Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38731969

RESUMO

This review offers a synthesis of the current understanding of the impact of low-dose thallium (Tl) on public health, specifically emphasizing its diverse effects on various populations and organs. The article integrates insights into the cytotoxic effects, genotoxic potential, and molecular mechanisms of thallium in mammalian cells. Thallium, a non-essential heavy metal present in up to 89 different minerals, has garnered attention due to its adverse effects on human health. As technology and metallurgical industries advance, various forms of thallium, including dust, vapor, and wastewater, can contaminate the environment, extending to the surrounding air, water sources, and soil. Moreover, the metal has been identified in beverages, tobacco, and vegetables, highlighting its pervasive presence in a wide array of food sources. Epidemiological findings underscore associations between thallium exposure and critical health aspects such as kidney function, pregnancy outcomes, smoking-related implications, and potential links to autism spectrum disorder. Thallium primarily exerts cellular toxicity on various tissues through mitochondria-mediated oxidative stress and endoplasmic reticulum stress. This synthesis aims to shed light on the intricate web of thallium exposure and its potential implications for public health, emphasizing the need for vigilant consideration of its risks.


Assuntos
Saúde Pública , Tálio , Humanos , Tálio/toxicidade , Animais , Exposição Ambiental/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos
2.
Environ Toxicol Chem ; 43(1): 87-96, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37750573

RESUMO

The mechanisms of acute (96-hour) and subchronic (28-day) toxicity of the waterborne trace metal thallium (Tl) to rainbow trout (Oncorhynchus mykiss) were investigated. Specifically, effects on branchial and renal ionoregulatory enzymes (sodium/potassium adenosine triphosphatase [ATPase; NKA] and proton ATPase) and hepatic oxidative stress endpoints (protein carbonylation, glutathione content, and activities of catalase and glutathione peroxidase) were examined. Fish (19-55 g) were acutely exposed to 0 (control), 0.9 (regulatory limit), 2004 (half the acute median lethal concentration), or 4200 (acute median lethal concentration) µg Tl L-1 or subchronically exposed to 0, 0.9, or 141 (an elevated environmental concentration) µg Tl L-1 . The only effect following acute exposure was a stimulation of renal H+ -ATPase activity at the highest Tl exposure concentration. Similarly, the only significant effect of subchronic Tl exposure was an inhibition of branchial NKA activity at 141 µg Tl L-1 , an effect that may reflect the interaction of Tl with potassium ion handling. Despite significant literature evidence for effects of Tl on oxidative stress, there were no effects of Tl on any such endpoint in rainbow trout, regardless of exposure duration or exposure concentration. Elevated basal levels of antioxidant defenses may explain this finding. These data suggest that ionoregulatory perturbance is a more likely mechanism of Tl toxicity than oxidative stress in rainbow trout but is an endpoint of relevance only at elevated environmental Tl concentrations. Environ Toxicol Chem 2024;43:87-96. © 2023 SETAC.


Assuntos
Oncorhynchus mykiss , Poluentes Químicos da Água , Animais , Oncorhynchus mykiss/metabolismo , Brânquias/metabolismo , Tálio/toxicidade , Poluentes Químicos da Água/metabolismo , Estresse Oxidativo , ATPase Trocadora de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/farmacologia , Potássio/metabolismo
3.
Toxicology ; 492: 153532, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37141935

RESUMO

Thallium (Tl) is a high-priority toxic metal that poses a severe threat to human health. The toxicity characteristics induced by Tl have been partially discussed. However, the immunotoxic effects of Tl exposure have remained largely unexplored. Our findings demonstrated that 50 ppm of Tl exposure for one week induced severe weight loss in mice, which was accompanied by appetite suppression. Moreover, although Tl exposure did not induce significant pathological damage to skeletal muscle and bone, Tl inhibited the expression of B cell development-related genes in the bone marrow. Additionally, Tl exposure increased B cell apoptosis and reduced its generation in the bone marrow. Analysis of B cells in the blood indicated that the percentage of B-2 cells decreased significantly, whereas B-2 cell proportions in the spleen did not. The percentage of CD4+ T cells in the thymus increased significantly, and the proportion of CD8+ T cells did not. Furthermore, although the proportion of the total CD4+ and CD8+ T cells was not significantly altered in the blood and spleen, Tl exposure promoted the migration of naïve CD4+ T cells and recent thymic emigrants (RTEs) from the thymus to the spleen. These results suggest that Tl exposure can affect B and T cell generation and migration, which provides new evidence for Tl-induced immunotoxicity.


Assuntos
Linfócitos B , Linfócitos T , Tálio , Tálio/toxicidade , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Animais , Camundongos , Movimento Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Timo/citologia , Timo/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos
4.
Environ Sci Technol ; 57(7): 2864-2876, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36745568

RESUMO

Element contamination, including that from heavy metals, is associated with gastrointestinal tumorigenesis, but the effects and mechanisms of crucial element exposure associated with colorectal cancer remain unclear. We profiled 56 elements by ICP-MS and used logistic regression, LASSO, BKMR, and GAM to identify colorectal cancer-relevant elements. A series of biochemical experiments were performed to demonstrate the cytotoxicity and the mechanisms of malignant transformation after metal exposure. Using an elementomics approach, we first found that the metal thallium (Tl) was positively correlated with many toxic metals and was associated with a significantly increased risk of colorectal cancer. Acute exposure to Tl induced cytotoxicity and cell death by accelerating the generation of reactive oxygen species and DNA damage. Chronic exposure to Tl led to the inhibition of cell death and thereby induced the malignant transformation of normal colon cells and xenograft tumor formation in nude mice. Furthermore, we describe the first identification of a significant metal quantitative trait locus for the novel colorectal cancer susceptibility locus rs1511625 near ATP13A3. Mechanistically, Tl increased the level of aberrant N6-methyladenosine (m6A) modification of ATP13A3 via the METLL3/METTL14/ALKBH5-ATP13A3 axis to promote colorectal tumorigenesis. This study provides a basis for the development of public health strategies for reducing metal exposure among populations at high risk for colorectal cancer.


Assuntos
Neoplasias Colorretais , Metais Pesados , Camundongos , Animais , Humanos , Camundongos Nus , Carcinogênese , Metais Pesados/toxicidade , Tálio/toxicidade , Neoplasias Colorretais/induzido quimicamente , Adenosina Trifosfatases , Proteínas de Membrana Transportadoras
5.
Neurotox Res ; 40(3): 814-824, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35476314

RESUMO

Thallium (Tl+) is a heavy metal that causes toxicity in several organs, including the brain. Its cytotoxic profile, combined with its affinity for tumor cells when used as a radioligand for labeling these cells, suggests its potential use as antitumor therapy. In this study, glioblastoma cell lines C6 (from rat) and U373 (from human) were exposed to increased concentrations of thallium(I) acetate (5, 10, 50, 100, or 200 µM) and several toxic endpoints were evaluated, including loss of confluence and morphological changes, loss of cell viability, changes in cell cycle, and apoptosis. Tl+ was detected in cells exposed to thallium(I) acetate, demonstrating efficient uptake mechanism. Confluence in both cell lines decreased in a concentration-dependent manner (50-200 µM), while morphological changes (cell shrinkage and decreased cell volume) were more evident at exposures to higher Tl+ concentrations. For both parameters, the effects of Tl+ were more prominent in C6 cells compared to U373 cells. The same trend was observed for cell viability, with Tl+ affecting this parameter in C6 cells at low concentrations, whereas U373 cells showed greater resistance, with significant changes observed only at the higher concentrations. C6 and U373 cells treated with Tl+ also showed morphological characteristics corresponding to apoptosis. The cytotoxic effects of Tl+ were also assessed in neural and astrocytic primary cultures from the whole rat brain. Primary neural and astrocytic cultures were less sensitive than C6 and U373 cells, showing changes in cell viability at 50 and 100 µM concentrations, respectively. Cell cycle in both brain tumor cell lines was altered by Tl+ in G1/G2 and S phases. In addition, when combined with temozolamide (500 µM), Tl+ elicited cell cycle alterations, increasing SubG1 population. Combined, our novel results characterize and validate the cytotoxic and antiproliferative effects of Tl+ in glioblastoma cells.


Assuntos
Antineoplásicos , Glioblastoma , Animais , Antineoplásicos/farmacologia , Apoptose , Técnicas de Cultura de Células , Ciclo Celular , Glioblastoma/metabolismo , Ratos , Tálio/toxicidade
6.
Fish Shellfish Immunol ; 118: 251-260, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34509627

RESUMO

A 60-day experiment was performed to assess the efficacy of dietary Astragalus membranaceus polysaccharides (ASP) in attenuation of sub-lethal thallium (Tl) toxicity in Nile tilapia. Six experimental groups (in triplicates) were designed where a fish group was raised in clean water and fed basal diet and served as control (CONT), two groups were fed the basal diet supplemented with 0.15% and 0.30% ASP (ASPL and ASPH), Tl-intoxicated group exposed to 1/10 of 96-h LC50 (= 41.9 µg/L), and two other groups were fed 0.15% and 0.30% ASP and concomitantly exposed to 41.9 µg Tl/L (ASPL-Tl and ASPH-Tl). At the end of the experiment, fish behavioral responses, clinical signs, survivability, growth, whole-body composition, intestinal digestive enzymes, serum biochemical parameters, hepatic antioxidative biomarkers, and transcription of stress and apoptosis genes were assessed. Results showed that the whole-body composition, intestinal α-amylase and protease enzymes, serum AST and blood urea levels, and hepatic GSH were not significantly different among groups (P > 0.05). The Tl-intoxicated fish group was off food, had darkened skin, showed restlessness and hyperexcitability, and high mortalities. FBW, WG, SGR and FI were significantly decreased alongside increase FCR in the Tl-exposed group. Tl exposure caused significant increases (P < 0.05) in intestinal lipase enzyme and serum indices such as ALT, creatinine, total cholesterol, triglycerides, glucose, and cortisol levels. Moreover, a significant decreases in hepatic CAT and SOD enzyme activities and significant increases in hepatic MDA contents were also noticed (P < 0.05). Furthermore, Tl exposure induced significant upregulation of hepatic HSP70 and apoptosis-related genes (p53 and caspase 3). Interestingly, dietary supplementation with ASP in ASPL-Tl and ASPH-Tl groups modulated the parameters mentioned above but still not reached the CONT values. Altogether, this study suggests that ASP could be beneficial in the modulation of sub-lethal Tl toxicity effects in Nile tilapia. Additionally, we can conclude that using natural feed supplements such as ASP in aquafeed might be necessary for maintaining the overall health performances of Nile tilapia.


Assuntos
Antioxidantes , Apoptose , Astragalus propinquus , Ciclídeos , Proteínas de Choque Térmico HSP70 , Polissacarídeos , Tálio , Ração Animal/análise , Animais , Antioxidantes/metabolismo , Apoptose/genética , Astragalus propinquus/química , Ciclídeos/genética , Ciclídeos/crescimento & desenvolvimento , Ciclídeos/metabolismo , Dieta/veterinária , Suplementos Nutricionais , Proteínas de Choque Térmico HSP70/genética , Polissacarídeos/farmacologia , Tálio/administração & dosagem , Tálio/toxicidade
7.
J Inorg Biochem ; 222: 111469, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34192625

RESUMO

Among transition and non-transition metals, thallium is a unique case of an element which, despite its known toxicity, provides interesting challenges through its biology and chemistry linked to diagnosis of human pathophysiologies. Poised to investigate in-depth the structural and electronic aspects of thallium involvement in physiological processes, the synthetic exploration of aqueous binary systems of Tl(I) with physiological binders from the family of hydroxycarboxylic acids (glycolic, lactic, mandelic and citric acid) was pursued in a pH-specific fashion. The isolated crystalline coordination polymers, emerging from that effort, were physicochemically characterized through elemental analysis, FT-IR, ESI-MS, 1H-/13C-NMR, and X-ray crystallography. The coordination environment of thallium in each molecular Tl(I) assembly, along with lattice dimensionality (2D3D), reflects the contributions of the ligands, collectively exemplifying interactions probed into though BVS and Hirshfeld surface analysis. The results portray a well-defined solid-state and solution profile for all species investigated, thereby providing the basis for their subsequent selection into in vitro biological studies involving the (patho)physiological cell lines 3T3-L1, Saos-2, C2C12, and MCF-7. Biotoxicity profiles, encompassing cell viability, morphology, and cell growth support clearly a concentration-, time-, and cell tissue-specific behavior for the chosen Tl(I) compounds in a structure-specific fashion. Collectively, the chemical experimental data support the biological results in formulating a structure-specific behavior for Tl(I)-hydroxycarboxylato species with respect to biotoxicity mechanisms in a (patho)physiological environment. The accrued knowledge stands as the foreground for further investigation into the relevant biological chemistry of Tl(I) and molecular technologies targeting its sequestration and removal from cellular media.


Assuntos
Ácidos Carboxílicos/toxicidade , Complexos de Coordenação/toxicidade , Polímeros/toxicidade , Tálio/toxicidade , Células 3T3-L1 , Animais , Ácidos Carboxílicos/síntese química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/síntese química , Humanos , Ligantes , Camundongos , Polímeros/síntese química , Tálio/química , Água/química
8.
J Pharmacol Toxicol Methods ; 110: 107087, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34153452

RESUMO

The aim of the study was to develop fast and accurate method for assessment of intracellular level of cadmium (Cd) and thallium (Tl), and to establish accumulation of the metals in the cells. HepG2 cells were treated with Cd or Tl (1.0 or 10.0 mg/L; 24 h) and level of Cd or Tl was assessed. ICP-MS was applied and the method was optimized and validated. Correlation coefficient (R2) for Cd was 0.9999 with intercept 0.0732 while for Tl was 1.00009 with intercept -0.1497, and limit of detection (LOD) for Cd was 0.020 µg/L and for Tl 0.097 µg/L. Both metals, Cd and Tl, accumulate in the cells in concentration-dependent manner. However, higher uptake of Cd in comparison to Tl was observed. Cells treated with the same concentration of the metal (1.0 mg/L) accumulated 10.0% of Cd and 1.0% of Tl. Higher uptake of Cd than Tl can explain higher toxicity of Cd toward HepG2 cells. Obtained results imply to the importance of monitoring the level of metals in the cells in order to connect changes at the molecular level with exposure to specific metal.


Assuntos
Cádmio , Tálio , Cádmio/toxicidade , Tálio/toxicidade
9.
Hum Exp Toxicol ; 39(6): 808-815, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32009467

RESUMO

CONTEXT: Heavy metals, including thallium and lead, are introduced to illicit drug users' body as a result of using drugs such as cocaine and heroin. OBJECTIVE: This study aimed to determine urine, blood, and hair thallium (Tl) concentrations in illicit opioid users along with the relevant clinical signs and symptoms consistent with thallotoxicosis and to compare them with the corresponding variables in the control non-opioid user group. MATERIALS AND METHODS: This case-control study was conducted on 50 illicit opioid users who had abused opioids continuously for more than a year, referred to Amirie Drug Abuse Treatment Clinic in Kashan, Iran. The control group included 50 non-opioid users. Thallium concentrations in urine, blood, and hair were assessed in both groups (n = 100) using electrothermal (graphite furnace) atomic absorption spectrometry (ET AAS, GF AAS). RESULTS: In the studied group, the median (interquartile range) concentrations of thallium in urine, blood, and hair were 54.8 ± 79.9 µg/L, 14.5 ± 11.1 µg/L, and 5.4 ± 3.7 µg/g, respectively; these values were 4.8 ± 5.2 µg/L, 2.5 ± 2.4 µg/L, and 1.4 ± 1.1 µg/g, respectively, in the control group. There were significant differences in urine, blood, and hair thallium concentrations between the study group and the control group (p < 0.001). There were significant correlations between duration of illicit opioid use and urine thallium concentrations (r = 0.394, p = 0.005) and hair thallium concentrations (r = 0.293, p = 0.039), but not with blood thallium concentrations (r = 0.246, p = 0.085). Urine and blood thallium concentrations of illicit opioid users with clinical signs and symptoms consistent with thallotoxicosis of weakness (p = 0.01), depression (p = 0.03), and headache (p = 0.03) were higher than users without these problems. DISCUSSION AND CONCLUSION: The results of the study showed that thallium concentrations in urine, blood, and hair in illicit opioid users were significantly higher than the comparable concentrations in the control group. This can be due to the use of illicit opioids adulterated with thallium. Also, this study showed long-term illicit opioid use may lead to thallium exposure. In addition, cigarette smoking was associated with increased thallium exposure.


Assuntos
Cabelo/química , Transtornos Relacionados ao Uso de Opioides , Tálio , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Analgésicos Opioides/toxicidade , Estudos de Casos e Controles , Feminino , Heroína/toxicidade , Humanos , Drogas Ilícitas/toxicidade , Irã (Geográfico)/epidemiologia , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/sangue , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/urina , Ópio/toxicidade , Tálio/análise , Tálio/sangue , Tálio/toxicidade , Tálio/urina , Adulto Jovem
10.
Sci Rep ; 9(1): 6905, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-31061518

RESUMO

Thallium is considered as an emergent contaminant owing to its potential use in the superconductor alloys. The monovalent thallium, Tl(I), is highly toxic to the animals as it can affect numerous metabolic processes. Here we observed that Tl(I) decreased protein synthesis and phosphorylated eukaryotic initiation factor 2α. Although Tl(I) has been shown to interact with the sulfhydryl groups of proteins and cause the accumulation of reactive oxygen species, it did not activate endoplasmic reticulum stress. Notably, the level of 60S ribosomal subunit showed significant under-accumulation after the Tl(I) treatment. Given that Tl(I) shares similarities with potassium in terms of the ionic charge and atomic radius, we proposed that Tl(I) occupies certain K+-binding sites and inactivates the ribosomal function. However, we observed neither activation of ribophagy nor acceleration of the proteasomal degradation of 60S subunits. On the contrary, the ribosome synthesis pathway was severely blocked, i.e., the impairment of rRNA processing, deformed nucleoli, and accumulation of 60S subunits in the nucleus were observed. Although p53 remained inactivated, the decreased c-Myc and increased p21 levels indicated the activation of nucleolar stress. Therefore, we proposed that Tl(I) interfered the ribosome synthesis, thus resulting in cell growth inhibition and lethality.


Assuntos
Nucléolo Celular/efeitos dos fármacos , Nucléolo Celular/metabolismo , Fator de Iniciação 2 em Eucariotos/biossíntese , Estresse Oxidativo/efeitos dos fármacos , Tálio/toxicidade , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fator de Iniciação 2 em Eucariotos/metabolismo , Células HEK293 , Humanos , Fosforilação/efeitos dos fármacos , Subunidades Ribossômicas Maiores de Eucariotos/efeitos dos fármacos , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Environ Int ; 127: 181-189, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30921669

RESUMO

BACKGROUND: Thallium (Tl) is a cumulative high toxicant in the environment, but few longitudinal studies have investigated the respiratory impairment of Tl exposure. OBJECTIVES: This study aimed to evaluate the effect of Tl and its interaction with smoking on lung function decline, and explore the potential mechanisms. METHODS: The baseline and follow-up lung functions were measured from a prospective cohort study of 1243 workers, who were followed from 2010 to 2014. Their baseline urinary levels of Tl were determined. We also measured the plasma C-reactive protein (CRP) and urinary 8-iso-prostaglandin-F2α (8-iso-PGF2α) in a randomly selected subcohort of 474 subjects. RESULTS: The results showed that a 2-fold increase in urinary Tl was associated with 29.81 mL (95%CI: 3.83-55.80) increased decline in forced expiratory volume in 1 s (FEV1). The effect was more pronounced among heavy-smokers (≥15 pack-years) [ß(95%CI) = 56.42 mL (9.66-103.19)]. In particular, compared to never-smokers with low Tl, heavy-smokers with high Tl had a separate 158.44 mL (95%CI: 54.88-262.00) and 4.58% (95%CI: 1.40-7.76) increased declines in FEV1 and percentage of predicted (ppFEV1), respectively. There was a significant interaction between Tl and smoking intensity on ppFEV1 decline (Pint = 0.034). More importantly, the increasing level of urinary Tl was correlated with elevated CRP and 8-iso-PGF2α. CONCLUSION: Our prospective cohort study identified that exposure to high Tl had a deleterious effect on lung function, and this effect may be enhanced by tobacco smoking. Increased inflammation may partly contribute to the joint effects of Tl and smoking on impaired lung function, but the biological mechanisms need further explorations.


Assuntos
Tálio/toxicidade , Fumar Tabaco , Adulto , Feminino , Volume Expiratório Forçado , Humanos , Estudos Longitudinais , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Testes de Função Respiratória
12.
Neurotox Res ; 33(3): 634-640, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29313218

RESUMO

The mechanisms by which the heavy metal thallium (Tl+) produces toxicity in the brain remain unclear. Herein, isolated synaptosomal/mitochondrial P2 crude fractions from adult rat brains were exposed to Tl+ (5-250 µM) for 30 min. Three toxic endpoints were evaluated: mitochondrial dysfunction, lipid peroxidation, and Na+/K+-ATPase activity inhibition. Concentration-response curves for two of these endpoints revealed the optimum concentration of Tl+ to induce damage in this preparation, 5 µM. Toxic markers were also estimated in preconditioned synaptosomes incubated in the presence of the N-methyl-D-aspartate receptor antagonist kynurenic acid (KYNA, 50 µM), the cannabinoid receptor agonist WIN 55,212-2 (1 µM), or the antioxidant S-allyl-L-cysteine (SAC, 100 µM). All these agents prevented Tl+ toxicity, though SAC did it with lower efficacy. Our results suggest that energy depletion, oxidative damage, and Na+/K+-ATPase activity inhibition account for the toxic pattern elicited by Tl+ in nerve terminals. In addition, the efficacy of the drugs employed against Tl+ toxicity supports an active role of excitatory/cannabinoid and oxidative components in the toxic pattern elicited by the metal.


Assuntos
Encéfalo/ultraestrutura , Mitocôndrias/efeitos dos fármacos , Sinaptossomos/efeitos dos fármacos , Tálio/toxicidade , Animais , Benzoxazinas/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Cisteína/análogos & derivados , Cisteína/farmacologia , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Cinurênico/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Morfolinas/farmacologia , Naftalenos/farmacologia , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/metabolismo
13.
Arch Toxicol ; 92(1): 195-211, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28866748

RESUMO

Thallium (Tl) is a toxic heavy metal that causes oxidative stress both in vitro and in vivo. In this work, we evaluated the production of oxygen (ROS)- and nitrogen (RNS)-reactive species in adherent PC12 (PC12adh) cells exposed for 0.5-6 h to Tl(I) or Tl(III) (10-100 µM). In this system, Tl(I) induced mostly H2O2 generation while Tl(III) induced H2O2 and ONOO·- generation. Both cations enhanced iNOS expression and activity, and decreased CuZnSOD expression but without affecting its activity. Tl(I) increased MnSOD expression and activity but Tl(III) decreased them. NADPH oxidase (NOX) activity remained unaffected throughout the period assessed. Oxidant levels returned to baseline values after 6 h of incubation, suggesting a response of the antioxidant defense system to the oxidative insult imposed by the cations. Tl also affected the glutathione-dependent system: while Tl(III) increased glutathione peroxidase (GPx) expression and activity, Tl(I) and Tl(III) decreased glutathione reductase (GR) expression. However, GR activity was mildly enhanced by Tl(III). Finally, thioredoxin-dependent system was evaluated. Only Tl(I) increased 2-Cys peroxiredoxins (2-Cys Prx) expression, although both cations increased their activity. Tl(I) increased cytosolic thioredoxin reductase (TrxR1) and decreased mitochondrial (TrxR2) expression. Tl(III) had a biphasic effect on TrxR1 expression and slightly increased TrxR2 expression. Despite of this, both cations increased total TrxR activity. Obtained results suggest that in Tl(I)-exposed PC12adh cells, there is an early response to oxidative stress mainly by GSH-dependent system while in Tl(III)-treated cells both GSH- and Trx-dependent systems are involved.


Assuntos
Antioxidantes/metabolismo , Glutationa/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Tálio/toxicidade , Tiorredoxinas/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Enzimas/metabolismo , Inativação Metabólica/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo , Tálio/administração & dosagem , Tálio/química , Testes de Toxicidade/métodos
14.
Bull Environ Contam Toxicol ; 99(5): 574-581, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28913548

RESUMO

Thallium is a rare-earth element, but widely distributed in water environments, posing a potential risk to our health. This study was designed to investigate the chronic effects of thallium based on physiological responses, gene expression, and changes in the activity of relevant enzymes in adult zebra fish exposed to thallium at low doses. The endpoints assessed include mRNA expression of metallothionein (MT)2 and heat shock protein HSP70; enzymatic activities of superoxide dismutase (SOD) and Na+/K+-ATPase; and the histopathology of gill, gonad, and liver tissues. The results showed significant increases in HSP70 mRNA expression following exposure to 100 ng/L thallium and in MT2 expression following exposure to 500 ng/L thallium. Significantly higher activities were observed for SOD in liver and Na+/K+-ATPase activity in gill in zebra fish exposed to thallium (20 and 100 ng/L, respectively) in comparison to control fish. Gill, liver, and gonad tissues displayed different degrees of damage. The overall results imply that thallium may cause toxicity to zebra fish at environmentally relevant aqueous concentrations.


Assuntos
Tálio/toxicidade , Poluentes Químicos da Água/toxicidade , Peixe-Zebra/fisiologia , Animais , Brânquias/metabolismo , Gônadas/metabolismo , Fígado/metabolismo , Metalotioneína/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Superóxido Dismutase/metabolismo , Testes de Toxicidade , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
15.
Arch Toxicol ; 91(3): 1157-1174, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27412756

RESUMO

We have reported recently that the proliferation of PC12 cells exposed to micromolar concentrations of Tl(I) or Tl(III) has different outcomes, depending on the absence (EGF- cells) or the presence (EGF+ cells) of epidermal growth factor (EGF) added to the media. In the current work, we investigated whether EGF supplementation could also modulate the extent of Tl(I)- or Tl(III)-induced cell apoptosis. Tl(I) and Tl(III) (25-100 µM) decreased cell viability in EGF- but not in EGF+ cells. In EGF- cells, Tl(I) decreased mitochondrial potential, enhanced H2O2 generation, and activated mitochondrial-dependent apoptosis. In addition, Tl(III) increased nitric oxide production and caused a misbalance between the anti- and pro-apoptotic members of Bcl-2 family. Tl(I) increased ERK1/2, JNK, p38, and p53 phosphorylation in EGF- cells. In these cells, Tl(III) did not affect ERK1/2 and JNK phosphorylation but increased p53 phosphorylation that was related to the promotion of cell senescence. In addition, this cation significantly activated p38 in both EGF- and EGF+ cells. The specific inhibition of ERK1/2, JNK, p38, or p53 abolished Tl(I)-mediated EGF- cell apoptosis. Only when p38 activity was inhibited, Tl(III)-mediated apoptosis was prevented in EGF- and EGF+ cells. Together, current results indicate that EGF partially prevents the noxious effects of Tl by preventing the sustained activation of MAPKs signaling cascade that lead cells to apoptosis and point to p38 as a key mediator of Tl(III)-induced PC12 cell apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Tálio/toxicidade , Animais , Apoptose/fisiologia , Sobrevivência Celular/efeitos dos fármacos , MAP Quinase Quinase 4/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células PC12/efeitos dos fármacos , Células PC12/metabolismo , Ratos , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Int J Toxicol ; 34(6): 505-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26350230

RESUMO

Thallium (Tl(+)) is a toxic heavy metal capable of increasing oxidative damage and disrupting antioxidant defense systems. Thallium invades the brain cells through potassium channels, increasing neuronal excitability, although until now the possible role of glutamatergic transmission in this event has not been investigated. Here, we explored the possible involvement of a glutamatergic component in the Tl(+)-induced toxicity through the N-methyl-d-aspartate (NMDA) receptor antagonist dizocilpine (MK-801) in rats. The effects of MK-801 (1 mg/kg, intraperitoneally [ip]) on early (24 hours) motor alterations, lipid peroxidation, reduced glutathione (GSH) levels, and GSH peroxidase activity induced by Tl(+) acetate (32 mg/kg, ip) were evaluated in adult rats. MK-801 attenuated the Tl(+)-induced hyperactivity and lipid peroxidation in the rat striatum, hippocampus and midbrain, and produced mild effects on other end points. Our findings suggest that glutamatergic transmission via NMDA receptors might be involved in the Tl(+)-induced altered regional brain redox activity and motor performance in rats.


Assuntos
Comportamento Animal/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Maleato de Dizocilpina/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Tálio/toxicidade , Animais , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Injeções Intraperitoneais , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Wistar , Transmissão Sináptica/efeitos dos fármacos
17.
Environ Toxicol ; 30(2): 232-41, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23996974

RESUMO

Thallium(I) is a highly toxic heavy metal; however, up to now, its mechanisms are poorly understood. The authors' previous studies showed that this compound could induce reactive oxygen species (ROS) formation, reduced glutathione (GSH) oxidation, membrane lipid peroxidation, and mitochondrial membrane potential (MMP) collapse in isolated rat hepatocyte. Because the liver is the storage site of thallium, it seems that the liver mitochondria are one of the important targets for hepatotoxicity. In this investigation, the effects of thallium on mitochondria were studied to investigate its mechanisms of toxicity. Mitochondria were isolated from rat liver and incubated with different concentrations of thallium (25-200 µM). Thallium(I)-treated mitochondria showed a marked elevation in oxidative stress parameters accompanied by MMP collapse when compared with the control group. These results showed that different concentrations of thallium (25-200 µM) induced a significant (P < 0.05) increase in mitochondrial ROS formation, ATP depletion, GSH oxidation, mitochondrial outer membrane rupture, mitochondrial swelling, MMP collapse, and cytochrome c release. In general, these data strongly supported that the thallium(I)-induced liver toxicity is a result of the disruptive effect of this metal on the mitochondrial respiratory complexes (I, II, and IV), which are the obvious causes of metal-induced ROS formation and ATP depletion. The latter two events, in turn, trigger cell death signaling via opening of mitochondrial permeability transition pore and cytochrome c expulsion.


Assuntos
Mitocôndrias Hepáticas/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Tálio/toxicidade , Trifosfato de Adenosina/metabolismo , Animais , Citocromos c/metabolismo , Glutationa/metabolismo , Peróxido de Hidrogênio/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Poro de Transição de Permeabilidade Mitocondrial , Dilatação Mitocondrial/efeitos dos fármacos , Permeabilidade , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
18.
Arch Toxicol ; 86(11): 1667-80, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22669516

RESUMO

The mechanisms that mediate thallium (Tl) toxicity are still not completely understood. The exposure of rat pheochromocytoma (PC12) cells to Tl(I) or Tl(III) activates both mitochondrial (Tl(I) and Tl(III)) and extrinsic (Tl(III)) pathways of apoptosis. In this work we evaluated the hypothesis that the effects of Tl(III) may be mediated by the damage to lysosomes, where it might be incorporated following the route of iron uptake. PC12 cells exposed for 3 h to 100 µM Tl(III) presented marked endosomal acidification, effect that was absent when cells were incubated in a serum-free medium and that was fully recovered when the latter was supplemented with transferrin. After 6 h of incubation the colocalization of cathepsins D and B with the lysosomal marker Lamp-1 was decreased together with an increase in the total activity of the enzymes. A permanent damage to lysosomes after 18 h of exposure was evidenced from the impairment of acridine orange uptake. Cathepsin D caused the cleavage of pro-apoptotic protein BID that is involved in the activation of the intrinsic pathway of apoptosis. Supporting that, BID cleavage and the activation of caspase 3 by Tl(III) were fully prevented when cells were preincubated with cathepsin D inhibitor (pepstatin A) and only partially prevented when cathepsin B inhibitor (E64d) was used. None of these inhibitors affected BID cleavage or caspase 3 activation in Tl(I)-treated cells. Together, experimental results support the role of Tl(III) uptake by the acidic cell compartments and their involvement in the early steps of Tl(III)-mediated PC12 cells apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Endossomos/metabolismo , Lisossomos/metabolismo , Tálio/toxicidade , Animais , Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspase 3/metabolismo , Catepsina B/antagonistas & inibidores , Catepsina B/metabolismo , Catepsina D/antagonistas & inibidores , Catepsina D/metabolismo , Compartimento Celular/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Proteínas de Membrana Lisossomal/metabolismo , Lisossomos/efeitos dos fármacos , Células PC12/efeitos dos fármacos , Pepstatinas/farmacologia , Ratos , Tálio/farmacologia
19.
Environ Toxicol Pharmacol ; 32(1): 107-12, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21787736

RESUMO

Heavy metals are ubiquitous in the environment and exposure through food and water as well as occupational sources can constitute a potential threat to human health. The mechanisms of heavy metal damage include the production of free radicals that alter mitochondrial activity, affecting cellular types like neurons and muscular fibres. We examined whether rats exposed subchronically via drinking water to low doses of heavy metals can produce alterations in muscle. Results showed that the proportion of ragged red fibres increased in muscle of rats exposed to lead and thallium, likewise slight changes in enzymatic activity of muscular fibres were also observed.


Assuntos
Cádmio/toxicidade , Chumbo/toxicidade , Músculos/efeitos dos fármacos , Tálio/toxicidade , Adenosina Trifosfatases/metabolismo , Animais , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Poluentes Ambientais/toxicidade , Masculino , Músculos/metabolismo , Músculos/patologia , NAD/metabolismo , Ratos , Ratos Wistar
20.
Environ Toxicol ; 25(5): 456-67, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20549620

RESUMO

Thallium (Tl) is a highly toxic heavy metal though up to now its mechanisms are poorly understood. In this study, we comparatively investigated the cytotoxic mechanisms of Tl(I) and Tl(III) in isolated rat hepatocytes. Both Tl(I) and Tl(III) cytotoxicities were associated with reactive oxygen species (ROS) formation, lipid peroxidation, collapse of mitochondrial membrane potential, activation of caspases cascade, lysosomal membrane leakiness, and cellular proteolysis. Hepatocyte glutathione (GSH) was also rapidly oxidized. GSH-depleted hepatocytes were more resistant to Tl(I)-induced cytotoxicity, ROS formation and lipid peroxidation. This suggests that Tl(I) is reductively activated by GSH. On the other hand, GSH-depleted hepatocytes were much more sensitive to Tl(III)-induced cytotoxicity, ROS formation, and lipid peroxidation. This suggests that GSH only plays an antioxidant role against Tl(III) cytotoxicity. Our results also showed that CYP2E1 involves in Tl(I) and Tl(III) oxidative stress cytotoxicity mechanism and both cations detoxified via methylation. In conclusion, both Tl(I) and Tl(III) cytotoxicities were associated with mutual mitochondrial/lysosomal injuries (cross-talk) initiated by increased ROS formation resulted from metal-CYP2E1 destructive interaction or metal-induced disruption of mitochondrial electron transfer chain.


Assuntos
Hepatócitos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Tálio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Cátions/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citocromo P-450 CYP2E1/metabolismo , Transporte de Elétrons , Glutationa/antagonistas & inibidores , Glutationa/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Masculino , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias Hepáticas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA