Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
J Lipid Res ; 64(11): 100458, 2023 11.
Article in English | MEDLINE | ID: mdl-37838304

ABSTRACT

Although pregnant women's fish consumption is beneficial for the brain development of the fetus due to the DHA in fish, seafood also contains methylmercury (MeHg), which adversely affects fetal brain development. Epidemiological studies suggest that high DHA levels in pregnant women's sera may protect the fetal brain from MeHg-induced neurotoxicity, but the underlying mechanism is unknown. Our earlier study revealed that DHA and its metabolite 19,20-dihydroxydocosapentaenoic acid (19,20-DHDP) produced by cytochrome P450s (P450s) and soluble epoxide hydrolase (sEH) can suppress MeHg-induced cytotoxicity in mouse primary neuronal cells. In the present study, DHA supplementation to pregnant mice suppressed MeHg-induced impairments of pups' body weight, grip strength, motor function, and short-term memory. DHA supplementation also suppressed MeHg-induced oxidative stress and the decrease in the number of subplate neurons in the cerebral cortex of the pups. DHA supplementation to dams significantly increased the DHA metabolites 19,20-epoxydocosapentaenoic acid (19,20-EDP) and 19,20-DHDP as well as DHA itself in the fetal and infant brains, although the expression levels of P450s and sEH were low in the fetal brain and liver. DHA metabolites were detected in the mouse breast milk and in human umbilical cord blood, indicating the active transfer of DHA metabolites from dams to pups. These results demonstrate that DHA supplementation increased DHA and its metabolites in the mouse pup brain and alleviated the effects of MeHg on fetal brain development. Pregnant women's intake of fish containing high levels of DHA (or DHA supplementation) may help prevent MeHg-induced neurotoxicity in the fetus.


Subject(s)
Methylmercury Compounds , Infant , Animals , Humans , Pregnancy , Female , Mice , Methylmercury Compounds/toxicity , Docosahexaenoic Acids/pharmacology , Brain , Oxidative Stress , Fetus
2.
Biol Pharm Bull ; 46(2): 292-300, 2023.
Article in English | MEDLINE | ID: mdl-36724957

ABSTRACT

Methylmercury (MeHg) is a well-known environmental pollutant that has harmful effects on the central nervous systems of humans and animals. The molecular mechanisms of MeHg-induced neurotoxicity at low concentrations are not fully understood. Here, we investigated the effects of low-concentration MeHg on the cell viability, Ca2+ homeostasis, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA2 levels, which determine Ca2+ permeability of AMPA receptors, in rat primary cortical neurons. Exposure of cortical neurons to 100 and 300 nM MeHg for 7 d resulted in a decrease in GluA2 levels, an increase in basal intracellular Ca2+ concentration, increased phosphorylation levels of extracellular signal-regulated kinase (ERK)1/2 and p38, and decreased cell viability. Moreover, glutamate stimulation exacerbated the decrease in cell viability and increased intracellular Ca2+ levels in MeHg-treated neurons compared to control neurons. MeHg-induced neuronal cell death was ameliorated by 1-naphthyl acetyl spermine, a specific antagonist of Ca2+-permeable, GluA2-lacking AMPA receptors. Our findings raise the possibility that decreased neuronal GluA2 levels and the subsequent increase in intracellular Ca2+ concentration may contribute to MeHg-induced neurotoxicity.


Subject(s)
Methylmercury Compounds , Receptors, AMPA , Animals , Rats , Calcium/metabolism , Cells, Cultured , Glutamic Acid/metabolism , Homeostasis , Methylmercury Compounds/metabolism , Neurons , Receptors, AMPA/metabolism
3.
Biochem Biophys Res Commun ; 592: 31-37, 2022 02 12.
Article in English | MEDLINE | ID: mdl-35016149

ABSTRACT

Tributyltin (TBT) is an environmental pollutant that remains in marine sediments and is toxic to mammals. For example, TBT elicits neurotoxic and immunosuppressive effects on rats. However, it is not entirely understood how TBT causes toxicity. Autophagy plays a pivotal role in protein quality control and eliminates aggregated proteins and damaged organelles. We previously reported that TBT dephosphorylates mammalian target of rapamycin (mTOR), which may be involved in enhancement of autophagosome synthesis, in primary cultures of cortical neurons. Autophagosomes can accumulate due to enhancement of autophagosome synthesis or inhibition of autophagic degradation, and we did not clarify whether TBT alters autophagic flux. Here, we investigated the mechanism by which TBT causes accumulation of autophagosomes in SH-SY5Y cells. TBT inhibited autophagy without affecting autophagosome-lysosome fusion before it caused cell death. TBT dramatically decreased the acidity of lysosomes without affecting lysosomal membrane integrity. TBT decreased the mature protein level of cathepsin B, and this may be related to the decrease in lysosomal acidity. These results suggest that TBT inhibits autophagic degradation by decreasing lysosomal acidity. Autophagy impairment may be involved in the mechanism underlying neuronal death and/or T-cell-dependent thymus atrophy induced by TBT.


Subject(s)
Autophagy , Lysosomes/metabolism , Trialkyltin Compounds/pharmacology , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydrolysis , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/metabolism
4.
Arch Biochem Biophys ; 715: 109099, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34856193

ABSTRACT

Xenobiotic-metabolizing enzymes (XMEs) expressed in the olfactory epithelium (OE) are known to metabolize odorants. Aldehyde oxidase (AOX) recognizes a wide range of substrates among which are substrates with aldehyde groups. Some of these AOX substrates are odorants, such as benzaldehyde and n-octanal. One of the mouse AOX isoforms, namely AOX2 (mAOX2), was shown to be specifically expressed in mouse OE but its role to metabolize odorants in this tissue remains unexplored. In this study, we investigated the involvement of mouse AOX isoforms in the oxidative metabolism of aldehyde-odorants in the OE. Mouse OE extracts effectively metabolized aromatic and aliphatic aldehyde-odorants. Gene expression analysis revealed that not only mAOX2 but also the mAOX3 isoform is expressed in the OE. Furthermore, evaluation of inhibitory effects using the purified recombinant enzymes led us to identify specific inhibitors of each isoform, namely chlorpromazine, 17ß-estradiol, menadione, norharmane, and raloxifene. Using these specific inhibitors, we defined the contribution of mAOX2 and mAOX3 to the metabolism of aldehyde-odorants in the mouse OE. Taken together, these findings demonstrate that mAOX2 and mAOX3 are responsible for the oxidation of aromatic and aliphatic aldehyde-odorants in the mouse OE, implying their involvement in odor perception.


Subject(s)
Aldehyde Oxidase/metabolism , Aldehyde Oxidoreductases/metabolism , Aldehydes/metabolism , Odorants , Olfactory Mucosa/metabolism , Aldehyde Oxidase/antagonists & inhibitors , Aldehyde Oxidoreductases/antagonists & inhibitors , Aldehydes/chemistry , Animals , Enzyme Inhibitors/pharmacology , Female , Male , Mice, Inbred C57BL , Olfactory Mucosa/drug effects , Oxidation-Reduction/drug effects , Smell/drug effects
5.
Neurochem Res ; 46(9): 2285-2296, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34081246

ABSTRACT

It has been demonstrated that peripheral inflammation induces cognitive dysfunction. Several histone deacetylase (HDAC) inhibitors ameliorate cognitive dysfunction in animal models of not only peripheral inflammation but also Alzheimer's disease. However, it is not clear which HDAC expressed in the central nervous system or peripheral tissues is involved in the therapeutic effect of HDAC inhibition on cognitive dysfunction. Hence, the present study investigated the effect of peripheral HDAC inhibition on peripheral inflammation-induced cognitive dysfunction. Suberoylanilide hydroxamic acid (SAHA), a pan-HDAC inhibitor that is mainly distributed in peripheral tissues after intraperitoneal administration, was found to prevent peripheral inflammation-induced cognitive dysfunction. Moreover, pretreatment with SAHA dramatically increased mRNA expression of interleukin-10, an anti-inflammatory cytokine, in peripheral and central tissues and attenuated peripheral inflammation-induced microglial activation in the CA3 region of the hippocampus. Minocycline, a macrophage/microglia inhibitor, also ameliorated cognitive dysfunction. Furthermore, as a result of treatment with liposomal clodronate, depletion of peripheral macrophages partially ameliorated the peripheral inflammation-evoked cognitive dysfunction. Taken together, these findings demonstrate that inhibition of peripheral HDAC plays a critical role in preventing cognitive dysfunction induced by peripheral inflammation via the regulation of anti-inflammatory cytokine production and the inhibition of microglial functions in the hippocampus. Thus, these findings could provide support for inhibition of peripheral HDAC as a novel therapeutic strategy for inflammation-induced cognitive dysfunction.


Subject(s)
Cognitive Dysfunction/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Microglia/drug effects , Vorinostat/therapeutic use , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Cytokines/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/chemically induced , Inflammation/complications , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides , Male , Mice , Microglia/metabolism
6.
Biochem Biophys Res Commun ; 509(1): 287-293, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30587336

ABSTRACT

Cytochrome P450 (CYP) 3A4 plays an important role in drug metabolism. Although transcriptional regulation of CYP3A expression by chemicals has been comprehensively studied, its post-translational regulation is not fully understood. We previously reported that acetaminophen (APAP) caused accumulation of functional CYP3A protein via inhibition of CYP3A protein degradation through reduction of glycoprotein 78 (gp78), an E3 ligase of the ubiquitin proteasome system. Furthermore, N-acetyl-m-aminophenol, a regioisomer of APAP causes CYP3A protein accumulation, whereas p-acetamidobezoic acid, in which a hydroxy group of APAP was substituted for a carboxy group, did not lead to the same effects. However, the mechanism underlying the reduction of gp78 protein expression by APAP has not yet been elucidated. In this study, we selected 32 compounds including a phenolic hydroxyl group such as APAP and explored the compounds that increased CYP3A enzyme activity to analyze their common mechanism. Four compounds, including salicylate, increased CYP3A enzyme activity and led to the accumulation of functional CYP3A protein similarly to APAP. APAP and salicylate activate p38 mitogen-activated protein kinase (p38 MAPK). gp78 is known to be phosphorylated by p38 MAPK; so, we investigated the relationship between p38 MAPK and CYP3A. APAP activated p38 MAPK, decreased gp78 protein expression, and subsequently induced CYP3A protein expression in a time-dependent manner. When SB203580, a p38 MAPK inhibitor, was co-administered with APAP, the inhibitory effects of APAP on CYP3A protein degradation were suppressed. In this study, we demonstrated the involvement of the p38 MAPK-gp78 pathway in suppressing CYP3A protein degradation by APAP. Salicylate derivatives may also suppress the CYP3A protein degradation.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cytochrome P-450 CYP3A/metabolism , Salicylates/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Proteolysis/drug effects , Rats, Sprague-Dawley
7.
Toxicol Appl Pharmacol ; 370: 133-144, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30880217

ABSTRACT

Liver resection is performed to remove tumors in patients with liver cancer, but the procedure's suitability depends on the regenerative ability of the liver. It is important to consider the effects of exogenous factors, such as diets, on liver regeneration for the recovery of function. The evaluation of drug metabolism during liver regeneration is also necessary because liver dysfunction is generally observed after the operation. Here, we investigated the influence of a purified diet (AIN-93G) on liver regeneration and changes in the mRNA expression of several cytochrome P450 (CYP) isoforms in the liver and small intestine using a two-thirds partial hepatectomy (PH) mouse model fed with a standard diet (MF) and a purified diet. Liver regeneration was significantly delayed in the purified diet group relative to that in the standard diet group. The liver Cyp2c55 and Cyp3a11 expression was increased at 3 day after PH especially in the purified diet group. Bile acid may partly cause the differences in liver regeneration and CYP expression between two types of diets. On the other hand, Cyp3a13 expression in the small intestine was transiently increased at day 1 after PH in both diet groups. The findings suggest that compensatory induction of the CYP expression occurred in the small intestine after attenuation of drug metabolism potential in the liver. The present results highlight the importance of the relationship between liver regeneration, drug metabolism, and exogenous factors for the effective treatment, including surgery and medication, in patients after liver resection or transplantation.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Diet , Hepatectomy , Intestines/enzymology , Liver Regeneration/physiology , Liver/enzymology , Animals , Bile Acids and Salts/blood , Cytochrome P-450 CYP3A/genetics , Gene Expression , Isoenzymes/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis
8.
Biol Pharm Bull ; 42(8): 1366-1375, 2019.
Article in English | MEDLINE | ID: mdl-31366871

ABSTRACT

Drug-induced liver injury (DILI) is a common side effect of several medications and is considered a major factor responsible for the discontinuation of drugs during their development. Cholestasis is a DILI that results from impairment of bile acid transporters, such as the bile salt export pump (BSEP), leading to accumulation of bile acids. Both in vitro and in vivo studies are required to predict the risk of drug-induced cholestasis. In the present study, we used chimeric mice with humanized liver as a model to study drug-induced cholestasis. Administration of a single dose of ketoconazole or rifampicin, known to potentially cause cholestasis by inhibiting BSEP, did not result in elevated levels of alkaline phosphatase (ALP), which are known hepatic biomarkers. The concentration of taurodeoxycholic acid increased in the liver after ketoconazole administration, whereas rifampicin resulted in increased tauromuricholic acid and taurocholic acid (TCA) levels in the liver and plasma. Furthermore, rifampicin resulted in an increase in the uniform distribution of a compound with m/z 514.3, presumed as TCA through imaging mass spectrometry. The mRNA levels of bile acid-related genes were also altered after treatment with ketoconazole or rifampicin. We believe these observations to be a part of a feedback mechanism to decrease bile acid concentrations. The changes in bile acid concentrations results may reflect the initial responses of the human body to cholestasis. Furthermore, these findings may contribute to the screening of drug candidates, thereby avoiding drug-induced cholestasis during clinical trials and drug development.


Subject(s)
Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Cholestasis/metabolism , Ketoconazole/adverse effects , Liver/drug effects , Rifampin/adverse effects , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Aspartate Aminotransferases/blood , Bile Acids and Salts/blood , Chemical and Drug Induced Liver Injury/blood , Cholestasis/blood , Cholestasis/chemically induced , Humans , Ketoconazole/blood , Ketoconazole/pharmacokinetics , Liver/metabolism , Male , Mice , Rifampin/blood , Rifampin/pharmacokinetics
9.
Arch Toxicol ; 92(1): 401-409, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28725974

ABSTRACT

Glutamate receptor 2 (GluA2/GluR2) is one of the four subunits of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR); an increase in GluA2-lacking AMPARs contributes to neuronal vulnerability to excitotoxicity because of the receptor's high Ca2+ permeability. Carbofuran is a carbamate pesticide used in agricultural areas to increase crop productivity. Due to its broad-spectrum action, carbofuran has also been used as an insecticide, nematicide, and acaricide. In this study, we investigated the effect of carbofuran on GluA2 protein expression. The 9-day treatment of rat primary cortical neurons with 1 µM and 10 µM carbofuran decreased GluA2 protein expression, but not that of GluA1, GluA3, or GluA4 (i.e., other AMPAR subunits). Decreased GluA2 protein expression was also observed on the cell surface membrane of 10 µM carbofuran-treated neurons, and these neurons showed an increase in 25 µM glutamate-triggered Ca2+ influx. Treatment with 50 µM glutamate, which did not affect the viability of control neurons, significantly decreased the viability of 10 µM carbofuran-treated neurons, and this effect was abolished by pre-treatment with 300 µM 1-naphthylacetylspermine, an antagonist of GluA2-lacking AMPAR. At a concentration of 100 µM, but not 1 or 10 µM, carbofuran significantly decreased acetylcholine esterase activity, a well-known target of this chemical. These results suggest that carbofuran decreases GluA2 protein expression and increases neuronal vulnerability to glutamate toxicity at concentrations that do not affect acetylcholine esterase activity.


Subject(s)
Carbofuran/toxicity , Cerebral Cortex/cytology , Glutamic Acid/toxicity , Neurons/drug effects , Receptors, AMPA/metabolism , Acetylcholinesterase/metabolism , Animals , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cerebral Cortex/embryology , Cholinesterase Inhibitors/toxicity , Female , GPI-Linked Proteins/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Pregnancy , Rats, Wistar , Spermine/analogs & derivatives , Spermine/pharmacology
10.
Biochem Biophys Res Commun ; 486(3): 639-644, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28341123

ABSTRACT

Cytochrome P450 (CYP) 3A subfamily members are known to metabolize various types of drugs, highlighting the importance of understanding drug-drug interactions (DDI) depending on CYP3A induction or inhibition. While transcriptional regulation of CYP3A members is widely understood, post-translational regulation needs to be elucidated. We previously reported that acetaminophen (APAP) induces CYP3A activity via inhibition of protein degradation and proposed a novel DDI concept. N-Acetyl-p-benzoquinone imine (NAPQI), the reactive metabolite of APAP formed by CYP, is known to cause adverse events related to depletion of intracellular reduced glutathione (GSH). We aimed to inspect whether NAPQI rather than APAP itself could cause the inhibitory effects on protein degradation. We found that N-acetyl-l-cysteine, the precursor of GSH, and 1-aminobenzotriazole, a nonselective CYP inhibitor, had no effect on CYP3A1/23 protein levels affected by APAP. Thus, we used APAP analogs to test CYP3A1/23 mRNA levels, protein levels, and CYP3A activity. We found N-acetyl-m-aminophenol (AMAP), a regioisomer of APAP, has the same inhibitory effects of CYP3A1/23 protein degradation, while p-acetamidobenzoic acid (PAcBA), a carboxy-substituted form of APAP, shows no inhibitory effects. AMAP and PAcBA cannot be oxidized to quinone imine forms such as NAPQI, so the inhibitory effects could depend on the specific chemical structure of APAP.


Subject(s)
Acetaminophen/pharmacology , Benzoquinones/pharmacology , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/genetics , Hepatocytes/drug effects , Imines/pharmacology , Acetaminophen/metabolism , Acetylcysteine/pharmacology , Animals , Benzoquinones/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inducers/metabolism , Cytochrome P-450 CYP3A Inhibitors/metabolism , Gene Expression Regulation , Glutathione/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Imines/metabolism , Male , Primary Cell Culture , Protein Stability/drug effects , Proteolysis/drug effects , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Triazoles/pharmacology
11.
Biol Pharm Bull ; 40(3): 303-309, 2017.
Article in English | MEDLINE | ID: mdl-28250271

ABSTRACT

Lead is a persistent environmental pollutant and exposure to high environmental levels causes various deleterious toxicities, especially to the central nervous system (CNS). The α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor that is devoid of the glutamate receptor 2 (GluR2) subunit is Ca2+-permeable, which increases the neuronal vulnerability to excitotoxicity. We have previously reported that long-term exposure of rat cortical neurons to lead acetate induces decrease of GluR2 expression. However, it is not clarified whether lead-induced GluR2 decrease is involved in neurotoxicity. Therefore, we investigated the contribution of GluR2 non-containing AMPA receptor to lead-induced neurotoxic events. Although the expression of four AMPA receptor subunits (GluR1, GluR2, GluR3, and GluR4) was decreased by lead exposure, the decrease in GluR2 expression was remarkable among four subunits. Lead-induced neuronal cell death was rescued by three glutamate receptor antagonists, 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX, a non-selective AMPA receptor blocker), MK-801 (N-methyl-D-aspartate (NMDA) receptor blocker), and 1-naphthyl acetyl spermine (NAS, a specific Ca2+-permeable AMPA receptor blocker). Lead exposure activated extracellular signal-regulated protein kinase (ERK) 1/2, which was significantly ameliorated by CNQX. In addition, lead exposure activated p38 mitogen-activated protein kinase (MAPK p38), and protein kinase C (PKC), which was partially ameliorated by CNQX. Our findings indicate that Ca2+-permeable AMPA receptors resulting from GluR2 decrease may be involved in lead-induced neurotoxicity.


Subject(s)
Brain/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutamic Acid/metabolism , Lead Poisoning, Nervous System/metabolism , Lead/adverse effects , Receptors, AMPA/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Brain/cytology , Calcium/metabolism , Cells, Cultured , Environmental Pollutants/adverse effects , Excitatory Amino Acid Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Protein Kinase C/metabolism , Protein Subunits , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Biol Pharm Bull ; 40(7): 1121-1124, 2017.
Article in English | MEDLINE | ID: mdl-28674256

ABSTRACT

Tributyltin (TBT), a common environmental contaminant, is widely used as an antifouling agent in paint. We previously reported that exposure of primary cortical neurons to TBT in vitro decreased the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit glutamate receptor 2 (GluR2) expression and subsequently increased neuronal vulnerability to glutamate. Therefore, to identify whether GluR2 expression also decreases after TBT exposure in vivo, we evaluated the changes in GluR2 expression in the mouse brain after prenatal or postnatal exposure to 10 and 25 ppm TBT through pellet diets. Although the mean feed intake and body weight did not decrease in TBT-exposed mice compared with that in control mice, GluR2 expression in the cerebral cortex and hippocampus decreased after TBT exposure during the prenatal period. These results indicate that a decrease in neuronal GluR2 may be involved in TBT-induced neurotoxicity, especially during the fetal period.


Subject(s)
Brain/drug effects , Prenatal Exposure Delayed Effects , Receptors, AMPA/metabolism , Trialkyltin Compounds/toxicity , Animals , Body Weight , Brain/metabolism , Feeding Behavior , Female , Mice , Pregnancy
13.
Arch Toxicol ; 91(2): 885-895, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27155986

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a persistent environmental contaminant. Although studies have described PFOS-induced neurotoxicity in animal brains and neuronal cells, the molecular mechanisms of PFOS-induced neurotoxicity based on the distribution properties, especially during developmental periods, have not been clarified. To clarify the mechanisms of PFOS-induced neuronal vulnerability during developmental periods, we examined changes in glutamate receptor 2 (GluR2) expression and related neurotoxicity in PFOS-treated primary cortical neurons and neonatal rat brains. Exposure of cortical neurons to 1 µM PFOS for 9 days resulted in decreased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluR2 expression, which subsequently enhanced vulnerability to glutamate by increasing intracellular Ca2+ concentrations. The brain-plasma ratio of PFOS in pups was approximately five times higher than that in dams, although there were no differences in liver-plasma ratio between dams and pups. GluR2 expression in pup cerebral cortex decreased after exposure to 2.0 mg/kg PFOS, and kainic acid induced histopathological abnormalities in PFOS-exposed pups. Our findings suggest that decreased neuronal GluR2 expression is involved in PFOS-induced neurotoxicity, especially during the fetal and neonatal periods.


Subject(s)
Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Neurons/drug effects , Receptors, AMPA/metabolism , Administration, Oral , Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/pharmacokinetics , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Female , Fluorocarbons/administration & dosage , Fluorocarbons/pharmacokinetics , Kainic Acid/pharmacology , Neurons/metabolism , Neurons/pathology , Pregnancy , Rats, Wistar , Receptors, AMPA/genetics , Tissue Distribution
14.
Int J Mol Sci ; 18(8)2017 Aug 11.
Article in English | MEDLINE | ID: mdl-28800112

ABSTRACT

Tributyltin (TBT), which has been widely used as an antifouling agent in paints, is a common environmental pollutant. Although the toxicity of high-dose TBT has been extensively reported, the effects of low concentrations of TBT are relatively less well studied. We have previously reported that low-concentration TBT decreases α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)-type glutamate receptor subunit 2 (GluR2) expression in cortical neurons and enhances neuronal vulnerability to glutamate. However, the mechanism of this TBT-induced GluR2 decrease remains unknown. Therefore, we examined the effects of TBT on the activity of transcription factors that control GluR2 expression. Exposure of primary cortical neurons to 20 nM TBT for 3 h to 9 days resulted in a decrease in GluR2 mRNA expression. Moreover, TBT inhibited the DNA binding activity of nuclear respiratory factor-1 (NRF-1), a transcription factor that positively regulates the GluR2. This result indicates that TBT inhibits the activity of NRF-1 and subsequently decreases GluR2 expression. In addition, 20 nM TBT decreased the expression of genes such as cytochrome c, cytochrome c oxidase (COX) 4, and COX 6c, which are downstream of NRF-1. Our results suggest that NRF-1 inhibition is an important molecular action of the neurotoxicity induced by low-concentration TBT.


Subject(s)
Environmental Pollutants/toxicity , NF-E2-Related Factor 1/metabolism , Neurons/drug effects , Receptors, AMPA/metabolism , Trialkyltin Compounds/toxicity , Animals , Cells, Cultured , Environmental Pollutants/pharmacology , HEK293 Cells , Humans , NF-E2-Related Factor 1/genetics , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, AMPA/genetics , Trialkyltin Compounds/pharmacology
15.
J Neurochem ; 139(2): 294-308, 2016 10.
Article in English | MEDLINE | ID: mdl-27309572

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder, but its underlying cause remains unknown. Although recent studies using PD-related neurotoxin MPP+ suggest autophagy involvement in the pathogenesis of PD, the effect of MPP+ on autophagic processes under mild exposure, which mimics the slow progressive nature of PD, remains largely unclear. We examined the effect of mild MPP+ exposure (10 and 200 µM for 48 h), which induces a more slowly developing cell death, on autophagic processes and the mechanistic differences with acute MPP+ toxicity (2.5 and 5 mM for 24 h). In SH-SY5Y cells, mild MPP+ exposure predominantly inhibited autophagosome degradation, whereas acute MPP+ exposure inhibited both autophagosome degradation and basal autophagy. Mild MPP+ exposure reduced lysosomal hydrolase cathepsin D activity without changing lysosomal acidity, whereas acute exposure decreased lysosomal density. Lysosome biogenesis enhancers trehalose and rapamycin partially alleviated mild MPP+ exposure induced impaired autophagosome degradation and cell death, but did not prevent the pathogenic response to acute MPP+ exposure, suggesting irreversible lysosomal damage. We demonstrated impaired autophagic degradation by MPP+ exposure and mechanistic differences between mild and acute MPP+ toxicities. Mild MPP+ toxicity impaired autophagosome degradation through novel lysosomal acidity-independent mechanisms. Sustained mild lysosomal damage may contribute to PD. We examined the effects of MPP+ on autophagic processes under mild exposure, which mimics the slow progressive nature of Parkinson's disease, in SH-SY5Y cells. This study demonstrated impaired autophagic degradation through a reduction in lysosomal cathepsin D activity without altering lysosomal acidity by mild MPP+ exposure. Mechanistic differences between acute and mild MPP+ toxicity were also observed. Sustained mild damage of lysosome may be an underlying cause of Parkinson's disease. Cover Image for this issue: doi: 10.1111/jnc.13338.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Autophagy/drug effects , Dopamine Agents/toxicity , Lysosomes/drug effects , 1-Methyl-4-phenylpyridinium/antagonists & inhibitors , Acids , Cathepsin D/metabolism , Cell Death/drug effects , Cell Line , Humans , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Phagosomes/drug effects , Sirolimus/pharmacology , Trehalose/pharmacology
16.
Biosci Biotechnol Biochem ; 80(6): 1164-7, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26967711

ABSTRACT

Serum is an important component of cell culture media. The present study demonstrates contamination of intracellular protein extract by bovine serum albumin from the culture media and illustrates how this contamination can cause the misinterpretation of western blot results. Preliminary experiments can prevent the misinterpretation of some experimental results, and optimization of the washing process may enable specific protein detection.


Subject(s)
Artifacts , Blood Proteins/analysis , Liquid-Liquid Extraction/methods , Serum Albumin, Bovine/analysis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Complex Mixtures/chemistry , Culture Media , Electrophoresis, Gel, Two-Dimensional , Gene Expression , HEK293 Cells , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
17.
Xenobiotica ; 45(7): 605-14, 2015.
Article in English | MEDLINE | ID: mdl-25733030

ABSTRACT

1. We used chimeric mice (PXB mice®), which were repopulated with human hepatocytes, to evaluate their predictabilities of human pharmacokinetics. 2. The relationships of total clearance (CLt) and the volume of distribution at steady state (Vdss) between that predicted from single-species allometric scaling (SSS) of PXB mice and the observed human values indicated good correlations for various drugs metabolized by cytochrome P450s (CYPs) and non-CYPs. 3. We examined the Dedrick plot with which the plasma concentration-time curves can exhibit superimposability using SSS of PXB mice for CLt and Vdss. The predicted plasma concentration-time curves using the complex Dedrick plot from PXB mice were generally superimposed with the observed human data. 4. However, the predicted curve of diazepam was not superimposable with the observed profile. Residual mouse hepatocytes in the livers of PXB mice may affect predictability of CLt of diazepam because significant discrepancy of in vitro intrinsic clearance in PXB mouse liver microsomes consisted of low and high replacement of human hepatocytes were observed. 5. The complex Dedrick plot with SSS from PXB mice is useful for predicting the plasma concentration-time curve in drug discovery, although there are some limitations.


Subject(s)
Pharmaceutical Preparations/blood , Pharmaceutical Preparations/metabolism , Animals , Child, Preschool , Chimera , Humans , Liver , Male , Mice , Species Specificity , Time Factors
18.
J Neurochem ; 130(6): 826-38, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24832624

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disease, but its pathogenesis remains elusive. A mutation in ubiquitin C-terminal hydrolase L1 (UCH-L1) is responsible for a form of genetic PD which strongly resembles the idiopathic PD. We previously showed that 1-(3',4'-dihydroxybenzyl)-1,2,3,4-tetrahydroisoquinoline (3',4'DHBnTIQ) is an endogenous parkinsonism-inducing dopamine derivative. Here, we investigated the interaction between 3',4'DHBnTIQ and UCH-L1 and its possible role in the pathogenesis of idiopathic PD. Our results indicate that 3',4'DHBnTIQ binds to UCH-L1 specifically at Cys152 in vitro. In addition, 3',4'DHBnTIQ treatment increased the amount of UCH-L1 in the insoluble fraction of SH-SY5Y cells and inhibited its hydrolase activity to 60%, reducing the level of ubiquitin in the soluble fraction of SH-SY5Y cells. Catechol-modified UCH-L1 as well as insoluble UCH-L1 were detected in the midbrain of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated PD model mice. Structurally as well as functionally altered UCH-L1 have been detected in the brains of patients with idiopathic PD. We suggest that conjugation of UCH-L1 by neurotoxic endogenous compounds such as 3',4'DHBnTIQ might play a key role in onset and progression of idiopathic PD. We investigated the interaction between ubiquitin C-terminal hydrolase L1 (UCH-L1) and the brain endogenous parkinsonism inducer 1-(3',4'-dihydroxybenzyl)-1,2,3,4-tetrahydroisoquinoline (3',4'DHBnTIQ). Our results indicate that 3',4'DHBnTIQ binds to UCH-L1 specifically at cysteine 152 and induces its aggregation. 3',4'DHBnTIQ also inhibits the hydrolase activity of UCH-L1. Catechol-modified as well as insoluble UCH-L1 were detected in the midbrains of MPTP-treated Parkinson's disease (PD) model mice. Conjugation of UCH-L1 by neurotoxic endogenous compounds like 3',4'DHBnTIQ might play a key role in onset and progression of PD.


Subject(s)
Dopamine/analogs & derivatives , Dopamine/metabolism , Neurotoxins/metabolism , Parkinson Disease/metabolism , Tretoquinol/analogs & derivatives , Ubiquitin Thiolesterase/metabolism , Animals , Blotting, Western , Catechols/chemistry , Catechols/pharmacology , Cell Line, Tumor , Cell Survival , Electrophoresis, Agar Gel , Escherichia coli/metabolism , Humans , Indicators and Reagents , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tretoquinol/metabolism , Tretoquinol/pharmacology , Ubiquitin Thiolesterase/chemistry
19.
Toxicol Appl Pharmacol ; 272(1): 137-46, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23743301

ABSTRACT

Organotin compounds, especially tributyltin chloride (TBT), have been widely used in antifouling paints for marine vessels, but exhibit various toxicities in mammals. The endoplasmic reticulum (ER) is a multifunctional organelle that controls post-translational modification and intracellular Ca(2+) signaling. When the capacity of the quality control system of ER is exceeded under stress including ER Ca(2+) homeostasis disruption, ER functions are impaired and unfolded proteins are accumulated in ER lumen, which is called ER stress. Here, we examined whether TBT causes ER stress in human neuroblastoma SH-SY5Y cells. We found that 700nM TBT induced ER stress markers such as CHOP, GRP78, spliced XBP1 mRNA and phosphorylated eIF2α. TBT also decreased the cell viability both concentration- and time-dependently. Dibutyltin and monobutyltin did not induce ER stress markers. We hypothesized that TBT induces ER stress via Ca(2+) depletion, and to test this idea, we examined the effect of TBT on intracellular Ca(2+) concentration using fura-2 AM, a Ca(2+) fluorescent probe. TBT increased intracellular Ca(2+) concentration in a TBT-concentration-dependent manner, and Ca(2+) increase in 700nM TBT was mainly blocked by 50µM dantrolene, a ryanodine receptor antagonist (about 70% inhibition). Dantrolene also partially but significantly inhibited TBT-induced GRP78 expression and cell death. These results suggest that TBT increases intracellular Ca(2+) concentration by releasing Ca(2+) from ER, thereby causing ER stress.


Subject(s)
Calcium/physiology , Endoplasmic Reticulum Stress/drug effects , Trialkyltin Compounds/toxicity , Blotting, Western , Calcium Signaling/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/metabolism , Dantrolene/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Microarray Analysis , Muscle Relaxants, Central/pharmacology , Polymerase Chain Reaction , Regulatory Factor X Transcription Factors , Transcription Factors/metabolism , X-Box Binding Protein 1
20.
J Toxicol Sci ; 48(3): 161-168, 2023.
Article in English | MEDLINE | ID: mdl-36858641

ABSTRACT

Tributyltin (TBT) is an environmental chemical, which was used as an antifouling agent for ships. Although its use has been banned, it is still persistently present in ocean sediments. Although TBT reportedly causes various toxicity in mammals, few studies on the mechanisms of biological response against TBT toxicity exist. The well-established Keap1-Nrf2 pathway is activated as a cytoprotective mechanism under stressful conditions. The relationship between TBT and the Keap1-Nrf2 pathway remains unclear. In the present study, we evaluated the effect of TBT on the Keap1-Nrf2 pathway. TBT reduced Keap1 protein expression in Neuro2a cells, a mouse neuroblastoma cell line, after 6 hr without altering mRNA expression levels. TBT also promoted the nuclear translocation of Nrf2, a transcription factor for antioxidant proteins, after 12 hr and augmented the expression of heme oxygenase 1, a downstream protein of Nrf2. Furthermore, TBT decreased Keap1 levels in mouse embryonic fibroblast (MEF) cells, with the knockout of Atg5, which is essential for macroautophagy, as well as in wild-type MEF cells. These results suggest that TBT activates the Keap1-Nrf2 pathway via the reduction in the Keap1 protein level in a macroautophagy-independent manner. The Keap1-Nrf2 pathway is activated by conformational changes in Keap1 induced by reactive oxygen species or electrophiles. Furthermore, any unutilized Keap1 protein is degraded by macroautophagy. Understanding the novel mechanism governing the macroautophagy-independent reduction in Keap1 by TBT may provide insights into the unresolved biological response mechanism against TBT toxicity and the activation mechanism of the Keap1-Nrf2 pathway.


Subject(s)
Kelch-Like ECH-Associated Protein 1 , Macroautophagy , NF-E2-Related Factor 2 , Trialkyltin Compounds , Animals , Mice , Fibroblasts , Trialkyltin Compounds/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL