Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 60: 128586, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35085721

ABSTRACT

Menadione (VK3) is used as a powerful inducer of cellular reactive oxygen species (ROS) for many years and displays the high anti-cancer activities in vivo. Recently, the development of mitochondria-targeted drugs has been more and more appreciated. Here, the thirteen derivatives of VK3 were synthesized, which could localize in mitochondria by the triphenylphosphonium (TPP) cation or the nitrogen-based cation. The results of cytotoxicity from six human cancer cell lines showed that the targeted compounds T1-T13 displayed higher activity than VK3 with the average IC50 value around 1 µM. The results of cytotoxicity indicated that the substitutes on C-2, the linear alkyl chains on C-3 and cation moiety all could affect the cytotoxicity. The mechanistic studies showed that five representative compounds (T2, T3, T5, T8 and T13) could localize in cellular mitochondria, elicit ROS burst and collapse mitochondrial membrane potential (ΔΨm), leading to cytochrome C release and apoptosis in MGC-803 cells. Particularly, they could obviously inhibit mitochondrial thioredoxin reductase TrxR2 expression, thus leading to aggravate cellular oxidative stress.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Mitochondria/drug effects , Thioredoxin Reductase 2/antagonists & inhibitors , Vitamin K 3/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cations/chemical synthesis , Cations/chemistry , Cations/pharmacology , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mitochondria/metabolism , Molecular Structure , Structure-Activity Relationship , Thioredoxin Reductase 2/metabolism , Vitamin K 3/chemical synthesis , Vitamin K 3/chemistry
2.
Bioorg Med Chem Lett ; 33: 127750, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33340662

ABSTRACT

Targeting specific mitochondrial alterations to kill cancer cells without affecting their normal counterparts emerges as a feasible strategy. Coumarin derivatives have demonstrated the potential anti-breast cancer activities. By coupling coumarin-3-carboxamide derivatives with mitochondria carrier triphenylphosphonium, mitocoumarins 15a-c were produced and tested as the anti-breast cancer fluorescence agents. Among them, 15b as the amide-based drug potently suppressed the cell growth in MCF-7, MDA-231, SK-BR-3 breast cancer cells with the IC50 values from 3.0 to 4.1 µM, including the lower cytotoxicity to normal MCF-10A cells with the IC50 value around 45.30 ± 2.45 µM. In mechanistic study for 15b in MDA-MB-231 cells, it could localize in mitochondria to elicit ROS burst and collapse Δψm. Besides, it could deplete GSH by an irreversible alkylation process and moderately inhibit mitochondrial thioredoxin reductase TrxR2, thus leading to aggravate cellular oxidative stress. This study reported 15b might be useful for the further development into a mitochondria-targeted anti-triple negative breast cancer drug.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coumarins/pharmacology , Fluorescent Dyes/pharmacology , Mitochondria/drug effects , Thioredoxin Reductase 2/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Mitochondria/metabolism , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Thioredoxin Reductase 2/metabolism
3.
Biochimie ; 162: 46-54, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30946948

ABSTRACT

Auranofin is a gold (I)-containing compound used for the treatment of rheumatic arthritis. Auranofin has anticancer activity in animal models and is approved for clinical trials for lung and ovarian carcinomas. Both the cytosolic and mitochondrial forms of the selenoprotein thioredoxin reductase (TrxR) are well documented targets of auranofin. Auranofin was recently reported to also inhibit proteasome activity at the level of the proteasome-associated deubiquitinases (DUBs) UCHL5 and USP14. We here set out to re-examine the molecular mechanism underlying auranofin cytotoxicity towards cultured cancer cells. The effects of auranofin on the proteasome were examined in cells and in vitro, effects on DUB activity were assessed using different substrates. The cellular response to auranofin was compared to that of the 20S proteasome inhibitor bortezomib and the 19S DUB inhibitor b-AP15 using proteomics. Auranofin was found to inhibit mitochondrial activity and to an induce oxidative stress response at IC50 doses. At 2-3-fold higher doses, auranofin inhibits proteasome processing in cells. At such supra-pharmacological concentrations USP14 activity was inhibited. Analysis of protein expression profiles in drug-exposed tumor cells showed that auranofin induces a response distinct from that of the 20S proteasome inhibitor bortezomib and the DUB inhibitor b-AP15, both of which induced similar responses. Our results support the notion that the primary mechanism of action of auranofin is TrxR inhibition and suggest that proteasome DUB inhibition is an off-target effect. Whether proteasome inhibition will contribute to the antineoplastic effect of auranofin in treated patients is unclear but remains a possibility.


Subject(s)
Antineoplastic Agents/pharmacology , Auranofin/pharmacology , Drug Repositioning , Selenoproteins/antagonists & inhibitors , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 2/antagonists & inhibitors , Bortezomib/pharmacology , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Mitochondria/drug effects , Oxidative Stress/drug effects , Piperidones/pharmacology , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology , Thioredoxin Reductase 2/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors
4.
Int J Pharm ; 555: 346-355, 2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30500459

ABSTRACT

Targeting mitochondrial redox homeostasis is an appealing methodology for cancer therapeutics because of the upregulated antioxidant capacity in drug resistance cases. By coupling triphenylamine (TPA) with an excellent fluorescent group BODIPY, a novel mitochondrial-targeted fluorescent probe, BODIPY-TPA (BTPA), was synthesized and characterized. Confocal microscopic colocalization imaging indicated that BTPA exhibited a subcellular mitochondrial distribution. Cytotoxicity experiments suggested that BTPA exhibited selective anticancer activity via the induction of mitochondrial dysfunction in BGC-823 cancer cells. BTPA induced alterations in mitochondrial redox homeostasis because of the electron-donating property of TPA and mitochondrial selectivity. In further studies, TrxR2 in the mitochondria was alternatively inhibited, which contributed to MtROS accumulation further attenuated PI3K/Akt signaling pathway. The resultant decline in mitochondrial antioxidant capacity aggravated mitochondrial oxidative stress, which is responsible for cytochrome C release and caspase-9 activation. NAC completely reversed BTPA-induced ROS-dependent mitochondrial-mediated intrinsic apoptosis. Therefore, BTPA was designed as a superior fluorescent cancer-imaging probe and a mitochondrial redox-targeting anticancer agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Boron Compounds/administration & dosage , Mitochondria/metabolism , Neoplasms/drug therapy , Aniline Compounds/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Apoptosis/drug effects , Boron Compounds/chemistry , Boron Compounds/pharmacology , Cell Line, Tumor , Cytochromes c/metabolism , Drug Resistance, Neoplasm , Humans , Microscopy, Confocal , Neoplasms/pathology , Oxidation-Reduction , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 2/antagonists & inhibitors
5.
Inorg Chem ; 56(22): 14237-14250, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-29095609

ABSTRACT

We report here on the synthesis of a series of mono- and dinuclear gold(I) complexes exhibiting sulfonated bis(NHC) ligands and novel hydroxylated mono(NHC) Au(I) compounds, which were also examined for their biological activities. Initial cell viability assays show strong antiproliferative activities of the hydroxylated mono(NHC) gold compounds (8 > 9 > 10) against 2008 human ovarian cancer cells even after 1 h incubation. In order to gain insight into the mechanism of biological action of the gold compounds, their effect on the pivotal cellular target seleno-enzyme thioredoxin reductase (TrxR), involved in the maintenance of intracellular redox balance, was investigated in depth. The compounds' inhibitory effects on TrxR and glutathione reductase (GR) were studied comparatively, using either the pure proteins or cancer cell extracts. The results show a strong and selective inhibitory effect of TrxR, specifically for the hydroxyl-functionalized NHC gold(I) complexes (8-10). Valuable information on the gold compounds' molecular reactivity with TrxR was gained using the BIAM (biotin-conjugated iodoacetamide) assay and performing competition experiments by mass spectrometry (MS). In good agreement, both techniques suggest the binding affinity of the mono(NHC) Au(I) complexes toward selenols and thiols. Notably, for the first time, bis-carbene formation from mono-carbenes in buffered solution could be observed by MS, which may provide new insights into the speciation mechanisms of bioactive Au(I) NHC complexes. Furthermore, the compounds' interactions with another relevant in cellulo target, namely telomeric G-quadruplex DNA-a higher-order DNA structure playing key roles in telomere function-was investigated by means of FRET melting assays. The lack of interactions with this type of nucleic acid secondary structure support the idea of selective targeting of the hydrophilic Au(I) NHC compounds toward proteins such as TrxR.


Subject(s)
Coordination Complexes/pharmacology , Gold/chemistry , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , DNA/metabolism , Drug Stability , G-Quadruplexes , Glutathione Reductase/antagonists & inhibitors , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Organogold Compounds/chemistry , Rats , Reactive Oxygen Species/metabolism , Silver/chemistry , Solubility
6.
Free Radic Biol Med ; 113: 530-538, 2017 12.
Article in English | MEDLINE | ID: mdl-29080841

ABSTRACT

Mitocurcumin is a derivative of curcumin, which has been shown to selectively enter mitochondria. Here we describe the anti-tumor efficacy of mitocurcumin in lung cancer cells and its mechanism of action. Mitocurcumin, showed 25-50 fold higher efficacy in killing lung cancer cells as compared to curcumin as demonstrated by clonogenic assay, flow cytometry and high throughput screening assay. Treatment of lung cancer cells with mitocurcumin significantly decreased the frequency of cancer stem cells. Mitocurcumin increased the mitochondrial reactive oxygen species (ROS), decreased the mitochondrial glutathione levels and induced strand breaks in the mitochondrial DNA. As a result, we observed increased BAX to BCL-2 ratio, cytochrome C release into the cytosol, loss of mitochondrial membrane potential and increased caspase-3 activity suggesting that mitocurcumin activates the intrinsic apoptotic pathway. Docking studies using mitocurcumin revealed that it binds to the active site of the mitochondrial thioredoxin reductase (TrxR2) with high affinity. In corroboration with the above finding, mitocurcumin decreased TrxR activity in cell free as well as the cellular system. The anti-cancer activity of mitocurcumin measured in terms of apoptotic cell death and the decrease in cancer stem cell frequency was accentuated by TrxR2 overexpression. This was due to modulation of TrxR2 activity to NADPH oxidase like activity by mitocurcumin, resulting in higher ROS accumulation and cell death. Thus, our findings reveal mitocurcumin as a potent anticancer agent with better efficacy than curcumin. This study also demonstrates the role of TrxR2 and mitochondrial DNA damage in mitocurcumin mediated killing of cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Thioredoxin Reductase 2/genetics , A549 Cells , Antineoplastic Agents, Phytogenic/chemistry , Binding Sites , Caspase 3/genetics , Caspase 3/metabolism , Curcumin/analogs & derivatives , Cytochromes c/genetics , Cytochromes c/metabolism , DNA Breaks, Single-Stranded , DNA, Mitochondrial , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Humans , Kinetics , Mitochondria/metabolism , Mitochondria/pathology , Molecular Docking Simulation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxidation-Reduction/drug effects , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Thermodynamics , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/chemistry , Thioredoxin Reductase 2/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
7.
Brain Pathol ; 27(3): 276-291, 2017 05.
Article in English | MEDLINE | ID: mdl-27117068

ABSTRACT

Preconditioning by brief ischemic episode induces tolerance to a subsequent lethal ischemic insult, and it has been suggested that reactive oxygen species are involved in this phenomenon. Thioredoxin 2 (Trx2), a small protein with redox-regulating function, shows cytoprotective roles against oxidative stress. Here, we had focused on the role of Trx2 in ischemic preconditioning (IPC)-mediated neuroprotection against oxidative stress followed by a subsequent lethal transient cerebral ischemia. Animals used in this study were randomly assigned to six groups; sham-operated group, ischemia-operated group, IPC plus (+) sham-operated group, IPC + ischemia-operated group, IPC + auranofin (a TrxR2 inhibitor) + sham-operated group and IPC + auranofin + ischemia-operated group. IPC was subjected to a 2 minutes of sublethal transient ischemia 1 day prior to a 5 minutes of lethal transient ischemia. A significant loss of neurons was found in the stratum pyramidale (SP) of the hippocampal CA1 region (CA1) in the ischemia-operated-group 5 days after ischemia-reperfusion; in the IPC + ischemia-operated-group, pyramidal neurons in the SP were well protected. In the IPC + ischemia-operated-group, Trx2 and TrxR2 immunoreactivities in the SP and its protein level in the CA1 were not significantly changed compared with those in the sham-operated-group after ischemia-reperfusion. In addition, superoxide dismutase 2 (SOD2) expression, superoxide anion radical ( O2-) production, denatured cytochrome c expression and TUNEL-positive cells in the IPC + ischemia-operated-group were similar to those in the sham-operated-group. Conversely, the treatment of auranofin to the IPC + ischemia-operated-group significantly increased cell damage/death and abolished the IPC-induced effect on Trx2 and TrxR2 expressions. Furthermore, the inhibition of Trx2R nearly cancelled the beneficial effects of IPC on SOD2 expression, O2- production, denatured cytochrome c expression and TUNEL-positive cells. In brief, this study shows that IPC conferred neuroprotection against ischemic injury by maintaining Trx2 and suggests that the maintenance or enhancement of Trx2 expression by IPC may be a legitimate strategy for therapeutic intervention of cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Ischemic Preconditioning , Neurons/metabolism , Neuroprotection/physiology , Thioredoxins/metabolism , Animals , Auranofin/pharmacology , Brain Ischemia/pathology , Brain Ischemia/prevention & control , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Cell Death/drug effects , Cell Death/physiology , Cytochromes c/metabolism , Enzyme Inhibitors/pharmacology , Gerbillinae , Ischemic Preconditioning/methods , Male , Neurons/drug effects , Neurons/pathology , Neuroprotection/drug effects , Oxidative Stress/physiology , Random Allocation , Superoxide Dismutase/metabolism , Superoxides/metabolism , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/metabolism
8.
J Biol Chem ; 291(19): 10021-31, 2016 May 06.
Article in English | MEDLINE | ID: mdl-27002142

ABSTRACT

Parthenolide (PTL), a major active sesquiterpene lactone from the herbal plant Tanacetum parthenium, has been applied in traditional Chinese medicine for centuries. Although PTL demonstrates potent anticancer efficacy in numerous types of malignant cells, the cellular targets of PTL have not been well defined. We reported here that PTL interacts with both cytosolic thioredoxin reductase (TrxR1) and mitochondrial thioredoxin reductase (TrxR2), two ubiquitous selenocysteine-containing antioxidant enzymes, to elicit reactive oxygen species-mediated apoptosis in HeLa cells. PTL selectively targets the selenocysteine residue in TrxR1 to inhibit the enzyme function, and further shifts the enzyme to an NADPH oxidase to generate superoxide anions, leading to reactive oxygen species accumulation and oxidized thioredoxin. Under the conditions of inhibition of TrxRs in cells, PTL does not cause significant alteration of cellular thiol homeostasis, supporting selective target of TrxRs by PTL. Importantly, overexpression of functional TrxR1 or Trx1 confers protection, whereas knockdown of the enzymes sensitizes cells to PTL treatment. Targeting TrxRs by PTL thus discloses an unprecedented mechanism underlying the biological activity of PTL, and provides deep insights to understand the action of PTL in treatment of cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Sesquiterpenes/pharmacology , Thioredoxin Reductase 1/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/chemistry , HeLa Cells , Humans , NADPH Oxidases/metabolism , Neoplasms/enzymology , Sesquiterpenes/chemistry , Superoxides/metabolism , Tanacetum parthenium/chemistry , Thioredoxin Reductase 1/metabolism , Thioredoxin Reductase 2/antagonists & inhibitors
9.
Sci Rep ; 6: 23071, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26975474

ABSTRACT

The mitochondrial thioredoxin system (NADPH, thioredoxin reductase, thioredoxin) is a major redox regulator. Here we have investigated the redox correlation between this system and the mitochondrial enzyme cyclophilin D. The peptidyl prolyl cis-trans isomerase activity of cyclophilin D was stimulated by the thioredoxin system, while it was decreased by cyclosporin A and the thioredoxin reductase inhibitor auranofin. The redox state of cyclophilin D, thioredoxin 1 and 2 and peroxiredoxin 3 was measured in isolated rat heart mitochondria and in tumor cell lines (CEM-R and HeLa) by redox Western blot analysis upon inhibition of thioredoxin reductase with auranofin, arsenic trioxide, 1-chloro-2,4-dinitrobenzene or after treatment with hydrogen peroxide. A concomitant oxidation of thioredoxin, peroxiredoxin and cyclophilin D was observed, suggesting a redox communication between the thioredoxin system and cyclophilin. This correlation was further confirmed by i) co-immunoprecipitation assay of cyclophilin D with thioredoxin 2 and peroxiredoxin 3, ii) molecular modeling and iii) depleting thioredoxin reductase by siRNA. We conclude that the mitochondrial thioredoxin system controls the redox state of cyclophilin D which, in turn, may act as a regulator of several processes including ROS production and pro-apoptotic factors release.


Subject(s)
Cyclophilins/metabolism , Mitochondria, Heart/metabolism , Peroxiredoxin III/metabolism , Thioredoxins/metabolism , Animals , Auranofin/pharmacology , Blotting, Western , Cell Line, Tumor , Peptidyl-Prolyl Isomerase F , Cyclophilins/antagonists & inhibitors , Cyclophilins/chemistry , Cyclosporine/pharmacology , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Mitochondria, Heart/genetics , Models, Molecular , Oxidants/metabolism , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Peroxiredoxin III/chemistry , Protein Binding/drug effects , Protein Domains , RNA Interference , Rats, Wistar , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/genetics , Thioredoxin Reductase 2/metabolism , Thioredoxins/chemistry
10.
J Biol Inorg Chem ; 20(6): 1005-20, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26202908

ABSTRACT

While N-heterocyclic carbenes (NHC) are ubiquitous ligands in catalysis for organic or industrial syntheses, their potential to form transition metal complexes for medicinal applications has still to be exploited. Within this frame, we synthesized new homo- and heterobimetallic complexes based on the Au(I)-NHC scaffold. The compounds were synthesized via a microwave-assisted method developed in our laboratories using Au(I)-NHC complexes carrying a pentafluorophenol ester moiety and another Au(I) phosphane complex or a bipyridine ligand bearing a pendant amine function. Thus, we developed two different methods to prepare homo- and heterobimetallic complexes (Au(I)/Au(I) or Au(I)/Cu(II), Au(I)/Ru(II), respectively). All the compounds were fully characterized by several spectroscopic techniques including far infrared, and were tested for their antiproliferative effects in a series of human cancer cells. They showed moderate anticancer properties. Their toxic effects were also studied ex vivo using the precision-cut tissue slices (PCTS) technique and initial results concerning their reactivity with the seleno-enzyme thioredoxin reductase were obtained.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Organogold Compounds/chemical synthesis , Thioglucosides/chemical synthesis , Thioglucosides/therapeutic use , Animals , Cell Line, Tumor , Copper , Glutathione Reductase/antagonists & inhibitors , Humans , Organogold Compounds/therapeutic use , Rats , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 2/antagonists & inhibitors
11.
Free Radic Biol Med ; 73: 95-105, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24816296

ABSTRACT

The thioredoxin system has essential functions in the maintenance of cellular redox homeostasis in the cytosol, nucleus, and mitochondria. Thioredoxin (Trx) and thioredoxin reductase (TrxR) are targets for mercury compounds in vitro and in vivo. This study aimed at understanding mechanistically how the mitochondrial and cytosolic thioredoxin systems were affected by mercurials, including the regulation of TrxR transcription. The effects of coexposure to selenite and mercurials on the thioredoxin system were also addressed. Results in HepG2 cells showed that TrxR1 expression was enhanced by Hg(2+), whereas exposure to MeHg decreased expression. Selenite exposure also increased the expression of TrxR1 and resulted in higher specific activity. Coexposure to 2 µM selenite and up to 5 µM Hg(2+) increased even further TrxR1 expression. This synergistic effect was not verified for MeHg, because TrxR1 expression and activity were reduced. Analysis of Nrf-2 translocation to the nucleus and TrxR mRNA suggests that induction of TrxR1 transcription was slower upon exposure to MeHg in comparison to Hg(2+). Subcellular fractions showed that MeHg affected the activity of the thioredoxin system equally in the mitochondria and cytosol, whereas Hg(2+) inhibited primarily the activity of TrxR2. The expression of TrxR2 was not upregulated by any treatment. These results show important differences between the mechanisms of toxicity of Hg(2+) and MeHg and stress the narrow range of selenite concentrations capable of antagonizing mercury toxicity. The results also highlight the relevance of the mitochondrial thioredoxin system (TrxR2 and Trx2) in the development of mercury toxicity.


Subject(s)
Mercury Compounds/toxicity , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 2/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation , Hep G2 Cells , Humans , Mitochondria/enzymology , Oxidation-Reduction , RNA, Messenger/biosynthesis , Selenium/metabolism , Thioredoxin Reductase 1/biosynthesis , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 2/biosynthesis , Thioredoxin Reductase 2/genetics , Thioredoxins/biosynthesis
12.
PLoS One ; 7(11): e50683, 2012.
Article in English | MEDLINE | ID: mdl-23226354

ABSTRACT

Mitochondria are considered major generators of cellular reactive oxygen species (ROS) which are implicated in the pathogenesis of neurodegenerative diseases such as Parkinson's disease (PD). We have recently shown that isolated mitochondria consume hydrogen peroxide (H2O2) in a substrate- and respiration-dependent manner predominantly via the thioredoxin/peroxiredoxin (Trx/Prx) system. The goal of this study was to determine the role of Trx/Prx system in dopaminergic cell death. We asked if pharmacological and lentiviral inhibition of the Trx/Prx system sensitized dopaminergic cells to mitochondrial dysfunction, increased steady-state H2O2 levels and death in response to toxicants implicated in PD. Incubation of N27 dopaminergic cells or primary rat mesencephalic cultures with the Trx reductase (TrxR) inhibitor auranofin in the presence of sub-toxic concentrations of parkinsonian toxicants paraquat; PQ or 6-hydroxydopamine; 6OHDA (for N27 cells) resulted in a synergistic increase in H2O2 levels and subsequent cell death. shRNA targeting the mitochondrial thioredoxin reductase (TrxR2) in N27 cells confirmed the effects of pharmacological inhibition. A synergistic decrease in maximal and reserve respiratory capacity was observed in auranofin treated cells and TrxR2 deficient cells following incubation with PQ or 6OHDA. Additionally, TrxR2 deficient cells showed decreased basal mitochondrial oxygen consumption rates. These data demonstrate that inhibition of the mitochondrial Trx/Prx system sensitizes dopaminergic cells to mitochondrial dysfunction, increased steady-state H2O2, and cell death. Therefore, in addition to their role in the production of cellular H2O2 the mitochondrial Trx/Prx system serve as a major sink for cellular H2O2 and its disruption may contribute to dopaminergic pathology associated with PD.


Subject(s)
Dopaminergic Neurons/cytology , Mitochondria/enzymology , Mitochondria/pathology , Oxidative Stress , Thioredoxin Reductase 2/deficiency , Animals , Auranofin/pharmacology , Cell Death/drug effects , Cell Death/genetics , Cell Line , Cell Respiration/drug effects , Cell Respiration/genetics , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Drug Synergism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Hydrogen Peroxide/metabolism , Lentivirus/genetics , Mesencephalon/cytology , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Oxidopamine/pharmacology , Paraquat/pharmacology , Peroxiredoxins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/genetics , Thioredoxin Reductase 2/metabolism
13.
Placenta ; 33(12): 1012-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23063346

ABSTRACT

Oxidative stress is a key feature in the pathogenesis of pre-eclampsia and antioxidants have been proposed as a potential therapy in the treatment of this important complication of pregnancy. In this report selenium supplementation was used to up-regulate the antioxidant enzymes glutathione peroxidase and thioredoxin reductase and the protective effect that this had on cellular metabolism during oxidative stress was examined. Bewo and Jeg-3 trophoblast cells were supplemented with organic and inorganic forms of selenium and 3 forms of peroxide in a range of doses were utilised to generate oxidative stress. Thioredoxin reductase and glutathione peroxidase activity were maximally expressed after supplementation with 100 nM NaSe and 500 nM SeMethionine. Application of H2O2 in the range of 200-400 µM for 24h resulted in significant (p<0.001) inhibition of cellular activity, an effect negated by Se supplementation. Tert-butyl H2O2 and cumene H2O2 concentrations between 30 and 50 uM similarly inhibited cellular activity and this could be significantly (p<0.001) reversed by Se supplementation. Auranofin, a specific inhibitor of thioredoxin reductase and glutathione peroxidase was used to prove that the protective effect generated by Se supplementation was due to up regulation of these enzymes. These studies provide direct evidence that selenium supplementation can up-regulate endogenous antioxidant systems and protects trophoblast cells from oxidative stress. This may inform the development of future therapies for pre-eclampsia and emphasises the importance of selenium adequacy during pregnancy.


Subject(s)
Enzyme Induction , Glutathione Peroxidase/metabolism , Oxidative Stress , Selenium/metabolism , Thioredoxin Reductase 1/metabolism , Thioredoxin Reductase 2/metabolism , Trophoblasts/metabolism , Auranofin/pharmacology , Cell Line , Dietary Supplements , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Female , Glutathione Peroxidase/antagonists & inhibitors , Glutathione Peroxidase/genetics , Humans , Osmolar Concentration , Oxidants/pharmacology , Pre-Eclampsia/prevention & control , Pregnancy , Protective Agents/chemistry , Protective Agents/metabolism , Protective Agents/therapeutic use , Selenium/chemistry , Selenium/therapeutic use , Selenomethionine/antagonists & inhibitors , Selenomethionine/metabolism , Selenomethionine/therapeutic use , Sodium Selenite/antagonists & inhibitors , Sodium Selenite/metabolism , Sodium Selenite/therapeutic use , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/genetics , Trophoblasts/drug effects , Glutathione Peroxidase GPX1
14.
J Biol Chem ; 286(38): 33669-77, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21832082

ABSTRACT

Respiring mitochondria produce H(2)O(2) continuously. When production exceeds scavenging, H(2)O(2) emission occurs, endangering cell functions. The mitochondrial peroxidase peroxiredoxin-3 reduces H(2)O(2) to water using reducing equivalents from NADPH supplied by thioredoxin-2 (Trx2) and, ultimately, thioredoxin reductase-2 (TrxR2). Here, the contribution of this mitochondrial thioredoxin system to the control of H(2)O(2) emission was studied in isolated mitochondria and cardiomyocytes from mouse or guinea pig heart. Energization of mitochondria by the addition of glutamate/malate resulted in a 10-fold decrease in the ratio of oxidized to reduced Trx2. This shift in redox state was accompanied by an increase in NAD(P)H and was dependent on TrxR2 activity. Inhibition of TrxR2 in isolated mitochondria by auranofin resulted in increased H(2)O(2) emission, an effect that was seen under both forward and reverse electron transport. This effect was independent of changes in NAD(P)H or membrane potential. The effects of auranofin were reproduced in cardiomyocytes; superoxide and H(2)O(2) levels increased, but similarly, there was no effect on NAD(P)H or membrane potential. These data show that energization of mitochondria increases the antioxidant potential of the TrxR2/Trx2 system and that inhibition of TrxR2 results in increased H(2)O(2) emission through a mechanism that is independent of changes in other redox couples.


Subject(s)
Hydrogen Peroxide/metabolism , Mitochondria, Heart/enzymology , Thioredoxin Reductase 2/metabolism , Animals , Auranofin/pharmacology , Dinitrochlorobenzene/pharmacology , Electron Transport/drug effects , Energy Metabolism/drug effects , Enzyme Assays , Glutathione/metabolism , Guinea Pigs , Mice , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidation-Reduction/drug effects , Peroxiredoxin III/metabolism , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxins/metabolism
15.
Free Radic Biol Med ; 50(6): 689-99, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21172426

ABSTRACT

The cytosolic and mitochondrial thioredoxin reductases (TrxR1 and TrxR2) and thioredoxins (Trx1 and Trx2) are key components of the mammalian thioredoxin system, which is important for antioxidant defense and redox regulation of cell function. TrxR1 and TrxR2 are selenoproteins generally considered to have comparable properties, but to be functionally separated by their different compartments. To compare their properties we expressed recombinant human TrxR1 and TrxR2 and determined their substrate specificities and inhibition by metal compounds. TrxR2 preferred its endogenous substrate Trx2 over Trx1, whereas TrxR1 efficiently reduced both Trx1 and Trx2. TrxR2 displayed strikingly lower activity with dithionitrobenzoic acid (DTNB), lipoamide, and the quinone substrate juglone compared to TrxR1, and TrxR2 could not reduce lipoic acid. However, Sec-deficient two-amino-acid-truncated TrxR2 was almost as efficient as full-length TrxR2 in the reduction of DTNB. We found that the gold(I) compound auranofin efficiently inhibited both full-length TrxR1 and TrxR2 and truncated TrxR2. In contrast, some newly synthesized gold(I) compounds and cisplatin inhibited only full-length TrxR1 or TrxR2 and not truncated TrxR2. Surprisingly, one gold(I) compound, [Au(d2pype)(2)]Cl, was a better inhibitor of TrxR1, whereas another, [(iPr(2)Im)(2)Au]Cl, mainly inhibited TrxR2. These compounds also inhibited TrxR activity in the cytoplasm and mitochondria of cells, but their cytotoxicity was not always dependent on the proapoptotic proteins Bax and Bak. In conclusion, this study reveals significant differences between human TrxR1 and TrxR2 in substrate specificity and metal compound inhibition in vitro and in cells, which may be exploited for development of specific TrxR1- or TrxR2-targeting drugs.


Subject(s)
Enzyme Inhibitors/pharmacology , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 1/metabolism , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/metabolism , Thioredoxins/metabolism , Animals , Antioxidants/metabolism , Cisplatin/metabolism , Cisplatin/pharmacology , Cytosol/enzymology , Dithionitrobenzoic Acid/metabolism , Humans , Mice , Mitochondria/enzymology , Naphthoquinones/metabolism , Organogold Compounds/pharmacology , Oxidation-Reduction , Recombinant Proteins/metabolism , Substrate Specificity , Thioctic Acid/analogs & derivatives , Thioctic Acid/metabolism , Thioredoxin Reductase 1/chemistry , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 2/chemistry , Thioredoxin Reductase 2/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
16.
Am J Physiol Heart Circ Physiol ; 298(1): H194-201, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19897710

ABSTRACT

The intracellular redox state is stringently maintained by thiol-based antioxidants to establish a balance for the physiological and pathophysiological roles of reactive oxygen species. The relative contributions of the thioredoxin (Trx) and glutathione/glutaredoxin systems to intracellular redox balance are incompletely understood, as are the consequences of altered thiol metabolism on endothelial nitric oxide (NO) synthase (eNOS) and NO-dependent pathways in the endothelium. We designed duplex small interfering RNA (siRNA) constructs to specifically "knock down" the expression of three key thiol-metabolizing enzymes in cultured aortic endothelial cells. Transfection of siRNA constructs targeting glutathione reductase (GR), cytosolic Trx reductase (TrxR1), or mitochondrial Trx reductase (TrxR2) significantly decreased the intracellular reduced glutathione-to-oxidized glutathione ratio. siRNA-mediated knockdown of either GR, TrxR1, or TrxR2 markedly suppressed VEGF-induced NO production (measured by an electrochemical NO sensor) and also blocked eNOS enzyme activity (using the [(3)H]arginine/[(3)H]citrulline assay). Pretreatment of endothelial cells with N,N'-bis(2-chloroethyl)-N-nitrosourea, an inhibitor of GR and TrxR, significantly decreased VEGF-induced NO production. siRNA-mediated TrxR2 knockdown led to a marked increase in hydrogen peroxide (H(2)O(2)) production in endothelial cells. In contrast, knockdown of GR or TrxR1 only slightly increased H(2)O(2) production. Supplementation of endothelial cells with tetrahydrobiopterin prevented the increase in H(2)O(2) generation seen with siRNA-mediated knockdown of GR. These studies show that the differential regulation of thiol-metabolizing proteins leads to critical changes in oxidative and nitrosative stress pathways. Greater understanding of the differential regulation of thiol-metabolizing proteins may lead to the development of new pharmacological targets for diseases associated with oxidative stress in the vascular wall.


Subject(s)
Biopterins/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide Synthase Type III/metabolism , Proteins/metabolism , Sulfhydryl Compounds/metabolism , Animals , Biopterins/analogs & derivatives , Cattle , Cells, Cultured , Disulfides/metabolism , Electrochemistry , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Glutathione Reductase/antagonists & inhibitors , Glutathione Reductase/genetics , Glutathione Reductase/metabolism , Hydrogen Peroxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Oxidation-Reduction , RNA, Small Interfering/pharmacology , Thioredoxin Reductase 1/antagonists & inhibitors , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/metabolism , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/genetics , Thioredoxin Reductase 2/metabolism , Transfection
17.
Biochemistry ; 48(26): 6213-23, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19366212

ABSTRACT

Mammalian thioredoxin reductase (TR) contains a rare selenocysteine (Sec) residue in a conserved redox-active tetrapeptide of sequence Gly-Cys(1)-Sec(2)-Gly. The high chemical reactivity of the Sec residue is thought to confer broad substrate specificity to the enzyme. In addition to utilizing thioredoxin (Trx) as a substrate, other substrates are protein disulfide isomerase, glutaredoxin, glutathione peroxidase, NK-lysin/granulysin, HIV Tat protein, H(2)O(2), lipid hydroperoxides, vitamin K, ubiquinone, juglone, ninhydrin, alloxan, dehydroascorbate, DTNB, lipoic acid/lipoamide, S-nitrosoglutathione, selenodiglutathione, selenite, methylseleninate, and selenocystine. Here we show that the Cys(2) mutant enzyme or the N-terminal reaction center alone can reduce Se-containing substrates selenocystine and selenite with only slightly less activity than the wild-type enzyme, in stark contrast to when Trx is used as the substrate when the enzyme suffers a 175-550-fold reduction in k(cat). Our data support the use of alternative mechanistic pathways for the Se-containing substrates that bypass a critical ring-forming step when Trx is the substrate. We also show that lipoic acid can be reduced through a Sec-independent mechanism that involves the N-terminal reaction center. These results show that the broad substrate specificity of the mammalian enzyme is not due to the presence of the rare Sec residue but is due to the catalytic power of the N-terminal reaction center. We hypothesize that the N-terminal reaction center can reduce substrates (i) with good leaving groups such as DTNB, (ii) that are highly electrophilic such as selenite, or (iii) that are activated by strain such as lipoic acid/lipoamide. We also show that the absence of Sec only changed the IC(50) for aurothioglucose by a factor of 1.7 in the full-length mammalian enzyme (83-142 nM), but surprisingly the truncated enzyme showed much stronger inhibition (25 nM). This contrasts with auranofin, where the absence of Sec more strongly perturbed inhibition.


Subject(s)
Selenium/chemistry , Selenocysteine/chemistry , Thioredoxin-Disulfide Reductase/chemistry , Amino Acid Substitution , Animals , Auranofin/chemistry , Aurothioglucose/chemistry , Biocatalysis , Caenorhabditis elegans/enzymology , Cystine/analogs & derivatives , Cystine/chemistry , Dinitrobenzenes/chemistry , Dithiothreitol/chemistry , Drosophila melanogaster/enzymology , Enzyme Inhibitors/chemistry , Gene Deletion , Glutathione/chemistry , Hydrogen-Ion Concentration , Kinetics , Mice , Models, Chemical , Organoselenium Compounds/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Selenocysteine/genetics , Sodium Selenite/chemistry , Substrate Specificity , Thioctic Acid/chemistry , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/chemistry , Thioredoxin Reductase 2/genetics , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/genetics
18.
J Med Chem ; 50(24): 5871-4, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-17975904

ABSTRACT

A novel "Keppler type" ruthenium(III) compound trans-[bis(2-amino 5-methylthiazole)tetrachlororuthenate(III)] 1, of potential interest as an anticancer agent, was designed, synthesized, and characterized. Its interactions with various proteins were analyzed, including the selenoenzyme thioredoxin reductase, an emerging target for anticancer metallodrugs. The selective inhibition of the cytosolic form of this selenoenzyme was documented, this being the first report of significant thioredoxin reductase inhibition by a ruthenium compound.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cytosol/enzymology , Organometallic Compounds/chemical synthesis , Thioredoxin Reductase 1/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Mitochondria/enzymology , Organometallic Compounds/chemistry , Rats , Thioredoxin Reductase 1/chemistry , Thioredoxin Reductase 2/antagonists & inhibitors , Thioredoxin Reductase 2/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...