Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
PLoS One ; 12(3): e0172998, 2017.
Article in English | MEDLINE | ID: mdl-28253308

ABSTRACT

It is well established that the formation of radical species centered on various atoms is involved in the mechanism leading to the development of several diseases or to the appearance of deleterious effects of toxic molecules. The detection of free radical is possible using Electron Paramagnetic Resonance (EPR) spectroscopy and the spin trapping technique. The classical EPR spin-trapping technique can be considered as a "hypothesis-driven" approach because it requires an a priori assumption regarding the nature of the free radical in order to select the most appropriate spin-trap. We here describe a "data-driven" approach using EPR and a cocktail of spin-traps. The rationale for using this cocktail was that it would cover a wide range of biologically relevant free radicals and have a large range of hydrophilicity and lipophilicity in order to trap free radicals produced in different cellular compartments. As a proof-of-concept, we validated the ability of the system to measure a large variety of free radicals (O-, N-, C-, or S- centered) in well characterized conditions, and we illustrated the ability of the technique to unambiguously detect free radical production in cells exposed to chemicals known to be radical-mediated toxic agents.


Subject(s)
Free Radicals/chemistry , Spin Labels , Electron Spin Resonance Spectroscopy , Humans , K562 Cells
2.
Free Radic Biol Med ; 99: 436-450, 2016 10.
Article in English | MEDLINE | ID: mdl-27591797

ABSTRACT

Regulation of ROS metabolism plays a major role in cellular adaptation to oxidative stress in cancer cells, but the molecular mechanism that regulates catalase, a key antioxidant enzyme responsible for conversion of hydrogen peroxide to water and oxygen, remains to be elucidated. Therefore, we investigated the transcriptional regulatory mechanism controlling catalase expression in three human mammary cell lines: the normal mammary epithelial 250MK primary cells, the breast adenocarcinoma MCF-7 cells and an experimental model of MCF-7 cells resistant against oxidative stress resulting from chronic exposure to H2O2 (Resox), in which catalase was overexpressed. Here we identify a novel promoter region responsible for the regulation of catalase expression at -1518/-1226 locus and the key molecules that interact with this promoter and affect catalase transcription. We show that the AP-1 family member JunB and retinoic acid receptor alpha (RARα) mediate catalase transcriptional activation and repression, respectively, by controlling chromatin remodeling through a histone deacetylases-dependent mechanism. This regulatory mechanism plays an important role in redox adaptation to chronic exposure to H2O2 in breast cancer cells. Our study suggests that cancer adaptation to oxidative stress may be regulated by transcriptional factors through chromatin remodeling, and reveals a potential new mechanism to target cancer cells.


Subject(s)
Catalase/genetics , Chromatin Assembly and Disassembly , Chromatin/metabolism , Gene Expression Regulation, Neoplastic , Retinoic Acid Receptor alpha/genetics , Transcription Factors/genetics , Adaptation, Physiological , Base Sequence , Catalase/metabolism , Cell Line , Cell Line, Tumor , Chromatin/chemistry , Chromatin/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , MCF-7 Cells , Oxidative Stress , Promoter Regions, Genetic , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Retinoic Acid Receptor alpha/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
3.
Life Sci ; 145: 57-65, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26687450

ABSTRACT

AIMS: Alterations in the expression of antioxidant enzymes are associated with changes in cancer cell sensitivity to chemotherapeutic drugs (menadione and ß-lapachone). Mechanisms of acquisition of resistance to pro-oxidant drugs were investigated using a model of oxidative stress-resistant MCF-7 breast cancer cells (Resox cells). MAIN METHODS: FISH experiments were performed in tumor biopsy and breast cancer cells to characterize the pattern of the NQO1 gene. SNP-arrays were conducted to detect chromosomal imbalances. Finally, the importance of NQO1 overexpression in the putative acquisition of either drug resistance or an increased sensitivity to quinones by cancer cells was investigated by immunoblotting and cytotoxicity assays. KEY FINDINGS: Genomic gain of the chromosomal band 16q22 was detected in Resox cells compared to parental breast cancer MCF-7 cells and normal human mammary epithelial 250MK cells. This genomic gain was associated with amplification of the NQO1 gene in one tumor biopsy as well as in breast cancer cell lines. Using different breast cell models, we found that NQO1 overexpression was a main determinant for a potential chemotherapy resistance or an increased sensitivity to quinone-bearing compounds. SIGNIFICANCE: Because NQO1 is frequently modified in tumors at genomic and transcriptomic levels, the impact of NQO1 modulation on breast cancer cell sensitivity places NQO1 as a potential link between cancer redox alterations and resistance to chemotherapy. Thus, the NQO1 gene copy number and NQO1 activity should be considered when quinone-bearing molecules are being utilized as potential drugs against breast tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast/drug effects , Drug Resistance, Neoplasm , NAD(P)H Dehydrogenase (Quinone)/genetics , Quinones/pharmacology , Up-Regulation , Antineoplastic Agents/chemistry , Breast/metabolism , Breast Neoplasms/genetics , Female , Gene Dosage , Genetic Loci , Humans , MCF-7 Cells , Polymorphism, Single Nucleotide , Quinones/chemistry
4.
Biochem Biophys Res Commun ; 466(3): 418-25, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26365353

ABSTRACT

Dihydroxynaphthyl aryl ketones 1-5 have been evaluated for their abilities to inhibit microtubule assembly and the binding to tubulin. Compounds 3, 4 and 5 displayed competitive inhibition against colchicine binding, and docking analysis showed that they bind to the tubulin colchicine-binding pocket inducing sheets instead of microtubules. Remarkable differences in biological activity observed among the assayed compounds seem to be related to the structure and position of the aryl substituent bonded to the carbonyl group. Compounds 2, 3 and 4, which contain a heterocyclic ring, presented higher affinity for tubulin compared to the carbocyclic analogue 5. Compound 4 showed the best affinity of the series, with an IC50 value of 2.1 µM for microtubule polymerization inhibition and a tubulin dissociation constant of 1.0 ± 0.2 µM, as determined by thermophoresis. Compound 4 was more efficacious in disrupting microtubule assembly in vitro than compound 5 although it contains the trimethoxyphenyl ring present in colchicine. Hydrogen bonds with Asn101 of α-tubulin seem to be responsible for the higher affinity of compound 4 respects to the others.


Subject(s)
Colchicine/metabolism , Ketones/metabolism , Microtubules/metabolism , Tubulin/metabolism , Animals , Binding Sites , Binding, Competitive , Chickens , Colchicine/pharmacology , Hydrogen Bonding , Ketones/chemistry , Ketones/pharmacology , Kinetics , Microtubules/drug effects , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology
5.
Free Radic Biol Med ; 87: 84-97, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26117330

ABSTRACT

Catalase is an important antioxidant enzyme that dismutates hydrogen peroxide into water and molecular oxygen. The catalase gene has all the characteristics of a housekeeping gene (no TATA box, no initiator element sequence, high GC content in promoter) and a core promoter that is highly conserved among species. We demonstrate in this review that within this core promoter, the presence of DNA binding sites for transcription factors, such as NF-Y and Sp1, plays an essential role in the positive regulation of catalase expression. Additional transcription factors, such as FoxO3a, are also involved in this regulatory process. There is strong evidence that the protein Akt/PKB in the PI3K signaling pathway plays a major role in the expression of catalase by modulating the activity of FoxO3a. Over the past decade, other transcription factors (PPARγ, Oct-1, etc.), as well as genetic, epigenetic, and posttranscriptional processes, have emerged as crucial contributors to the regulation of catalase expression. Altered expression levels of catalase have been reported in cancer tissues compared to their normal counterparts. Deciphering the molecular mechanisms that regulate catalase expression could, therefore, be of crucial importance for the future development of pro-oxidant cancer chemotherapy.


Subject(s)
Catalase/biosynthesis , DNA-Binding Proteins/genetics , Neoplasms/genetics , Transcription, Genetic , Antioxidants/metabolism , Binding Sites , Catalase/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/pathology , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
6.
Biochem Pharmacol ; 91(2): 168-80, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25058527

ABSTRACT

Hepatocellular carcinoma is one of the most frequent tumor types worldwide and oxidative stress represents a major risk factor in pathogenesis of liver diseases leading to HCC. Nuclear factor erythroid 2-related factor (Nrf2) is a transcription factor activated by oxidative stress that governs the expression of many genes which constitute the antioxidant defenses of the cell. In addition, oxidative stress activates AMP-activated protein kinase (AMPK), which has emerged in recent years as a kinase that controls the redox-state of the cell. Since both AMPK and Nrf2 are involved in redox homeostasis, we investigated whether there was a crosstalk between the both signaling systems in hepatocarcinoma cells. Here, we demonstrated that AMPK activator AICAR, in contrary to the A769662 allosteric activator, induces Nrf2 activation and concomitantly modulates the basal redox state of the hepatocarcinoma cells. When the expression of Nrf2 is knocked down, AICAR failed to induce its effect on redox state. These data highlight a major role of Nrf2 signaling pathway in mediating the AICAR effect on basal oxidative state. Furthermore, we demonstrated that AICAR metabolization by the cell is required to induce Nrf2 activation while, the silencing of AMPK does not have any effect on Nrf2 activation. This suggests that AICAR-induced Nrf2 activation is independent of AMPK activity. In conclusion, we identified AICAR as a potent modulator of the redox state of human hepatocarcinoma cells, via the Nrf2 signaling pathway and in an AMPK-independent mechanism.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases/genetics , Active Transport, Cell Nucleus/physiology , Aminoimidazole Carboxamide/pharmacology , Biphenyl Compounds , Carcinoma, Hepatocellular/etiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Liver Neoplasms/etiology , NF-E2-Related Factor 2/genetics , Phosphorylation , Pyrones/pharmacology , Thiophenes/pharmacology
7.
Biochem Pharmacol ; 89(2): 217-23, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24630930

ABSTRACT

Catalase is an antioxidant enzyme that catalyzes mainly the transformation of hydrogen peroxide into water and oxygen. Although catalase is frequently down-regulated in tumors the underlying mechanism remains unclear. Few transcription factors have been reported to directly bind the human catalase promoter. Among them FoxO3a has been proposed as a positive regulator of catalase expression. Therefore, we decided to study the role of the transcription factor FoxO3a and the phosphatidylinositol-3 kinase (PI3K) signaling pathway, which regulates FoxO3a, in the expression of catalase. To this end, we developed an experimental model of mammary breast MCF-7 cancer cells that acquire resistance to oxidative stress, the so-called Resox cells, in which catalase is overexpressed as compared with MCF-7 parental cell line. In Resox cells, Akt expression is decreased but its phosphorylation is enhanced when compared with MCF-7 cells. A similar profile is observed for FoxO3a, with less total protein but more phosphorylated FoxO3a in Resox cells, correlating with its higher Akt activity. The modulation of FoxO3a expression by knockdown and overexpression strategies did not affect catalase expression, neither in MCF-7 nor in Resox cells. Inhibition of PI3K and mTOR by LY295002 and rapamycin, respectively, decreases the phosphorylation of downstream targets (i.e. GSK3ß and p70S6K) and leads to an increase of catalase expression only in MCF-7 but not in Resox cells. In conclusion, FoxO3a does not appear to play a critical role in the regulation of catalase expression in both cancer cells. Only MCF-7 cells are sensitive and dependent on PI3K/Akt/mTOR signaling.


Subject(s)
Catalase/biosynthesis , Gene Expression Regulation, Enzymologic , Phosphatidylinositol 3-Kinase/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
8.
Free Radic Biol Med ; 67: 292-303, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24269898

ABSTRACT

Emerging evidence indicates that oxidative stress instigates the formation of ubiquitin (Ub) aggregates, substrates of autophagy, through a process requiring the ubiquitin binding adaptors p62/SQSTM1 and NBR1. Here, we have investigated the role of p62 and NBR1 in cell survival after hypericin-mediated photodynamic therapy (Hyp-PDT), a procedure known to incite robust reactive oxygen species (ROS)-based endoplasmic reticulum stress and autophagy pathways. We found that Hyp-PDT stimulated the formation of p62- and NBR1-associated Ub aggregates in normal and cancer cells, which were ultimately removed by autophagy, through a mechanism partially regulated by p38(MAPK). In line with this, genetic or pharmacological p38(MAPK) inhibition reduced p62 and NBR1 levels and aggregate formation and impaired Nrf2 activation, thus increasing photo-oxidative stress and cell death. p62-deficient cells, or cells lacking p62 and with reduced levels of NBR1 (through siRNA knockdown), also displayed reduced aggregate formation but exhibited attenuated ROS levels, reduced caspase activation, and improved survival after Hyp-PDT. The increased resistance to photo-oxidative stress exhibited by cells lacking p62 and/or NBR1 was overruled by the inhibition of p38(MAPK), which restored cytotoxic ROS levels, thus indicating the relevance of this signal in the control of cell viability. Taken together these findings provide evidence that in photodynamically treated cells a p38(MAPK)-regulated pathway coordinates the p62/NBR1-mediated clearance of cytosolic aggregates and mitigates PDT-induced proteotoxicity. They also reveal that a functional p38(MAPK)-Nrf2 signal is required to keep ROS levels in check and protect against PDT-induced proteotoxicity, independent of aggregate formation.


Subject(s)
Autophagy/genetics , Fibroblasts/metabolism , NF-E2-Related Factor 2/genetics , Proteins/genetics , RNA-Binding Proteins/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Anthracenes , Autophagy/drug effects , Autophagy/radiation effects , Cell Line , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/radiation effects , Gene Expression Regulation , Gene Knockout Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Light , Mice , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Perylene/analogs & derivatives , Perylene/pharmacology , Protein Aggregates , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Proteolysis , RNA-Binding Proteins/metabolism , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction , Ubiquitin/genetics , Ubiquitin/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Biochem Pharmacol ; 86(2): 200-9, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23688501

ABSTRACT

Chronic alcohol consumption is a well-known risk factor for liver disease. Progression of alcohol-induced liver disease (ALD) is a multifactorial process that involves a number of genetic, nutritional and environmental factors. Experimental and clinical studies increasingly show that oxidative damage induced by ethanol contributes in many ways to the pathogenesis of alcohol hepatoxicity. Oxidative stress appears to activate AMP-activated protein kinase (AMPK) signaling system, which has emerged in recent years as a kinase that controls the redox-state and mitochondrial function. This review focuses on the most recent insights concerning the activation of AMPK by reactive oxygen species (ROS), and describes recent evidences supporting the hypothesis that AMPK signaling pathways play an important role in promoting cell viability under conditions of oxidative stress, such as during alcohol exposure. We suggest that AMPK activation by ROS can promote cell survival by inducing autophagy, mitochondrial biogenesis and expression of genes involved in antioxidant defense. Hence, increased intracellular concentrations of ROS may represent a general mechanism for enhancement of AMPK-mediated cellular adaptation, including maintenance of redox homeostasis. On the other hand, AMPK inhibition in the liver by ethanol appears to play a key role in the development of steatosis induced by chronic alcohol consumption. Although more studies are needed to assess the functions of AMPK during oxidative stress, AMPK may be a possible therapeutic target in the particular case of alcohol-induced liver disease.


Subject(s)
Adenylate Kinase/metabolism , Liver Diseases, Alcoholic/enzymology , Oxidative Stress , Enzyme Activation , Humans , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Reactive Oxygen Species/metabolism
10.
Biochem Biophys Res Commun ; 430(3): 883-8, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23261463

ABSTRACT

Pharmacological doses of ascorbate were evaluated for its ability to potentiate the toxicity of sodium orthovanadate (Na(3)VO(4)) in tumor cells. Cytotoxicity, inhibition of cell proliferation, generation of ROS and DNA fragmentation were assessed in T24 cells. Na(3)VO(4) was cytotoxic against T24 cells (EC(50)=5.8 µM at 24 h), but in the presence of ascorbate (100 µM) the EC(50) fell to 3.3 µM. Na(3)VO(4) plus ascorbate caused a strong inhibition of cell proliferation (up to 20%) and increased the generation of ROS (4-fold). Na(3)VO(4) did not directly cleave plasmid DNA, at this aspect no synergism was found occurring between Na(3)VO(4) and ascorbate once the resulting action of the combination was no greater than that of both substances administered separately. Cells from Ehrlich ascites carcinoma-bearing mice were used to determine the activity of antioxidant enzymes, the extent of the oxidative damage and the type of cell death. Na(3)VO(4) alone, or combined with ascorbate, increased catalase activity, but only Na(3)VO(4) plus ascorbate increased superoxide dismutase activity (up to 4-fold). Oxidative damage on proteins and lipids was higher due to the treatment done with Na(3)VO(4) plus ascorbate (2-3-fold). Ascorbate potentiated apoptosis in tumor cells from mice treated with Na(3)VO(4). The results indicate that pharmacological doses of ascorbate enhance the generation of ROS induced by Na(3)VO(4) in tumor cells causing inhibition of proliferation and apoptosis. Apoptosis induced by orthovanadate and ascorbate is closer related to inhibition on Bcl-xL and activation of Bax. Our data apparently rule out a mechanism of cell demise p53-dependent or related to Cdk2 impairment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ascorbic Acid/pharmacology , Cell Proliferation/drug effects , Reactive Oxygen Species/metabolism , Vanadates/pharmacology , Animals , Cell Line, Tumor , DNA/drug effects , DNA Fragmentation , Drug Synergism , Humans , Male , Mice , Mice, Inbred BALB C , Plasmids/drug effects , bcl-2-Associated X Protein/agonists , bcl-X Protein/antagonists & inhibitors
11.
Curr Top Med Chem ; 12(19): 2094-102, 2012.
Article in English | MEDLINE | ID: mdl-23167798

ABSTRACT

Hsp90 is a chaperone that plays a key function in cancer cells by stabilizing proteins responsible of cell growth and survival. Disruption of the Hsp90 chaperone machinery leads to the proteasomal degradation of its client proteins. Hsp90 appears then as an attractive target for the development of new anticancer molecules. We have shown that ascorbate- driven menadione-redox cycling inhibits Hsp90 activity by provoking an N-terminal cleavage of the protein, inducing the degradation of several of its client proteins. Since the mechanism involves an oxidative stress, we explored the effect of a series of diverse donor-acceptor 3-acyl-2-phenylamino 1,4-naphthoquinones on Hsp90 integrity, in the presence of ascorbate. Results show that quinone-derivatives that bear two electroactive groups (namely quinone and nitro) exhibit the highest inhibitory activity (Hsp90 cleavage and cell death). The biological activity of the series mainly relies on their redox capacity and their lipophilicity, which both modulate the ability of these compounds to induce a cytotoxic effect in K562 cells. As observed with other redox cycling quinones, the protein cleavage is blocked in the presence of N-terminal Hsp90 inhibitors suggesting that the availability or occupancy of nucleotide binding site in the N-terminal pocket of Hsp90 plays a critical role. In addition the survival of cancer cells and their metabolic and redox homeostasis were strongly impaired by the presence of ascorbate. Since these effects were similar to that obtained by ascorbate/menadione and they were blocked by the antioxidant N-acetylcyteine (NAC), it appears that oxidative stress is a major component of this cytotoxicity.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Naphthoquinones/pharmacology , Adenosine Triphosphate/metabolism , Blotting, Western , Glutathione/metabolism , HSP90 Heat-Shock Proteins/physiology , Humans , K562 Cells , Magnetic Resonance Spectroscopy , Mass Spectrometry , Spectrophotometry, Infrared
12.
PLoS One ; 7(10): e46571, 2012.
Article in English | MEDLINE | ID: mdl-23082126

ABSTRACT

Cancer can be envisioned as a metabolic disease driven by pressure selection and intercellular cooperativeness. Together with anaerobic glycolysis, the Warburg effect, formally corresponding to uncoupling glycolysis from oxidative phosphorylation, directly participates in cancer aggressiveness, supporting both tumor progression and dissemination. The transcription factor hypoxia-inducible factor-1 (HIF-1) is a key contributor to glycolysis. It stimulates the expression of glycolytic transporters and enzymes supporting high rate of glycolysis. In this study, we addressed the reverse possibility of a metabolic control of HIF-1 in tumor cells. We report that lactate, the end-product of glycolysis, inhibits prolylhydroxylase 2 activity and activates HIF-1 in normoxic oxidative tumor cells but not in Warburg-phenotype tumor cells which also expressed lower basal levels of HIF-1α. These data were confirmed using genotypically matched oxidative and mitochondria-depleted glycolytic tumor cells as well as several different wild-type human tumor cell lines of either metabolic phenotype. Lactate activates HIF-1 and triggers tumor angiogenesis and tumor growth in vivo, an activity that we found to be under the specific upstream control of the lactate transporter monocarboxylate transporter 1 (MCT1) expressed in tumor cells. Because MCT1 also gates lactate-fueled tumor cell respiration and mediates pro-angiogenic lactate signaling in endothelial cells, MCT1 inhibition is confirmed as an attractive anticancer strategy in which a single drug may target multiple tumor-promoting pathways.


Subject(s)
Glycolysis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactic Acid/pharmacology , Animals , Basigin/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , Mice , Mice, Nude , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Oxidation-Reduction/drug effects , Phenotype , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/metabolism , Protein Stability/drug effects , Symporters/antagonists & inhibitors , Symporters/metabolism
13.
PLoS One ; 7(7): e40795, 2012.
Article in English | MEDLINE | ID: mdl-22848402

ABSTRACT

Hsp90 is an essential chaperone that is necessary for the folding, stability and activity of numerous proteins. In this study, we demonstrate that free radicals formed during oxidative stress conditions can cleave Hsp90. This cleavage occurs through a Fenton reaction which requires the presence of redox-active iron. As a result of the cleavage, we observed a disruption of the chaperoning function of Hsp90 and the degradation of its client proteins, for example, Bcr-Abl, RIP, c-Raf, NEMO and hTert. Formation of Hsp90 protein radicals on exposure to oxidative stress was confirmed by immuno-spin trapping. Using a proteomic analysis, we determined that the cleavage occurs in a conserved motif of the N-terminal nucleotide binding site, between Ile-126 and Gly-127 in Hsp90ß, and between Ile-131 and Gly-132 in Hsp90α. Given the importance of Hsp90 in diverse biological functions, these findings shed new light on how oxidative stress can affect cellular homeostasis.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Iron/metabolism , Oxidative Stress/physiology , Proteolysis , Reactive Oxygen Species/metabolism , Amino Acid Motifs , HSP90 Heat-Shock Proteins/chemistry , Homeostasis/physiology , Humans , Iron/chemistry , K562 Cells , Oxidation-Reduction , Reactive Oxygen Species/chemistry
14.
PLoS One ; 7(6): e37971, 2012.
Article in English | MEDLINE | ID: mdl-22761662

ABSTRACT

The gut microbiota has recently been proposed as a novel component in the regulation of host homeostasis and immunity. We have assessed for the first time the role of the gut microbiota in a mouse model of leukemia (transplantation of BaF3 cells containing ectopic expression of Bcr-Abl), characterized at the final stage by a loss of fat mass, muscle atrophy, anorexia and inflammation. The gut microbial 16S rDNA analysis, using PCR-Denaturating Gradient Gel Electrophoresis and quantitative PCR, reveals a dysbiosis and a selective modulation of Lactobacillus spp. (decrease of L. reuteri and L. johnsonii/gasseri in favor of L. murinus/animalis) in the BaF3 mice compared to the controls. The restoration of Lactobacillus species by oral supplementation with L. reuteri 100-23 and L. gasseri 311476 reduced the expression of atrophy markers (Atrogin-1, MuRF1, LC3, Cathepsin L) in the gastrocnemius and in the tibialis, a phenomenon correlated with a decrease of inflammatory cytokines (interleukin-6, monocyte chemoattractant protein-1, interleukin-4, granulocyte colony-stimulating factor, quantified by multiplex immuno-assay). These positive effects are strain- and/or species-specific since L. acidophilus NCFM supplementation does not impact on muscle atrophy markers and systemic inflammation. Altogether, these results suggest that the gut microbiota could constitute a novel therapeutic target in the management of leukemia-associated inflammation and related disorders in the muscle.


Subject(s)
Disease Models, Animal , Inflammation Mediators/metabolism , Inflammation/prevention & control , Lactobacillus/physiology , Leukemia, Experimental/complications , Muscular Atrophy/prevention & control , Acute Disease , Animals , Cells, Cultured , Dietary Supplements , Female , Fusion Proteins, bcr-abl/genetics , Gastrointestinal Tract/microbiology , Inflammation/etiology , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/microbiology , Liver Neoplasms/pathology , Metagenome , Mice , Mice, Inbred BALB C , Muscular Atrophy/etiology , Precursor Cells, B-Lymphoid/transplantation , Splenic Neoplasms/metabolism , Splenic Neoplasms/microbiology , Splenic Neoplasms/pathology
15.
Radiother Oncol ; 105(1): 64-71, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22682746

ABSTRACT

BACKGROUND AND PURPOSE: The relevance of Mitogen Activated Protein Kinase (MAPK) inhibitors as co-treatments for radiation therapy is investigated, with special focus on a potential link between the MAPK pathway and tumor hypoxia, which is a critical determinant for response to therapy. MATERIALS AND METHODS: The effects of two MAPK inhibitors, Sorafenib and PD0325901, were monitored daily using in vivo EPR (Electron Paramagnetic Resonance) oximetry in FSaII and TLT tumor models in order to identify a window of reoxygenation, during which tumor blood flow, oxygen consumption and radiation sensitivity were assessed. RESULTS: Reoxygenation was shown after two days of treatments with Sorafenib or PD0325901 in two tumor models, which was further successfully exploited with Sorafenib for improving the radiation response of FSaII tumors by a factor of 1.5. The increase in tumor oxygenation was shown to be the result of two major factors: (i) an increase in blood flow for Sorafenib, that might be linked to its anti-angiogenic effect (vascular normalization), and (ii) a decrease in oxygen consumption for Sorafenib and PD0325901, due to an alteration of the mitochondrial activity. CONCLUSION: We evidenced tumor reoxygenation in vivo following MAPK inhibition and suggest a rationale for the combination of radiation therapy with Sorafenib.


Subject(s)
Neoplasms, Experimental/radiotherapy , Oxygen Consumption/drug effects , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/physiology , Angiogenesis Inducing Agents , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Blotting, Western , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Electron Spin Resonance Spectroscopy , Fibrosarcoma/metabolism , Fibrosarcoma/radiotherapy , Glutathione/analysis , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/radiotherapy , Mice , Neoplasms, Experimental/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , Sorafenib
16.
PLoS One ; 7(3): e33418, 2012.
Article in English | MEDLINE | ID: mdl-22428047

ABSTRACT

Switching to a glycolytic metabolism is a rapid adaptation of tumor cells to hypoxia. Although this metabolic conversion may primarily represent a rescue pathway to meet the bioenergetic and biosynthetic demands of proliferating tumor cells, it also creates a gradient of lactate that mirrors the gradient of oxygen in tumors. More than a metabolic waste, the lactate anion is known to participate to cancer aggressiveness, in part through activation of the hypoxia-inducible factor-1 (HIF-1) pathway in tumor cells. Whether lactate may also directly favor HIF-1 activation in endothelial cells (ECs) thereby offering a new druggable option to block angiogenesis is however an unanswered question. In this study, we therefore focused on the role in ECs of monocarboxylate transporter 1 (MCT1) that we previously identified to be the main facilitator of lactate uptake in cancer cells. We found that blockade of lactate influx into ECs led to inhibition of HIF-1-dependent angiogenesis. Our demonstration is based on the unprecedented characterization of lactate-induced HIF-1 activation in normoxic ECs and the consecutive increase in vascular endothelial growth factor receptor 2 (VEGFR2) and basic fibroblast growth factor (bFGF) expression. Furthermore, using a variety of functional assays including endothelial cell migration and tubulogenesis together with in vivo imaging of tumor angiogenesis through intravital microscopy and immunohistochemistry, we documented that MCT1 blockers could act as bona fide HIF-1 inhibitors leading to anti-angiogenic effects. Together with the previous demonstration of MCT1 being a key regulator of lactate exchange between tumor cells, the current study identifies MCT1 inhibition as a therapeutic modality combining antimetabolic and anti-angiogenic activities.


Subject(s)
Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1/metabolism , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Symporters/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Movement/physiology , Endothelial Cells/physiology , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/metabolism , Humans , Immunohistochemistry , Lactic Acid/pharmacology , Luciferases , Mice , Nuclear Magnetic Resonance, Biomolecular , Polymerase Chain Reaction , RNA Interference , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Free Radic Biol Med ; 52(6): 993-1002, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22245095

ABSTRACT

Targeting the altered redox status of cancer cells is emerging as an interesting approach to potentiate chemotherapy. However, to maximize the effectiveness of this strategy and define the correct chemotherapeutic associations, it is important to understand the biological consequences of chronically exposing cancer cells to reactive oxygen species (ROS). Using an H(2)O(2)-generating system, we selected a ROS-resistant MCF-7 breast cancer cell line, namely Resox cells. By exploring different survival pathways that are usually induced during oxidative stress, we identified a constitutive overexpression of the endoplasmic reticulum chaperone, GRP94, in these cells, whereas levels of its cytoplasmic homolog HSP90, or GRP78, were not modified. This overexpression was not mediated by constitutive unfolded protein response (UPR) activation. The increase in GRP94 is tightly linked to an increase in cell proliferation and migration capacities, as shown by GRP94-silencing experiments. Interestingly, we also observed that GRP94 silencing inhibits migration and proliferation of the highly aggressive MDA-MB-231 cells. By immunohistochemistry, we showed that GRP94 expression was higher in recurrent human breast cancers than in their paired primary neoplasias. Similar to the situation in the Resox cells, this increase was not associated with an increase in UPR activation in recurrent tumors. In conclusion, this study suggests that GRP94 overexpression may be a hallmark of aggressiveness and recurrence in breast cancers.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Oxidative Stress , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Survival , Endoplasmic Reticulum Chaperone BiP , Female , HSP70 Heat-Shock Proteins/genetics , Humans , Hydrogen Peroxide/metabolism , Membrane Proteins/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Recurrence , Up-Regulation
18.
Invest New Drugs ; 30(3): 1003-11, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21465233

ABSTRACT

Several phenylaminopyrimidoisoquinolinequinones (APIQs) were tested for their cytotoxicity against different cancer cell lines (K562, T24, HepG2) in the presence or absence of ascorbate. Ascorbate enhanced the toxic effects of quinones with first half-wave potential E(I) (1/2) values in the range of -480 to -660 mV. Phenylaminoquinones that were unsubstituted at position 6 exhibited greater cytotoxic activity than did their 6-methyl-substituted analogues. Two groups of compounds were further selected, namely 8-10 and 20-22, to study the cellular mechanisms involved in quinone cytotoxicity. Indeed, these compounds have the same range of redox potentials but differed considerably in their capacity to induce cell death. In the presence of ascorbate, the cell demise induced by compounds 8-10 was not caspase-3 dependent, as shown by the lack of activation of caspase-3 and the absence of cleaved PARP fragments. In addition, an index of ER stress (eIF2α phosphorylation) was activated by these compounds. Quinones 8-10 decreased the cellular capacity to reduce MTT dye and caused marked ATP depletion. Taken together, our results show that ascorbate enhances quinone redox-cycling and leads to ROS formation that inhibits cell proliferation and provokes caspase-independent cell death. Interestingly, we also observed that quinone 8 had a rather selective effect given that freshly isolated peripheral blood leukocytes from human healthy donors were more resistant than human leukemia K562 cells.


Subject(s)
Ascorbic Acid/pharmacology , Cytostatic Agents/pharmacology , Necrosis/chemically induced , Quinolones/pharmacology , Adult , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Humans , L-Lactate Dehydrogenase/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Male , Middle Aged , Necrosis/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Oxidative Stress/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism
19.
Cancer Res ; 72(2): 482-90, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22139377

ABSTRACT

Arsenic trioxide (As(2)O(3)) is an effective therapeutic against acute promyelocytic leukemia and certain solid tumors. Because As(2)O(3) inhibits mitochondrial respiration in leukemia cells, we hypothesized that As(2)O(3) might enhance the radiosensitivity of solid tumors by increasing tumor oxygenation [partial pressure of oxygen (pO(2))] via a decrease in oxygen consumption. Two murine models of radioresistant hypoxic cancer were used to study the effects of As(2)O(3). We measured pO(2) and the oxygen consumption rate in vivo by electron paramagnetic resonance oximetry and (19)fluorine-MRI relaxometry. Tumor perfusion was assessed by Patent blue staining. In both models, As(2)O(3) inhibited mitochondrial respiration, leading to a rapid increase in pO(2). The decrease in oxygen consumption could be explained by an observed decrease in glutathione in As(2)O(3)-treated cells, as this could increase intracellular reactive oxygen species that can disrupt mitochondrial membrane potential. When tumors were irradiated during periods of As(2)O(3)-induced augmented oxygenation, radiosensitivity increased by 2.2-fold compared with control mice. Notably, this effect was abolished when temporarily clamped tumors were irradiated. Together, our findings show that As(2)O(3) acutely increases oxygen consumption and radiosensitizes tumors, providing a new rationale for clinical investigations of As(2)O(3) in irradiation protocols to treat solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Carcinoma, Lewis Lung/drug therapy , Liver Neoplasms, Experimental/drug therapy , Oxides/pharmacology , Oxygen Consumption/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Arsenic Trioxide , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/radiotherapy , Combined Modality Therapy , Disease Models, Animal , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/radiotherapy , Male , Mice , Mice, Inbred C57BL
20.
Biochem Pharmacol ; 82(11): 1540-8, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21843513

ABSTRACT

Cancer cells generally exhibit high levels of reactive oxygen species (ROS) that stimulate cell proliferation and promote genetic instability. Since this biochemical difference between normal and cancer cells represents a specific vulnerability that can be selectively targeted for cancer therapy, various ROS-generating agents are currently in clinical trials, either as single agents or in combination with standard therapy. However, little is known about the potential consequences of an increased oxidative stress for the efficacy of standard chemotherapeutic agents. In this context, we have assessed the influence of an oxidative stress generated by the combination of ascorbate and the redox-active quinone menadione on the capacity of melphalan, a common alkylating agent, to induce apoptosis in a chronic myelogenous leukemia cell line. Our data show that oxidative stress did not inhibit but rather promoted cancer cell killing by melphalan. Interestingly, we observed that, in the presence of oxidative stress, the type of cell death shifted from a caspase-3 dependent apoptosis to necrosis because of an ATP depletion which prevented caspase activation. Taken together, these data suggest that ROS-generating agents could be useful in combination with standard chemotherapy, even if all the molecular consequences of such an addition remain to be determined.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Ascorbic Acid/pharmacology , Melphalan/pharmacology , Vitamin K 3/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Drug Interactions , Humans , K562 Cells , Necrosis , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...