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










Publication year range
1.
Sci Rep ; 11(1): 10935, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035383

ABSTRACT

Bacterial infection of pressure ulcers (PUs) are a notable source of hospitalization for individuals with diabetes. This study evaluated the safety profile and efficacy of auranofin to treat diabetic PUs infected with methicillin-resistant Staphylococcus aureus (MRSA). PUs were infected with MRSA in diabetic TALLYHO/JngJ mice and then treated with topical auranofin (2%), topical mupirocin (2%), or oral clindamycin (30 mg/kg) for four days. PUs were harvested post-treatment to enumerate bacterial burden and determine expression of cytokines/growth factors. Landrace cross pigs were exposed topically to auranofin (1%, 2%, and 3%) for 4-14 days and evaluated for signs of localized or systemic toxicity. Auranofin eradicated MRSA in PUs within four days (7.92-log10 reduction) in contrast to mupirocin (2.15-log10 reduction) and clindamycin (0.73-log10 reduction). Additionally, auranofin treatment resulted in decreased expression of pro-inflammatory cytokines and increased expression of biomarkers associated with re-epithelization of wounded tissue, confirmed with histopathologic analysis. No significant histopathologic lesions were present on porcine skin sites exposed to topical auranofin. Additionally, minimal accumulation of plasma gold and no systemic toxicity was observed in pigs exposed to topical auranofin. Auranofin appears to be a potent and safe topical agent to further investigate for treatment of mild-to-moderate MRSA-infected diabetic PUs.


Subject(s)
Auranofin/therapeutic use , Diabetes Complications/drug therapy , Pressure Ulcer/drug therapy , Skin/drug effects , Staphylococcal Skin Infections/drug therapy , Animals , Anti-Bacterial Agents/therapeutic use , Auranofin/adverse effects , Auranofin/toxicity , Clindamycin/therapeutic use , Female , Male , Methicillin-Resistant Staphylococcus aureus , Mice , Mupirocin/therapeutic use , Pressure Ulcer/etiology , Skin/pathology , Sus scrofa
2.
J Appl Toxicol ; 37(5): 602-610, 2017 05.
Article in English | MEDLINE | ID: mdl-27813112

ABSTRACT

Auranofin (AF) is used in clinic for the treatment of rheumatoid arthritis, repurposing of AF as an anticancer drug has just finished a phase I/II clinical trial, but the developmental toxicity of AF remains obscure. This study focused on its developmental toxicity by using zebrafish embryos. Zebrafish embryos were exposed to different concentrations (1, 2.5, 5, 10 µm) of AF from 2 h post-fertilization (hpf) to 72 hpf. At 72 hpf, two major developmental defects caused by AF were found, namely severe pericardial edema and hypopigmentation, when embryos were exposed to concentrations higher than 2.5 µm. Biochemical detection of oxidative stress enzyme combined with expressions of a series of genes related to oxidative stress, cardiac, metal stress and pigment formation were subsequently tested. The superoxide dismutase activity was decreased while malondialdehyde content was accumulated by AF treatment. The expression of oxidative stress-related genes (sod1, gpx1a, gst), pigment-related genes (mitfb, trp-1a) and one metal stress-related gene ctr1 were all decreased by AF exposure. The expressions of cardiac-related genes (amhc, vmhc) and one metal-related gene hsp70 were found to be significantly upregulated by AF exposure. These findings indicated the potential developmental toxicity of AF on zebrafish early development. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Antirheumatic Agents/toxicity , Auranofin/toxicity , Embryonic Development/drug effects , Zebrafish , Animals , Edema/chemically induced , Edema/pathology , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Heart/drug effects , Hypopigmentation/chemically induced , Hypopigmentation/genetics , Hypopigmentation/pathology , Malondialdehyde/metabolism , Metals/toxicity , Oxidative Stress/drug effects , Stress, Physiological/drug effects , Superoxide Dismutase/metabolism , Teratogens
3.
Redox Biol ; 6: 648-655, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26584358

ABSTRACT

Exposure to environmental stressors during embryo development can have long-term effects on the adult organism. This study used the thioredoxin reductase inhibitor auranofin to investigate the consequences of oxidative stress during zebrafish development. Auranofin at low doses triggered upregulation of the antioxidant genes gstp1 and prdx1. As the dose was increased, acute developmental abnormalities, including cerebral hemorrhaging and jaw malformation, were observed. To determine whether transient disruption of redox homeostasis during development could have long-term consequences, zebrafish embryos were exposed to a low dose of auranofin from 6-24 hours post fertilization, and then raised to adulthood. The adult fish were outwardly normal in their appearance with no gross physical differences compared to the control group. However, these adult fish had reduced odds of breeding and a lower incidence of egg fertilization. This study shows that a suboptimal early life environment can reduce the chances of reproductive success in adulthood.


Subject(s)
Embryo, Nonmammalian/metabolism , Fertility , Oxidative Stress , Reproduction , Animals , Auranofin/toxicity , Embryonic Development , Female , Male , Metabolic Networks and Pathways , Phenotype , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Zebrafish , Zebrafish Proteins/antagonists & inhibitors
4.
Redox Biol ; 5: 319-327, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26114584

ABSTRACT

Transforming growth factor ß-activated kinase 1 (TAK1) is critical for survival of many KRAS mutated colorectal cancer cells, and TAK1 inhibition with 5Z-7-oxozeaenol has been associated with oxidative stress leading to tumor cell killing. When SW 620 and HCT 116 human colon cancer cells were treated with 5µM 5Z-7-oxozeaenol, cell viability, growth, and clonogenic survival were significantly decreased. Consistent with TAK1 inhibition being causally related to thiol-mediated oxidative stress, 10mM N-acetylcysteine (NAC) partially reversed the growth inhibitory effects of 5Z-7-oxozeaenol. In addition, 5Z-7-oxozeaenol also increased steady-state levels of H2DCFDA oxidation as well as increased levels of total glutathione (GSH) and glutathione disulfide (GSSG). Interestingly, depletion of GSH using buthionine sulfoximine did not significantly potentiate 5Z-7-oxozeaenol toxicity in either cell line. In contrast, pre-treatment of cells with auranofin (Au) to inhibit thioredoxin reductase activity significantly increased levels of oxidized thioredoxin as well as sensitized cells to 5Z-7-oxozeaenol-induced growth inhibition and clonogenic cell killing. These results were confirmed in SW 620 murine xenografts, where treatment with 5Z-7-oxozeaenol or with Au plus 5Z-7-oxozeaenol significantly inhibited growth, with Au plus 5Z-7-oxozeaenol trending toward greater growth inhibition compared to 5Z-7-oxozeaenol alone. These results support the hypothesis that thiol-mediated oxidative stress is causally related to TAK1-induced colon cancer cell killing. In addition, these results support the hypothesis that thioredoxin metabolism is a critical target for enhancing colon cancer cell killing via TAK1 inhibition and could represent an effective therapeutic strategy in patients with these highly resistant tumors.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Thioredoxins/metabolism , ras Proteins/genetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Auranofin/chemistry , Auranofin/therapeutic use , Auranofin/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Glutathione/metabolism , HCT116 Cells , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice , Mice, Nude , Mutation , Oxidative Stress/drug effects , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Transplantation, Heterologous , Zearalenone/analogs & derivatives , Zearalenone/chemistry , Zearalenone/therapeutic use , Zearalenone/toxicity , ras Proteins/metabolism
5.
Mol Biochem Parasitol ; 201(1): 16-25, 2015 May.
Article in English | MEDLINE | ID: mdl-26024834

ABSTRACT

Previously, we have studied the effect of the gold-compound auranofin (AF) on both thioredoxin-glutathione reductasa (TGR) activity and viability of Taenia crassiceps cysticerci. It was demonstrated that micromolar concentrations of AF were high enough to fully inhibit TGR and kill the parasites. In this work, the dynamics of changes in the glutathione pool of T. crassiceps cysticerci following the addition of AF, was analyzed. A dose-dependent decrease in the internal glutathione concentration, concomitant with an increase in ROS production was observed. These changes were simultaneous with the formation of glutathione-protein complexes and the export of glutathione disulfide (GSSG) to the culture medium. Incubation of cysticerci in the presence of both AF and N-acetyl cysteine (NAC) prevents all the above changes, maintaining cysticerci viability. By contrast, the presence of both AF and buthionine sulfoximine (BSO) resulted in a potentiation of the effects of the gold compound, jeopardizing cysticerci viability. These results suggest the lethal effect of AF on T. crassiceps cysticerci, observed at micromolar concentrations, can be explained as a consequence of major changes in the glutathione status, which results in a significant increase in the oxidative stress of the parasites.


Subject(s)
Auranofin/toxicity , Glutathione/analysis , Oxidants/toxicity , Oxidative Stress , Taenia/chemistry , Taenia/drug effects , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Buthionine Sulfoximine/metabolism , Cysticercus/chemistry , Cysticercus/drug effects , Cysticercus/physiology , Reactive Oxygen Species/analysis , Survival Analysis , Taenia/physiology
6.
J Inorg Biochem ; 142: 28-38, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25306263

ABSTRACT

Gold(I) complexes are an important tool in the arsenal of established approaches for treating rheumatoid arthritis (RA), while some recent studies have suggested that gold nanoparticles (Au NPs) may also be therapeutically efficacious. These observations prompted the current biological studies involving gold(I) anti-RA agents and Au NPs, which are aimed towards improving our knowledge of how they work. The cytotoxicity of auranofin, aurothiomalate, aurothiosulfate and Au NPs towards RAW264.7 macrophages was evaluated using the MTT assay, with the former compound proving to be the most toxic. The extent of cellular uptake of the various gold agents was determined using graphite furnace atomic absorption spectrometry, while their distribution within macrophages was examined using microprobe synchrotron radiation X-ray fluorescence spectroscopy. The latter technique showed accumulation of gold in discrete regions of the cell, and co-localisation with sulfur in the case of cells treated with aurothiomalate or auranofin. Electrospray ionization mass spectrometry was used to characterize thioredoxin reductase (TrxR) in which the penultimate selenocysteine residue was replaced by cysteine. Mass spectra of solutions of TrxR and aurothiomalate, aurothiosulfate or auranofin showed complexes containing bare gold atoms bound to the protein, or protein adducts containing gold atoms retaining some of their initial ligands. These results support TrxR being an important target of gold(I) drugs used to treat RA, while the finding that Au NPs are incorporated into macrophages, but elicit little toxicity, indicates further exploration of their potential for treatment of RA is warranted.


Subject(s)
Gold , Macrophages/drug effects , Metal Nanoparticles/toxicity , Thioredoxin-Disulfide Reductase/metabolism , Auranofin/metabolism , Auranofin/toxicity , Gold/analysis , Gold Sodium Thiomalate/metabolism , Gold Sodium Thiomalate/toxicity , Macrophages/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Spectrometry, X-Ray Emission/methods , Spectrophotometry, Atomic/methods , Thioredoxin-Disulfide Reductase/chemistry
7.
Chem Commun (Camb) ; 50(72): 10343-54, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-24931774

ABSTRACT

The fascinating biological activity of gold coordination compounds has led to the development of a wide range of complexes. The precise biological action of such species is often poorly understood and the ability to map gold distribution in cellular environments is key. This article discusses the recent progress in luminescent Au(I) complexes whilst considering their utility in bioimaging and therapeutics.


Subject(s)
Coordination Complexes/chemistry , Gold/chemistry , Animals , Auranofin/chemistry , Auranofin/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/toxicity , Crystallography, X-Ray , Heterocyclic Compounds/chemistry , Humans , Mice , Microscopy, Confocal , Molecular Conformation , Spectrometry, Fluorescence
8.
Metallomics ; 6(2): 233-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24287583

ABSTRACT

The reaction of Auoxo6, a dinuclear gold(III) complex, with the model protein bovine pancreatic ribonuclease is explored here by X-ray diffraction and ESI mass spectrometry. Data provide clues on the processes of adduct formation and of enzyme inhibition and, inductively, on the likely mode of action of this metallodrug.


Subject(s)
Gold/metabolism , Gold/toxicity , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/metabolism , Animals , Auranofin/chemistry , Auranofin/toxicity , Cattle , Cell Death/drug effects , Crystallography, X-Ray , Gold/chemistry , Hydrolysis/drug effects , Models, Molecular , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Spectrometry, Mass, Electrospray Ionization
9.
BMB Rep ; 46(1): 59-64, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23351386

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) and telomerase are considered attractive targets for anticancer therapy. The in vitro anticancer activity of the gold(I) compound auranofin was investigated using MDA-MB 231 human breast cancer cells, in which STAT3 is constitutively active. In cell culture, auranofin inhibited growth in a dose-dependent manner, and N-acetyl-L-cysteine (NAC), a scavenger of reactive oxygen species (ROS), markedly blocked the effect of auranofin. Incorporation of 5-bromo-2'-deoxyuridine into DNA and anchorage-independent cell growth on soft agar were decreased by auranofin treatment. STAT3 phosphorylation and telomerase activity were also attenuated in cells exposed to auranofin, but NAC pretreatment restored STAT3 phosphorylation and telomerase activity in these cells. These findings indicate that auranofin exerts in vitro antitumor effects in MDA-MB 231 cells and its activity involves inhibition of STAT3 and telomerase. Thus, auranofin shows potential as a novel anticancer drug that targets STAT3 and telomerase.


Subject(s)
Antineoplastic Agents/toxicity , Auranofin/toxicity , Cell Proliferation/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , Telomerase/antagonists & inhibitors , Acetylcysteine/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Female , Gold/chemistry , Humans , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Telomerase/metabolism
10.
Free Radic Biol Med ; 43(12): 1574-83, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18037123

ABSTRACT

The present study investigated the impact of ifosfamide (IFO) on renal thioredoxin reductase (TrxR) activity. In mice treated with IFO for 6 h, TrxR activity significantly decreased in a dose-dependent manner. Subsequently, acute renal failure (ARF) occurred dose-dependently. Like IFO, the well-established TrxR-specific inhibitor auranofin suppresfssed renal TrxR activity and generated ARF too. TrxR was inactivated by IFO preferentially over other antioxidant parameters at 6 h; however, it recovered nearly to normal levels within 12 h. When auranofin was administered at 6 h after IFO treatment, the recovery at 12 h was sharply attenuated. Consequently, ARF was pronouncedly exacerbated. IFO within its maximum tolerated dose did not considerably deplete renal glutathione. However, escalating IFO dose strikingly attacked both the thioredoxin and the glutathione systems, resulting in lethality, which implies that glutathione depletion sensitizes IFO-induced nephrotoxicity and cosuppression of both systems causes more severe toxicological consequences than suppressing the thioredoxin system alone. Indeed, combining IFO with buthionine sulfoximine, an inhibitor of glutathione synthesis, induced much more severe ARF than IFO alone did. Taken together, inhibition of renal TrxR activity can be considered as a pivotal mechanism of IFO-induced ARF, and individuals with lower levels of renal glutathione are at high risk of incurring ARF after IFO treatment.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/enzymology , Ifosfamide/toxicity , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/toxicity , Antioxidants/metabolism , Apoptosis/drug effects , Auranofin/administration & dosage , Auranofin/toxicity , Buthionine Sulfoximine/administration & dosage , Buthionine Sulfoximine/toxicity , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Free Radicals/metabolism , Glutathione/metabolism , Ifosfamide/administration & dosage , Kidney/drug effects , Kidney/enzymology , Kidney/pathology , Male , Mice , Oxidation-Reduction
11.
Toxicol In Vitro ; 20(6): 882-90, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16510263

ABSTRACT

UNLABELLED: Thioredoxin reductase (TrxR) reduces thioredoxin (Trx), thereby contributing to cellular redox balance, facilitating the synthesis of deoxy-ribose sugars for DNA synthesis, and regulating redox-sensitive gene expression. Auranofin is a gold compound that potently inhibits TrxR. This inhibition is one suspected mechanism of auranofin's therapeutic benefit in the treatment of rheumatoid arthritis. The use of other gold compounds to treat cancer or inflammatory disease may rely on their ability to inhibit TrxR. In the current study, we tested the hypothesis that a variety of gold compounds may inhibit TrxR. METHODS: We exposed rat-TrxR1 to auranofin, gold sodium thiomalate, sodium aurothiosulfate, triphenyl phosphine gold chloride, or gold acetate, and measured TrxR activity ex vivo. We then compared TrxR1 inhibitory levels of gold compounds to those that inhibited mitochondrial activity of THP1 monocytes and OSC2 epithelial cells, estimated by succinate dehydrogenase activity. RESULTS: All gold compounds inhibited TrxR1 at concentrations ranging from 5 to 4000 nM (50% inhibitory concentration). The oxidation state of gold did not correlate with inhibitory potency, but ligand configuration was important. Au(I)-phosphine compounds (triphenyl phosphine gold chloride and auranofin) were the most potent inhibitors of TrxR. All TrxR1 inhibitory concentrations were sublethal to mitochondrial activity in both THP1 and OSC2 cells. CONCLUSIONS: Diverse types of gold compounds may be effective inhibitors of TrxR1 at concentrations that do not suppress cellular mitochondrial function. Inhibition may be optimized to some degree by altering the ligand configuration of the compounds. These results support future study of a variety of Au compounds for therapeutic development as inhibitors of TrxR1.


Subject(s)
Cytosol/enzymology , Enzyme Inhibitors/toxicity , Gold/toxicity , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Auranofin/toxicity , Cell Line , Dithionitrobenzoic Acid/metabolism , Gold Compounds/toxicity , Gold Sodium Thiomalate/toxicity , Humans , NADP/metabolism , Rats , Thioredoxin Reductase 1
12.
Toxicol In Vitro ; 20(5): 625-33, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16377126

ABSTRACT

UNLABELLED: The toxicity of anti-rheumatic gold compounds has limited their use and development, yet both the toxicological and therapeutic actions of these compounds remain unclear. In the current study, we tested the hypothesis that intracellular reactive oxygen species (ROS) induced by Au(I) or Au(III) compounds mediate their ability to suppress mitochondrial activity. METHODS: Human THP1 monocytes were exposed to HAuCl(4) x 3H(2)O (Au(III)), or the anti-rheumatic compounds auranofin (AF) or gold sodium thiomalate (GSTM) for 6-72 h, after which mitochondrial activity (succinate dehydrogenase) was measured. To assess the role of cellular redox status as a mediator of mitochondrial suppression, monocytes were pre-treated with a pro-oxidant (t-butyl hydroquinone, t-BHQ) or antioxidant (N-acetyl cysteine, NAC ). ROS levels were measured 0-24h post-gold addition to determine their role as mediators of mitochondrial activity suppression. RESULTS: AF was the most potent inhibitor of mitochondrial activity, followed by Au(III) and GSTM. Only Au(III) induced intracellular ROS; no ROS formation was observed in response to AF or GSTM exposure. Although anti- and pro-oxidants had some effects on mitochondrial suppression of Au compounds, collectively the data do not support redox effects or ROS formation as major mediators of Au-compound mitochondrial suppression. CONCLUSIONS: Our results do not indicate that ROS and redox effects play major roles in mediating the cytotoxicity of AF, GSTM or Au(III).


Subject(s)
Gold Compounds/toxicity , Mitochondria/drug effects , Monocytes/drug effects , Reactive Oxygen Species/metabolism , Auranofin/toxicity , Cells, Cultured , Gold Sodium Thiomalate/toxicity , Humans , Mitochondria/metabolism , Monocytes/metabolism
13.
Toxicol In Vitro ; 19(3): 365-71, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15713543

ABSTRACT

The objective of this study was to quantify the ability of sublethal concentrations of several gold compounds to differentially modulate the monocytic secretion of key cytokines that are important in the etiology of rheumatic diseases. Human THP1 monocytic cells were exposed to the anti-rheumatic drugs auranofin (AF), gold sodium thiomalate (GSTM) or HAuCl4 (Au(III)) for 24-72 h. Succinate dehydrogenase (SDH) activity of the monocytes was used to determine sublethal concentrations. Monocytes were then exposed to sublethal concentrations of gold compounds for 72 h, and the activator lipopolysaccharide (LPS) was added (or not) to cultures for the last 6h. The secretion of IL6, IL8, IL10, and TNFalpha were measured in cell supernatants using ELISA. Cytokine secretion was compared among concentrations and gold compounds. SDH experiments established a sublethal concentration range of 0-75 microM for GSTM and Au(III) and 0-0.5 microM for AF. In cytokine experiments, none of the compounds alone activated secretion of any of the cytokines, but all differentially (50-440%, p<0.05) increased LPS-induced secretion of IL6 and IL8 over TNFalpha and IL10. In conclusion, sublethal concentrations of AF, GSTM, and Au(III) all may differentially modulate activation of monocytic cells, and this differential modulation may be important in the mechanisms of action of these compounds.


Subject(s)
Antirheumatic Agents/toxicity , Auranofin/toxicity , Chlorides/toxicity , Cytokines/metabolism , Gold Compounds/toxicity , Gold Sodium Thiomalate/toxicity , Monocytes/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , Monocytes/metabolism , Succinate Dehydrogenase/metabolism
14.
Clin Exp Pharmacol Physiol ; 31(5-6): 289-94, 2004.
Article in English | MEDLINE | ID: mdl-15191400

ABSTRACT

Auranofin, an antirheumatic gold compound, is an inhibitor of selenocysteine enzymes, such as thioredoxin reductase and glutathione peroxidase. These enzymes play an important role in protecting cardiac tissue from oxidative stress generated during ischaemia-reperfusion. Auranofin (100 mg/kg) was administered to rats and their hearts were subjected to an in vitro model of ischaemia-reperfusion. The activity of thioredoxin reductase and glutathione peroxidase was determined in liver and heart tissues in an attempt to correlate enzymatic activity with heart recovery after ischaemia-reperfusion. There was significantly less thioredoxin reductase activity in rat liver extracts, whereas the level of glutathione activity remained unchanged, demonstrating that the dose of auranofin used was able to selectively inhibit one of these enzyme systems. Rats administered auranofin displayed significantly impaired recovery from ischaemic insult. The end diastolic pressure was increased, whereas the rate pressure product was significantly decreased. The level of postischaemic apoptosis was also assessed by examining caspase-3 activity in tissue homogenates. Auranofin significantly increased the degree of postischaemic apoptosis, leading to poor postischaemic recovery.


Subject(s)
Apoptosis/drug effects , Auranofin/pharmacology , Heart/drug effects , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/chemically induced , Animals , Apoptosis/physiology , Auranofin/toxicity , Heart/physiology , In Vitro Techniques , Male , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/physiopathology , Rats , Rats, Wistar
15.
Arthritis Rheum ; 33(10): 1449-61, 1990 Oct.
Article in English | MEDLINE | ID: mdl-1977391

ABSTRACT

We performed 2 metaanalyses of placebo-controlled and comparative clinical trials to examine the relative efficacy and toxicity of methotrexate (MTX), injectable gold, D-penicillamine (DP), sulfasalazine (SSZ), auranofin (AUR), and antimalarial drugs, the second-line drugs most commonly used to treat rheumatoid arthritis (RA). For the efficacy study, we applied a set of inclusion criteria and focused on trials which provided information on tender joint count, erythrocyte sedimentation rate, or grip strength. We found 66 clinical trials that contained 117 treatment groups of interest, and for each drug, we combined the treatment groups. For each outcome, results showed that AUR tended to be weaker than other second-line drugs. The results of the 3 outcome measures were synthesized into a composite measure of outcomes, and AUR was significantly weaker than MTX (P = 0.006), injectable gold (P less than 0.0001), DP (P less than 0.0001), and SSZ (P = 0.009) and was slightly, but not significantly, weaker than antimalarial agents (P = 0.11). We also found heterogeneity among antimalarial agents, in that patients treated with chloroquine did better than those treated with hydroxychloroquine. We found little difference in efficacy between MTX, injectable gold, DP, and SSZ. A power analysis showed that a trial should contain at least 170 patients per treatment group to successfully differentiate between more effective and less effective (e.g., AUR) second-line drugs. None of the reported interdrug comparative trials we reviewed were this large. For the toxicity study, our inclusion criteria captured RA trials which reported the proportion of patients who discontinued therapy because of drug toxicity and the total proportion who dropped out. We found 71 clinical trials that contained 129 treatment groups. The average proportion who dropped out and the average proportion who dropped out because of drug toxicity were computed for each drug. Overall, 30.2% of the patients in these trials dropped out; 50% of them did so because of drug toxicity. Injectable gold had higher toxicity rates (P less than 0.05) and higher total dropout rates (P less than 0.01) than any other drug; 30% of gold-treated patients dropped out because of side effects versus 15% of all trial patients. Antimalarial drugs and AUR had relatively low rates of toxicity; the rate for MTX was imprecise because of discrepancies between trials. Thus, of the commonly used second-line drugs, AUR is the weakest, and injectable gold is the most toxic. Agents introduced in the future will be compared with these drugs.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Antimalarials/standards , Arthritis, Rheumatoid/drug therapy , Auranofin/standards , Antimalarials/toxicity , Auranofin/toxicity , Gold/standards , Gold/toxicity , Humans , Meta-Analysis as Topic , Methotrexate/standards , Methotrexate/toxicity , Penicillamine/standards , Penicillamine/toxicity , Sulfasalazine/standards , Sulfasalazine/toxicity
16.
J Rheumatol ; 16(6): 749-56, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2778756

ABSTRACT

Auranofin (AF) in concentrations between 3 x 10(-7) and 3 x 10(-6) mol/l stimulated bone resorption in cultured neonatal mouse calvariae significantly with 1 x 10(-6) mol/l being most potent. Complete inhibition by 5 x 10(-7) mol/l indomethacin and increased medium concentrations of prostaglandin (PG) E2 and 6-keto-PGF1 alpha after 72 h indicate a PG mediated mechanism. Morphology revealed active osteoclasts. Cytotoxic effects were observed with 3 x 10(-6) and 1 x 10(-5) mol/l AF with osteocytes and osteoblasts being considerably more sensitive than osteoclasts. The latter concentrations inhibited bone resorption stimulated by parathyroid hormone (PTH) 1,25-dihydroxyvitamin D3, PGE2, thrombin and interleukin 1. The stimulatory effect of AF on PG production and subsequent bone resorption could limit its therapeutic usefulness.


Subject(s)
Auranofin/pharmacology , Bone Resorption/drug effects , Bone and Bones/drug effects , Prostaglandins/biosynthesis , Animals , Auranofin/toxicity , Bone and Bones/ultrastructure , Cells, Cultured , Mice , Skull
17.
Fundam Appl Toxicol ; 11(2): 277-84, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3220206

ABSTRACT

Auranofin, a gold-containing compound, was administered to Charles River CD-1 mice for 18 months to assess its possible carcinogenicity. The mice were dosed orally with 1.0, 3.0, or 6.0 (increased to 9.0 on Day 294) mg/kg/day. Each dose group and each of two control groups contained 110 males and 110 females. Survival was greater than 70% at the end of the study. No effect of the treatment on neoplastic or nonneoplastic lesions was found. This is in contrast to the results reported in rats. Auranofin in rats produced a heavy metal nephropathy characterized by acute coagulative necrosis, subacute renal cortical fibrosis, chronic cytomegaly and karyomegaly, and finally renal cortical neoplasia (adenomas and adenocarcinomas). The lack of effect of auranofin on tumor incidence in mice suggests the findings in rats may be species specific.


Subject(s)
Auranofin/toxicity , Carcinogens , Administration, Oral , Animals , Auranofin/administration & dosage , Body Weight/drug effects , Carcinogenicity Tests , Eating/drug effects , Female , Kidney Cortex/anatomy & histology , Kidney Cortex/drug effects , Male , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Sex Factors , Time Factors
18.
J Pharmacol Exp Ther ; 241(2): 540-6, 1987 May.
Article in English | MEDLINE | ID: mdl-3106619

ABSTRACT

Two gold compounds, gold sodium thiomalate (AuTM) and auranofin (AF) are presently in clinical use in therapy of rheumatoid arthritis. The effects of varying doses of AF administered to rats by either the p.o. or the i.p. route on heme metabolism were determined. Twenty four hours after a single dose of AF, decreases in the sulfhydryl-containing enzymes, delta-aminolevulinic acid dehydratase and ferrochelatase activities were observed in the liver and kidneys. These decreases in heme biosynthetic enzymes were accompanied by decreases in cytochrome P-450-dependent enzymic activities and increases in microsomal heme oxygenase activity. These changes were observed with AF dosages as low as 5 mg/kg, with maximal changes occurring at a p.o. dose of about 15 mg of AF per kg and an i.p. dose of 5 to 10 mg of AF per kg. Dose-response studies with AuTM showed that maximal changes in heme metabolism occur at a lower dose of AF than of AuTM, even though AF was administered p.o. and AuTM was administered parenterally. In addition, the kidneys appeared to be more susceptible to the inhibitory effects of the two chrysotherapeutic agents than did the liver. The present studies demonstrate the p.o. drug AF affects heme metabolism in a manner similar to that reported previously with the parenterally administered AuTM.


Subject(s)
Auranofin/toxicity , Gold Sodium Thiomalate/toxicity , Heme/metabolism , Kidney/metabolism , Liver/metabolism , Oxygenases/metabolism , Animals , Benzopyrene Hydroxylase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Ethylmorphine-N-Demethylase/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Kidney/drug effects , Liver/drug effects , Male , Porphobilinogen Synthase/metabolism , Rats , Rats, Inbred Strains
19.
Mol Pharmacol ; 31(1): 21-6, 1987 Jan.
Article in English | MEDLINE | ID: mdl-3807890

ABSTRACT

Metallothioneins (MTs) are low molecular weight, thiol-rich, metal-binding proteins. Auranofin (AF) is a gold compound active in the treatment of rheumatoid arthritis. The effects of AF on regulation of MT gene expression in Chinese hamster ovary cells were studied. AF-resistant cells accumulated substantial amounts of MT mRNA and protein, whereas no induction was observed in AF-sensitive cells. Cells capable of inducing MT in the presence of AF were much less sensitive to AF-mediated cytotoxicity. Induction of MT by low concentrations of Cd protected cells from subsequently administered doses of AF. The level of protection correlated with the level of induced MT. These findings indicate that MT plays a central role in the mechanisms underlying cellular resistance to gold compounds.


Subject(s)
Auranofin/pharmacology , Metallothionein/biosynthesis , Animals , Auranofin/antagonists & inhibitors , Auranofin/toxicity , Cadmium/pharmacology , Cadmium/toxicity , Cell Survival/drug effects , Cricetinae , Drug Resistance , Gene Expression Regulation/drug effects , Metallothionein/genetics , RNA, Messenger/genetics
20.
Toxicol Pathol ; 15(3): 283-93, 1987.
Article in English | MEDLINE | ID: mdl-3317768

ABSTRACT

Investigating the immunotoxic potential of candidate drugs as part of a preclinical safety evaluation poses several problems. These include the need for practical, validated tests, the difficulty in establishing the toxicologic significance of positive findings, and a poor understanding of the predictive value such findings hold for drug effects in man. A key component of this investigation is the toxicologic profile generated through preclinical toxicity and safety studies. As this "routine" assessment becomes increasingly comprehensive and sophisticated, most toxicologically significant drug-associated effects are revealed. Such findings may serve as "triggers" for investigating possible immune mechanisms. Decisions to test specifically for immunotoxicity may also be influenced by the molecular structure and pharmacologic profile of the compound, as well as the intended use of the drug. Examples of such indications and follow-up studies are discussed in this review. We are presently poorly equipped to effectively screen drugs indiscriminately for an immunotoxic potential. We are better prepared, however, to investigate whether a drug-associated change is due to an adverse effect on the immune system. This problem-oriented approach to immunotoxicology challenges us as diagnosticians and immunopathologists, and requires a close working relationship among the toxicologic pathologist, the basic immunologist, the immunopharmacologist, and the clinician.


Subject(s)
Immune System/drug effects , Toxicology , Animals , Auranofin/toxicity , Cefamandole/analogs & derivatives , Cefamandole/toxicity , Cefonicid , Humans , Immune System/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...