Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Free Radic Res ; 52(10): 1158-1169, 2018 Oct.
Article in English | MEDLINE | ID: mdl-31282788

ABSTRACT

Selenium (Se) containing organic compounds, such as ebselen (Ebs) and diphenyl diselenide [(PhSe)2], have been used as pharmacological agents due to their antioxidant properties. Tellurium (Te) does not have any biological function in mammals, but Te-containing organic compounds, such as diphenyl ditelluride [(PhTe)2], has been used both as an antioxidant or neurotoxic agent. At high concentrations, these compounds cause toxicity by oxidising thiol and selenol groups of proteins. Here, we analysed whether these compounds could modulate reactive species (RS) production, apoptosis and antioxidant gene expression profile of some selenoproteins and antioxidant enzymes or transcription factors in leukocytes isolated from human blood. Since no data is available about their accumulation in isolated leukocytes, we determine their concentration in the cells by CG-MS. Apoptosis (propidium iodide) and RS production (dichloro fluorescein) were determined by flow cytometry. The expression of CAT, SOD1, GPX3, GPX4, TRXR1, and NFLE2L2 genes were analysed by RT-PCR. (PhTe)2 was the only compound able to increase apoptosis rate. (PhSe)2 altered the expression of CAT and SOD1, and this was associated with a high RS production. All compounds decreased the expression of GPX3 but did not alter GPX4 and TRXR1 expression. All compounds decreased NFE2L2 expression (Ebs > (PhTe)2> (PhSe)2). We hypothesise that the toxicity induced by these organochalcogens is not directly related to their ability of inducing RS production.


Subject(s)
Antioxidants/metabolism , Apoptosis/drug effects , Chalcogens/toxicity , Gene Expression Regulation/drug effects , Leukocytes/drug effects , Reactive Oxygen Species/metabolism , Carbonic Anhydrases/genetics , Gene Expression Profiling , Healthy Volunteers , Humans , Leukocytes/metabolism , Selenoproteins/genetics , Superoxide Dismutase-1/genetics , Transcription Factors/genetics
2.
Toxicol Sci ; 160(1): 30-46, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29036705

ABSTRACT

Considering a novel series of zidovudine (AZT) derivatives encompassing selenoaryl moieties promising candidates as therapeutics, we examined the toxicities elicited by AZT and derivatives 5'-(4-Chlorophenylseleno)zidovudine (SZ1); 5'-(Phenylseleno)zidovudine (SZ2); and 5'-(4-Methylphenylseleno)zidovudine (SZ3) in healthy cells and in mice. Resting and stimulated cultured human peripheral blood mononuclear cells (PBMCs) were treated with the compounds at concentrations ranging from 10 to 200 µM for 24 and/or 72 h. Adult mice received a single injection of compounds (100 µmol/kg, s.c.) and 72 h after administration, hepatic/renal biomarkers were analyzed. Resting and stimulated PBMCs exposed to SZ1 displayed loss of viability, increased reactive species production, disruption in cell cycle, apoptosis and increased transcript levels and production of pro-inflammatory cytokines. In a mild way, most of these effects were also induced by SZ2. AZT and SZ3 did not cause significant toxicity towards resting PBMCs. Differently, both compounds elicited apoptosis and S phase arrest in stimulated cells. AZT and derivatives administration did not change the body weight and plasma biochemical markers in mice. However, the absolute weight and organ-to-body weight ratio of liver, kidneys and spleen were altered in AZT, SZ1-, and SZ2-treated mice. Our results highlighted the involvement of derivatives SZ1 and SZ2 in redox and immunological dyshomeostasis leading to activation of apoptotic signaling pathways in healthy cells under different division phases. On the other hand, the derivative SZ3 emerged as a promising candidate for further viral infection/antitumor studies as a new effective therapy with low toxicity for immune cells and after acute in vivo treatment.


Subject(s)
Antineoplastic Agents/toxicity , Chalcogens/toxicity , Leukocytes, Mononuclear/drug effects , Zidovudine/toxicity , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Behavior, Animal/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Organ Size/drug effects , Reactive Oxygen Species/metabolism , Risk Assessment , S Phase Cell Cycle Checkpoints/drug effects , Zidovudine/analogs & derivatives
4.
Neurotox Res ; 24(2): 109-18, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23224748

ABSTRACT

Organochalcogens, such as organoselenium and organotellurium compounds, can be neurotoxic to rodents. Since mitochondrial dysfunction plays a pivotal role in neurological disorders, the present study was designed to test the hypothesis that rat brain mitochondrial complexes (I, II, I-III, II-III and IV) could be molecular targets of organochalcogens. The results show that organochalcogens caused statistically significant inhibition of mitochondrial complex I activity, which was prevented by preincubation with NADH and fully blunted by reduced glutathione (GSH). Mitochondrial complex II activity remained unchanged in response to (PhSe)2 treatment. Ebs and (PhTe)2 caused a significant concentration-dependent inhibition of complex II that was also blunted by GSH. Mitochondrial complex IV activity was not modified by organochalcogens. Collectively, Ebs, (PhSe)2 and (PhTe)2 were more effective inhibitors of brain mitochondrial complex I than of complex II, whereas they did not affect complex IV. These observations are consistent with organochalcogens inducing mitochondrial complex I and II inhibition via their thiol-oxidase-like activity, with Ebs, (PhSe)2 and (PhTe)2 effectively oxidising critical thiol groups of these complexes.


Subject(s)
Brain/drug effects , Chalcogens/toxicity , Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex I/antagonists & inhibitors , Mitochondrial Membranes/drug effects , Organoselenium Compounds/toxicity , Animals , Brain/enzymology , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Male , Mitochondrial Membranes/enzymology , Rats , Rats, Wistar
5.
Toxicol Sci ; 117(1): 133-43, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20573786

ABSTRACT

Ebselen (Ebs) and diphenyl diselenide [(PhSe)(2)] readily oxidize thiol groups. Here we studied mitochondrial swelling changes in mitochondrial potential (Deltapsim), NAD(P)H oxidation, reactive oxygen species production, protein aggregate formation, and oxygen consumption as ending points of their in vitro toxicity. Specifically, we tested the hypothesis that organochalchogens toxicity could be associated with mitochondrial dysfunction via oxidation of vicinal thiol groups that are known to be involved in the regulation of mitochondrial permeability (Petronilli et al. J. Biol. Chem., 269; 16638; 1994). Furthermore, we investigated the possible mechanism(s) by which these organochalchogens could disrupt liver mitochondrial function. Ebs and (PhSe)(2) caused mitochondrial depolarization and swelling in a concentration-dependent manner. Furthermore, both organochalchogens caused rapid oxidation of the mitochondrial pyridine nucleotides (NAD(P)H) pool, likely reflecting the consequence and not the cause of increased mitochondrial permeability (Costantini, P., Chernyak, B. V., Petronilli, V., and Bernardi, P. (1996). Modulation of the mitochondrial permeability transition pore (PTP) by pyridine nucleotides and dithiol oxidation at two separate sites. J. Biol. Chem. 271, 6746-6751). The organochalchogens-induced mitochondrial dysfunction was prevented by the reducing agent dithiothreitol (DTT). Ebs- and (PhSe)(2)-induced mitochondrial depolarization and swelling were unchanged by ruthenium red (4microM), butylated hydroxytoluene (2.5microM), or cyclosporine A (1microM). N-ethylmaleimide enhanced the organochalchogens-induced mitochondrial depolarization, without affecting the magnitude of the swelling response. In contrast, iodoacetic acid did not modify the effects of Ebs or (PhSe)(2) on the mitochondria. Additionally, Ebs and (PhSe)(2) decreased the basal 2' 7' dichlorofluorescin diacetate (H(2)-DCFDA) oxidation and oxygen consumption rate in state 3 and increased it during the state 4 of oxidative phosphorylation and induced the formation of protein aggregates, which were prevented by DTT. However, DTT failed to reverse the formation of protein aggregates, when it was added after a preincubation of liver mitochondria with Ebs or (PhSe)(2). Similarly, DTT did not reverse the Ebs- or (PhSe)(2)-induced Deltapsim collapse or swelling, when it was added after a preincubation period of mitochondria with chalcogenides. These results show that Ebs and (PhSe)(2) can effectively induce mitochondrial dysfunction and suggest that effects of these compounds are associated with mitochondrial thiol groups oxidation. The inability of cyclosporine A to reverse the Ebs- and (PhSe)(2)-induced mitochondrial effects suggests that the redox-regulated mitochondrial permeability transition (MPT) pore was mechanistically regulated in a manner that is distinct from the classical MPT pore.


Subject(s)
Chalcogens/toxicity , Mitochondria, Liver/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Sulfhydryl Compounds/metabolism , Animals , Butylated Hydroxytoluene/pharmacology , Cyclosporins/pharmacology , Ethylmaleimide/pharmacology , Iodoacetic Acid/pharmacology , Male , Mitochondria, Liver/metabolism , Mitochondrial Permeability Transition Pore , NADP/metabolism , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
6.
Toxicol In Vitro ; 21(4): 639-45, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17321101

ABSTRACT

The purpose of this study was to investigate the possible involvement of the glutamatergic system in the neurotoxicity of diorganylchalcogenides or organochalcogenides from slices of cerebral cortex in different ages of development: 12- and 60-day-old rats. Glutamate uptake was evaluated in cortical slices of 12 and 60 days old rats. Cortex slices were incubated with three different organochalcogenides with or without reduced glutathione or dithiothreitol. At 100 microM, ebselen, diphenyl diselenide (PhSe)2 and diphenyl ditelluride (PhTe)2 in vitro inhibited the [3H]glutamate uptake in both age. Both 60-day-old rats and for 12-day-old rats, GSH and DTT prevented the (PhTe)2-induced inhibition of glutamate uptake but did not protect the inhibition caused by ebselen and (PhSe)2. These findings suggest that the neurotoxicity of organochalcogenides could be related to their effects on brain glutamate uptake, conceivably involving a redox modulation of reactive amino acids from the glutamate transporter proteins.


Subject(s)
Antidotes/pharmacology , Azoles/toxicity , Brain Chemistry/drug effects , Chalcogens/toxicity , Dithiothreitol/pharmacology , Glutamic Acid/metabolism , Glutathione/pharmacology , Organoselenium Compounds/toxicity , Aging/physiology , Animals , Benzene Derivatives/toxicity , Brain/drug effects , Brain/growth & development , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , In Vitro Techniques , Isoindoles , Male , Organometallic Compounds/toxicity , Rats , Rats, Wistar
7.
Toxicol In Vitro ; 21(1): 139-45, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17084588

ABSTRACT

Previous literature reports have demonstrated that a number of human diseases, including inflammation and cancer, can be caused by environmental and occupational exposure to toxic compounds, via DNA damage, protein modifications, or lipid peroxidation. The present study was undertaken to screen the toxicity of a variety of chalcogens using erythrocytes as a model of cell injury. The toxicity of these compounds was evaluated via quantification of hemolysis and lipid peroxidation. The present investigation shows that diphenyl ditelluride and phenyl tellurides are toxic to erythrocytes. The organoselenium compounds were not toxic to erythrocytes even when tested at high concentrations and with a hematocrit of 45%. The hemolytic effect of tellurides was not positively correlated with thiobarbituric acid-reactive substance (TBARS) production suggesting that lipid peroxidation is not involved in the hemolysis provoked by organotellurium compounds. The results suggest that chalcogen compounds may be toxic to human erythrocytes, depending on their structure.


Subject(s)
Chalcogens/toxicity , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , In Vitro Techniques , Indicators and Reagents , Lipid Peroxidation/drug effects , Organic Chemicals/toxicity , Oxidants/toxicity , Thiobarbituric Acid Reactive Substances
8.
Bioorg Med Chem ; 13(23): 6394-403, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16084725

ABSTRACT

Several thio and seleno analogues of tetramethylrosamine (TMR) were prepared. Thio derivatives of TMR have absorption maxima near 570 nm, while seleno derivatives of TMR have absorption maxima near 580 nm. The 3- or 4-N,N-dimethylaminophenyl substituent in the 9-position greatly increases internal conversion, which lowers quantum yields for fluorescence and the generation of singlet oxygen. Thio and seleno analogues of TMR are effective photosensitizers against chemosensitive AUXB1 cells in vitro and against multidrug-resistant CR1R12 cells in vitro, which have been treated with verapamil. The CR1R12 cells accumulated significantly lower concentrations of the photosensitizers relative to the AUXB1 cells presumably due to the expression of P-glycoprotein (Pgp) in the CR1R12 cells. Following treatment with 5 x 10(-5) M verapamil, the uptake in CR1R12 cells of several fluorescent thio analogues of TMR is comparable to that observed for the chemosensitive AUXB1 cells.


Subject(s)
Chalcogens/chemistry , Chalcogens/pharmacology , Drug Resistance, Multiple , Heterocyclic Compounds, 3-Ring/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Chalcogens/chemical synthesis , Chalcogens/toxicity , Cricetinae , Molecular Structure , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/toxicity , Rhodamines , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...