Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Arch Toxicol ; 96(2): 545-558, 2022 02.
Article in English | MEDLINE | ID: mdl-35001170

ABSTRACT

The functional roles of protein modification by small ubiquitin-like modifier (SUMO) proteins are not well understood compared to ubiquitination. Promyelocytic leukemia (PML) proteins are good substrates for SUMOylation, and PML-nuclear bodies (PML-NBs) may function as a platform for the PML SUMOylation. PML proteins are rapidly modified both with SUMO2/3 and SUMO1 after exposure to arsenite (As3+) and SUMOylated PML are further ubiquitinated and degraded by proteasomes. However, effects of As3+ on SUMO dynamics on PML-NBs are not well investigated. In the present study, we report that (1) the number and size of PML-NBs were regulated by SUMO E1-activating enzyme, (2) SUMO2/3 co-localized with PML irrespective of As3+ exposure and was restricted to PML-nuclear bodies (PML-NBs) via covalent binding in response to As3+, and (3) As3+-induced biochemical changes in PML were not modulated by ubiquitin-proteasome system (UPS) in THP-1 cells. Undifferentiated and differentiated THP-1 cells responded to As3+ similarly and PML proteins were changed from the detergent soluble to the insoluble form and further SUMOylated with SUMO2/3 and SUMO1. ML792, a SUMO E1 inhibitor, decreased the number of PML-NBs and reciprocally increased the size irrespective of exposure to As3+, which itself slightly decrease both the number and size of PML-NBs. TAK243, a ubiquitin E1 inhibitor, did not change the PML-NBs, while SUMOylated proteins accumulated in the TAK243-exposed cells. Proteasome inhibitors did not change the As3+-induced SUMOylation levels of PML. Co-localization and further restriction of SUMO2/3 to PML-NBs were confirmed by PML-transfected CHO-K1 cells. Collectively, SUMOylation regulates PML-NBs and As3+ restricts SUMO dynamics on PML by changing its solubility.


Subject(s)
Arsenites/pharmacology , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/metabolism , Sumoylation/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Esters/pharmacology , Humans , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Small Ubiquitin-Related Modifier Proteins/metabolism , Solubility , Sulfides/pharmacology , Sulfonamides/pharmacology , Sulfonic Acids/pharmacology , THP-1 Cells , Ubiquitins/metabolism
2.
Int J Mol Sci ; 24(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36614034

ABSTRACT

Recent epidemiological studies have reported significantly increasing hospital admission rates for mental disorders such as anxiety and depression, not only in adults but also in children and adolescents, indicating more research is needed for evaluation of the etiology and possible reduction and prevention of these disorders. The aim of the present study was to examine the associations between perinatal exposure to traffic-related air pollutants and anxiety-like behaviors and alterations in neurological and immunological markers in adulthood using a rat model. Sprague Dawley pregnant rats were exposed to clean air (control), diesel exhaust (DE) 101 ± 9 µg/m3 or diesel exhaust origin secondary organic aerosol (DE-SOA) 118 ± 23 µg/m3 from gestational day 14 to postnatal day 21. Anxiety-related behavioral tests including open field tests, elevated plus maze, light/dark transition tests and novelty-induced hypophagia were performed on 10-week-old rats. The hippocampal expression of neurotransmitters, neurotrophic factors, and inflammatory molecular markers was examined by real-time RT-PCR. Anxiety-like behaviors were observed in both male and female rat offspring exposed to DE or DE-SOA. Moreover, serotonin receptor (5HT1A), dopamine receptor (Drd2), brain-derived neurotrophic factor and vascular endothelial growth factor A mRNAs were significantly decreased, whereas interleukin-1ß, cyclooxygenase-2, heme oxygenase-1 mRNAs and microglial activation were significantly increased in both male and female rats. These findings indicate that brain developmental period exposure to traffic-related air pollutants may induce anxiety-like behaviors via modulation of neurotransmitters, neurotrophic factors, and immunological molecular markers, triggering neuroinflammation and microglia activation in rats.


Subject(s)
Air Pollutants , Vehicle Emissions , Pregnancy , Rats , Animals , Male , Female , Vehicle Emissions/toxicity , Vascular Endothelial Growth Factor A , Rats, Sprague-Dawley , Anxiety/etiology , Neurotransmitter Agents
3.
Int J Mol Sci ; 22(2)2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33430368

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social communication, poor social interactions, and repetitive behaviors. We aimed to examine autism-like behaviors and related gene expressions in rats exposed to diesel exhaust (DE)-origin secondary organic aerosol (DE-SOA) perinatally. Sprague-Dawley pregnant rats were exposed to clean air (control), DE, and DE-SOA in the exposure chamber from gestational day 14 to postnatal day 21. Behavioral phenotypes of ASD were investigated in 10~13-week-old offspring using a three-chambered social behavior test, social dominance tube test, and marble burying test. Prefrontal cortex was collected to examine molecular analyses including neurological and immunological markers and glutamate concentration, using RT-PCR and ELISA methods. DE-SOA-exposed male and female rats showed poor sociability and social novelty preference, socially dominant behavior, and increased repetitive behavior. Serotonin receptor (5-HT(5B)) and brain-derived neurotrophic factor (BDNF) mRNAs were downregulated whereas interleukin 1 ß (IL-ß) and heme oxygenase 1 (HO-1) mRNAs were upregulated in the prefrontal cortex of male and female rats exposed to DE-SOA. Glutamate concentration was also increased significantly in DE-SOA-exposed male and female rats. Our results indicate that perinatal exposure to DE-SOA may induce autism-like behavior by modulating molecules such as neurological and immunological markers in rats.


Subject(s)
Air Pollutants/toxicity , Autism Spectrum Disorder/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Vehicle Emissions/toxicity , Aerosols/toxicity , Animals , Autism Spectrum Disorder/chemically induced , Brain-Derived Neurotrophic Factor/genetics , Female , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Humans , Interleukin-1beta/genetics , Male , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Rats , Rats, Sprague-Dawley , Receptors, Serotonin/genetics
4.
Arch Toxicol ; 94(8): 2587-2601, 2020 08.
Article in English | MEDLINE | ID: mdl-32435915

ABSTRACT

Arsenic is a well-known environmental carcinogen and chronic exposure to arsenic through drinking water has been reported to cause skin, bladder and lung cancers, with arsenic metabolites being implicated in the pathogenesis. In contrast, arsenic trioxide (As2O3) is an effective therapeutic agent for the treatment of acute promyelocytic leukemia, in which the binding of arsenite (iAsIII) to promyelocytic leukemia (PML) protein is the proposed initial step. These findings on the two-edged sword characteristics of arsenic suggest that after entry into cells, arsenic reaches the nucleus and triggers various nuclear events. Arsenic is reduced, conjugated with glutathione, and methylated in the cytosol. These biotransformations, including the production of reactive metabolic intermediates, appear to determine the intracellular dynamics, target organs, and biological functions of arsenic.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenic Poisoning/etiology , Arsenic Trioxide/pharmacology , Arsenicals/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Animals , Antineoplastic Agents/metabolism , Arsenic Poisoning/metabolism , Arsenic Trioxide/metabolism , Arsenicals/metabolism , Biotransformation , Humans , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Risk Assessment , Toxicity Tests
5.
Toxicol Appl Pharmacol ; 360: 150-159, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30292834

ABSTRACT

Promyelocytic leukemia (PML) and a suite of other proteins form nuclear bodies (NBs) where SUMOylation of PML and tumor suppression events occur in response to arsenite (As3+) treatment. Soluble PML is rapidly modified to the insoluble form in response to As3+, yet the relationship between the solubility change and nuclear localization of PML and PML-nuclear body (PML-NB) proteins remained elusive. We have investigated differences in the solubility change of well-known PML-NB proteins such as death-associated protein 6 (DAXX), SUMO, and PML in genetically engineered HEK293, and Jurkat and HL60 cells. The solubility of PML and SUMO2/3 monomers in RIPA solution decreased in 2 h in response to As3+. Live image analysis of GFP-PML revealed that extranuclear PML was insoluble in RIPA irrespective of the As3+-treatment and PML in PML-NBs, which was soluble in the untreated cells, was converted to insoluble forms by As3+. The solubility of DAXX was not changed by As3+, even though PML and DAXX co-localized completely in the subcellular compartments. Murine double mutant 2 (MDM2), which is known to interacts with intranuclear PML, did not affect the As3+-induced solubility change of PML. These results indicate that As3+ selectively reorganizes PML and SUMO2/3 monomers into insoluble forms in PML-NBs, and then PML SUMOylation proceeds.


Subject(s)
Arsenites/pharmacology , Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/metabolism , SUMO-1 Protein/metabolism , Cell Line , Cell Line, Tumor , Cell Nucleus/drug effects , HEK293 Cells , HL-60 Cells , Humans , Jurkat Cells , Leukemia, Promyelocytic, Acute/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Solubility , Sumoylation/drug effects , Tumor Suppressor Proteins/metabolism
6.
J Appl Toxicol ; 37(4): 471-478, 2017 04.
Article in English | MEDLINE | ID: mdl-27624343

ABSTRACT

Silver is commonly used as a disinfectant, and chronic exposure to silver may cause argyria, resulting in a gray-blue discoloration of human skin. However, the mechanism for cellular toxicity of silver has not been well explained. We studied the mode of cell death, the ratio of glutathione disulfide/glutathione, induction of metallothionein and activation of mitogen-activated protein kinases in J774.1 cells together with activation of antioxidant responsive element and nuclear factor-κB in CHO cells following exposure to silver ion (Ag+ ) to investigate the mechanism by which Ag+ causes lethal effects. Ag+ increased phosphorylation levels of extracellular signal-regulated, c-Jun N-terminal and p38 mitogen-activated protein kinases and remarkably increased the ratio of glutathione disulfide/glutathione in both a time- and concentration-dependent manner. Luciferase reporter gene assays revealed that antioxidant responsive element and nuclear factor-κB were activated following exposure to Ag+ . In addition, exposure to Ag+ increased the mRNA and protein levels of metallothionein. We investigated whether or not Ag+ killed J774.1 cells by inducing apoptosis. Ag+ increased the activity of caspase-3/7 which was abrogated by caspase 3 and pan-caspase inhibitors. However, these inhibitors did not ameliorate the cytotoxic effects of Ag+ , suggesting that Ag+ causes oxidative stress, which leads to necrotic rather than apoptotic cell death in J774.1 cells by decreasing functional sulfhydryl groups including glutathione in the cells. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Lung/cytology , Macrophages/drug effects , Oxidative Stress/drug effects , Silver/toxicity , Animals , Apoptosis/drug effects , CHO Cells , Caspase Inhibitors/pharmacology , Caspases/biosynthesis , Caspases/genetics , Cell Line , Cell Survival/drug effects , Cricetulus , Genes, Reporter/drug effects , Glutathione/metabolism , Lung/drug effects , Metallothionein/metabolism , Mice , Necrosis , Silver/chemistry
7.
Toxicol Appl Pharmacol ; 287(3): 191-201, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26049103

ABSTRACT

Promyelocytic leukemia (PML), which is a tumor suppressor protein that nevertheless plays an important role in the maintenance of leukemia initiating cells, is known to be biochemically modified by As(3+). We recently developed a simple method to evaluate the modification of PML by As(3+) resulting in a change in solubility and the covalent binding of small ubiquitin-like modifier (SUMO). Here we semi-quantitatively investigated the SUMOylation of PML using HEK293 cells which were stably transfected with PML-VI (HEK-PML). Western blot analyses indicated that PML became insoluble in cold RadioImmunoPrecipitation Assay (RIPA) lysis buffer and was SUMOylated by both SUMO2/3 and SUMO1 by As(3+). Surprisingly SUMO1 monomers were completely utilized for the SUMOylation of PML. Antimony (Sb(3+)) but not bismuth (Bi(3+)), Cu(2+), or Cd(2+) biochemically modified PML similarly. SUMOylated PML decreased after removal of As(3+) from the culture medium. However, unSUMOylated PML was still recovered in the RIPA-insoluble fraction, suggesting that SUMOylation is not requisite for changing the RIPA-soluble PML into the RIPA-insoluble form. Immunofluorescence staining of As(3+)-exposed cells indicated that SUMO2/3 was co-localized with PML in the nuclear bodies. However, some PML protein was present in peri-nuclear regions without SUMO2/3. Functional Really Interesting New Gene (RING)-deleted mutant PML neither formed PML nuclear bodies nor was biochemically modified by As(3+). Conjugation with intracellular glutathione may explain the accessibility of As(3+) and Sb(3+) to PML in the nuclear region evading chelation and entrapping by cytoplasmic proteins such as metallothioneins.


Subject(s)
Arsenites/toxicity , Nuclear Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Sodium Compounds/toxicity , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Antimony/toxicity , Chlorides/toxicity , Glutathione/metabolism , HEK293 Cells , Humans , Nuclear Proteins/chemistry , Promyelocytic Leukemia Protein , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Solubility , Sumoylation , Time Factors , Transcription Factors/chemistry , Transfection , Tumor Suppressor Proteins/chemistry , Ubiquitins/metabolism
8.
Arch Toxicol ; 89(1): 73-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24760326

ABSTRACT

Long fibers, such as asbestos and carbon nanotubes (CNTs), are more potent activators of inflammatory and genotoxicity than short or tangled fibers. Fibrous particles trigger interleukin (IL)-1ß secretion and cause inflammatory diseases through NLRP3 inflammasomes in phagocytotic cells. However, the mechanism involved in fibrous particle-induced inflammation has not been well documented. In this study, we focused on GTPase effector Rho-kinases (ROCK1, and 2), which are known to be involved in a wide range of cellular functions such as adhesion, regulation of cytoskeleton, and phagocytosis. We examined whether ROCKs are associated with multi-walled CNT (MWCNT)- or asbestos-induced IL-1ß secretion in human monocytic THP-1 cells using a selective inhibitor and small interfering RNA. THP-1 cells were differentiated to macrophages by PMA and were exposed to MWCNTs, crocidolite asbestos or lipopolysaccharide (LPS) in the presence or absence of Y27632 (ROCK inhibitor) or Z-YVAD (caspase-1 inhibitor). Exposure of the cells to MWCNTs or asbestos provoked IL-1ß secretion, but this secretion was suppressed by both Y27632 and Z-YVAD, whereas LPS-induced IL-1ß secretion was inhibited only by Z-YVAD and not by Y27632. siRNA designed for knockdown of both ROCK1 and ROCK2 suppressed MWCNT- and asbestos-induced IL-1ß secretion, but did not change LPS-induced IL-1ß secretion. Moreover, Y27632 suppressed pro-IL-1ß protein levels and the release of activated-cathepsin B and activated-caspase-1 induced by MWCNTs or asbestos. In contrast, LPS-induced pro-IL-1ß protein was not suppressed by Y27632. These results suggest that ROCKs are involved in fibrous particle-induced inflammasome responses in THP-1 cells.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Monocytes/immunology , Phagocytosis/immunology , rho-Associated Kinases/metabolism , Asbestos/toxicity , Cell Culture Techniques , Cell Line , Humans , Interleukin-1beta/immunology , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein , Nanotubes, Carbon/toxicity
9.
Arch Toxicol ; 88(1): 145-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23963510

ABSTRACT

Although carbon nanotubes (CNTs) are promising nanomaterials, their potential carcinogenicity is a major concern. We previously established a genetic method of analyzing genotoxicity of chemical compounds, where we evaluated their cytotoxic effect on the DT40 lymphoid cell line comparing DNA-repair-deficient isogenic clones with parental wild-type cells. However, application of our DT40 system for the cytotoxic and genotoxic evaluation of nanomaterials seemed to be difficult, because DT40 cells only poorly internalized nanoparticles. To solve this problem, we have constructed a chimeric gene encoding a trans-membrane receptor consisting of the 5' region of the transferrin receptor (TR) gene (to facilitate internalization of nanoparticles) and the 3' region of the macrophage receptor with collagenous structure (MARCO) gene (which is a receptor for environmental particles). We expressed the resulting MARCO-TR chimeric receptor on DNA-repair-proficient wild-type cells and mutants deficient in base excision repair (FEN1 (-/-)) and translesion DNA synthesis (REV3 (-/-)). We demonstrated that the chimera mediates uptake of particles such as fluorescence-tagged polystyrene particles and multi-walled carbon nanotubes (MWCNTs), with very poor uptake of those particles by DT40 cells not expressing the chimera. MWCNTs were cytotoxic and this effect was greater in FEN1 (-/-)and REV3 (-/-) cells than in wild-type cells. Furthermore, MWCNTs induced greater oxidative damage (measured as 8-OH-dG formation) and a larger number of mitotic chromosomal aberrations in repair-deficient cells compared to repair-proficient cells. Taken together, our novel assay system using the chimeric receptor-expressing DT40 cells provides a sensitive method to screen for genotoxicity of CNTs and possibly other nanomaterials.


Subject(s)
B-Lymphocytes/drug effects , Mutagenicity Tests/methods , Nanotubes, Carbon/toxicity , Receptors, Immunologic/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Cell Line/drug effects , Chickens , Chromosome Aberrations , DNA Repair/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Polystyrenes/pharmacokinetics , Receptors, Immunologic/genetics , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
10.
Toxicol Appl Pharmacol ; 273(3): 590-9, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24135626

ABSTRACT

Inorganic arsenite (iAs(3+)) is a two-edged sword. iAs(3+) is a well-known human carcinogen; nevertheless, it has been used as a therapeutic drug for acute promyelocytic leukemia (APL), which is caused by a fusion protein comprising retinoic acid receptor-α and promyelocytic leukemia (PML). PML, a nuclear transcription factor, has a RING finger domain with densely positioned cysteine residues. To examine PML-modulated cellular responses to iAs(3+), CHO-K1 and HEK293 cells were each used to establish cell lines that expressed ectopic human PML. Overexpression of PML increased susceptibility to iAs(3+) in CHO-K1 cells, but not in HEK293 cells. Exposure of PML-transfected cells to iAs(3+) caused PML to change from a soluble form to less soluble forms, and this modification of PML was observable even with just 0.1 µM iAs(3+) (7.5 ppb). Western blot and immunofluorescent microscopic analyses revealed that the biochemical changes of PML were caused at least in part by conjugation with small ubiquitin-like modifier proteins (SUMOylation). A luciferase reporter gene was used to investigate whether modification of PML was caused by oxidative stress or activation of antioxidant response element (ARE) in CHO-K1 cells. Modification of PML protein occurred faster than activation of the ARE in response to iAs(3+), suggesting that PML was not modified as a consequence of oxidative stress-induced ARE activation.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenites/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Animals , Antioxidant Response Elements , CHO Cells , Cricetulus , HEK293 Cells , Humans , Leukemia, Promyelocytic, Acute/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oxidative Stress/drug effects , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
J Pathol ; 228(3): 366-77, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22864872

ABSTRACT

Exposure to asbestos is a risk for malignant mesothelioma (MM) in humans. Among the commercially used types of asbestos (chrysotile, crocidolite, and amosite), the carcinogenicity of chrysotile is not fully appreciated. Here, we show that all three asbestos types similarly induced MM in the rat peritoneal cavity and that chrysotile caused the earliest mesothelioma development with a high fraction of sarcomatoid histology. The pathogenesis of chrysotile-induced mesothelial carcinogenesis was closely associated with iron overload: repeated administration of an iron chelator, nitrilotriacetic acid, which promotes the Fenton reaction, significantly reduced the period required for carcinogenesis; massive iron deposition was found in the peritoneal organs with high serum ferritin; and homozygous deletion of the CDKN2A/2B/ARF tumour suppressor genes, the most frequent genomic alteration in human MM and in iron-induced rodent carcinogenesis, was observed in 92.6% of the cases studied with array-based comparative genomic hybridization. The induced rat MM cells revealed high expression of mesoderm-specific transcription factors, Dlx5 and Hand1, and showed an iron regulatory profile of active iron uptake and utilization. These data indicate that chrysotile is a strong carcinogen when exposed to mesothelia, acting through the induction of local iron overload. Therefore, an intervention to remove local excess iron might be a strategy to prevent MM after asbestos exposure.


Subject(s)
Asbestos, Serpentine/adverse effects , Biomarkers, Tumor/metabolism , Iron Overload/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Mesothelioma/chemically induced , Mesothelioma/metabolism , Peritoneal Neoplasms/chemically induced , Peritoneal Neoplasms/metabolism , Animals , Asbestos, Serpentine/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Transformation, Neoplastic/drug effects , DNA Copy Number Variations/drug effects , DNA, Neoplasm/drug effects , Disease Models, Animal , Homeodomain Proteins/metabolism , Iron/metabolism , Lung Neoplasms/pathology , Male , Mesothelioma/pathology , Mesothelioma, Malignant , Nitrilotriacetic Acid/pharmacology , Peritoneal Neoplasms/pathology , Rats , Rats, Inbred BN , Rats, Inbred F344 , Signal Transduction/drug effects , Transcription Factors/metabolism
12.
Arch Toxicol ; 87(6): 969-79, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22811022

ABSTRACT

Arsenic is a worldwide environmental pollutant and a human carcinogen. It is well recognized that the toxicity of arsenicals largely depends on the oxidoreduction states (trivalent or pentavalent) and methylation levels (monomethyl, dimethyl, and trimethyl) that are present during the process of metabolism in mammals. However, presently, the specifics of the metabolic pathway of inorganic arsenicals have yet to be confirmed. In mammals, there are two possible mechanisms that have been proposed for the metabolic pathway of inorganic arsenicals, oxidative methylation, and glutathione conjugation. Oxidative methylation, which was originally proposed in fungi, is based on findings that arsenite (iAs(III)) is sequentially converted to monomethylarsonic acid (MMA(V)) and dimethylarsinic acid (DMA(V)) in both humans and in laboratory animals such as mice and rats. However, recent in vitro observations have demonstrated that arsenic is only methylated in the presence of glutathione (GSH) or other thiol compounds, which strongly suggests that arsenic is methylated in trivalent forms. The glutathione conjugation mechanism is supported by findings that have shown that most intracellular arsenicals are trivalent and excreted from cells as GSH conjugates. Since non-conjugated trivalent arsenicals are highly reactive with thiol compounds and are easily converted to less toxic corresponding pentavalent arsenicals, the arsenic-glutathione conjugate stability may be the most important factor for determining the toxicity of arsenicals. In addition, "being a non-anionic form" also appears to be a determinant of the toxicity of oxo-arsenicals or thioarsenicals. The present review discusses both the metabolism of arsenic and the toxicity of arsenic metabolites.


Subject(s)
Arsenic Poisoning/etiology , Arsenic Poisoning/metabolism , Arsenicals/adverse effects , Arsenicals/metabolism , Environmental Pollutants/adverse effects , Environmental Pollutants/metabolism , Animals , Biological Transport , Biotransformation , Glutathione/metabolism , Humans , Intestinal Absorption , Methylation , Oxidation-Reduction , Risk Assessment , Tissue Distribution
13.
Arch Toxicol ; 87(7): 1245-56, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23479079

ABSTRACT

Hydrogen sulfide (H2S) is a toxic gaseous substance, and accidental exposure to high concentrations of H2S has been reported to be lethal to humans. Inhaled and absorbed H2S is partially dissolved within the circulation and causes toxic effects on lymphocytes. However, the mechanisms involved in H2S toxicity have not been well documented. In this study, we examined the cellular uptake and injury of sulfide-exposed human T lymphocytes (Jurkat). Cells were exposed to a H2S donor, sodium hydroxysulfide (NaHS), at pH 6.0, 7.0, or 8.0 for 1 h at 37 °C in a sealed conical tube to avoid the loss of dissolved H2S gas. Cytotoxicity and cellular sulfide concentrations increased dramatically as the pH of the NaHS solution decreased. Sulfide enhanced the cleavage of caspase-3 and poly (ADP-ribose) polymerase and induced early cellular apoptosis. A pan-caspase inhibitor reduced sulfide-induced apoptosis. These results indicate that sulfide induces pH-dependent and caspase-dependent apoptosis. We also found that blebbing of the plasma membrane occurred in sulfide-exposed cells. Both ROCK-1 and ROCK-2 (Rho kinases) were activated by sulfide, and sulfide-induced cell blebbing was suppressed by a ROCK inhibitor, suggesting that a Rho pathway is involved in sulfide-induced blebbing in lymphocytes.


Subject(s)
Apoptosis/drug effects , Cell Membrane/drug effects , Hydrogen Sulfide/toxicity , Sulfides/toxicity , T-Lymphocytes/drug effects , rho-Associated Kinases/metabolism , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Membrane/enzymology , Cell Membrane/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Hydrogen Sulfide/metabolism , Hydrogen-Ion Concentration , Jurkat Cells , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sulfides/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/pathology , rho-Associated Kinases/antagonists & inhibitors
14.
J Toxicol Sci ; 48(2): 75-86, 2023.
Article in English | MEDLINE | ID: mdl-36725023

ABSTRACT

Quaternary ammonium compounds, including benzalkonium chloride (BAC) and cetylpyridinium chloride (CPC), are widely used as disinfectants. Increased use of inhalable products containing BAC or CPC has raised concerns for lung toxicity. This study sought to elucidate the microstructure of plasma membrane damage caused by BAC and CPC and the subsequent mechanism by which the damage is mediated, as assessed using two human pulmonary epithelial cell lines (A549 and BEAS-2B). Scanning electron microscopic observation showed that exposure to BAC or CPC for 3 hr reduced the length and density of microvilli on the plasma membrane in A549 cells. Analysis of cell cycle distribution following plasma membrane damage revealed that BAC and CPC promote G0/G1 cell cycle arrest in both cell lines. The protein levels of Cdc6, an essential regulator of DNA replication during G1/S transition, are decreased significantly and dose dependently by BAC or CPC exposure. CPC and BAC decreased the Cdc6 levels that had been increased by a PI3K agonist in A549 cells, and levels of phosphorylated AKT were reduced in response to BAC or CPC. Conversely, exposure to equivalent concentrations of pyridinium chloride (lacking a hydrocarbon tail) induce no changes. These results suggest that plasma membrane damage triggered by BAC or CPC causes Cdc6-dependent G0/G1 cell cycle arrest in pulmonary cells. These effects are attributable to the long alkyl chains of BAC and CPC. The reduction of Cdc6 following plasma membrane damage may be caused, at least in part, by diminished signaling via the PI3K/AKT pathway.


Subject(s)
Benzalkonium Compounds , Cetylpyridinium , Humans , Benzalkonium Compounds/toxicity , Cetylpyridinium/toxicity , Cetylpyridinium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Lung , Epithelial Cells , Cell Cycle Checkpoints , Cell Membrane , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacology , Cell Cycle Proteins/metabolism
15.
Toxicol Appl Pharmacol ; 259(1): 96-103, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22209804

ABSTRACT

The toxicity of carbon nanotubes (CNTs), a highly promising nanomaterial, is similar to that of asbestos because both types of particles have a fibrous shape and are biopersistent. Here, we investigated the characteristics of macrophage receptor with collagenous structure (MARCO), a membrane receptor expressed on macrophages that recognizes environmental or unopsonized particles, and we assessed whether and how MARCO was involved in cellular uptake of multi-walled CNTs (MWCNTs). MARCO-transfected Chinese hamster ovary (CHO-K1) cells took up polystyrene beads irrespective of the particle size (20nm-1µm). In the culture of MARCO-transfected CHO-K1 cells dendritic structures were observed on the bottom of culture dishes, and the edges of these dendritic structures were continually renewed as the cell body migrated along the dendritic structures. MWCNTs were first tethered to the dendritic structures and then taken up by the cell body. MWCNTs appeared to be taken up via membrane ruffling like macropinocytosis, rather than phagocytosis. The cytotoxic EC(50) value of MWCNTs in MARCO-transfected CHO-K1 cells was calculated to be 6.1µg/mL and transmission electron microscopic observation indicated that the toxicity of MWCNTs may be due to the incomplete inclusion of MWCNTs by the membrane structure.


Subject(s)
Cell Membrane , Nanotubes, Carbon/toxicity , Receptors, Immunologic/metabolism , Actins/metabolism , Animals , Blotting, Western , CHO Cells , Cell Adhesion , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Survival/drug effects , Cricetinae , Cricetulus , Green Fluorescent Proteins/genetics , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nanotubes, Carbon/chemistry , Phagocytosis , Plasmids , Protein Binding , Receptors, Immunologic/genetics , Transfection
16.
Toxicol Appl Pharmacol ; 262(3): 355-62, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22659509

ABSTRACT

Recently, our laboratory reported that exposure to nanoparticle-rich diesel exhaust (NRDE) for 3 months impaired hippocampus-dependent spatial learning ability and up-regulated the expressions of memory function-related genes in the hippocampus of female mice. However, whether NRDE affects the hippocampus-dependent non-spatial learning ability and the mechanism of NRDE-induced neurotoxicity was unknown. Female BALB/c mice were exposed to clean air, middle-dose NRDE (M-NRDE, 47 µg/m(3)), high-dose NRDE (H-NRDE, 129 µg/m(3)), or filtered H-NRDE (F-DE) for 3 months. We then investigated the effect of NRDE exposure on non-spatial learning ability and the expression of genes related to glutamate neurotransmission using a novel object recognition test and a real-time RT-PCR analysis, respectively. We also examined microglia marker Iba1 immunoreactivity in the hippocampus using immunohistochemical analyses. Mice exposed to H-NRDE or F-DE could not discriminate between familiar and novel objects. The control and M-NRDE-exposed groups showed a significantly increased discrimination index, compared to the H-NRDE-exposed group. Although no significant changes in the expression levels of the NMDA receptor subunits were observed, the expression of glutamate transporter EAAT4 was decreased and that of glutamic acid decarboxylase GAD65 was increased in the hippocampus of H-NRDE-exposed mice, compared with the expression levels in control mice. We also found that microglia activation was prominent in the hippocampal area of the H-NRDE-exposed mice, compared with the other groups. These results indicated that exposure to NRDE for 3 months impaired the novel object recognition ability. The present study suggests that genes related to glutamate metabolism may be involved in the NRDE-induced neurotoxicity observed in the present mouse model.


Subject(s)
Nanoparticles/toxicity , Recognition, Psychology/drug effects , Vehicle Emissions/toxicity , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 4/analysis , Female , Gene Expression/drug effects , Glutamate Decarboxylase/analysis , Hippocampus/chemistry , Hippocampus/drug effects , Inhalation Exposure/adverse effects , Learning/drug effects , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction
17.
Nihon Eiseigaku Zasshi ; 67(3): 375-82, 2012 May.
Article in Japanese | MEDLINE | ID: mdl-22781011

ABSTRACT

Among the symposia organized by various study groups in the Japanese Society of Hygiene (JSH), the Study Group on Fibrous and Particulate Substances (SGFPS) presented a symposium entitled "The frontline of nanoparticle research" chaired by Professor Takemi Otsuki (Kawasaki Medical School, Japan) and Dr. Seishiro Hirano (National Institute for Environmental Studies, Japan) on 26 March, 2012, as a part of the program of the 82nd Annual Meeting of JSH in Kyoto, Japan. Special features consist of three presentations given at the above-mentioned symposium. In this article, we introduce the progress of the Study Group on Fibrous and Particulate Substances (SGFPS) from the initial symposium entitled "Asbestos: Science and Society" held at the 76th Annual Meeting of JSH at Ube, Japan to the last above-mentioned symposium in Kyoto. The health-related issues caused by exposure to fibrous materials such as asbestos and also particulated substances such as nanoparticles will be lasting in the future and researchers including our study group have to make their best efforts to resolve these problems and to reduce health impairments due to exposure to environmental fibrous and particulated substances.


Subject(s)
Asbestos/toxicity , Nanoparticles/toxicity , Humans , Mesothelioma/chemically induced
18.
Nihon Eiseigaku Zasshi ; 67(3): 383-9, 2012 May.
Article in Japanese | MEDLINE | ID: mdl-22781012

ABSTRACT

Silver (Ag) possesses a well-known antibacterial activity and has been used for medical treatment and cosmetics such as wound dressing and deodorant powders. Occupational Safety and Health Administration (OSHA) and Mine Safety and Health Administration (MSHA) proposed that the permissible exposure limit (PEL) for both metallic and most soluble Ag compounds should be 0.01 mg/m3. Argyria and argyrosis are known to be caused by deposition of insoluble Ag in the dermis and cornea/conjunctiva. However, the metabolic behavior and biological roles of Ag have not been well characterized in mammals. Ag can be absorbed into the systemic circulation from drinking water, and also through parenteral routes such as inhalation and dermal exposure. Experimental studies have demonstrated that Ag+ induces and binds to metallothionein I and II (MTs), which are cysteine-rich proteins, in cells. MTs are major cytoplasmic metal binding proteins and thereby reduce cellular damage caused by toxic heavy metals including Ag. Profiles of Ag distribution in MTs and other Ag-binding proteins can be determined using high performance liquid chromatography-inductively coupled plasma mass spectrometry (HPLC-ICP-MS). This technique directly provides information on the intracellular behavior of Ag, which is important for elucidating the mechanism underlying Ag toxicity. Silver nanoparticles (AgNPs) are also commercially used mainly as antimicrobial agents. Despite the widespread use of AgNPs, relatively few studies have been undertaken to evaluate the health effects of AgNP exposure. In the present paper, we discuss the absorption, toxicodynamics, and metabolism of both Ag and AgNPs in mammals and their health effects.


Subject(s)
Environmental Exposure , Silver/toxicity , Animals , Humans , Metallothionein/analysis , Nanoparticles/toxicity , Silver/pharmacokinetics
19.
PLoS One ; 17(5): e0268835, 2022.
Article in English | MEDLINE | ID: mdl-35594310

ABSTRACT

Promyelocytic leukemia (PML) proteins are involved in the pathogenesis of acute promyelocytic leukemia (APL). Trivalent arsenic (As3+) is known to cure APL by binding to cysteine residues of PML and enhance the degradation of PML-retinoic acid receptor α (RARα), a t(15;17) gene translocation product in APL cells, and restore PML-nuclear bodies (NBs). The size, number, and shape of PML-NBs vary among cell types and during cell division. However, topological changes of PML-NBs in As3+-exposed cells have not been well-documented. We report that As3+-induced solubility shift underlies rapid SUMOylation of PML and late agglomeration of PML-NBs. Most PML-NBs were toroidal and granular dot-like in GFPPML-transduced CHO-K1 and HEK293 cells, respectively. Exposure to As3+ and antimony (Sb3+) greatly reduced the solubility of PML and enhanced SUMOylation within 2 h in the absence of changes in the number and size of PML-NBs. However, the prolonged exposure to As3+ and Sb3+ resulted in agglomeration of PML-NBs. Exposure to bismuth (Bi3+), another Group 15 element, did not induce any of these changes. ML792, a SUMO activation inhibitor, reduced the number of PML-NBs and increased the size of the NBs, but had little effect on the As3+-induced solubility change of PML. These results warrant the importance of As3+- or Sb3+-induced solubility shift of PML for the regulation intranuclear dynamics of PML-NBs.


Subject(s)
Arsenic , Leukemia, Promyelocytic, Acute , Nuclear Bodies , Arsenic/metabolism , Arsenic/pharmacology , Cell Nucleus/metabolism , HEK293 Cells , Humans , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Bodies/chemistry , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Solubility , Transcription Factors/metabolism
20.
Biol Open ; 11(6)2022 06 15.
Article in English | MEDLINE | ID: mdl-35579421

ABSTRACT

Promyelocytic leukemia (PML) nuclear bodies (PML-NBs), a class of membrane-less cellular organelles, participate in various biological activities. PML-NBs are known as the core-shell-type nuclear body, harboring 'client' proteins in their core. Although multiple membrane-less organelles work in the oocyte nucleus, PML-NBs have been predicted to be absent from oocytes. Here, we show that some well-known PML clients (but not endogenous PML) co-localized with small ubiquitin-related modifier (SUMO) protein in the nucleolus and peri-centromeric heterochromatin of maturing oocytes. In oocytes devoid of PML-NBs, endogenous PML protein localized in the vicinity of chromatin. During and after meiotic resumption, PML co-localized with SUMO gathering around chromosomes. To examine the benefit of the PML-NB-free intranuclear milieu in oocytes, we deliberately assembled PML-NBs by microinjecting human PML-encoding plasmids into oocytes. Under conditions of limited SUMO availability, assembled PML-NBs tended to cluster. Upon proteotoxic stress, SUMO delocalized from peri-centromeric heterochromatin and co-localized with SC35 (a marker of nuclear speckles)-positive large compartments, which was disturbed by pre-assembled PML-NBs. These observations suggest that the PML-NB-free intranuclear environment helps reserve SUMO for emergent responses by redirecting the flux of SUMO otherwise needed to maintain PML-NB dynamics.


Subject(s)
Heterochromatin , Leukemia , Animals , Heterochromatin/genetics , Humans , Mice , Nuclear Bodies , Oocytes/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL