Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 326
Filter
1.
Cell ; 180(2): 296-310.e18, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31978346

ABSTRACT

Mitochondria and lysosomes are functionally linked, and their interdependent decline is a hallmark of aging and disease. Despite the long-standing connection between these organelles, the function(s) of lysosomes required to sustain mitochondrial health remains unclear. Here, working in yeast, we show that the lysosome-like vacuole maintains mitochondrial respiration by spatially compartmentalizing amino acids. Defects in vacuole function result in a breakdown in intracellular amino acid homeostasis, which drives age-related mitochondrial decline. Among amino acids, we find that cysteine is most toxic for mitochondria and show that elevated non-vacuolar cysteine impairs mitochondrial respiration by limiting intracellular iron availability through an oxidant-based mechanism. Cysteine depletion or iron supplementation restores mitochondrial health in vacuole-impaired cells and prevents mitochondrial decline during aging. These results demonstrate that cysteine toxicity is a major driver of age-related mitochondrial deterioration and identify vacuolar amino acid compartmentation as a cellular strategy to minimize amino acid toxicity.


Subject(s)
Cysteine/toxicity , Iron/metabolism , Mitochondria/metabolism , Amino Acids/metabolism , Cellular Senescence/physiology , Cysteine/metabolism , Homeostasis , Lysosomes/metabolism , Mitochondria/physiology , Mitophagy/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuoles/metabolism
2.
Arch Toxicol ; 97(2): 523-545, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36576512

ABSTRACT

Environmental or occupational exposure of humans to trichloroethylene (TCE) has been associated with different extrahepatic toxic effects, including nephrotoxicity and neurotoxicity. Bioactivation of TCE via the glutathione (GSH) conjugation pathway has been proposed as underlying mechanism, although only few mechanistic studies have used cell models of human origin. In this study, six human derived cell models were evaluated as in vitro models representing potential target tissues of TCE-conjugates: RPTEC/TERT1 (kidney), HepaRG (liver), HUVEC/TERT2 (vascular endothelial), LUHMES (neuronal, dopaminergic), human induced pluripotent stem cells (hiPSC) derived peripheral neurons (UKN5) and hiPSC-derived differentiated brain cortical cultures containing all subtypes of neurons and astrocytes (BCC42). A high throughput transcriptomic screening, utilizing mRNA templated oligo-sequencing (TempO-Seq), was used to study transcriptomic effects after exposure to TCE-conjugates. Cells were exposed to a wide range of concentrations of S-(1,2-trans-dichlorovinyl)glutathione (1,2-DCVG), S-(1,2-trans-dichlorovinyl)-L-cysteine (1,2-DCVC), S-(2,2-dichlorovinyl)glutathione (2,2-DCVG), and S-(2,2-dichlorovinyl)-L-cysteine (2,2-DCVC). 1,2-DCVC caused stress responses belonging to the Nrf2 pathway and Unfolded protein response in all the tested models but to different extents. The renal model was the most sensitive model to both 1,2-DCVC and 1,2-DCVG, with an early Nrf2-response at 3 µM and hundreds of differentially expressed genes at higher concentrations. Exposure to 2,2-DCVG and 2,2-DCVC also resulted in the upregulation of Nrf2 pathway genes in RPTEC/TERT1 although at higher concentrations. Of the three neuronal models, both the LUHMES and BCC42 showed significant Nrf2-responses and at higher concentration UPR-responses, supporting recent hypotheses that 1,2-DCVC may be involved in neurotoxic effects of TCE. The cell models with the highest expression of γ-glutamyltransferase (GGT) enzymes, showed cellular responses to both 1,2-DCVG and 1,2-DCVC. Little to no effects were found in the neuronal models from 1,2-DCVG exposure due to their low GGT-expression. This study expands our knowledge on tissue specificity of TCE S-conjugates and emphasizes the value of human cell models together with transcriptomics for such mechanistic studies.


Subject(s)
Induced Pluripotent Stem Cells , Trichloroethylene , Humans , Cysteine/toxicity , Cysteine/metabolism , Trichloroethylene/toxicity , Trichloroethylene/metabolism , Transcriptome , NF-E2-Related Factor 2/metabolism , Induced Pluripotent Stem Cells/metabolism , Glutathione/metabolism , Phenotype
3.
Chem Res Toxicol ; 35(2): 293-302, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35076219

ABSTRACT

Emodin (EMD) is a major ingredient of Polygonum multiflorum Thunb. (PMT), which has shown adverse liver reactions. Despite multiple pharmacological activities, EMD is reported to show various toxicities. Our early study demonstrated the reactivity of EMD to glutathione. This study aimed to determine the covalent interaction of hepatic protein with EMD and the correlation of the protein modification with hepatotoxicity induced by EMD. EMD-derived protein adduction was detected in an incubation mixture containing mouse liver homogenates and EMD. Such protein adduction was also observed in hepatic protein obtained from mice exposed to EMD. The protein covalent binding occurred in time- and dose-dependent manners. Pre-treatment of l-buthionine-sulfoximine significantly potentiated EMD-induced adduction and hepatotoxicity caused by EMD and lipopolysaccharide co-treatment. As expected, EMD-derived protein modification was observed in mouse primary hepatocytes treated with EMD. The increase in EMD exposure concentration intensified EMD-derived protein adduction and increased EMD-induced cell death. The susceptibility of hepatocytes to EMD cytotoxicity and the intensity of EMD-induced protein adduction were attenuated by the co-treatment of hepatocytes with N-acetyl cysteine. A good association of protein modification with hepatotoxicity induced by EMD was illustrated, which facilitates the understanding of the mechanism of hepatotoxicity induced by EMD.


Subject(s)
Cysteine/toxicity , Emodin/toxicity , Hepatocytes/drug effects , Proteins/chemistry , Animals , Binding Sites/drug effects , Cells, Cultured , Cysteine/chemistry , Emodin/chemistry , Fallopia multiflora/chemistry , Hepatocytes/metabolism , Male , Mice , Mice, Inbred Strains , Molecular Structure
4.
Mar Drugs ; 19(5)2021 May 15.
Article in English | MEDLINE | ID: mdl-34063509

ABSTRACT

Marine biodiversity has been yielding promising novel bioproducts from venomous animals. Despite the auspices of conotoxins, which originated the paradigmatic painkiller Prialt, the biotechnological potential of gastropod venoms remains to be explored. Marine bioprospecting is expanding towards temperate species like the dogwhelk Nucella lapillus, which is suspected to secrete immobilizing agents through its salivary glands with a relaxing effect on the musculature of its preferential prey, Mytilus sp. This work focused on detecting, localizing, and testing the bioreactivity of cysteine-rich proteins and peptides, whose presence is a signature of animal venoms and poisons. The highest content of thiols was found in crude protein extracts from the digestive gland, which is associated with digestion, followed by the peribuccal mass, where the salivary glands are located. Conversely, the foot and siphon (which the gastropod uses for feeding) are not the main organs involved in toxin secretion. Ex vivo bioassays with Mytilus gill tissue disclosed the differential bioreactivity of crude protein extracts. Secretions from the digestive gland and peribuccal mass caused the most significant molecular damage, with evidence for the induction of apoptosis. These early findings indicate that salivary glands are a promising target for the extraction and characterization of bioactive cysteine-rich proteinaceous toxins from the species.


Subject(s)
Bodily Secretions/chemistry , Cysteine/chemistry , Cysteine/toxicity , Gastropoda/chemistry , Animal Structures/anatomy & histology , Animal Structures/chemistry , Animals , Bivalvia/anatomy & histology , Cysteine/analysis , DNA Damage/drug effects , Gastropoda/anatomy & histology , Gastropoda/metabolism , Gills/anatomy & histology , Marine Toxins/analysis , Marine Toxins/chemistry , Marine Toxins/toxicity , Salivary Glands/chemistry
5.
Int J Mol Sci ; 23(1)2021 Dec 30.
Article in English | MEDLINE | ID: mdl-35008820

ABSTRACT

Mercury is a heavy metal toxicant that is prevalent throughout the environment. Organic forms of mercury, such as methylmercury (MeHg), can cross the placenta and can lead to lasting detrimental effects in the fetus. The toxicological effects of MeHg on the placenta itself have not been clearly defined. Therefore, the purpose of the current study was to assess the transport of MeHg into placental syncytiotrophoblasts and to characterize the mechanisms by which MeHg exerts its toxic effects. Cultured placental syncytiotrophoblasts (BeWo) were used for these studies. The transport of radioactive MeHg was measured to identify potential mechanisms involved in the uptake of this compound. The toxicological effects of MeHg on BeWo cells were determined by assessing visible pathological change, autophagy, mitochondrial viability, and oxidative stress. The findings of this study suggest that MeHg compounds are transported into BeWo cells primarily by sodium-independent amino acid carriers and organic anion transporters. The MeHg altered mitochondrial function and viability, decreased mitophagy and autophagy, and increased oxidative stress. Exposure to higher concentrations of MeHg inhibited the ability of cells to protect against MeHg-induced injury. The findings show that MeHg is directly toxic to syncytiotrophoblasts and may lead to disruptions in the fetal/maternal transfer of nutrients and wastes.


Subject(s)
Cysteine/analogs & derivatives , Methylmercury Compounds/metabolism , Methylmercury Compounds/toxicity , Autophagy/drug effects , Biological Transport/drug effects , Biomarkers/metabolism , Cell Line , Cell Shape/drug effects , Cell Survival/drug effects , Cysteine/metabolism , Cysteine/toxicity , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Methionine/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Oxidative Stress/drug effects , Substrate Specificity/drug effects , Time Factors , Tritium/metabolism
6.
Chem Res Toxicol ; 33(6): 1339-1355, 2020 06 15.
Article in English | MEDLINE | ID: mdl-31951115

ABSTRACT

Trichloroethylene (TCE) is a widespread environmental contaminant following decades of use as an industrial solvent, improper disposal, and remediation challenges. Consequently, TCE exposure continues to constitute a risk to human health. Despite epidemiological evidence associating exposure with adverse birth outcomes, the effects of TCE and its metabolite S-(1, 2-dichlorovinyl)-L-cysteine (DCVC) on the placenta remain undetermined. Flexible and efficient macronutrient and energy metabolism pathway utilization is essential for placental cell physiological adaptability. Because DCVC is known to compromise cellular energy status and disrupt energy metabolism in renal proximal tubular cells, this study investigated the effects of DCVC on cellular energy status and energy metabolism pathways in placental cells. Human extravillous trophoblast cells, HTR-8/SVneo, were exposed to 5-20 µM DCVC for 6 or 12 h. After establishing concentration and exposure duration thresholds for DCVC-induced cytotoxicity, targeted metabolomics was used to evaluate overall energy status and metabolite concentrations from energy metabolism pathways. The data revealed glucose metabolism perturbations including a time-dependent accumulation of glucose-6-phosphate+frutose-6-phosphate (G6P+F6P) as well as independent shunting of glucose intermediates that diminished with time, with modest energy status decline but in the absence of significant changes in ATP concentrations. Furthermore, metabolic profiling suggested that DCVC stimulated compensatory utilization of glycerol, lipid, and amino acid metabolism to provide intermediate substrates entering downstream in the glycolytic pathway or the tricarboxylic acid cycle. Lastly, amino acid deprivation increased susceptibility to DCVC-induced cytotoxicity. Taken together, these results suggest that DCVC caused metabolic perturbations necessitating adaptations in macronutrient and energy metabolism pathway utilization to maintain adequate ATP levels.


Subject(s)
Cysteine/analogs & derivatives , Energy Metabolism/drug effects , AMP-Activated Protein Kinases/metabolism , Amino Acids/metabolism , Cell Line , Cell Survival/drug effects , Cysteine/toxicity , Glucose/metabolism , Glycerol/metabolism , Humans , Lipid Metabolism/drug effects , Nutrients/metabolism , Phosphofructokinase-1/metabolism , Solvents/metabolism , Trichloroethylene/metabolism
7.
J Am Chem Soc ; 141(2): 849-857, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30541274

ABSTRACT

Nanoformulations that can respond to the specific tumor microenvironment (TME), such as a weakly acidic pH, low oxygen, and high glutathione (GSH), show promise for killing cancer cells with minimal invasiveness and high specificity. In this study, we demonstrate self-assembled copper-amino acid mercaptide nanoparticles (Cu-Cys NPs) for in situ glutathione-activated and H2O2-reinforced chemodynamic therapy for drug-resistant breast cancer. After endocytosis into tumor cells, the Cu-Cys NPs could first react with local GSH, induce GSH depletion, and reduce Cu2+ to Cu+. Subsequently, the generated Cu+ would react with local H2O2 to generate toxic hydroxyl radicals (·OH) via a Fenton-like reaction, which has a fast reaction rate in the weakly acidic TME, that are responsible for tumor-cell apoptosis. Due to the high GSH and H2O2 concentration in tumor cells, which sequentially triggers the redox reactions, Cu-Cys NPs exhibited relatively high cytotoxicity to cancer cells, whereas normal cells were left alive. The in vivo results also proved that Cu-Cys NPs efficiently inhibited drug-resistant breast cancer without causing obvious systemic toxicity. As a novel copper mercaptide nanoformulation responsive to the TME, these Cu-Cys NPs may have great potential in chemodynamic cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Copper/therapeutic use , Cysteine/therapeutic use , Metal Nanoparticles/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Copper/chemistry , Copper/toxicity , Cysteine/chemistry , Cysteine/toxicity , Female , Glutathione/chemistry , Glutathione/metabolism , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydroxyl Radical/metabolism , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Oxidation-Reduction , Xenograft Model Antitumor Assays
8.
Anal Chem ; 91(14): 8987-8993, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31265249

ABSTRACT

Although attractive for their low toxicity, CuInS2/ZnS core/shell quantum dots (CIS/ZnS QDs) still suffer from poor luminescence efficiency and poor water solubility. Herein, two amino acids (AAs), i.e., cysteine (Cys) and threonine (Thr), are used to tune the properties of CIS/ZnS QDs by capping them in both core and shell. It is found that Thr can regulate the density of Cys on the surface of QDs, thus causing a synergistic effect on the enhancement of photoluminescence (PL) intensity. Capping in the shell mainly leads to the enhancement of PL intensity, and capping in the core results in a red-shift of PL wavelength. Accordingly, a new kind of near-infrared region CIS/ZnS QDs with improved optical properties has been prepared. In addition, the Cys- and Thr-capped CIS/ZnS QDs possess outstanding water solubility and biocompatibility. In this work, the QDs are further employed in Cd2+ determination and multicolor imaging, indicating their potential applications. Relying on the enhancement of PL intensity via cation exchange, the Cys- and Thr-capped CIS/ZnS QDs can sense Cd2+ sensitively. Notably, because ZnS shells of the QDs will not be affected by Zn2+, the analytical method can discriminate Cd2+ from Zn2+ depending on the inherent characteristics of QDs. Moreover, intercellular Cd2+ can also be evaluated by the bright PL from the QDs, and the QDs can achieve multicolor imaging. Overall, this work demonstrates that various properties of QDs may be tuned by capping with AAs, and AA-capped QDs are of great value in advanced biosensing and bioimaging.


Subject(s)
Cadmium/analysis , Cysteine/chemistry , Fluorescent Dyes/chemistry , Quantum Dots/chemistry , Threonine/chemistry , Copper/chemistry , Copper/toxicity , Cysteine/toxicity , Fluorescent Dyes/toxicity , Indium/chemistry , Indium/toxicity , Limit of Detection , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Quantum Dots/toxicity , Solubility , Sulfides/chemistry , Sulfides/toxicity , Threonine/toxicity , Water/chemistry , Zinc Compounds/chemistry , Zinc Compounds/toxicity
9.
Drug Metab Dispos ; 47(8): 809-817, 2019 08.
Article in English | MEDLINE | ID: mdl-31097424

ABSTRACT

Vildagliptin (VG), a dipeptidyl peptidase-4 inhibitor, is used for treating type 2 diabetes. On rare occasions, VG causes liver injury as an adverse reaction. One case report suggested the involvement of immune responses in the hepatotoxicity, but the underlying mechanisms are unknown. We recently reported that VG binds covalently in vitro to l-cysteine to produce a thiazoline acid metabolite, M407, implying that the covalent binding may trigger the immune-mediated hepatotoxicity. There was no evidence, however, that such a thiazoline acid metabolite was formed in vivo. In the present study, we administered a single oral dose of VG to male Sprague-Dawley rats, and detected M407 in plasma. The sum of urinary and fecal excretions of M407 reached approximately 2% of the dose 48 hours postdosing. Using bile duct-cannulated rats, we demonstrated that M407 was secreted into bile as a glucuronide, designated as M583. Another newly identified thiazoline metabolite of VG, the cysteinylglycine conjugate M464, was detected in urine, feces, and bile. The formation of M464 was confirmed by in vitro incubation of VG with glutathione even in the absence of metabolic enzymes. A glutathione adduct against the nitrile moiety M611 was also detected in vitro but not in vivo. In summary, we found three new thiazoline-containing thiol adduct metabolites in VG-administered rats. Nonenzymatic covalent binding of VG would likely occur in humans, and it may be relevant to predicting adverse reactions.


Subject(s)
Cysteine/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Glutathione/metabolism , Sulfhydryl Compounds/metabolism , Vildagliptin/pharmacokinetics , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Cysteine/chemistry , Cysteine/toxicity , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Glutathione/chemistry , Glutathione/toxicity , Humans , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/toxicity , Vildagliptin/administration & dosage , Vildagliptin/adverse effects
10.
Chem Res Toxicol ; 32(3): 447-455, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30681327

ABSTRACT

Hydropersulfides and related polysulfides have recently become topics of significant interest due to their physiological prevalence and proposed biological functions. Currently, examination of the effects of hydropersulfide treatment on cells is difficult due to their lack of inherent stability with respect to disproportionation. Herein, it is reported that the treatment of a variety of cell types with cysteine trisulfide (also known as thiocystine; Cys-SSS-Cys), results in an increase in intracellular hydropersulfide levels (e.g., cysteine hydropersulfide; Cys-SSH, and glutathione hydropersulfide; GSSH). Thus, Cys-SSS-Cys represents a possible pharmacological agent for examining the effects of hydropersulfides on cell function/viability. It has also been found that cells with increased intracellular hydropersulfide levels can export Cys-SSH into the extracellular media. Interestingly, the Cys-SSH is the major hydropersulfide exported by cells, although GSSH is the predominant intracellular species. The possible implications of cellular export are discussed.


Subject(s)
Cysteine/metabolism , Cysteine/toxicity , Sulfides/metabolism , Sulfides/toxicity , 3T3 Cells , Animals , Cell Line , Cell Survival/drug effects , Cysteine/chemistry , Humans , Mice , Molecular Structure , Sulfides/chemistry , Tetrazolium Salts/pharmacology
11.
Toxicol Appl Pharmacol ; 338: 30-42, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29129777

ABSTRACT

Trichloroethylene (TCE), a prevalent environmental contaminant, is a potent renal and hepatic toxicant through metabolites such as S-(1, 2-dichlorovinyl)-l-cysteine (DCVC). However, effects of TCE on other target organs such as the placenta have been minimally explored. Because elevated apoptosis and lipid peroxidation in placenta have been observed in pregnancy morbidities involving poor placentation, we evaluated the effects of DCVC exposure on apoptosis and lipid peroxidation in a human extravillous trophoblast cell line, HTR-8/SVneo. We exposed the cells in vitro to 10-100µM DCVC for various time points up to 24h. Following exposure, we measured apoptosis using flow cytometry, caspase activity using luminescence assays, gene expression using qRT-PCR, and lipid peroxidation using a malondialdehyde quantification assay. DCVC significantly increased apoptosis in time- and concentration-dependent manners (p<0.05). DCVC also significantly stimulated caspase 3, 7, 8 and 9 activities after 12h (p<0.05), suggesting that DCVC stimulates the activation of both the intrinsic and extrinsic apoptotic signaling pathways simultaneously. Pre-treatment with the tBID inhibitor Bl-6C9 partially reduced DCVC-stimulated caspase 3 and 7 activity, signifying crosstalk between the two pathways. Additionally, DCVC treatment increased lipid peroxidation in a concentration-dependent manner. Co-treatment with the antioxidant peroxyl radical scavenger (±)-α-tocopherol attenuated caspase 3 and 7 activity, suggesting that lipid peroxidation mediates DCVC-induced apoptosis in extravillous trophoblasts. Our findings suggest that DCVC-induced apoptosis and lipid peroxidation in extravillous trophoblasts could contribute to poor placentation if similar effects occur in vivo in response to TCE exposure, indicating that further studies into this mechanism are warranted.


Subject(s)
Apoptosis/drug effects , Cysteine/analogs & derivatives , Lipid Peroxidation/drug effects , Placenta/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle Proteins/physiology , Cells, Cultured , Cysteine/toxicity , Female , Humans , NF-kappa B p50 Subunit/physiology , Nuclear Proteins/physiology , Placenta/cytology , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First , Reactive Oxygen Species/metabolism
12.
J Toxicol Environ Health A ; 81(1-3): 37-52, 2018.
Article in English | MEDLINE | ID: mdl-29190187

ABSTRACT

Trichloroethylene (TCE) is a ubiquitous environmental toxicant that is a liver and kidney carcinogen. Conjugation of TCE with glutathione (GSH) leads to formation of nepthrotoxic and mutagenic metabolites postulated to be critical for kidney cancerdevelopment; however, relatively little is known regarding their tissue levels as previous analytical methods for their detection lacked sensitivity. Here, an LC-MS/MS-based method for simultaneous detection of S-(1,2-dichlorovinyl)-glutathione (DCVG), S-(1,2-dichlorovinyl)-L-cysteine (DCVC), and N-acetyl-S-(1,2-dichlorovinyl)-L-cysteine (NAcDCVC) in multiple mouse tissues was developed. This analytical method is rapid, sensitive (limits of detection (LOD) 3-30 fmol across metabolites and tissues), and robust to quantify all three metabolites in liver, kidneys, and serum. The method was used to characterize inter-tissue and inter-strain variability in formation of conjugative metabolites of TCE. Single oral dose of TCE (24, 240 or 800 mg/kg) was administered to male mice from 20 inbred strains of Collaborative Cross. Inter-strain variability in the levels of DCVG, DCVC, and NAcDCVC (GSD = 1.6-2.9) was observed. Whereas NAcDCVC was distributed equally among analyzed tissues, highest levels of DCVG were detected in liver and DCVC in kidneys. Evidence indicated that inter-strain variability in conjugative metabolite formation of TCE might affect susceptibility to adverse health effects and that this method might aid in filling data gaps in human health assessment of TCE.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine/analogs & derivatives , Glutathione/analogs & derivatives , Glutathione/metabolism , Glutathione/toxicity , Trichloroethylene/metabolism , Trichloroethylene/toxicity , Acetylcysteine/metabolism , Acetylcysteine/toxicity , Animals , Cysteine/metabolism , Cysteine/toxicity , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Signal-To-Noise Ratio , Tissue Distribution
13.
Article in English | MEDLINE | ID: mdl-28418776

ABSTRACT

Many pyrrolizidine alkaloids (PAs) are hepatotoxic, genotoxic, and carcinogenic phytochemicals. Metabolism of PAs in vivo generates four (±)-6,7-dihydro-7-hydroxy-1-hydroxymethyl-5H-pyrrolizine (DHP)-DNA adducts that have been proposed to be responsible for PA-induced liver tumor formation in rats. In this present study, we determined that the same set of DHP-DNA adducts was formed upon the incubation of 7-glutathione-DHP and 7-cysteine-DHP with cultured human hepatocarcinoma HepG2 cells. These results suggest that 7-glutathione-DHP and 7-cysteine-DHP are reactive metabolites of PAs that can bind to cellular DNA to form DHP-DNA adducts in HepG2 cells, and can potentially initiate liver tumor formation.


Subject(s)
Carcinogens/toxicity , Cysteine/analogs & derivatives , Glutathione/analogs & derivatives , Pyrroles/toxicity , Pyrrolizidine Alkaloids/toxicity , Animals , Cysteine/metabolism , Cysteine/toxicity , DNA Adducts , Glutathione/metabolism , Glutathione/toxicity , Pyrrolizidine Alkaloids/metabolism , Rats , Rats, Inbred F344
14.
Amino Acids ; 48(1): 149-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26306843

ABSTRACT

High intake of dietary cysteine is extremely toxic to animals and the underlying mechanism remains largely unknown. This study was conducted to test the hypothesis that excessive L-cysteine induces cell death by activating endoplasmic reticulum (ER) stress and mitogen-activated protein kinase (MAPK) signaling in intestinal porcine epithelial cells. Jejunal enterocytes were cultured in the presence of 0-10 mmol/L L-cysteine. Cell viability, morphologic alterations, mRNA levels for genes involved in ER stress, protein abundances for glucose-regulated protein 78, C/EBP homologous protein (CHOP), alpha subunit of eukaryotic initiation factor-2 (eIF2α), extracellular signal-regulated kinase (ERK1/2), p38 MAPK, and c-Jun N-terminal protein kinase (JNK1/2) were determined. The results showed that L-cysteine (5-10 mmol/L) reduced cell viability (P < 0.05) and led to vacuole-like cell death in intestinal porcine epithelial cells. These adverse effects of L-cysteine were not affected by the autophagy inhibitor 3-methyladenine. The protein abundances for CHOP, phosphorylated (p)-eIF2α, p-JNK1/2, p-p38 MAPK, and the spliced form of XBP-1 mRNA were enhanced (P < 0.05), whereas those for p-ERK1/2 were reduced (P < 0.05). Collectively, excessive L-cysteine induces vacuole-like cell death via the activation of ER stress and MAPK signaling in small intestinal epithelial cells. These signaling pathways may be potential targets for developing effective strategies to prevent the toxicity of dietary cysteine.


Subject(s)
Apoptosis , Cysteine/toxicity , Endoplasmic Reticulum Stress , Enterocytes/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Intestines/cytology , Vacuoles/metabolism , Animals , Cell Survival , Cysteine/metabolism , Enterocytes/metabolism , Intestinal Mucosa/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Swine , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
15.
Environ Sci Technol ; 49(13): 8041-7, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26018638

ABSTRACT

Various factors have been invoked to explain the toxicity of silver nanoparticles (AgNP) to microorganisms including particle size and the nature of stabilizing coatings as well as the amount of dissolved silver occurring in AgNP suspensions. In this study we have assessed the effects of nine differently coated AgNP (chitosan, lactate, polyvinylpyrrolidone, polyethelene glycol, gelatin, sodium dodecylbenzenesulfonate, citrate, dexpanthenol, and carbonate) and AgNO3 on the photosynthesis of the freshwater algae Chlamydomonas reinhardtii. We have thus examined how AgNP effects on algae relate to particle size, measured dissolved silver (Agd), and bioavailable silver (Agbioav). Agbioav was indirectly estimated in toxicity experiments by cysteine-silver complexation at the EC50. The EC50 calculated as a function of measured Agd concentrations showed for some coatings values similar to that of dissolved Ag, whereas other coated AgNP displayed lower EC50 values. In all cases, excess cysteine completely prevented effects on photosynthetic yield, confirming the role of Agd as a cause of the observed effect on the photosynthesis. Toxicity was related neither to particle size nor to the coatings. For all differently coated AgNP suspensions, the EC50 values calculated as a function of Agbioav were comparable to the value of AgNO3. Depending on the coatings Agbioav was comparable to or higher than measured Agd.


Subject(s)
Chlamydomonas reinhardtii/drug effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Photosynthesis/drug effects , Silver/toxicity , Benzenesulfonates/chemistry , Benzenesulfonates/toxicity , Carbonates/chemistry , Carbonates/toxicity , Chitosan/chemistry , Chitosan/toxicity , Chlamydomonas reinhardtii/physiology , Citrates/chemistry , Citrates/toxicity , Cysteine/pharmacology , Cysteine/toxicity , Gelatin/chemistry , Gelatin/toxicity , Lactates/chemistry , Lactates/toxicity , Pantothenic Acid/analogs & derivatives , Pantothenic Acid/chemistry , Pantothenic Acid/toxicity , Particle Size , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity , Povidone/toxicity , Silver/pharmacokinetics , Silver Nitrate/pharmacokinetics , Toxicity Tests/methods
16.
Bull Environ Contam Toxicol ; 94(4): 407-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25638440

ABSTRACT

Dietary mercury exposure is associated with suppressed immune responsiveness in birds. This study examined the immune-responsiveness of domestic zebra finches (Taeniopygia guttata) experimentally exposed to mercury through their diet. We used the phytohemagglutinin (PHA) skin-swelling test to assay the effect of two modes of mercury exposure. Some finches received exposure to mercury only after reaching sexual maturity, while others were maintained on a mercury-dosed diet throughout life, including development. Each bird received one of five dietary concentrations of methylmercury cysteine (0.0, 0.3, 0.6, 1.2 or 2.4 ppm). In contrast to a study on wild songbirds at a mercury-contaminated site, we detected no relationship between mercury level and immunological response to PHA, regardless of mode of exposure. This result represents the first major difference found by our laboratory between wild birds exposed to environmental mercury and captive birds experimentally exposed to mercury.


Subject(s)
Cysteine/analogs & derivatives , Environmental Exposure , Environmental Pollutants/toxicity , Finches/immunology , Methylmercury Compounds/toxicity , Animals , Birds , Cysteine/metabolism , Cysteine/toxicity , Environmental Pollutants/metabolism , Finches/metabolism , Mercury , Methylmercury Compounds/metabolism , Phytohemagglutinins/immunology , Skin Tests , Songbirds/immunology , Songbirds/metabolism
17.
Analyst ; 139(5): 954-63, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24343373

ABSTRACT

Because of microbial resistance to conventional antibiotics, there is increasing interest in silver, including silver nanoparticles (nano-Ag), in antimicrobial applications. However, questions remain regarding the relative roles of nano-Ag particles, versus Ag(+) ions released from nano-Ag dissolution, in imparting bacterial toxicity. Here, we developed a novel nano-Ag that, based on its cysteine cap, was expected to dissolve slowly and thus potentially allow for differentiating nanoparticle, versus ionic, effects of Ag. The nano-Ag was systematically tested for its differential toxicity to Escherichia coli and Pseudomonas aeruginosa. Bacterial growth, reactive oxygen species (ROS) generation, particle dissolution, cellular electron transfer activity, and cell membrane damage and potential were evaluated. In minimal growth medium, E. coli and P. aeruginosa growth were slowed at 100 mg L(-1) (0.93 mM) and 5 mg L(-1) (0.046 mM), respectively; P. aeruginosa was completely inhibited at and above 10 mg L(-1) (0.093 mM). For both strains, toxicity was associated with ROS and cell membrane damage. Based on comparisons to AgNO3 exposures, toxicity from nano-Ag was due to Ag(+) ions and not intact nano-Ag, even though nanoparticle dissolution was less than 2% in minimal growth medium. Because of their stability and slow Ag(+) ion release, the cysteine-capped nano-Ag particles here are useful to antimicrobial applications. Additionally, our systematic approach to evaluating toxicity, membrane damage, and ROS generation can be applied with other nanomaterials and bacteria.


Subject(s)
Cysteine/toxicity , Escherichia coli/drug effects , Metal Nanoparticles/toxicity , Pseudomonas aeruginosa/drug effects , Silver/toxicity , Dose-Response Relationship, Drug , Escherichia coli/chemistry , Escherichia coli/ultrastructure , Microscopy, Electron, Transmission/methods , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/ultrastructure , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Spectrophotometry, Infrared/methods , Spectrophotometry, Ultraviolet/methods
18.
J Toxicol Environ Health A ; 77(1-3): 46-56, 2014.
Article in English | MEDLINE | ID: mdl-24555646

ABSTRACT

Methylmercury (MeHg) is a highly toxic environmental contaminant that produces neurological and developmental impairments in animals and humans. Although its neurotoxic properties have been widely reported, the molecular mechanisms by which MeHg enters the cells and exerts toxicity are not yet completely understood. Taking into account that MeHg is found mostly bound to sulfhydryl-containing molecules such as cysteine in the environment and based on the fact that the MeHg-cysteine complex (MeHg-S-Cys) can be transported via the L-type neutral amino acid carrier transport (LAT) system, the potential beneficial effects of L-methionine (L-Met, a well known LAT substrate) against MeHg (administrated as MeHg-S-Cys)-induced neurotoxicity in mice were investigated. Mice were exposed to MeHg (daily subcutaneous injections of MeHg-S-Cys, 10 mg Hg/kg) and/or L-Met (daily intraperitoneal injections, 250 mg/kg) for 10 consecutive days. After treatments, the measured hallmarks of toxicity were mostly based on behavioral parameters related to motor performance, as well as biochemical parameters related to the cerebellar antioxidant glutathione (GSH) system. MeHg significantly decreased motor activity (open-field test) and impaired motor performance (rota-rod task) compared with controls, as well as producing disturbances in the cerebellar antioxidant GSH system. Interestingly, L-Met administration did not protect against MeHg-induced behavioral and cerebellar changes, but rather increased motor impairments in animals exposed to MeHg. In agreement with this observation, cerebellar levels of mercury (Hg) were higher in animals exposed to MeHg plus L-Met compared to those only exposed to MeHg. However, this event was not observed in kidney and liver. These results are the first to demonstrate that L-Met enhances cerebellar deposition of Hg in mice exposed to MeHg and that this higher deposition may be responsible for the greater motor impairment observed in mice simultaneously exposed to MeHg and L-Met.


Subject(s)
Cerebellum/chemistry , Cysteine/analogs & derivatives , Environmental Pollutants/toxicity , Methionine/pharmacology , Methylmercury Compounds/toxicity , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Psychomotor Performance/drug effects , Animals , Antioxidants/metabolism , Biomarkers/metabolism , Cerebellum/metabolism , Cysteine/administration & dosage , Cysteine/pharmacokinetics , Cysteine/toxicity , Drug Administration Schedule , Environmental Pollutants/administration & dosage , Environmental Pollutants/pharmacokinetics , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Injections, Intraperitoneal , Male , Methionine/administration & dosage , Methylmercury Compounds/administration & dosage , Methylmercury Compounds/pharmacokinetics , Mice , Neuroprotective Agents/administration & dosage , Random Allocation
19.
Pharm Dev Technol ; 18(6): 1288-93, 2013.
Article in English | MEDLINE | ID: mdl-22356486

ABSTRACT

Acetaldehyde is a known mutagenic substance and has been classified as a group-one carcinogen by the WHO. It is possible to bind acetaldehyde locally in the gastrointestinal (GI) tract with the semi-essential amino acid l-cysteine, which reacts covalently with acetaldehyde and forms compound 2-methyl-thiozolidine-4-carboxylic acid (MTCA). The Caco-2 cell line was used to determine the permeation of l-cysteine and MTCA, as well as the possible cell toxicity of both substances. Neither of the substances permeated through the Caco-2 cells at the concentrations used in this study, and only the highest concentration of MTCA affected the viability of the cells in the MTT (3-[4,5-dimethylthiazol-2yl]-2,5-diphenyltetrazolium bromide) test. These results showed that when l-cysteine is administered in formulations releasing it locally in the lower parts of GI tract, it is not absorbed but can react with acetaldehyde, and that neither l-cysteine nor MTCA is harmful to the cells when present locally in the upper parts of GI tract. This study also shows that MTCA is sensitive at a lower pH of 5.5. Since stable MTCA is desired in different parts of the GI tract, this observation raises concern over the influence of lower pH on l-cysteine-containing product ability to bind and eliminate carcinogenic acetaldehyde.


Subject(s)
Cysteine/pharmacokinetics , Cysteine/toxicity , Thiazolidines/pharmacokinetics , Thiazolidines/toxicity , Acetaldehyde/pharmacokinetics , Caco-2 Cells , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Permeability
20.
Toxicol Lett ; 383: 75-88, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37353095

ABSTRACT

Bioactivation of trichloroethylene (TCE) via glutathione conjugation is associated with several adverse effects in the kidney and other extrahepatic tissues. Of the three regioisomeric conjugates formed, S-(1,2-trans-dichlorovinyl)-glutathione (1,2-trans-DCVG), S-(1,2-cis-dichlorovinyl)-glutathione and S-(2,2-dichlorovinyl)-glutathione, only 1,2-trans-DCVG and its corresponding cysteine-conjugate, 1,2-trans-DCVC, have been subject to extensive mechanistic studies. In the present study, the metabolism and cellular effects of 1,2-cis-DCVG, the major regioisomer formed by rat liver fractions, and 1,2-cis-DCVC were investigated for the first time using RPTEC/TERT1-cells as in vitro renal model. In contrast to 1,2-trans-DCVG/C, the cis-regioisomers showed minimal effects on cell viability and mitochondrial respiration. Transcriptomics analysis showed that both 1,2-cis-DCVC and 1,2-trans-DCVC caused Nrf2-mediated antioxidant responses, with 3 µM as lowest effective concentration. An ATF4-mediated integrated stress response and p53-mediated responses were observed starting from 30 µM for 1,2-trans-DCVC and 125 µM for 1,2-cis-DCVC. Comparison of the metabolism of the DCVG regioisomers by LC/MS showed comparable rates of processing to their corresponding DCVC. No detectable N-acetylation was observed in RPTEC/TERT1 cells. Instead, N-glutamylation of DCVC to form N-γ-glutamyl-S-(dichlorovinyl)-L-cysteine was identified as a novel route of metabolism. The results suggest that 1,2-cis-DCVC may be of less toxicological concern for humans than 1,2-trans-DCVC, considering its lower intrinsic toxicity and lower rate of formation by human liver fractions.


Subject(s)
Cysteine , Trichloroethylene , Rats , Animals , Humans , Cysteine/toxicity , Cysteine/metabolism , Kidney/metabolism , Glutathione/metabolism , Trichloroethylene/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL