Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Appl Toxicol ; 43(9): 1379-1392, 2023 09.
Article in English | MEDLINE | ID: mdl-37002789

ABSTRACT

The novel and numerous psychoactive compounds derived from the analgesic prescription drug N-phenyl-N-[1-(2-phenylethyl)piperidin-4-yl]propanamide (fentanyl) have been illegally abused as recreational drugs and caused numerous fatalities. Because some psychoactive/psychotropic drugs are known to be hepatotoxic in humans and experimental animals, the cytotoxic effects and mechanisms of 4-fluoroisobutyrylfentanyl (4F-iBF), 4-chloroisobutyrylfentanyl (4Cl-iBF), and the parent compound isobutyrylfentanyl (iBF) were studied in freshly isolated rat hepatocytes. 4F-iBF caused not only concentration (0-2.0 mM)- and time (0-3 h)-dependent cell death accompanied by the depletion of cellular ATP and reduced glutathione (GSH) and protein thiol levels but also the accumulation of oxidized glutathione. Of these fentanyls examined, 4Cl-iBF/4F-iBF-induced cytotoxicity with the loss of mitochondrial membrane potential at concentrations of 0.5 and 1.0 mM and the production of reactive oxygen species (ROS) at 0.5 mM were greater than those induced by iBF. The pretreatment of hepatocytes with N-acetyl-l-cysteine as a precursor of cellular GSH ameliorated, at least in part, cytotoxicity accompanied by insufficient ATP levels, the loss of mitochondrial membrane potential, and generation of ROS caused by 4Cl-iBF/4F-iBF, whereas pretreatment with diethyl maleate as a GSH depletor enhanced fentanyl-induced cytotoxicity accompanied by the rapid loss of cellular GSH. Taken collectively, these results indicate that the onset of cytotoxic effects caused by these fentanyls is partially attributable to cellular energy stress as well as oxidative stress.


Subject(s)
Glutathione , Hepatocytes , Humans , Rats , Animals , Reactive Oxygen Species/metabolism , Rats, Inbred F344 , Cells, Cultured , Glutathione/metabolism , Fentanyl/toxicity , Adenosine Triphosphate/metabolism
2.
Naunyn Schmiedebergs Arch Pharmacol ; 396(1): 149-159, 2023 01.
Article in English | MEDLINE | ID: mdl-36269341

ABSTRACT

New synthetic opioids continue to emerge in the illicit market, and among them, fentanyl analogues pose a serious threat to the public health with their abuse and trafficking. We investigated the toxicity of fentanyl analogues on the liver and kidneys mediated by the µ-opioid receptor (MOR). Our study focused on 4-fluoro-isobutyrylfentanyl (4F-iBF), which is classified as a "narcotic" in Japan; structurally similar analogues 4-chloro-isobutyrylfentanyl (4Cl-iBF) and isobutyrylfentanyl (iBF) were also investigated. Rats that were intraperitoneally administered 4F-iBF (5 mg/kg (12.3 µmol/kg)) or iBF (12.3 µmol/kg) displayed hepatic and renal ischemic-like damage, but 4Cl-iBF (12.3 µmol/kg) did milder renal damage only. We found that the agonist activity of 4F-iBF, at MORs was approximately 7.2 times that of 4Cl-iBF, and that pretreatment with MOR antagonist naltrexone (0.8 mg/kg) alleviated liver and kidney injuries caused by 4F-iBF. These results suggested that 4F-iBF might cause ischemic damage to the liver and kidneys, induced by respiratory depression mediated by MORs. Furthermore, to elucidate the metabolism of fentanyl analogues, we investigated the change over time in the amount of 4F-iBF, 4Cl-iBF, iBF (6.15 µmol/kg, respectively), and their respective metabolites in serum after intraperitoneal administration to rats. The results showed that in 24-h post-dose serum, 4Cl-iBF and iBF were substantially eliminated while 4F-iBF remained at about 30% of the maximum level, and each of the N-dephenylethylated metabolites of 4F-iBF, 4Cl-iBF, and iBF was detected in 2-h post-dose serum. The results from this study revealed information on the hepatic and renal toxicities and metabolism related to fentanyl analogues.


Subject(s)
Analgesics, Opioid , Fentanyl , Rats , Animals , Fentanyl/toxicity , Analgesics, Opioid/toxicity , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Liver
3.
J Appl Toxicol ; 40(2): 234-244, 2020 02.
Article in English | MEDLINE | ID: mdl-31633820

ABSTRACT

Thioxanthone and its analogues, 2- or 4-isopropylthioxanthone, 2-chlorothioxanthone, 2,4-diethylthioxanthone (DETX) and xanthone, are used as photoinitiators of ultraviolet (UV) light-initiated curable inks. As these photoinitiators were found in numerous food/beverage products packaged in cartons printed with UV-cured inks, the cytotoxic effects and mechanisms of these compounds were studied in freshly isolated rat hepatocytes. The toxicity of DETX was greater than that of other compounds. DETX elicited not only concentration (0-2.0 mm)- and time (0-3 hours)-dependent cell death accompanied by the depletion of cellular adenosine triphosphate (ATP), and reduced glutathione (GSH) and protein thiol levels, but also the accumulation of GSH disulfide and malondialdehyde. Pretreatment of hepatocytes with either fructose at a concentration of 10 mm or N-acetyl-l-cysteine (NAC) at a concentration of 5.0 mm ameliorated DETX (1 mm)-induced cytotoxicity. Further, the exposure of hepatocytes to DETX resulted in the induction of reactive oxygen species (ROS) and loss of mitochondrial membrane potential, both of which were partially prevented by the addition of NAC. These results indicate that: (1) DETX-induced cytotoxicity is linked to mitochondrial failure and depletion of cellular GSH; (2) insufficient cellular ATP levels derived from mitochondrial dysfunction were, at least in part, ameliorated by the addition of fructose; and (3) GSH loss and/or ROS formation was prevented by NAC. Taken collectively, these results suggest that the onset of toxic effects caused by DETX may be partially attributable to cellular energy stress as well as oxidative stress.


Subject(s)
Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured/drug effects , Hepatocytes/drug effects , Light , Thioxanthenes/toxicity , Xanthones/toxicity , Animals , Rats , Rats, Inbred F344
4.
J Appl Toxicol ; 38(2): 284-291, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28949027

ABSTRACT

Psychoactive compounds, N-methyl-5-(2-aminopropyl)benzofuran (5-MAPB) and 3,4-methylenedioxy-N-methamphetamine (MDMA), are known to be hepatotoxic in humans and/or experimental animals. As previous studies suggested that these compounds elicited cytotoxicity via mitochondrial dysfunction and/or oxidative stress in rat hepatocytes, the protective effects of fructose and N-acetyl-l-cysteine (NAC) on 5-MAPB- and MDMA-induced toxicity were studied in rat hepatocytes. These drugs caused not only concentration-dependent (0-4 mm) and time-dependent (0-3 hours) cell death accompanied by the depletion of cellular levels of adenosine triphosphate (ATP) and glutathione (reduced form; GSH) but also an increase in the oxidized form of GSH. The toxic effects of 5-MAPB were greater than those of MDMA. Pretreatment of hepatocytes with either fructose at a concentration of 10 mm or NAC at a concentration of 2.5 mm prevented 5-MAPB-/MDMA-induced cytotoxicity. In addition, the exposure of hepatocytes to 5-MAPB/MDMA caused the loss of mitochondrial membrane potential, although the preventive effect of fructose was weaker than that of NAC. These results suggest that: (1) 5-MAPB-/MDMA-induced cytotoxicity is linked to mitochondrial failure and depletion of cellular GSH; (2) insufficient cellular ATP levels derived from mitochondrial dysfunction were ameliorated, at least in part, by the addition of fructose; and (3) GSH loss via oxidative stress was prevented by NAC. Taken collectively, these results indicate that the onset of toxic effects caused by 5-MAPB/MDMA may be partially attributable to cellular energy stress as well as oxidative stress.


Subject(s)
Acetylcysteine/pharmacology , Benzofurans/toxicity , Fructose/pharmacology , Hepatocytes/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Propylamines/toxicity , Psychotropic Drugs/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Energy Metabolism/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects , Rats, Inbred F344
5.
J Appl Toxicol ; 37(3): 243-252, 2017 03.
Article in English | MEDLINE | ID: mdl-27291301

ABSTRACT

The novel psychoactive compounds derived from amphetamine have been illegally abused as recreational drugs, some of which are known to be hepatotoxic in humans and experimental animals. The cytotoxic effects and mechanisms of 5-(2-aminopropyl)benzofuran (5-APB) and N-methyl-5-(2-aminopropyl)benzofuran (5-MAPB), both of which are benzofuran analogues of amphetamine, and 3,4-methylenedioxy-N-methamphetamine (MDMA) were studied in freshly isolated rat hepatocytes. 5-MAPB caused not only concentration-dependent (0-4.0 mm) and time-dependent (0-3 h) cell death accompanied by the depletion of cellular ATP and reduced glutathione and protein thiol levels, but also accumulation of oxidized glutathione. Of the other analogues examined at a concentration of 4 mm, 5-MAPB/5-APB-induced cytotoxicity with the production of reactive oxygen species and loss of mitochondrial membrane potential was greater than that induced by MDMA. In isolated rat liver mitochondria, the benzofurans resulted in a greater increase in the rate of state 4 oxygen consumption than did MDMA, with a decrease in the rate of state 3 oxygen consumption. Furthermore, the benzofurans caused more of a rapid mitochondrial swelling dependent on the mitochondrial permeability transition than MDMA. 5-MAPB at a weakly toxic level (1 mm) was metabolized slowly: levels of 5-MAPB and 5-APB were approximately 0.9 mm and 50 µm, respectively, after 3 h incubation. Taken collectively, these results indicate that mitochondria are target organelles for the benzofuran analogues and MDMA, which elicit cytotoxicity through mitochondrial failure, and the onset of cytotoxicity may depend on the initial and/or residual concentrations of 5-MAPB rather than on those of its metabolite 5-APB. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Benzofurans/toxicity , Designer Drugs/toxicity , Hepatocytes/drug effects , Methamphetamine/analogs & derivatives , Propylamines/toxicity , Animals , Benzofurans/metabolism , Biotransformation , Cell Survival/drug effects , Cells, Cultured , Designer Drugs/metabolism , Dose-Response Relationship, Drug , Hepatocytes/metabolism , Hepatocytes/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Methamphetamine/metabolism , Methamphetamine/toxicity , Mitochondria, Liver/drug effects , Mitochondrial Membranes/drug effects , Permeability , Propylamines/metabolism , Rats, Inbred F344 , Reactive Oxygen Species/metabolism
6.
J Appl Toxicol ; 35(12): 1465-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25809591

ABSTRACT

DNA damage and cytotoxicity induced by a hydroxylated fullerene [C60 (OH)24 ], which is a spherical nanomaterial and/or a water-soluble fullerene derivative, and their protection by sulfhydryl compounds were studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to C60 (OH)24 at a concentration of 50 µM caused time (0 to 3 h)-dependent cell death accompanied by the formation of cell surface blebs, the loss of cellular levels of ATP and reduced glutathione, accumulation of glutathione disulfide, and induction of DNA fragmentation assayed using alkali single-cell agarose-gel electrophoresis. C60 (OH)24 -induced cytotoxicity was effectively prevented by pretreatment with sulfhydryl compounds. N-acetyl-L-cysteine (NAC), L-cysteine and L-methionine, at a concentration of 2.5 mM, ameliorated cell death, accompanied by a decrease in cellular ATP levels, formation of cell surface blebs, induction of reactive oxygen species (ROS) and loss of mitochondrial membrane potential caused by C60 (OH)24 . In addition, DNA fragmentation caused by C60 (OH)24 was also inhibited by NAC, whereas an antioxidant ascorbic acid did not affect C60 (OH)24 -induced cell death and DNA damage in rat hepatocytes. Taken collectively, these results indicate that incubation of rat hepatocytes with C60 (OH)24 elicits DNA damage, suggesting that nuclei as well as mitochondria are target sites of the hydroxylated fullerene; and induction of DNA damage and oxidative stress is ameliorated by an increase in cellular GSH levels, suggesting that the onset of toxic effects may be partially attributable to a thiol redox-state imbalance caused by C60 (OH)24 .


Subject(s)
DNA Damage/drug effects , Fullerenes/toxicity , Hepatocytes/drug effects , Membrane Potential, Mitochondrial/drug effects , Organelles/drug effects , Sulfhydryl Compounds/pharmacology , Acetylcysteine/pharmacology , Animals , Ascorbic Acid/pharmacology , Cell Culture Techniques , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Survival/drug effects , Cells, Cultured , Cysteine/pharmacology , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Male , Methionine/pharmacology , Organelles/metabolism , Organelles/ultrastructure , Rats, Inbred F344 , Reactive Oxygen Species/metabolism
7.
Arch Toxicol ; 88(1): 115-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23877122

ABSTRACT

The effects of N-acetyl-L-cysteine (NAC) on cytotoxicity caused by a hydroxylated fullerene [C60(OH)24], which is known a nanomaterial and/or a water-soluble fullerene derivative, were studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to C60(OH)24 at a concentration of 0.1 mM caused time (0-3 h)-dependent cell death accompanied by the formation of cell blebs, loss of cellular ATP, and reduced glutathione (GSH) and protein thiol levels, as well as the accumulation of glutathione disulfide and malondialdehyde (MDA), indicating lipid peroxidation. Despite this, C60(OH)24-induced cytotoxicity was effectively prevented by NAC pretreatment ranging in concentrations from 1 to 5 mM. Further, the loss of mitochondrial membrane potential (MMP) and generation of oxygen radical species in hepatocytes incubated with C60(OH)24 were inhibited by pretreatment with NAC, which caused increases in cellular and/or mitochondrial levels of GSH, accompanied by increased levels of cysteine via enzymatic deacetylation of NAC. On the other hand, severe depletion of cellular GSH levels caused by diethyl maleate at a concentration of 1.25 mM led to the enhancement of C60(OH)24-induced cell death accompanied by a rapid loss of ATP. Taken collectively, these results indicate that pretreatment with NAC ameliorates (a) mitochondrial dysfunction linked to the depletion of ATP, MMP, and mitochondrial GSH level and (b) induction of oxidative stress assessed by reactive oxygen species generation, losses of intracellular GSH and protein thiol levels, and MDA formation caused by C60(OH)24, suggesting that the onset of toxic effects is at least partially attributable to a thiol redox-state imbalance as well as mitochondrial dysfunction related to oxidative phosphorylation.


Subject(s)
Acetylcysteine/pharmacology , Fullerenes/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Animals , Cells, Cultured , Cysteine/metabolism , Glutathione/metabolism , Hydroxylation , Male , Maleates/pharmacology , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Protective Agents/pharmacology , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism
8.
Arch Toxicol ; 85(11): 1429-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21365311

ABSTRACT

The cytotoxic effects of hydroxylated fullerenes, also termed fullerenols or fullerols [C(60)(OH)( n )], which are known nanomaterials and water-soluble fullerene derivatives, were studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to C(60)(OH)(24) caused not only concentration (0-0.25 mM)- and time (0-3 h)-dependent cell death accompanied by the formation of cell blebs, loss of cellular ATP, reduced glutathione (GSH), and protein thiol levels, but also the accumulation of glutathione disulfide and malondialdehyde, indicating lipid peroxidation. Of the other analogues examined, the cytotoxic effects of C(60)(OH)(12) and fullerene C(60) at a concentration of 0.125 mM were less than those of C(60)(OH)(24). The loss of mitochondrial membrane potential and generation of oxygen radical species in hepatocytes incubated with C(60)(OH)(24) were greater than those with C(60)(OH)(12) and fullerene C(60). In the oxygen consumption of mitochondria isolated from rat liver, the ratios of state-3/state-4 respiration were more markedly decreased by C(60)(OH)(24) and C(60)(OH)(12) compared with C(60). In addition, C(60)(OH)(24) and C(60)(OH)(12) resulted in the induction of the mitochondrial permeability transition (MPT), and the effects of C(60)(OH)(12) were less than those of C(60)(OH)(24). Taken collectively, these results indicate that (a) mitochondria are target organelles for fullerenols, which elicit cytotoxicity through mitochondrial failure related to the induction of the MPT, mitochondrial depolarization, and inhibition of ATP synthesis in the early stage and subsequently oxidation of GSH and protein thiols, and lipid peroxidation through oxidative stress at a later stage; and (b) the toxic effects of fullerenols may depend on the number of hydroxyl groups participating in fullerene in rat hepatocytes.


Subject(s)
Cytotoxins/toxicity , Fullerenes/toxicity , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Animals , Cell Death , Cell Respiration/drug effects , Cells, Cultured , Glutathione/metabolism , Glutathione Disulfide/metabolism , Hepatocytes/cytology , Hydroxylation , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Oxidative Stress , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism
9.
Chem Biol Interact ; 188(3): 393-403, 2010 Dec 05.
Article in English | MEDLINE | ID: mdl-20833158

ABSTRACT

The cytotoxic effects and biotransformation of harmine and harmaline, which are known ß-carboline alkaloids and potent hallucinogens, were studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to harmine caused not only concentration (0-0.50mM)- and time (0-3h)-dependent cell death accompanied by the formation of cell blebs and the loss of cellular ATP, reduced glutathione, and protein thiols but also the accumulation of glutathione disulfide. Of the other analogues examined, the cytotoxic effects of harmaline and harmol (a metabolite of harmine) at a concentration of 0.5mM were less than those of harmine. The loss of mitochondrial membrane potential and generation of oxygen radical species in hepatocytes treated with harmine were greater than those with harmaline and harmol. In the oxygen consumption of mitochondria isolated from rat liver, the ratios of state-3/state-4 respiration of these ß-carbolines were decreased in a concentration-dependent manner. In addition, harmine resulted in the induction of the mitochondrial permeability transition (MPT), and the effects of harmol and harmaline were less than those of harmine. At a weakly toxic level of harmine (0.25mM), it was metabolized to harmol and its monoglucuronide and monosulfate conjugates, and the amounts of sulfate rather than glucuronide predominantly increased with time. In the presence of 2,5-dichloro-4-nitrophenol (50µM; an inhibitor of sulfotransferase), harmine-induced cytotoxicity was enhanced, accompanied by decrease in the amount of harmol-sulfate conjugate, due to an increase in the amount of unconjugated harmol and the inhibition of harmine loss. Taken collectively, these results indicate that (a) mitochondria are target organelles for harmine, which elicits cytotoxicity through mitochondrial failure related to the induction of the MPT, mitochondrial depolarization, and inhibition of ATP synthesis; and (b) the toxic effects of harmine are greater than those of either its metabolite harmol or its analogue harmaline, suggesting that the onset of harmine-induced cytotoxicity may depend on the initial and/or residual concentrations of harmine rather than on those of its metabolites.


Subject(s)
Harmaline/metabolism , Harmaline/toxicity , Harmine/metabolism , Harmine/toxicity , Hepatocytes/cytology , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Biotransformation , Cell Respiration/drug effects , Glutathione/metabolism , Glutathione Disulfide/metabolism , Hallucinogens/metabolism , Hallucinogens/toxicity , Harmine/analogs & derivatives , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Malondialdehyde/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Nitrophenols/pharmacology , Permeability/drug effects , Rats , Sulfhydryl Compounds/metabolism
10.
Chem Biol Interact ; 180(1): 89-97, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19428348

ABSTRACT

The biotransformation and cytotoxic effects of hydroxychavicol (HC; 1-allyl-3,4-dihydroxybenzene), which is a catecholic component in piper betel leaf and a major intermediary metabolite of safrole in rats and humans, was studied in freshly isolated rat hepatocytes. The exposure of hepatocytes to HC caused not only concentration (0.25-1.0mM)- and time (0-3h)-dependent cell death accompanied by the loss of cellular ATP, adenine nucleotide pools, reduced glutathione, and protein thiols, but also the accumulation of glutathione disulfide and malondialdehyde, indicating lipid peroxidation. At a concentration of 1mM, the cytotoxic effects of safrole were less than those of HC. The loss of mitochondrial membrane potential and generation of oxygen radical species assayed using 2',7'-dichlorodihydrofluoresein diacetate (DCFH-DA) in hepatocytes treated with HC were greater than those with safrole. HC at a weakly toxic level (0.25 and/or 0.50mM) was metabolized to monoglucuronide, monosulfate, and monoglutathione conjugates, which were identified by mass spectra and/or (1)H nuclear magnetic resonance spectra. The amounts of sulfate rather than glucuronide or glutathione conjugate predominantly increased, accompanied by a loss of the parent compound, with time. In hepatocytes pretreated with either diethyl maleate or salicylamide, HC-induced cytotoxicity was enhanced, accompanied by a decrease in the formation of these conjugates and by the inhibition of HC loss. Taken collectively, our results indicate that (a) mitochondria are target organelles for HC, which elicits cytotoxicity through mitochondrial failure related to mitochondrial membrane potential at an early stage and subsequently lipid peroxidation through oxidative stress at a later stage; (b) the onset of cytotoxicity depends on the initial and residual concentrations of HC rather than those of its metabolites; (c) the toxicity of HC is greater than that of safrole, suggesting the participation of a catecholic intermediate in safrole cytotoxicity in rat hepatocytes.


Subject(s)
Anti-Infective Agents/toxicity , Eugenol/analogs & derivatives , Hepatocytes/drug effects , Safrole/chemistry , Animals , Anti-Infective Agents/metabolism , Apoptosis/drug effects , Biotransformation , Cells, Cultured , Chromatography, High Pressure Liquid , Eugenol/metabolism , Eugenol/toxicity , Male , Mitochondria/drug effects , Mitochondria/enzymology , Oxidative Stress/drug effects , Rats , Safrole/metabolism , Spectrometry, Mass, Electrospray Ionization
11.
Arch Toxicol ; 83(1): 69-80, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18553070

ABSTRACT

The amphetamine-derived designer drugs have been illegally used worldwide as recreational drugs, some of which are known to be hepatotoxic in humans. To compare their cytotoxic effects, 3,4-methylenedioxy-N-methamphetamine (MDMA) and its related analogues, N-methyl-1-(3,4-methylenedioxyphenyl)-2-butanamine (MBDB), 3,4-(methylenedioxyphenyl)-2-butanamine (BDB) and 2-methylamino-1-(3,4-methylenedioxyphenyl)-propane-1-one (methylone) were studied in freshly isolated rat hepatocytes. MBDB caused not only concentration (0-4.0 mM)- and time (0-2 h)-dependent cell death accompanied by the formation of cell blebs, and the loss of cellular ATP and adenine nucleotide pools, and reduced glutathione levels, but also the accumulation of oxidized glutathione. Of the other analogues examined, the cytotoxicity of MBDB and BDB was greater than that of MDMA and methylone, suggesting that hepatotoxicity is generally induced by these drugs. In addition, DNA damage and the induction of reactive oxygen species were greater after the incubation of hepatocytes with MBDB (2 and 4 mM) than after that with MDMA. In isolated liver mitochondria, MBDB/BDB resulted in a greater increase in the rate of state 4 oxygen consumption than did MDMA/methylone, indicating an uncoupling effect and a decrease in the rate of state 3 oxygen consumption in a concentration dependent manner. Furthermore, MBDB resulted in mitochondrial swelling dependent on the mitochondrial permeability transition (MPT); the effect of MDMA was less than that of MBDB. Taken collectively, these results suggest that (1) the onset of cytotoxicity caused by designer drugs such as MBDB and MDMA is linked to mitochondrial failure dependent upon the induction of the MPT accompanied by mitochondrial depolarization and depletion of ATP through uncoupling of oxidative phosphorylation in rat hepatocytes, and (2) MBDB and MDMA elicit DNA damage, suggesting that nuclei as well as mitochondria are target sites of these compounds.


Subject(s)
Hepatocytes/drug effects , Mitochondria, Liver/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , 3,4-Methylenedioxyamphetamine/administration & dosage , 3,4-Methylenedioxyamphetamine/analogs & derivatives , 3,4-Methylenedioxyamphetamine/toxicity , Adenosine Triphosphate/metabolism , Animals , Cell Death/drug effects , DNA Damage/drug effects , Hallucinogens/administration & dosage , Hallucinogens/toxicity , Hepatocytes/metabolism , Male , Methamphetamine/administration & dosage , Methamphetamine/analogs & derivatives , Methamphetamine/toxicity , Mitochondria, Liver/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , Oxidative Phosphorylation/drug effects , Rats , Rats, Inbred F344 , Time Factors
12.
Biochem Pharmacol ; 75(7): 1504-14, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18261712

ABSTRACT

Nobiletin is a citrus polymethoxylated flavonoid extracted from Citrus depressa, and has several reported biological effects. In this study, we investigated the effect of nobiletin on bacterial lipopolysaccharide (LPS)-induced expression of tissue factor (TF), a trigger protein for the blood coagulation cascade, and studied the possible mechanism of TF transcriptional regulation. THP-1 monocytic cells stimulated with LPS showed an increased expression of both TF protein and mRNA levels. However, pretreatment with nobiletin resulted in inhibition of LPS-induced expression of both TF protein and mRNA in a dose-dependent manner. Electrophoretic mobility shift assays revealed that binding of nuclear proteins from LPS-stimulated THP-1 cells to the NF-kappaB or AP-1 binding motif was increased as compared to non-stimulated control cells. Such increased binding activities were significantly reduced by pretreatment with nobiletin. Binding activity of nuclear proteins to the Sp1 binding motif was observed irrespective of LPS stimulation, but Sp1 activation was inhibited by nobiletin treatment of the cells. Treatment of THP-1 cells with Sp1-specific small interfering RNA (Sp1 siRNA) abolished the ability of LPS to induce TF activity. A similar reduction in the level of TF mRNA was also observed upon treatment of cells with Sp1 siRNA. These studies reveal that constitutive Sp1 activation is an essential event for transcriptional activation of TF, and nobiletin prevents LPS-induced TF expression by inhibiting NF-kappaB, AP-1, and Sp1 activation.


Subject(s)
Flavones/pharmacology , Monocytes/metabolism , NF-kappa B/antagonists & inhibitors , Sp1 Transcription Factor/antagonists & inhibitors , Thromboplastin/biosynthesis , Transcription Factor AP-1/antagonists & inhibitors , Animals , Cells, Cultured , Citrus , Flavones/isolation & purification , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Lipopolysaccharides/pharmacology , Monocytes/drug effects , NF-kappa B/metabolism , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rabbits , Sp1 Transcription Factor/metabolism , Thromboplastin/genetics , Transcription Factor AP-1/metabolism
13.
Mutat Res ; 649(1-2): 114-25, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-17913570

ABSTRACT

Some environmental estrogen-like compounds, such as bisphenol A (BPA), 4-nonylphenol (NP), 4-octylphenol (OP), propyl p-hydroxybenzoate (P-PHBA), and butyl p-hydroxybenzoate (B-PHBA), synthetic estrogen, diethylstilbestrol (DES), and natural estrogen, 17beta-estradiol (E2), were studied for their genotoxicity in CHO-K1 cells using sister-chromatid exchange (SCE), chromosome aberration (CA), and DNA strand break (comet) assays. Six of the chemicals, excluding E2, caused DNA migration in the comet assay and induced SCEs at one or more of the highest doses. Among the chemicals, OP produced an especially high incidence of SCEs. Structural CA was induced by five of the chemicals, excluding OP and NP, and BPA, E2, and DES also induced aneuploid cells. E2 and DES particularly increased the rate of polyploidy at high doses. The incidence of colchicine-mitosis-like (c-mitotic) figures suggesting spindle disrupting effects was also detected with five of the chemicals, excluding OP and NP, and six of the chemicals, excluding E2, caused endoreduplication (ERD), a form of nuclear polyploidization induced by block of cell cycle at G2 phase, at one or more high doses. Our present results suggest that OP and NP cause repairable DNA damage, including SCEs, and do not result in CA, while the damage caused by DES, BPA, P-PHBA, and B-PHBA results in the induction of CAs together with SCEs probably because of imperfect repair. We are unable to explain the observation that the DNA damage caused by E2 resulted in CA induction but not DNA migration or SCE induction, except for speculating that the DNA damage is different from that caused by DES and the estrogen-like chemicals. Our findings also suggest that E2, DES and BPA have aneuploidogenic properties, and that the former two of chemicals also are polyploidy-inducing agents.


Subject(s)
Chromosome Aberrations/drug effects , DNA Breaks, Double-Stranded/drug effects , Estrogens/toxicity , Sister Chromatid Exchange/drug effects , Animals , Benzhydryl Compounds , CHO Cells , Comet Assay , Cricetinae , Cricetulus , Diethylstilbestrol/toxicity , Dose-Response Relationship, Drug , Estradiol/toxicity , Estradiol Congeners/toxicity , Estrogens/chemistry , Mutagenicity Tests , Parabens/toxicity , Phenols/toxicity
14.
Chem Biol Interact ; 164(1-2): 93-101, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17056023

ABSTRACT

The relationship between cytotoxicity induced by N-nitrosofenfluramine and mitochondrial or glycolytic adenosine triphosphate (ATP) synthesis-dependent intracellular bioenergetics was studied in isolated rat hepatocytes. The supplementation of fructose, an ATP-generating glycolytic substrate, to hepatocyte suspensions prevented N-nitrosofenfluramine-induced cell injury accompanied by the formation of cell blebs, abrupt loss of intracellular ATP and reduced glutathione and mitochondrial membrane potential (DeltaPsi), and the accumulation of oxidized glutathione and malondialdehyde, indicating lipid peroxidation, during a 2h incubation period. Fructose (1-20mM) resulted in concentration-dependent protection against the cytotoxicity of N-nitrosofenfluramine at a concentration of 0.6mM, a low toxic dose. Pretreatment with xylitol, another glycolytic substrate, at concentration of 15mM also prevented the cytotoxicity caused by the nitroso compound, but neither glucose nor sucrose exhibited protective effects. In addition, fructose inhibited N-nitrosofenfluramine (0.5 and 0.6mM)-induced DNA damage, as evaluated in the comet assay, indicating that nuclei as well as mitochondria are target sites of the compound. These results indicate that (a) the onset of N-nitrosofenfluramine-induced cytotoxicity in rat hepatocytes is linked to mitochondrial failure, and that (b) the insufficient supply of ATP in turn limits the activities of all energy-requiring reactions and consequently leads to acute cell death.


Subject(s)
Adenosine Triphosphate/pharmacology , Fenfluramine/analogs & derivatives , Glycolysis , Hepatocytes/drug effects , Membrane Potentials/drug effects , Mitochondria, Liver/drug effects , Animals , Cell Death/drug effects , Cells, Cultured , Comet Assay , DNA Damage , Dose-Response Relationship, Drug , Fenfluramine/metabolism , Fenfluramine/toxicity , Glutathione/metabolism , Hepatocytes/metabolism , Male , Membrane Potentials/physiology , Mitochondria, Liver/metabolism , Rats , Rats, Inbred F344 , Time Factors
15.
Eur J Pharmacol ; 529(1-3): 33-9, 2006 Jan 04.
Article in English | MEDLINE | ID: mdl-16325799

ABSTRACT

The role of mitochondrial membrane permeability transition in N-nitrosofenfluramine-induced cell injury was studied in mitochondria and hepatocytes isolated from rat liver. Mitochondrial permeability transition has been proposed as a common final pathway in acute cell death through mitochondrial dysfunction. In isolated mitochondria, N-nitrosofenfluramine (0.25 to 1.0 mM) in the presence of Ca(2+) (50 microM) elicited a concentration-dependent induction of mitochondrial swelling dependent on mitochondrial permeability transition and the release of cytochrome c, both of which were prevented by pretreatment with a specific inhibitor of mitochondrial permeability transition, cyclosporin A (0.2 microM). The effects of N-nitrosofenfluramine on mitochondria were more potent than those of fenfluramine, which is a sympathomimetic amine with anorectic action. The pretreatment of isolated hepatocytes with cyclosporin A (2 microM) partially but not completely prevented N-nitrosofenfluramine (0.6 mM; a low toxic dose)-induced cell death, loss of cellular ATP, formation of cell blebs and decrease in mitochondrial membrane potential. These results suggest that the onset of N-nitrosofenfluramine-induced cytotoxicity is linked to mitochondrial failure dependent upon induction of mitochondrial permeability transition accompanied by mitochondrial depolarization, the release of cytochrome c and depletion of intracellular ATP through uncoupling of oxidative phosphorylation.


Subject(s)
Dietary Supplements/toxicity , Fenfluramine/analogs & derivatives , Hepatocytes/drug effects , Ion Channels/metabolism , Mitochondria, Liver/drug effects , Animals , Cell Death/drug effects , Cells, Cultured , Cyclosporine/pharmacology , Cytochromes c/metabolism , Fenfluramine/toxicity , Hepatocytes/metabolism , Male , Membrane Potentials/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Rats , Rats, Inbred F344
16.
Arch Toxicol ; 79(6): 312-20, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15696257

ABSTRACT

The cytotoxic effects of fenfluramine, an appetite suppressant, and its N-nitroso derivative, N-nitrosofenfluramine, have been studied in freshly isolated rat hepatocytes and isolated hepatic mitochondria. Exposure of hepatocytes to N-nitrosofenfluramine caused not only concentration (0.25-1.0 mmol L(-1)) and time (0-3 h)-dependent cell death accompanied by the loss of cellular ATP, adenine nucleotide pools, reduced glutathione (GSH), and protein thiols, but also the accumulation of oxidized glutathione and malondialdehyde (MDA), indicating lipid peroxidation. There was a time lag for the onset of the accumulation of MDA after the rapid depletion of ATP. Supplementation of the hepatocyte suspensions with N-acetylcysteine (4 mmol L(-1)), a precursor of intracellular GSH, partially inhibited N-nitrosofenfluramine (1 mmol L(-1))-induced cytotoxicity. In comparative effects based on cell viability and rhodamine 123 retention, an index of mitochondrial membrane potential, fenfluramine was less toxic than N-nitrosofenfluramine. In mitochondria isolated from rat liver, N-nitrosofenfluramine caused an increase in the rate of state-4 oxygen consumption, indicating an uncoupling effect, and a decrease in the rate of state-3 oxygen consumption in a concentration-dependent manner. These results indicate that (a) mitochondria are target organelles for N-nitrosofenfluramine, which elicits cytotoxicity through mitochondrial dysfunction related to membrane potential and/or oxidative phosphorylation at an early stage and subsequently lipid peroxidation at a later stage; and (b) the toxicity of N-nitrosofenfluramine is greater than that of fenfluramine, suggesting participation of the nitroso group in the toxicity.


Subject(s)
Fenfluramine/analogs & derivatives , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Adenine Nucleotides/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fenfluramine/toxicity , Glutathione/metabolism , Hepatocytes/metabolism , Male , Malondialdehyde/metabolism , Membrane Potentials/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/physiology , Rats , Rats, Inbred F344 , Sulfhydryl Compounds/metabolism , Time Factors
17.
Circ J ; 68(11): 982-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15502376

ABSTRACT

BACKGROUND: The relationship between abdominal visceral fat accumulation and lacunar infarcts has not been previously investigated in Japanese men. METHODS AND RESULTS: The subjects were 637 middle-aged (40-64 years) and 222 elderly (65-79 years) men who participated in a health checkup program from 1999 to 2003. The association between lacunar infarcts identified by magnetic resonance imaging and cardiovascular risk factors, including abdominal visceral fat accumulation evaluated by computed tomography, was examined. The prevalence of lacunar infarcts was 4.9%. Hypertension was associated with lacunar infarcts among both the middle-aged men [age-adjusted odds ratio (OR)=2.9 (95% confidence interval (CI): 1.1-7.8)] and the elderly men [OR=5.1 (95%CI: 1.4-19.0)]. Abdominal visceral fat accumulation was slightly associated with lacunar infarcts among middle-aged men, but not among elderly men: OR in the highest (>or=117 cm(2)) vs lowest (or=143 cm(2)) was still slightly associated with lacunar infarcts after adjustment for age, hypertension, drinking and smoking among middle-aged men [OR=2.7 (95%CI: 0.8-9.1)]. CONCLUSIONS: This cross-sectional study suggests that abdominal visceral fat accumulation is a possible risk factor of lacunar infarcts, in addition to hypertension, in middle-aged Japanese men.


Subject(s)
Abdomen , Adipose Tissue/diagnostic imaging , Asian People , Brain Infarction/diagnosis , Brain Infarction/ethnology , Magnetic Resonance Imaging , Tomography, X-Ray Computed , Adult , Aged , Brain/pathology , Brain Infarction/epidemiology , Cardiovascular Diseases/etiology , Cross-Sectional Studies , Humans , Japan/epidemiology , Male , Middle Aged , Prevalence , Risk Factors , Viscera
18.
Toxicology ; 200(2-3): 123-33, 2004 Aug 05.
Article in English | MEDLINE | ID: mdl-15212809

ABSTRACT

The metabolism and action of chlorpropham (isopropyl N-(3-chlorophenyl)carbamate; CIPC, a post-harvest agent) and its metabolites were studied in freshly isolated rat hepatocytes and isolated rat hepatic mitochondria, respectively. The exposure of hepatocytes to CIPC caused a concentration (0.25-1.0 mM)- and time (0-3h)-dependent cell death accompanied by loss of cellular ATP and adenine nucleotides. CIPC at a weakly toxic level (0.5 mM) was metabolized to isopropyl N-(3-chloro-4-hydroxyphenyl)carbamate (4OH-CIPC) and subsequently to its glucuronide and sulfate conjugates (major metabolites) or alternatively to a minor metabolite 3-chloroaniline (3CA). The addition of SKF-525A (50 microM), an inhibitor of microsomal monooxygenase, enhanced the CIPC (0.5 mM)-induced cytotoxicity accompanied by loss of ATP and 4OH-CIPC and inhibited the decrease in the concentration of the parent compound. CIPC led to a strong decrease in cellular ATP content compared to its metabolites, 4OH-CIPC and 3CA. On the other hand, the exposure of isolated hepatic mitochondria to CIPC reduced State 3 respiration with a FAD-linked substrate (succinate plus rotenone) and/or with a NAD+ -linked substrate (pyruvate plus malate), whereas State 3 respiration with ascorbate plus tetramethyl-p-phenylendiamine (cytochrome oxidase-linked respiration) was not affected markedly by CIPC. Further, the addition of CIPC caused an increase in the rate of State 4 oxygen consumption, indicating an uncoupling effect, and a decrease in the rate of State 3 oxygen consumption in a concentration-dependent manner, respectively. In contrast, the addition of neither 4OH-CIPC nor 3CA markedly affected the rate of states 3 and/or 4 oxygen consumption. These results indicate that CIPC-induced cytotoxicity is mediated by the parent compound rather than by its metabolites such as 4OH-CIPC and 3CA, and that the toxicity is associated with a rapid depletion of ATP via impairment of mitochondrial function related to oxidative phosphorylation.


Subject(s)
Chlorpropham/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Herbicides/toxicity , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Adenine Nucleotides/metabolism , Animals , Biotransformation , Cell Death/drug effects , Cell Separation , Chlorpropham/metabolism , Chromatography, High Pressure Liquid , Enzyme Inhibitors/toxicity , Herbicides/metabolism , In Vitro Techniques , Kinetics , Male , Mass Spectrometry , Mixed Function Oxygenases/antagonists & inhibitors , Oxygen Consumption/drug effects , Proadifen/pharmacology , Rats , Rats, Inbred F344
19.
Environ Sci Technol ; 38(8): 2389-96, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15116845

ABSTRACT

Monitoring of bisphenol A [BPA; 2,2-bis(4-hydroxyphenyl)propane] and its biological metabolites [4,4'-dihydroxy-alphamethylstilbene (DHMS), 2,2-bis(4-hydroxyphenyl)-1-propanol (BPA-OH), 2,2-bis(4-hydroxyphenyl)propanoic acid (BPA-COOH), and 2-(3,4-dihydroxyphenyl)-2-(4-hydroxyphenyl)propane (3-OH-BPA)] in river waters was performed by solid-phase extraction and GC/MS determination. The concentrations of BPA, BPA-COOH, BPA-OH, and 3-OH-BPA in the river water ranged from 2 to 230 (8.8 x 10(-12) to 1.0 x 10(-9) M), from 5 to 75 (1.9 x 10(-11) to 2.9 x 10(-10) M), from 3 to 16 (1.2 x 10(-11) to 6.6 x 10(-11) M), and from 3 to 11 (1.2 x 10(-11) to 4.5 x 10(-11) M) ng L(-1), respectively. DHMS, an intermediate in the main degradation pathway of BPA, was not detected in any water sample. Under the aerobic conditions in the river water, BPA disappeared within 8 d of incubation, but BPA-COOH, BPA-OH, and tetraol remained in the supernatant after 14 d of incubation. For the xeno-estrogenic activity of BPA and the metabolites, their ability to bind to recombinant human estrogen receptor alpha in competition with fluorescence-labeled 17beta-estradiol was measured. Fifty percent inhibitory concentrations (IC50) of BPA, DHMS, 3-OH-BPA, and BPA-OH were approximately 1 x 10(-5), 1 x 10(-6), 3 x 10(-5), and 1 x 10(-2) M, respectively. In human cultured MCF-7 breast cancer cells, BPA increased cell numbers in a dose-dependent manner at concentrations from 10(-7) to 10(-5) M. For the BPA metabolites, DOHMS, 3-OH-BPA, and BPA-COOH caused the cells proliferation at concentrations from 10(-9) to 10(-6), from 10(-7) to 10(-6), and from 10(-5) to 10(-4) M, respectively. BPA-OH did not cause MCF-7 cells proliferation. These results indicate that BPA is mainly metabolized through oxidative rearrangement by bacteria in the river water, and intermediate bisphenols via minor metabolic pathways exist in river water. The presence of the bisphenols having the xeno-estrogenic effect suggests the necessity of monitoring those in river water, in the effluent waters from sewage plants, or in landfill leachate.


Subject(s)
Estrogens, Non-Steroidal/metabolism , Estrogens, Non-Steroidal/pharmacology , Phenols/metabolism , Phenols/pharmacology , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/pharmacology , Bacteria , Benzhydryl Compounds , Environmental Monitoring , Estradiol/pharmacology , Estrogens, Non-Steroidal/analysis , Humans , In Vitro Techniques , Oxidation-Reduction , Phenols/analysis , Receptors, Estrogen/drug effects , Rivers , Sewage/chemistry , Water Pollutants, Chemical/analysis
20.
Biochem Pharmacol ; 66(1): 63-73, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12818366

ABSTRACT

The metabolism and action of trans-anethole (anethole) and the estrogen-like activity of the compound and its metabolites were studied in freshly isolated rat hepatocytes and cultured MCF-7 human breast cancer cells, respectively. The incubation of hepatocytes with anethole (0.25-2.0mM) caused a concentration- and time-dependent cell death accompanied by losses of cellular ATP and adenine nucleotide pools. Anethole at a weakly toxic level (0.5mM) was metabolized to 4-methoxycinnamic acid (4MCA), 4-hydroxy-1-propenylbenzene (4OHPB), and the monosulfate conjugate of 4OHPB; the levels of 4OHPB sulfate and 4MCA reached approximately 20 and 200 microM within 2 hr, respectively, whereas that of free unconjugated 4OHPB was less than approximately 0.5 microM. At a moderately toxic concentration (1.0mM), unconjugated 4OHPB reached approximately 10 microM, followed by abrupt loss of 3'-phosphoadenosine 5'-phosphosulphate (PAPS). Based on cell viability and adenine nucleotide levels, 4OHPB was more toxic than anethole and 4MCA. The addition of 2,6-dichloro-4-nitrophenol (50 microM), an inhibitor of sulfotransferase, enhanced the anethole-induced cytotoxicity associated with losses of ATP, PAPS, and 4OHPB sulfate, and symmetrically increased the unconjugated 4OHPB concentration. 4OHPB as well as diethylstilbestrol (DES) and bisphenol A (BPA), which are known xenoestrogenic compounds, competitively displaced 17beta-estradiol bound to the estrogen receptor alpha in a concentration-dependent manner; IC(50) values of these compounds were approximately 1 x 10(-5), 1 x 10(-8) and 5 x 10(-5)M, respectively. 4OHPB also caused a concentration (10(-8) to 10(-6)M)-dependent proliferation of MCF-7 cells, whereas neither anethole nor 4MCA (10(-9) to 10(-5)M) affected cell proliferation. However, at higher concentrations (>10(-4)M), 4OHPB rather than anethole and 4MCA was cytotoxic. These results suggest that the biotransformation of anethole induces a cytotoxic effect at higher concentrations in rat hepatocytes and an estrogenic effect at lower concentrations in MCF-7 cells based on the concentrations of the hydroxylated intermediate, 4OHPB.


Subject(s)
Anisoles/metabolism , Biotransformation , Hepatocytes/metabolism , Pentobarbital/analogs & derivatives , Allylbenzene Derivatives , Animals , Anisoles/pharmacology , Binding, Competitive , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Cinnamates/pharmacology , Drug Interactions , Estrogen Receptor alpha , Hepatocytes/drug effects , Humans , Male , Pentobarbital/pharmacology , Rats , Rats, Inbred F344 , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Sulfotransferases/antagonists & inhibitors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...