Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 130
1.
Biol Pharm Bull ; 47(1): 104-111, 2024.
Article En | MEDLINE | ID: mdl-38171771

White matter lesions induced by chronic cerebral hypoperfusion can cause vascular dementia; however, no appropriate treatments are currently available for these diseases. In this study, we investigated lipid peroxidation, which has recently been pointed out to be associated with cerebrovascular disease and vascular dementia, as a therapeutic target for chronic cerebral hypoperfusion. We used ethoxyquin, a lipid-soluble antioxidant, in a neuronal cell line and mouse model of the disease. The cytoprotective effect of ethoxyquin on glutamate-stimulated HT-22 cells, a mouse hippocampal cell line, was comparable to that of a ferroptosis inhibitor. In addition, the administration of ethoxyquin to bilateral common carotid artery stenosis model mice suppressed white matter lesions, blood-brain barrier disruption, and glial cell activation. Taken together, we propose that the inhibition of lipid peroxidation may be a useful therapeutic approach for chronic cerebrovascular disease and the resulting white matter lesions.


Brain Ischemia , Carotid Stenosis , Cerebrovascular Disorders , Dementia, Vascular , White Matter , Animals , Mice , Dementia, Vascular/complications , Ethoxyquin/metabolism , Ethoxyquin/pharmacology , Ethoxyquin/therapeutic use , White Matter/metabolism , White Matter/pathology , Brain Ischemia/pathology , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/complications , Cerebrovascular Disorders/metabolism , Disease Models, Animal , Carotid Stenosis/complications , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Mice, Inbred C57BL
2.
J Cardiovasc Pharmacol ; 80(5): 690-699, 2022 11 01.
Article En | MEDLINE | ID: mdl-35881422

ABSTRACT: Doxorubicin (DOX) is an effective anti-cancer agent for various malignancies. Nevertheless, it has a side effect of cardiotoxicity, referred to as doxorubicin-induced cardiomyopathy (DIC), that is associated with a poorer prognosis. This cardiotoxicity limits the clinical use of DOX as a therapeutic agent for malignancies. Recently, ferroptosis, a form of regulated cell death induced by the accumulation of lipid peroxides, has been recognized as a major pathophysiology of DIC. Ethoxyquin is a lipophilic antioxidant widely used for food preservation and thus may be a potential therapeutic drug for preventing DIC. However, the efficacy of ethoxyquin against ferroptosis and DIC remains to be fully elucidated. Here, we investigated the inhibitory action of ethoxyquin against GPx4-deficient ferroptosis and its therapeutic efficacy against DOX-induced cell death in cultured cardiomyocytes and cardiotoxicity in a murine model of DIC. In cultured cardiomyocytes, ethoxyquin treatment effectively prevented GPx4-deficient ferroptosis. Ethoxyquin also prevented DOX-induced cell death, accompanied by the suppression of malondialdehyde (MDA) and mitochondrial lipid peroxides, which were induced by DOX. Furthermore, ethoxyquin significantly prevented DOX-induced cell death without any suppression of caspase cleavages representing apoptosis. In DIC mice, ethoxyquin treatment ameliorated cardiac impairments, such as contractile dysfunction and myocardial atrophy, and lung congestion. Ethoxyquin also suppressed serum lactate dehydrogenase and creatine kinase activities, decreased the levels of lipid peroxides such as MDA and acrolein, inhibited cardiac fibrosis, and reduced TUNEL-positive cells in the hearts of DIC mice. Collectively, ethoxyquin is a competent antioxidant for preventing ferroptosis in DIC and can be its prospective therapeutic drug.


Cardiomyopathies , Ferroptosis , Mice , Animals , Cardiotoxicity/prevention & control , Antioxidants/therapeutic use , Ethoxyquin/metabolism , Ethoxyquin/pharmacology , Ethoxyquin/therapeutic use , Lipid Peroxides/metabolism , Lipid Peroxides/pharmacology , Oxidative Stress , Doxorubicin/toxicity , Myocytes, Cardiac , Apoptosis , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Cardiomyopathies/metabolism
3.
Sci Rep ; 11(1): 10749, 2021 05 24.
Article En | MEDLINE | ID: mdl-34031437

Ethoxyquin (EQ), a quinolone-based antioxidant, has demonstrated neuroprotective properties against several neurotoxic drugs in a phenotypic screening and is shown to protect axons in animal models of chemotherapy-induced peripheral neuropathy. We assessed the effects of EQ on peripheral nerve function in the db/db mouse model of type II diabetes. After a 7 week treatment period, 12-week-old db/db-vehicle, db/+ -vehicle and db/db-EQ treated animals were evaluated by nerve conduction, paw withdrawal against a hotplate, and fiber density in hindlimb footpads. We found that the EQ group had shorter paw withdrawal latency compared to vehicle db/db group. The EQ group scored higher in nerve conduction studies, compared to vehicle-treated db/db group. Morphology studies yielded similar results. To investigate the potential role of mitochondrial DNA (mtDNA) deletions in the observed effects of EQ, we measured total mtDNA deletion burden in the distal sciatic nerve. We observed an increase in total mtDNA deletion burden in vehicle-treated db/db mice compared to db/+ mice that was partially prevented in db/db-EQ treated animals. These results suggest that EQ treatment may exert a neuroprotective effect in diabetic neuropathy. The prevention of diabetes-induced mtDNA deletions may be a potential mechanism of the neuroprotective effects of EQ in diabetic neuropathy.


Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/prevention & control , Ethoxyquin/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Neuropathies/etiology , Diabetic Neuropathies/genetics , Disease Models, Animal , Ethoxyquin/pharmacology , Mice , Mutation , Neural Conduction/drug effects , Neuroprotective Agents/pharmacology , Sciatic Nerve/chemistry , Sciatic Nerve/drug effects
4.
Arch Razi Inst ; 76(6): 1765-1776, 2021 12.
Article En | MEDLINE | ID: mdl-35546987

The current experiment aimed to assess the effect of the synthetic antioxidants ethoxyquin (EQ) and/or butylated hydroxytoluene (BHT) on the liver function tests, hematological parameters, and liver histoarchitecture in rats. A total of 50 male Sprague-Dawley rats were divided into five groups of 10 animals per group. The first group served as the control and did not receive any treatments, and the second group served as the vehicle control and was orally administrated 1 ml of corn oil day after day for consecutive 45 and 90 days. The third group (EQ) was orally administered 1 ml of EQ dissolved in corn oil day after day for consecutive 45 and 90 days in a dose of 1/5 LD50, and the fourth group (BHT) was orally received 1 ml of BHT dissolved in corn oil day after day for consecutive 45 and 90 days in a dose of 1/5 LD50. The fifth group (combination group) was orally administered both EQ and BHT at the same doses and durations described above. The present results showed that the final body weight was significantly decreased in the EQ- or BHT-treated group particularly at 90 days of exposure to both compounds. Furthermore, the liver weight was significantly elevated in EQ, BHT, and co-exposed groups at 45 and 90 days of exposure, compared to the control group. Moreover, EQ, BHT, and their co-exposure caused a significant elevation in the levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzymes, as well as total bilirubin at 45 and 90 days of exposure. On the other hand, there was no significant change in the total albumin. Hemoglobin value, red blood cells, white blood cells, platelets, and differential leucocyte count at 45 and 90 days of exposure were significantly decreased. Histopathological significant findings in the liver were observed as vascular congestions, vacuolations, hydropic degenerations, lipidosis, and swelling, particularly in the co-exposed group for 90 days. These findings confirmed the hepatotoxic potential of EQ and BHT; therefore, it is recommended to control and limit the utilization of such chemicals.


Butylated Hydroxytoluene , Ethoxyquin , Animals , Butylated Hydroxytoluene/toxicity , Corn Oil/pharmacology , Ethoxyquin/pharmacology , Liver , Male , Rats , Rats, Sprague-Dawley , Toluene/pharmacology
5.
PLoS One ; 14(1): e0211128, 2019.
Article En | MEDLINE | ID: mdl-30682099

Ethoxyquin (EQ; 6-Ethoxy-2,2,4-trimethyl-1,2-dihydroquinoline) has been used as an antioxidant in feed components for pets, livestock and aquaculture. However, possible risks of EQ used in aquafeed for fish health have not yet been characterized. The present study investigated the toxicity and dose-response of subchronic dietary EQ exposure at doses ranging from 41 to 9666 mg EQ/kg feed in Atlantic salmon (Salmo salar L.). Feed at concentrations higher than 1173 mg EQ/kg were rejected by the fish, resulting in reduced feed intake and growth performance. No mortality was observed in fish exposed to any of the doses. A multi-omic screening of metabolome and proteome in salmon liver indicated an effect of dietary EQ on bioenergetics pathways and hepatic redox homeostasis in fish fed concentrations above 119 mg EQ/kg feed. Increased energy expenditure associated with an upregulation of hepatic fatty acid ß-oxidation and induction and carbohydrate catabolic pathways resulted in a dose-dependent depletion of intracytoplasmic lipid vacuoles in liver histological sections, decreasing whole body lipid levels and altered purine/pyrimidine metabolism. Increased GSH and TBARS in the liver indicated a state of oxidative stress, which was associated with activation of the NRF2-mediated oxidative stress response and glutathione-mediated detoxification processes. However, no oxidative DNA damage was observed. As manifestation of altered energy metabolism, the depletion of liver intracytoplasmic lipid vacuoles was considered the critical endpoint for benchmark dose assessment, and a BMDL10 of 243 mg EQ/kg feed was derived as a safe upper limit of EQ exposure in Atlantic salmon.


Eating/drug effects , Energy Metabolism/drug effects , Ethoxyquin/pharmacology , Lipid Metabolism/drug effects , Liver/metabolism , Salmo salar/metabolism , Animal Feed , Animals , DNA Damage , Dose-Response Relationship, Drug
6.
Fish Physiol Biochem ; 45(1): 43-61, 2019 Feb.
Article En | MEDLINE | ID: mdl-29980882

Firstly, a linoleic and linolenic acid emulsion and fish feeds were incubated with graded levels of ethoxyquin (EQ) and petroleum ether extract, ethyl acetate extract (EAE), ethanol extract and aqueous extract of Angelica sinensis. The results showed that EQ and extracts of Angelica sinensis (EAs) inhibited lipid oxidation in material above. Of all of the examined EAs, EAE showed the strongest protective effects against the lipid oxidation. Moreover, EAE at high concentrations showed a stronger inhibitory effect on lipid oxidation than that of EQ. Next, 7 experimental diets that respectively supplemented 0.0, 0.2, 0.8 and 3.2 g kg-1 of EQ and EAE were fed to 280 juvenile red carp (Cyprinus carpio var. xingguonensis) with seven treatment groups for 30 days. The results indicated that dietary EAE improved growth performance in carp. Moreover, dietary EAE increased the activities of trypsin, lipase, alpha-amylase, alkaline phosphatase, glutamate-oxaloacetate transaminase and glutamate-pyruvate transaminase (GPT) and decreased plasma ammonia content in carp. Meanwhile, dietary EAE reduced the levels of malondialdehyde and raised the activities of anti-superoxide anion, anti-hydroxyl radical, superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase and the content of reduced glutathione in the hepatopancreas and intestine of carp. However, with the exception of GPT, dietary EQ got the opposite results to dietary EAE in carp. These results revealed that dietary EAE improved the digestive, absorptive and antioxidant capacities in fish. However, dietary EQ inhibited the digestive, absorptive and antioxidant capacities in fish. So, EAE could be used as a natural antioxidant for replacing EQ in fish feeds.


Angelica sinensis/chemistry , Animal Feed/analysis , Carps/growth & development , Ethoxyquin/pharmacology , Lipid Peroxidation/drug effects , Plant Extracts/pharmacology , Animal Nutritional Physiological Phenomena , Animals , Antioxidants/metabolism , Diet/veterinary , Digestion/drug effects , Gene Expression Regulation/drug effects , Plant Extracts/chemistry , Random Allocation
7.
Sci Rep ; 6: 28861, 2016 06 28.
Article En | MEDLINE | ID: mdl-27350330

Ethoxyquin was recently identified as a neuroprotective compound against toxic neuropathies and efficacy was demonstrated against paclitaxel-induced neurotoxicity in vivo. In this study we examined the efficacy of ethoxyquin in preventing neurotoxicity of cisplatin in rodent models of chemotherapy-induced peripheral neuropathy and explored its mechanism of action. Ethoxyquin prevented neurotoxicity of cisplatin in vitro in a sensory neuronal cell line and primary rat dorsal root ganglion neurons. In vivo, chronic co-administration of ethoxyquin partially abrogated cisplatin-induced behavioral, electrophysiological and morphological abnormalities. Furthermore, ethoxyquin did not interfere with cisplatin's ability to induce tumor cell death in ovarian cancer cell line in vitro and in vivo. Finally, ethoxyquin reduced the levels of two client proteins (SF3B2 and ataxin-2) of a chaperone protein, heat shock protein 90 (Hsp90) when co-administered with cisplatin in vitro. These results implied that the neuroprotective effect of ethoxyquin is mediated through these two client proteins of Hsp90. In fact, reducing levels of SF3B2 in tissue-cultured neurons was effective against neurotoxicity of cisplatin. These findings suggest that ethoxyquin or other compounds that inhibit chaperone activity of Hsp90 and reduce levels of its client protein, SF3B2 may be developed as an adjuvant therapy to prevent neurotoxicity in cisplatin-based chemotherapy protocols.


Cisplatin/toxicity , Ethoxyquin/pharmacology , Neurons/drug effects , Neuroprotection/drug effects , Animals , Antineoplastic Agents/toxicity , Ataxin-2/metabolism , Axons/drug effects , Axons/physiology , Cell Line , Cells, Cultured , Female , Ganglia, Spinal/cytology , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice, Nude , Neurons/metabolism , Neuroprotective Agents/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/prevention & control , RNA Splicing Factors/metabolism , Rats , Xenograft Model Antitumor Assays
8.
Appl Environ Microbiol ; 82(2): 747-55, 2016 01 15.
Article En | MEDLINE | ID: mdl-26590271

Thiabendazole (TBZ), imazalil (IMZ), ortho-phenylphenol (OPP), diphenylamine (DPA), and ethoxyquin (EQ) are used in fruit-packaging plants (FPP) with the stipulation that wastewaters produced by their application would be depurated on site. However, no such treatment systems are currently in place, leading FPP to dispose of their effluents in agricultural land. We investigated the dissipation of those pesticides and their impact on soil microbes known to have a key role on ecosystem functioning. OPP and DPA showed limited persistence (50% dissipation time [DT50], 0.6 and 1.3 days) compared to TBZ and IMZ (DT50, 47.0 and 150.8 days). EQ was rapidly transformed to the short-lived quinone imine (QI) (major metabolite) and the more persistent 2,4-dimethyl-6-ethoxyquinoline (EQNL) (minor metabolite). EQ and OPP exerted significant inhibition of potential nitrification, with the effect of the former being more persistent. This was not reflected in the abundance (determined by quantitative PCR [qPCR]) of the amoA gene of ammonia-oxidizing bacteria (AOB) and archaea (AOA). Considering the above discrepancy and the metabolic pattern of EQ, we further investigated the hypothesis that its metabolites and not only EQ were toxic to ammonia oxidizers. Potential nitrification, amoA gene abundance, and amoA gene transcripts of AOB and AOA showed that QI was probably responsible for the inhibition of nitrification. Our findings have serious ecological and practical implications for soil productivity and N conservation in agriculturally impacted ecosystems and stress the need to include metabolites and RNA-based methods when the soil microbial toxicity of pesticides is assessed.


Ammonia/metabolism , Antioxidants/pharmacology , Bacteria/drug effects , Ethoxyquin/pharmacology , Food Preservatives/pharmacology , Soil Microbiology , Soil Pollutants/pharmacology , Wastewater/chemistry , Antioxidants/metabolism , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Ethoxyquin/metabolism , Food Packaging , Food Preservatives/metabolism , Fruit/chemistry , Industrial Waste/analysis , Oxidation-Reduction , Soil Pollutants/metabolism , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/pharmacology
9.
Ann Neurol ; 74(6): 893-904, 2013 Dec.
Article En | MEDLINE | ID: mdl-23955554

OBJECTIVE: Peripheral neurotoxicity is a major dose-limiting side effect of many chemotherapeutic drugs. Currently there are no effective disease-modifying therapies for chemotherapy-induced peripheral neuropathies, but these side effects of chemotherapy are potentially ideal targets for development of neuroprotective therapies, because candidate drugs can be co- or preadministered before the injury to peripheral axons takes place. METHODS: We used a phenotypic drug screening approach to identify ethoxyquin as a potential neuroprotective drug and carried out additional biochemical experiments to identify its mechanism of action. RESULTS: We validated the screening results with ethoxyquin and its derivatives and showed that they prevented paclitaxel-induced peripheral neuropathy without blocking paclitaxel's ability to kill tumor cells. Furthermore, we demonstrated that ethoxyquin acts by modulating the chaperone activity of heat shock protein 90 (Hsp90) and blocking the binding of 2 of its client proteins, ataxin-2 and Sf3b2. Ethoxyquin-induced reduction in levels of both of these proteins resulted in prevention of axonal degeneration caused by paclitaxel. INTERPRETATION: Ethoxyquin and its novel derivatives as well as other classes of small molecules that act as Hsp90 modulators may offer a new opportunity for development of drugs to prevent chemotherapy-induced axonal degeneration.


Drug Evaluation, Preclinical/methods , Ethoxyquin/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Axons/drug effects , Cell Line , Male , Mice , Nerve Degeneration/chemically induced , Neurons/drug effects , Paclitaxel/adverse effects , Paclitaxel/antagonists & inhibitors
10.
Poult Sci ; 90(4): 922-30, 2011 Apr.
Article En | MEDLINE | ID: mdl-21406381

The objective of the present study was to evaluate the effect of antioxidant inclusion and oil quality on broiler performance, meat quality, shelf life, and tissue oxidative status. Ross 308 male broilers were allotted to a randomized complete block design in a 2 × 2 factorial arrangement. Factors consisted of antioxidant (ethoxyquin and propyl gallate) inclusion at 2 levels (0 or 135 mg/kg) and oil quality (fresh soybean oil, control diet peroxide value <1 mEq/kg, or oxidized soybean oil, diet peroxide value 7 mEq/kg). Each treatment included 12 pen replicates comprising 24 birds for a total of 1,152 birds on trial allotted to 48 pens. On the final day of the study, 1 bird from each pen was killed by cervical dislocation and used for determination of tissue oxidative status. Another 5 broilers from each pen were processed at a commercial slaughtering facility. Immediately after processing, carcasses were transported to the University of Illinois Meat Science Laboratory (Urbana) for further analysis. With the exception of 2 responses (liver vitamin A and serum vitamin A), no interactions were found between antioxidant inclusion and oil quality. Body weight and weight gain were increased by dietary antioxidant inclusion (P < 0.001) and fresh oil (P < 0.001). Feed intake was increased in broilers fed the antioxidant (P = 0.047) and fresh oil (P = 0.062). Antioxidant inclusion had no effect on G:F (P = 0.18). Antioxidant supplementation had no effect on carcass weight (P = 0.202), dressing percentage (P = 0.906), breast yield (P = 0.708), or breast ultimate pH (P = 0.625) and had minimal effect on breast color. Antioxidant supplementation (P = 0.057) reduced breast thiobarbituric acid reactive substances after 7 d of display. Fresh oil decreased liver thiobarbituric acid reactive substances, whereas antioxidant inclusion increased serum and liver vitamin A and E concentration. The presence of an antioxidant in the feed protects lipids from further oxidizing, therefore increasing broiler performance and improving shelf life when using oxidized oil.


Antioxidants/pharmacology , Chickens/growth & development , Lipid Metabolism/drug effects , Meat/analysis , Soybean Oil/pharmacology , Animals , Body Weight/physiology , Chickens/metabolism , Ethoxyquin/pharmacology , Least-Squares Analysis , Male , Propyl Gallate/pharmacology , Random Allocation , Thiobarbituric Acid Reactive Substances/metabolism , Vitamin A/blood , Vitamin E/blood
11.
Chem Biol Interact ; 191(1-3): 185-91, 2011 May 30.
Article En | MEDLINE | ID: mdl-21277289

Aldo-keto reductase 1B10 (AKR1B10) is an aldose reductase-like oxidoreductase of human origin. The expression of AKR1B10 is highly induced in the cells of various cancers such as lung non-small-cell carcinoma and hepatocellular carcinoma. Since the enzyme exhibits broad substrate specificities toward various xenobiotics such as anti-tumor drugs or various endogenous compounds such as retinaldehyde, AKR1B10 may play an important role in tumor progression or drug resistance. However, very little is known about its gene regulation. In this study, we investigated the regulation of AKR1B10 expression. A -3282bp of the 5'-flanking fragment of AKR1B10 gene was isolated from A549 lung carcinoma cells. This region contains several putative regulatory motifs such as AP-1, NF-κB and antioxidant response element. In addition, a complex polymorphic microsatellite with repetitive sequences enriched with C and T was found. However, luciferase reporter assay revealed that the microsatellite polymorphism did not influence the basal promoter activity. We found that an antioxidant ethoxyquin induced the AKR1B10 expression based on RT-PCR analysis and luciferase reporter assay. Since ethoxyquin is known to activate the gene expression mediated through transcription factor Nrf2, the involvement of Nrf2 was examined. Forced expression of dominant-negative Nrf2 mutant suppressed the ethoxyquin-induced AKR1B10 expression, and co-introduction of Nrf2 expression plasmid into the cells significantly augmented the luciferase reporter activity. Deletion analysis revealed that Nrf2-regulating cis-element(s) lay within -539bp of the 5'-flanking region. These results suggest that Nrf2 is one of the major factors involved in the AKR1B10 gene regulation.


Aldehyde Reductase/genetics , Gene Expression Regulation, Enzymologic , NF-E2-Related Factor 2/metabolism , Aldo-Keto Reductases , Base Sequence , Cell Line, Tumor , Ethoxyquin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Microsatellite Repeats/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics
12.
Drug Metab Dispos ; 38(2): 341-6, 2010 Feb.
Article En | MEDLINE | ID: mdl-19920056

The aldo-keto reductase (AKR) phase I drug metabolism enzyme superfamily is implicated in detoxification or bioactivation of a wide variety of carbonyl-bearing compounds. In this study, we have used antibodies raised against purified recombinant rat AKR isoforms 1A3, 1B4, 1C9, 1D2, and 7A1 to characterize the expression profile of these superfamily members in the rat and define their localization by immunohistochemistry. Western blotting showed that AKR1A3, AKR1B4, and AKR1C9 are ubiquitously expressed, whereas AKR1D2 and AKR7A1 are present in liver, adrenal gland, and kidney, with the latter also present in testis, spleen, and stomach. Immunohistochemical analysis of the kidney demonstrated the localization of AKR1A3 in proximal convoluted tubules, AKR1B4 in the loop of Henle, and AKR1C9 in the pars recta S3 segment of proximal tubules. We also report localization of AKR1B4 in the adrenal gland (parenchymal cells of the zona reticularis) and testis (Sertoli cells and late spermatids), of AKR1D2 in the liver (hepatocyte nuclei), and of AKR7A1 in the pancreatic duct and bronchiolar epithelium. Previous studies have shown that expression of AKR7A1 is induced in response to dietary administration of the phenolic antioxidants butylated hydroxyanisole and ethoxyquin. Here we identify AKR1B13 and AKR1D2 as further inducible members of the rat AKR superfamily.


Antioxidants/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Oxidoreductases/genetics , Oxidoreductases/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Animals , Butylated Hydroxyanisole/pharmacology , Ethoxyquin/pharmacology , Female , Immunohistochemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/drug effects , Liver/metabolism , Male , Organ Specificity , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Wistar , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction
13.
Chem Biol Interact ; 182(1): 22-8, 2009 Nov 10.
Article En | MEDLINE | ID: mdl-19695238

Identifying agents that block tumor initiation is a goal of cancer prevention. The ability of a chemically varied group of agents to induce various drug metabolizing genes in livers of rats was examined. Sprague-Dawley rats were treated for 7 days with various agents in the diet or by gavage. The agents examined, which might be expected to respond via specific nuclear receptors (CAR, AhR) as well as antioxidant response elements (AREs), included Phase I/II inducers [5,6-benzoflavone (BF, 5000mg/kg diet), diallyl sulfide (DAS, 500mg/kg BW/day), ethoxyquin (EXO, 300mg/kg BW/day) and phenobarbital (PB, 500mg/kg diet)] or pure Phase II inducers [1,2-dithiol-3-thione (DTT, 500mg/kg diet), and cyclopentadithiolthione (CPDTT, 175mg/kg BW/day)]. Liver RNA expression was analyzed employing oligonucleotide microarrays. The agents yielded unique expression profiles. In genes with known AREs, the induction ratios (Levels Treated/Levels Controls) were: quinone oxidoreductase (BF, 8:1; DTT, 3.2:1; CPDTT, 3:1; DAS, 1.8:1; Exo, 1.7:1), glutatione transferase Pi (DTT, 36:1; CPDTT, 34:1; EXO, 8:1; DAS, 5:1; BF, 2.5:1), and aldehyde keto reductase 7A3 (AFAR) (DTT and CPDTT, 14:1; DAS, 6:1; EXO, 4:1; PB, 1.5:1). When the search included a wider variety of Phase II drug metabolizing enzymes, no clear pattern was observed. Agent induced gene expression and preventive activity in published carcinogen induced tumor models showed limited correlation; questioning whether measuring the induction of one or two genes (e.g., quinone reductase) is a surrogate for overall Phase II inducing (antioxidant) and potential anti-tumor activity.


Anticarcinogenic Agents/pharmacology , Antioxidants/physiology , Enzyme Induction/drug effects , Liver/enzymology , Allyl Compounds/pharmacology , Animals , Ethoxyquin/pharmacology , Female , Gene Expression Regulation/drug effects , Glutathione Transferase/biosynthesis , Glutathione Transferase/genetics , Liver/drug effects , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/genetics , Oligonucleotide Array Sequence Analysis , Phenobarbital/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Response Elements/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sulfhydryl Compounds/pharmacology , Sulfides/pharmacology , Thiones/pharmacology , Thiophenes/pharmacology , beta-Naphthoflavone/pharmacology
14.
Chem Biol Interact ; 162(3): 268-73, 2006 Sep 25.
Article En | MEDLINE | ID: mdl-16959229

Four newly synthesized salts of ethoxyquin (EQ: 1,2-dihydro-6-ethoxy-2,2,4-trimethylquinoline), an antioxidant used in animal feeds, were evaluated with the use of the comet assay performed on human lymphocytes: ethoxyquin ascorbate, ethoxyquin hexanoate, ethoxyquin salicylate and ethoxyquin salt of Trolox C (6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid). In the study the abilities of these compounds to cause DNA fragmentation and to protect against H2O2-induced DNA damage were analysed. The obtained results were compared with those noted earlier for EQ. After EQ salts treatments (1-25 microM) the genotoxic effects were observed, but the genotoxic potentials of the compounds studied were lower than that of EQ. On the other hand, EQ salts, similarly to EQ, effectively protected the cells from oxidative effect of H2O2. EQ hexanoate was the most effective and its antioxidant activity was even slightly higher than that of EQ. We suggest that it is worth further detailed studies to estimate its usefulness as a preservative.


Antioxidants/pharmacology , Comet Assay , Ethoxyquin/pharmacology , Ethoxyquin/toxicity , Mutagens/toxicity , DNA Damage/drug effects , DNA Damage/genetics , Female , Humans , Hydrogen Peroxide/pharmacology , Lymphocytes/drug effects , Salts/pharmacology
15.
Water Res ; 39(17): 4251-63, 2005 Oct.
Article En | MEDLINE | ID: mdl-16199076

The potential inhibitory effect of ethoxyquin, an antioxidant commonly used as a preservative in the food processing industry (e.g., for stabilizing dissolved air flotation residuals), was evaluated at concentrations up to 300 mg/L using a mixed, mesophilic (35 degrees C) methanogenic culture and dextrin, peptone and methanol as the carbon source. A batch assay conducted with a range of ethoxyquin concentrations did not result in any inhibition up to an ethoxyquin concentration of 75 mg/L, but severe inhibition of methanogenesis was observed at concentrations higher than 150 mg/L. Ethoxyquin addition to a batch reactor with the same mixed, methanogenic culture, at ethoxyquin concentrations gradually increasing over 100 days, resulted in a transient and a complete inhibition of methanogenesis at ethoxyquin concentrations of 150 and 300 mg/L, respectively. Acidogens were not significantly impacted, whereas aceticlastic and methanol degrading methylotrophic methanogens were impacted the most. Acclimation of the methanogenic culture to ethoxyquin was not observed over an incubation period of more than 100 days. Long-term (>100 days) incubation at sub-inhibitory ethoxyquin concentrations did not result in ethoxyquin biotransformation. Similarly, ethoxyquin biotransformation was not evident over an 8-day aeration period in a laboratory-scale activated sludge reactor operated under fully aerobic conditions. Ethoxyquin phase distribution tests conducted with the mixed, methanogenic culture at 1.61 g/L volatile solids concentration and nominal ethoxyquin concentrations equal to or higher than 300 mg/L resulted in solid phase/liquid phase ethoxyquin ratios equal to or higher than 1.0. The combined effect of ethoxyquin recalcitrance under anaerobic conditions along with its phase distribution, which favors biosolids, will result in ethoxyquin accumulation in anaerobic treatment systems used by the food processing industry. Such accumulation may pose concerns relative to inhibitory effects in these treatment systems and the disposal of ethoxyquin-bearing biosolids.


Antioxidants/chemistry , Antioxidants/pharmacology , Ethoxyquin/chemistry , Ethoxyquin/pharmacology , Methane/biosynthesis , Solubility , Spectrophotometry, Ultraviolet
16.
Acta Pol Pharm ; 62(2): 111-5, 2005.
Article En | MEDLINE | ID: mdl-16161351

In our study ethoxyquin (EQ) and its two complexes with flavonoids were obtained from ethoxyquin (1,2-dihydro-6-ethoxy-2,2,4-trimethylquinoline, EQ) and quercetin (EQ-Q, 1:1) or rutin (EQ-R, 1:1). Cytotoxicity of the tested compounds was studied using the trypan blue exclusion method and the properties of the studied compounds were also analyzed with the TUNEL method evaluating their ability to induce apoptosis. It was shown that EQ induced apoptosis in cultured human lymphocytes, especially at 0.25 and 0.5 mM concentrations. The same effects were also observed after the incubation of lymphocytes with EQ-Q and EQ-R, but the numbers of apoptotic cells observed were lower than for EQ.


Antioxidants/chemical synthesis , Ethoxyquin/chemical synthesis , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Ethoxyquin/metabolism , Ethoxyquin/pharmacology , Food Preservatives/chemical synthesis , Humans , In Situ Nick-End Labeling , Lymphocytes/cytology , Lymphocytes/drug effects , Preservatives, Pharmaceutical/chemical synthesis , Time Factors
17.
Chem Biol Interact ; 155(3): 140-7, 2005 Aug 15.
Article En | MEDLINE | ID: mdl-16045903

Reduction of naphthoquinones by DT-diaphorase is often described as a detoxification reaction. This is true for some naphthoquinone derivatives, such as alkyl and di-alkyl naphthoquinones, but the situation with other substances, such as 2-hydroxy-1,4-naphthoquinone, is more complex. In the present study, the effect of several substances that are known to increase tissue activities of DT-diaphorase on the toxicity of 2-amino-1,4-naphthoquinone has been investigated. Like 2-hydroxy-1,4-naphthoquinone, the 2-amino-derivative was found to cause both haemolytic anaemia and renal tubular necrosis in rats. Again like 2-hydroxy-1,4-naphthoquinone, the severity of the haemolysis induced by the 2-amino derivative was increased in animals pre-treated with inducers of DT-diaphorase, but the degree of nephrotoxicity was decreased. With these substances, therefore, DT-diaphorase both activates and detoxifies the quinone, depending on the target organ. It is not possible to generalize with regard to the effects of modulation of tissue levels of DT-diaphorase on naphthoquinone toxicity in vivo, since this may change not only the severity of the toxic effects, but also the target organ specificity. In evaluating the possible therapeutic applications of such compounds, the possibility of toxic effects upon the blood and kidney must be borne in mind. In man, renal damage by compounds such as 2-hydroxy- and 2-amino-1,4-naphthoquinone may be a particular problem, because of the low level of DT-diaphorase in human liver.


Hemolysis/drug effects , Kidney/drug effects , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Naphthoquinones/toxicity , Anemia, Hemolytic/chemically induced , Animals , Biotransformation/drug effects , Butylated Hydroxyanisole/pharmacology , Butylated Hydroxytoluene/pharmacology , Dimethyl Fumarate , Disulfiram/pharmacology , Enzyme Induction/drug effects , Ethoxyquin/pharmacology , Female , Fumarates/pharmacology , Inactivation, Metabolic , Kidney/metabolism , Kidney Tubular Necrosis, Acute/chemically induced , Naphthoquinones/chemistry , Naphthoquinones/pharmacokinetics , Rats , Rats, Sprague-Dawley
18.
Cell Mol Biol Lett ; 10(1): 15-21, 2005.
Article En | MEDLINE | ID: mdl-15809676

In our study, we analyzed the cytotoxicity of ethoxyquin (EQ) and its two salts, ethoxyquin hydrochloride (EQ-HCL) and ethoxyquin phosphate (EQ-P). It was shown that EQ was the most cytotoxic compound (IC(50) = 0.09 mM), while the lowest cytotoxic effect was observed for EQ-P (IC(50) = 0.8 mM). The properties of ethoxyquin and its salts were also analyzed with the TUNEL method, which evaluates their ability to induce apoptosis. It was shown that EQ induced apoptosis in cultured human lymphocytes, especially at concentrations of 0.25 and 0.5 mM.


Antioxidants/pharmacology , Antioxidants/toxicity , Apoptosis/drug effects , Ethoxyquin/pharmacology , Ethoxyquin/toxicity , Dose-Response Relationship, Drug , Humans , In Situ Nick-End Labeling , Lymphocytes/drug effects
19.
Mol Cell Proteomics ; 3(2): 167-75, 2004 Feb.
Article En | MEDLINE | ID: mdl-14676331

We are using a proteomic approach that combines two-dimensional electrophoresis and tandem mass spectrometry to detect and identify proteins that are differentially expressed in a cell line that is resistant to oxidative stress. The resistant cell line (OC14 cells) was developed previously through chronic exposure of a parent cell line (HA1 cells) to increasing hydrogen peroxide concentrations. Biochemical analyses of this system by other investigators have identified elevated content and activity of several classical antioxidant proteins that have established roles in oxidative stress resistance, but do not provide a complete explanation of this resistance. The proteomics studies described here have identified the enzyme aldose reductase (AR) as 4-fold more abundant in the resistant OC14 cells than in the HA1 controls. Based on this observation, the role of AR in the resistant phenotype was investigated by using a combination of AR induction with ethoxyquin and AR inhibition with Alrestatin to test the cytotoxicity of two oxidation-derived aldehydes: acrolein and glycolaldehyde. The results show that AR induction in HA1 cells provides protection against both acrolein- and glycolaldehyde-induced cytotoxicity. Furthermore, glutathione depletion sensitizes the cells to the acrolein-induced toxicity, but not the glycolaldehyde-induced toxicity, while AR inhibition sensitizes the cells to both acrolein- and glycolaldehyde-induced. These observations are consistent with a significant role for AR in the oxidative stress-resistant phenotype. These studies also illustrate the productive use of proteomic methods to investigate the molecular mechanisms of oxidative stress.


Aldehyde Reductase/metabolism , Antioxidants/metabolism , Cell Survival/drug effects , Oxidative Stress/physiology , Proteome/metabolism , Acrolein/toxicity , Amino Acid Sequence , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Ethoxyquin/pharmacology , Gas Chromatography-Mass Spectrometry , Glutaral/toxicity , Humans , Hydrogen Peroxide/toxicity , Isoquinolines/pharmacology , Molecular Sequence Data
20.
Genomics ; 83(1): 119-29, 2004 Jan.
Article En | MEDLINE | ID: mdl-14667815

Aldehyde reductase is involved in the reductive detoxification of reactive aldehydes that can modify cellular macromolecules. To analyze the mechanism of basal regulation of aldehyde reductase expression, we cloned the murine gene and adjacent regulatory region and compared it to the human gene. The mouse enzyme exhibits substrate specificity similar to that of the human enzyme, but with a 2-fold higher catalytic efficiency. In contrast to the mouse gene, the human aldehyde reductase gene has two alternatively spliced transcripts. A fragment of 57 bp is sufficient for 25% of human promoter activity and consists of two elements. The 3' element binds transcription factors of the Sp1 family. Gel-shift assays and chromatin immunoprecipitation as well as deletion/mutation analysis reveal that selenocysteine tRNA transcription activating factor (STAF) binds to the 5' element and drives constitutive expression of both mouse and human aldehyde reductase. Aldehyde reductase thus becomes the fourth protein-encoding gene regulated by STAF. The human, but not the mouse, promoter also binds C/EBP homologous protein (CHOP), which competes with STAF for the same binding site. Transfection of the human promoter into ethoxyquin-treated mouse 3T3 cells induces a 3.5-fold increase in promoter activity and a CHOP-C/EBP band appears on gel shifts performed with the 5' probe from the human aldehyde reductase promoter. Induction is attenuated in similar transfection studies of the mouse promoter. Mutation of the CHOP-binding site in the human promoter abolishes CHOP binding and significantly reduces ethoxyquin induction, suggesting that CHOP mediates stimulated expression in response to antioxidants in the human. This subtle difference in the human promoter suggests a further evolution of the promoter toward responsiveness to exogenous stress and/or toxins.


Aldehyde Reductase/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , 3T3 Cells , Aldehyde Reductase/metabolism , Animals , Base Sequence , Binding Sites/genetics , Blotting, Northern , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Cloning, Molecular , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Ethoxyquin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Genes/genetics , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Molecular Sequence Data , Mutation , Precipitin Tests , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Trans-Activators/genetics , Transcription Factor CHOP , Transcription Factors/genetics
...