RNA Interference
20.
Ther Apher Dial ; 15(2): 135-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21426504

ABSTRACT

Uremic syndrome is attributed to the progressive retention of a large number of compounds, such as indoxyl sulfate, which under physiological conditions are excreted by the kidneys. Previous in vitro studies have demonstrated that uremic indoxyl sulfate concentrations induce a weak increase in the proliferation of both rat and human vascular aortic smooth muscle cells (hVASMC) after short term exposition to the toxin (i.e. 24 h). In the present study, we evaluated indoxyl sulfate effects on the proliferation of hVASMC at three different concentrations after long-term exposure (seven days). In contrast to previously published studies, we observed a dose-dependent and significant inhibitory effect of this toxin on hVASMC proliferation. We also demonstrated that indoxyl sulfate inhibits epidermal growth factor-induced hVASMC proliferation after long-term exposure. Indoxyl sulfate effects were associated with a dose-dependent induction of intracellular reactive oxygen species and up-regulation of p21 and p27 protein expression. Chronic exposure to indoxyl sulfate produces a significant inhibitory effect on hVASMC proliferation. The relevance of these findings must be evaluated by further studies, particularly in an in vivo setting.


Subject(s)
Cell Proliferation/drug effects , Indican/metabolism , Muscle, Smooth, Vascular/drug effects , Reactive Oxygen Species/metabolism , Aorta/drug effects , Aorta/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Humans , Indican/administration & dosage , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Time Factors , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...