Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-37230210

RESUMO

In nature, arsenic is mostly found in the form of inorganic compounds. Inorganic arsenic compounds have a variety of uses and are currently used in the manufacture of pesticides, preservatives, pharmaceuticals, etc. While inorganic arsenic is widely used, arsenic pollution is increasing worldwide. Public hazards caused by arsenic contamination of drinking water and soil are becoming increasingly evident. Epidemiological and experimental studies have linked inorganic arsenic exposure to the development of many diseases, including cognitive impairment, cardiovascular failure, cancer, etc. Several mechanisms have been proposed to explain the effects caused by arsenic, such as oxidative damage, DNA methylation, and protein misfolding. Understanding the toxicology and potential molecular mechanisms of arsenic can help mitigate its harmful effects. Therefore, this paper reviews the multiple organ toxicity of inorganic arsenic in animals, focusing on the various toxicity mechanisms of arsenic-induced diseases in animals. In addition, we have summarized several drugs that can have therapeutic effects on arsenic poisoning in pursuit of reducing the harm of arsenic contamination from different pathways.


Assuntos
Intoxicação por Arsênico , Arsênio , Arsenicais , Água Potável , Animais , Arsênio/toxicidade , Arsênio/análise , Intoxicação por Arsênico/tratamento farmacológico , Intoxicação por Arsênico/metabolismo , Poluição Ambiental
2.
Hum Exp Toxicol ; 41: 9603271221121313, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35968550

RESUMO

Increasing evidence supports the role of arsenic in dysregulated immune and inflammation responses, while, safe and effective treatments have not been fully examined. Rosa roxburghii Tratt (RRT), a traditional Chinese edible fruit with potential immunoregulatory activities, was considered as a dietary supplement to explore its protective effects and possible mechanism in arsenic-induced dysregulated inflammation responses. We enrolled 209 arsenicosis patients and 41 controls to obtain baseline data, including the degree of arsenic poisoning prior to the RRT juice (RRTJ) intervention. Then, based on criteria of inclusion and exclusion and the principle of voluntary participation, 106 arsenicosis patients who volunteered to receive treatment were divided into RRTJ (n = 53) and placebo (n = 53) groups randomly. After three months follow-up, 89 subjects (46 and 43 of the RRTJ and placebo groups, respectively) completed the study and were examined for the effects and possible mechanisms of RRTJ on the Th17 cells-related pro-inflammatory responses in peripheral blood mononuclear cells (PBMCs). The PBMCs had higher levels of Th17 and Th17-related inflammatory cytokines IL-17, IL-6, and RORγt. Furthermore, the gene expressions of STAT3 and SOCS3 in PBMCs increased and decreased, respectively. Conversely, RRTJ decreased the number of Th17 cells, secretion of IL-17, IL-6, RORγt, and relative mRNA levels of STAT3, and increased the transcript levels of SOCS3. This study provides limited evidence that possible immunomodulatory effects of RRTJ on the critical regulators, IL-6 and STAT3, of the Th17 cells in arsenicosis patients, which indicated that IL-6/STAT3 pathway might appear as a potential therapeutic target in arsenicosis.


Assuntos
Intoxicação por Arsênico , Arsênio , Fitoterapia , Preparações de Plantas , Rosa , Arsênio/toxicidade , Intoxicação por Arsênico/genética , Intoxicação por Arsênico/metabolismo , Intoxicação por Arsênico/terapia , Sucos de Frutas e Vegetais , Humanos , Inflamação/induzido quimicamente , Interleucina-17/metabolismo , Interleucina-6 , Leucócitos Mononucleares/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Preparações de Plantas/metabolismo , Preparações de Plantas/uso terapêutico , Rosa/metabolismo
3.
Chem Res Toxicol ; 35(6): 916-934, 2022 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-35575307

RESUMO

Arsenic toxicity is a major concern due to its deleterious consequences for human health. Rapid industrialization also has weakened the quality of the environment by introducing pollutants that may disrupt balanced ecosystems, adversely and irreversibly impacting humans, plants, and animals. Arsenic, an important toxicant among all environmental hazards, can lead to several detrimental effects on cells and organs, impacting the overall quality of life. Nevertheless, arsenic also has a rich history as a chemotherapeutic agent used in ancient days for the treatment of diseases such as malaria, cancer, plague, and syphilis when other chemotherapeutic agents were yet to be discovered. Arsenicosis-mediated disorders remain a serious problem due to the lack of effective therapeutic options. Initially, chelation therapy was used to metabolically eliminate arsenic by forming a complex, but adverse effects limited their pharmacological use. More recently, plant-based products have been found to provide significant relief from the toxic effects of arsenic poisoning. They act by different mechanisms affecting various cellular processes. Phytoconstituents such as curcumin, quercetin, diallyl trisulfide, thymoquinone, and others act via various molecular pathways, primarily by attenuating oxidative damage, membrane damage, DNA damage, and proteinopathies. Nonetheless, most of the phytochemicals reviewed here protect against the adverse effects of metal or metalloid exposure, supporting their consideration as alternatives to chelation therapy. These agents, if used prophylactically and in conjunction with other chemotherapeutic agents, may provide an effective approach for management of arsenic toxicity. In a few instances, such strategies like coadministration of phytochemicals with a known chelating agent have led to more pronounced elimination of arsenic from the body with lesser off-site adverse effects. This is possible because combination treatment ensures the use of a reduced dose of chelating agent with a phytochemical without compromising treatment. Thus, these therapies are more practical than conventional therapeutic agents in ameliorating arsenic-mediated toxicity. This review summarizes the potential of phytochemicals in alleviating arsenic toxicity on the basis of available experimental and clinical evidence.


Assuntos
Intoxicação por Arsênico , Arsênio , Animais , Arsênio/metabolismo , Arsênio/toxicidade , Intoxicação por Arsênico/tratamento farmacológico , Intoxicação por Arsênico/metabolismo , Quelantes , Ecossistema , Compostos Fitoquímicos/farmacologia , Compostos Fitoquímicos/uso terapêutico , Qualidade de Vida
4.
Molecules ; 27(3)2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35164302

RESUMO

Neurotoxicity is a serious health problem of patients chronically exposed to arsenic. There is no specific treatment of this problem. Oxidative stress has been implicated in the pathological process of neurotoxicity. Polyphenolics have proven antioxidant activity, thereby offering protection against oxidative stress. In this study, we have isolated the polyphenolics from Acacia nilotica and investigated its effect against arsenic-induced neurotoxicity and oxidative stress in mice. Acacia nilotica polyphenolics prepared from column chromatography of the crude methanol extract using diaion resin contained a phenolic content of 452.185 ± 7.879 mg gallic acid equivalent/gm of sample and flavonoid content of 200.075 ± 0.755 mg catechin equivalent/gm of sample. The polyphenolics exhibited potent antioxidant activity with respect to free radical scavenging ability, total antioxidant activity and inhibition of lipid peroxidation. Administration of arsenic in mice showed a reduction of acetylcholinesterase activity in the brain which was counteracted by Acacia nilotica polyphenolics. Similarly, elevation of lipid peroxidation and depletion of glutathione in the brain of mice was effectively restored to normal level by Acacia nilotica polyphenolics. Gallic acid methyl ester, catechin and catechin-7-gallate were identified in the polyphenolics as the major active compounds. These results suggest that Acacia nilotica polyphenolics due to its strong antioxidant potential might be effective in the management of arsenic induced neurotoxicity.


Assuntos
Acacia , Antioxidantes/uso terapêutico , Intoxicação por Arsênico/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Polifenóis/uso terapêutico , Acacia/química , Animais , Antioxidantes/química , Arsênio/toxicidade , Intoxicação por Arsênico/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Polifenóis/química
5.
Toxicol Appl Pharmacol ; 431: 115738, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34619159

RESUMO

Millions of people worldwide are exposed to arsenic, a metalloid listed as one of the top chemical pollutants of concern to human health. Epidemiological and experimental studies link arsenic exposure to the development of cancer and other diseases. Several mechanisms have been proposed to explain the effects induced by arsenic. Notably, arsenic and its metabolites interact with proteins by direct binding to individual cysteine residues, cysteine clusters, zinc finger motifs, and RING finger domains. Consequently, arsenic interactions with proteins disrupt the functions of proteins and may lead to the development and progression of diseases. In this review, we focus on current evidence in the literature that implicates the interaction of arsenic with proteins as a mechanism of arsenic toxicity. Data show that arsenic-protein interactions affect multiple cellular processes and alter epigenetic regulation, cause endocrine disruption, inhibit DNA damage repair mechanisms, and deregulate gene expression, among other adverse effects.


Assuntos
Intoxicação por Arsênico/etiologia , Arsenicais/efeitos adversos , Disruptores Endócrinos/efeitos adversos , Poluentes Ambientais/efeitos adversos , Proteínas/metabolismo , Animais , Intoxicação por Arsênico/genética , Intoxicação por Arsênico/metabolismo , Arsenicais/metabolismo , Cisteína , Reparo do DNA/efeitos dos fármacos , Disruptores Endócrinos/metabolismo , Poluentes Ambientais/metabolismo , Epigênese Genética/efeitos dos fármacos , Humanos , Ligação Proteica , Proteínas/genética , Domínios RING Finger , Medição de Risco , Dedos de Zinco
6.
BMC Pharmacol Toxicol ; 22(1): 19, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33827703

RESUMO

BACKGROUND: Arsenic poisoning affects millions of people. The inorganic forms of arsenic are more toxic. Treatment for arsenic poisoning relies on chelation of extracellularly circulating arsenic molecules by 2,3-dimecaptosuccinic acid (DMSA). As a pharmacological intervention, DMSA is unable to chelate arsenic molecules from intracellular spaces. The consequence is continued toxicity and cell damage in the presence of DMSA. A two-pronged approach that removes extracellular arsenic, while protecting from the intracellular arsenic would provide a better pharmacotherapeutic outcome. In this study, Coenzyme Q10 (CoQ10), which has been shown to protect from intracellular organic arsenic, was administered separately or with DMSA; following oral exposure to sodium meta-arsenite (NaAsO2) - a very toxic trivalent form of inorganic arsenic. The aim was to determine if CoQ10 alone or when co-administered with DMSA would nullify arsenite-induced toxicity in mice. METHODS: Group one represented the control; the second group was treated with NaAsO2 (15 mg/kg) daily for 30 days, the third, fourth and fifth groups of mice were given NaAsO2 and treated with 200 mg/kg CoQ10 (30 days) and 50 mg/kg DMSA (5 days) either alone or in combination. RESULTS: Administration of CoQ10 and DMSA resulted in protection from arsenic-induced suppression of RBCs, haematocrit and hemoglobin levels. CoQ10 and DMSA protected from arsenic-induced alteration of WBCs, basophils, neutrophils, monocytes, eosinophils and platelets. Arsenite-induced dyslipidemia was nullified by administration of CoQ10 alone or in combination with DMSA. Arsenite induced a drastic depletion of the liver and brain GSH; that was significantly blocked by CoQ10 and DMSA alone or in combination. Exposure to arsenite resulted in significant elevation of liver and kidney damage markers. The histological analysis of respective organs confirmed arsenic-induced organ damage, which was ameliorated by CoQ10 alone or when co-administered with DMSA. When administered alone, DMSA did not prevent arsenic-driven tissue damage. CONCLUSIONS: Findings from this study demonstrate that CoQ10 and DMSA separately or in a combination, significantly protect against arsenic-driven toxicity in mice. It is evident that with further pre-clinical and clinical studies, an adjunct therapy that incorporates CoQ10 alongside DMSA may find applications in nullifying arsenic-driven toxicity.


Assuntos
Antídotos/uso terapêutico , Intoxicação por Arsênico/tratamento farmacológico , Arsenitos/toxicidade , Quelantes/uso terapêutico , Substâncias Protetoras/uso terapêutico , Compostos de Sódio/toxicidade , Succímero/uso terapêutico , Ubiquinona/análogos & derivados , Animais , Intoxicação por Arsênico/sangue , Intoxicação por Arsênico/metabolismo , Intoxicação por Arsênico/patologia , Células Sanguíneas/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Quimioterapia Combinada , Glutationa/metabolismo , Hematócrito , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Ubiquinona/uso terapêutico
8.
Food Chem Toxicol ; 145: 111706, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32871193

RESUMO

Activation of the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor E2 related factor 2 (Nrf2) system plays a role in repression of xenobiotic toxicity. The Coriandrum sativum L. leaf extract (CSLE) contains various aliphatic electrophiles such as (E)-2-decenal and (E)-2-dodecenal. In the present study, we examined the activation of Nrf2 coupled to chemical modification of Keap1 mediated by (E)-2-alkenals in CSLE, and the protective role of CSLE and (E)-2-alkenals against inorganic arsenite (iAsIII) cytotoxicity. Ultra-performance liquid chromatography-elevated collision energy mass spectrometry analysis revealed that (E)-2-decenal modified recombinant Keap1 at Cys241, Cys249, Cys257 and His274. Exposure of HepG2 cells to CSLE, (E)-2-decenal, or (E)-2-dodecenal upregulated Nrf2-related downstream signaling such as expression of phase-II xenobiotic-metabolizing enzymes and phase-III transporters involved in cytoprotection against iAsIII. Pretreatment with CSLE or (E)-2-butenal, a prototype of (E)-2-alkenal, prior to iAsIII exposure suppressed accumulation of iAsIII significantly and reduced iAsIII-induced cytotoxicity in cells. Oral administration of CSLE to C57BL/6 mice upregulated downstream proteins of Nrf2 and reduced accumulation of arsenic in liver tissue. The present study indicates that CSLE containing (E)-2-alkenals activates Nrf2, leading to a reduction in arsenic accumulation in vivo.


Assuntos
Intoxicação por Arsênico/tratamento farmacológico , Arsênio/toxicidade , Coriandrum/química , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Extratos Vegetais/administração & dosagem , Animais , Antioxidantes/administração & dosagem , Intoxicação por Arsênico/genética , Intoxicação por Arsênico/metabolismo , Feminino , Células Hep G2 , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , Folhas de Planta/química , Transdução de Sinais/efeitos dos fármacos
9.
Biomed Res Int ; 2020: 9414196, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32802886

RESUMO

BACKGROUND: Arsenic is a toxic metalloid widely present in nature, and arsenic poisoning in drinking water is a serious global public problem. Glutathione is an important reducing agent that inhibits arsenic-induced oxidative stress and participates in arsenic methylation metabolism. Therefore, glutathione plays an important role in regulating arsenic toxicity. In recent years, a large number of studies have shown that arsenic can regulate glutathione synthesis in many ways, but there are many contradictions in the research results. At present, the mechanism of the effect of arsenic on glutathione synthesis has not been elucidated. OBJECTIVE: We will conduct a meta-analysis to illustrate the effects of arsenic on GSH synthesis precursors Glu, Cys, Gly, and rate-limiting enzyme γ-GCS in mammalian models, as well as the regulation of p38/Nrf2 of γ-GCS subunit GCLC, and further explore the molecular mechanism of arsenic affecting glutathione synthesis. RESULTS: This meta-analysis included 30 studies in vivo and 58 studies in vitro, among which in vivo studies showed that arsenic exposure could reduce the contents of GSH (SMD = -2.86, 95% CI (-4.45, -1.27)), Glu (SMD = -1.11, 95% CI (-2.20,-0.02)), and Cys (SMD = -1.48, 95% CI (-2.63, -0.33)), with no statistically significant difference in p38/Nrf2, GCLC, and GCLM. In vitro studies showed that arsenic exposure increased intracellular GSH content (SMD = 1.87, 95% CI (0.18, 3.56)) and promoted the expression of p-p38 (SMD = 4.19, 95% CI (2.34, 6.05)), Nrf2 (SMD = 4.60, 95% CI (2.34, 6.86)), and GCLC (SMD = 1.32, 95% CI (0.23, 2.41)); the p38 inhibitor inhibited the expression of Nrf2 (SMD = -1.27, 95% CI (-2.46, -0.09)) and GCLC (SMD = -5.37, 95% CI (-5.37, -2.20)); siNrf2 inhibited the expression of GCLC, and BSO inhibited the synthesis of GSH. There is a dose-dependent relationship between the effects of exposure on GSH in vitro. Conclusions. These indicate the difference between in vivo and in vitro studies of the effect of arsenic on glutathione synthesis. In vivo studies have shown that arsenic exposure can reduce glutamate and cysteine levels and inhibit glutathione synthesis, while in vitro studies have shown that chronic low-dose arsenic exposure can activate the p38/Nrf2 pathway, upregulate GCLC expression, and promote glutathione synthesis.


Assuntos
Intoxicação por Arsênico/metabolismo , Arsênio/toxicidade , Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Humanos
10.
Artigo em Inglês | MEDLINE | ID: mdl-32698366

RESUMO

Chronic arsenic (As) exposure is a critical public health issue. The As metabolism can be influenced by many factors. The objective of this study is to verify if these factors influence As metabolism in four Italian areas affected by As pollution. Descriptive analyses were conducted on 271 subjects aged 20-49 in order to assess the effect of each factor considered on As methylation. Percentages of metabolites of As in urine, primary and secondary methylation indexes were calculated as indicators for metabolic capacity. The results indicate that women have a better methylation capacity (MC) than men, and drinking As-contaminated water from public aqueducts is associated with poorer MC, especially in areas with natural As pollution. In areas with anthropogenic As pollution occupational exposure is associated with a higher MC while smoking with a poorer MC. Dietary habits and genetic characteristics are probably implicated in As metabolism. BMI, alcohol consumption and polymorphism of the AS3MT gene seem not to influence As MC. Arsenic metabolism may be affected by various factors and in order to achieve a comprehensive risk assessment of As-associated disease, it is crucial to understand how these factors contribute to differences in As metabolism.


Assuntos
Intoxicação por Arsênico/metabolismo , Arsênio/metabolismo , Exposição Ambiental/efeitos adversos , Poluentes Ambientais/metabolismo , Adulto , Arsênio/análise , Intoxicação por Arsênico/etiologia , Poluentes Ambientais/efeitos adversos , Feminino , Humanos , Itália , Masculino , Metilação , Pessoa de Meia-Idade , Exposição Ocupacional , Poluição da Água , Adulto Jovem
11.
Arch Toxicol ; 94(8): 2587-2601, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32435915

RESUMO

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


Assuntos
Antineoplásicos/farmacologia , Intoxicação por Arsênico/etiologia , Trióxido de Arsênio/farmacologia , Arsenicais/farmacologia , Leucemia Promielocítica Aguda/tratamento farmacológico , Animais , Antineoplásicos/metabolismo , Intoxicação por Arsênico/metabolismo , Trióxido de Arsênio/metabolismo , Arsenicais/metabolismo , Biotransformação , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Medição de Risco , Testes de Toxicidade
12.
Biomolecules ; 10(2)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32033229

RESUMO

: High arsenic (As) levels in food and drinking water, or under some occupational conditions, can precipitate chronic toxicity and in some cases cancer. Millions of people are exposed to unacceptable amounts of As through drinking water and food. Highly exposed individuals may develop acute, subacute, or chronic signs of poisoning, characterized by skin lesions, cardiovascular symptoms, and in some cases, multi-organ failure. Inorganic arsenite(III) and organic arsenicals with the general formula R-As2+ are bound tightly to thiol groups, particularly to vicinal dithiols such as dihydrolipoic acid (DHLA), which together with some seleno-enzymes constitute vulnerable targets for the toxic action of As. In addition, R-As2+-compounds have even higher affinity to selenol groups, e.g., in thioredoxin reductase that also possesses a thiol group vicinal to the selenol. Inhibition of this and other ROS scavenging seleno-enzymes explain the oxidative stress associated with arsenic poisoning. The development of chelating agents, such as the dithiols BAL (dimercaptopropanol), DMPS (dimercapto-propanesulfonate) and DMSA (dimercaptosuccinic acid), took advantage of the fact that As had high affinity towards vicinal dithiols. Primary prevention by reducing exposure of the millions of people exposed to unacceptable As levels should be the prioritized strategy. However, in acute and subacute and even some cases with chronic As poisonings chelation treatment with therapeutic dithiols, in particular DMPS appears promising as regards alleviation of symptoms. In acute cases, initial treatment with BAL combined with DMPS should be considered.


Assuntos
Antídotos/uso terapêutico , Intoxicação por Arsênico/tratamento farmacológico , Arsênio/toxicidade , Quelantes/uso terapêutico , Animais , Antídotos/química , Antídotos/farmacologia , Arsênio/efeitos adversos , Intoxicação por Arsênico/etiologia , Intoxicação por Arsênico/metabolismo , Arsenicais/efeitos adversos , Quelantes/química , Quelantes/farmacologia , Dimercaprol/análogos & derivados , Dimercaprol/farmacologia , Dimercaprol/uso terapêutico , Água Potável/efeitos adversos , Humanos , Modelos Moleculares , Exposição Ocupacional/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Succímero/química , Succímero/farmacologia , Succímero/uso terapêutico , Unitiol/química , Unitiol/farmacologia , Unitiol/uso terapêutico , Poluentes Químicos da Água/efeitos adversos , Poluentes Químicos da Água/toxicidade
13.
Hum Exp Toxicol ; 39(1): 37-46, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31775542

RESUMO

Chronic exposure to arsenic may result in the manifestation of damage in multiple organs or systems of the body. Arsenic-induced renal dysfunction has been determined, but their pathogenesis has not been fully examined. In this study, we measured the expression levels of miR-191 in plasma, the contents of pro-inflammatory (interleukin (IL)-6 and tumor necrosis factor alpha) and anti-inflammatory (IL-2 and transforming growth factor beta) cytokines, and renal dysfunction indicators (blood urea nitrogen, blood creatinine, uric acid, and cystatin C) in serum from control and arsenic poisoning populations and analyzed the relationship between the miR-191, cytokines, and renal dysfunction indicators. The results clearly show the alteration of miR-191 expression was significantly associated with arsenic-induced renal dysfunction. Overall, the association of miR-191, inflammatory response and renal dysfunction, is clearly supported by the current findings. In other words, miR-191 is involved in renal dysfunction in exposed populations by regulating inflammatory response caused by coal-burning arsenic. The study provides a scientific basis for further studies of the causes of the arsenic-induced renal dysfunction, the biological role of miR-191, and targeted prevention strategies.


Assuntos
Arsênio/toxicidade , Carvão Mineral/análise , Exposição Ambiental/efeitos adversos , Poluentes Ambientais/toxicidade , Nefropatias/induzido quimicamente , MicroRNAs/metabolismo , Arsênio/química , Intoxicação por Arsênico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Exposição Ambiental/análise , Poluentes Ambientais/química , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/genética , Regulação para Cima
14.
Metab Brain Dis ; 34(6): 1595-1606, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31422513

RESUMO

Both arsenic (As) and obesity are associated with brain disorders. However, long term studies to evaluate their concomitant adverse effects on the brain functions are lacking. Present study was conducted to evaluate the long term co-exposure of As and high fat diet (HFD) on memory and brain mitochondrial function in mice. Male mice were randomly divided into 7 groups fed with HFD or ordinary diet (OD) and instantaneously exposed to As (25 or 50 ppm) in drinking water for, 4, 8, 12, 16 or 20 weeks. Step-down passive avoidance method was used for memory assessment and post exposure various parameters including mitochondrial damage, level of reactive oxygen species (ROS), malondialdeid (MDA) and glutathione (GSH) were determined. Results indicated that the retention latency decreased in As (25 and 50 ppm) and HFD received mice after 12 and 16 weeks respectively. Same results were observed at significantly shorter duration (8th week) when As was administered along with HFD as compared to control group. In the HFD alone fed mice increased the mitochondrial membrane damage, levels of ROS and MDA were observed while GSH contents decreased significantly. Concomitant administration of HFD and As amplified those mentioned toxic effects (p < 0.001). In conclusion, our findings demonstrated that the simultaneous HFD and As impaired memory at least three times more than exposing each one alone. These toxic effects could be due to the mitochondria originated oxidative stress along with the depleted antioxidant capacity of the brain of mice.


Assuntos
Intoxicação por Arsênico/metabolismo , Arsenitos/farmacologia , Dieta Hiperlipídica/efeitos adversos , Transtornos da Memória/metabolismo , Memória/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Compostos de Sódio/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Glutationa/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Camundongos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
15.
Int J Mol Sci ; 20(14)2019 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-31336801

RESUMO

Arsenic (As) contamination affects hundreds of millions of people globally. Although the number of patients with chronic As exposure is large, the symptoms and long-term clinical courses of the patients remain unclear. In addition to reviewing the literature on As contamination and toxicity, we provide useful clinical information on medical care for As-exposed patients. Further, As metabolite pathways, toxicity, speculated toxicity mechanisms, and clinical neurological symptoms are documented. Several mechanisms that seem to play key roles in As-induced neurotoxicity, including oxidative stress, apoptosis, thiamine deficiency, and decreased acetyl cholinesterase activity, are described. The observed neurotoxicity predominantly affects peripheral nerves in sensory fibers, with a lesser effect on motor fibers. A sural nerve biopsy showed the axonal degeneration of peripheral nerves mainly in small myelinated and unmyelinated fibers. Exposure to high concentrations of As causes severe central nervous system impairment in infants, but no or minimal impairment in adults. The exposure dose-response relationship was observed in various organs including neurological systems. The symptoms caused by heavy metal pollution (including As) are often nonspecific. Therefore, in order to recognize patients experiencing health problems caused by As, a multifaceted approach is needed, including not only clinicians, but also specialists from multiple fields.


Assuntos
Arsênio/toxicidade , Síndromes Neurotóxicas/etiologia , Fatores Etários , Animais , Arsênio/metabolismo , Intoxicação por Arsênico/metabolismo , Relação Dose-Resposta a Droga , Poluentes Ambientais/toxicidade , Poluição Ambiental , Humanos , Redes e Vias Metabólicas , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/metabolismo , Especificidade de Órgãos , Estresse Oxidativo
16.
Food Chem Toxicol ; 123: 28-41, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30342114

RESUMO

Arsenic is a ubiquitous contaminant, with typical human dietary intake below 1 µg/kg bw/d and extreme drinking water exposures up to ∼50 µg/kg bw/d. The formation and binding of trivalent metabolites are central to arsenic toxicity and strong human evidence suggests special concern for early life exposures in the etiology of adult diseases, especially cancer. This study measured the metabolism and disposition of arsenite in neonatal mice to understand the role of maturation in metabolic activation and detoxification of arsenic. Many age-related differences were observed after gavage administration of arsenite, with consistent evidence in blood and tissues for higher exposures to trivalent arsenic species in neonatal mice related to the immaturity of metabolic and/or excretory functions. The evidence for greater tissue binding of arsenic species in young mice is consistent with enhanced susceptibility to toxicity based on metabolic and toxicokinetic differences alone. Lactational transfer from arsenite-dosed dams to suckling mice was minimal, based on no dosing-related changes in the levels of arsenic species in pup blood or milk collected from the dams. Animal models evaluating whole-life exposure to inorganic arsenic must use direct dosing in early neonatal life to predict accurately potential toxicity from early life exposures in children.


Assuntos
Intoxicação por Arsênico/metabolismo , Arsenicais/metabolismo , Arsenitos/metabolismo , Leite/química , Compostos de Sódio/metabolismo , Animais , Intoxicação por Arsênico/fisiopatologia , Arsenicais/química , Arsenitos/química , Feminino , Contaminação de Alimentos , Humanos , Lactação , Masculino , Camundongos , Leite/metabolismo , Compostos de Sódio/química , Distribuição Tecidual , Toxicocinética
17.
J Toxicol Environ Health A ; 81(18): 893-912, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30230972

RESUMO

The aim of this study was to establish a process for deriving a chemical-specific mode of action (MOA) from chemical-agnostic adverse outcome pathway (AOPs), using inorganic arsenic (iAs) as a case study. The AOP developed for this case study are related to disruption of cellular signaling by chemicals that strongly bind to vicinal dithiols in cellular proteins, leading to disruption of inflammatory and oxidative stress signaling along with inhibition of the DNA damage responses. The proposed MOA for iAs incorporates this AOP, overlaid on a background of increasing oxidative stress and/or co-exposure to mutagenic chemicals or radiation. The most challenging aspect of developing a MOA from AOP is the incorporation of metabolism and dose-response, neither of which may be considered in the development of an AOP. The cellular responses to relatively low concentrations (below 100 parts per billion) of iAs in drinking water appear to be secondary to binding of trivalent arsenite and its trivalent metabolite, monomethyl arsenous acid to key cellular vicinal dithiols in target tissues, resulting in a co-carcinogenic MOA. The proposed AOP may also be applied to non-cancer endpoints, enabling an integrated approach to conducting a risk assessment for iAs.


Assuntos
Rotas de Resultados Adversos , Arsenicais/efeitos adversos , Intoxicação por Arsênico/metabolismo , Arsenicais/metabolismo , Humanos , Medição de Risco/métodos
18.
Acta toxicol. argent ; 26(1): 32-44, mayo 2018. ilus
Artigo em Espanhol | LILACS | ID: biblio-973614

RESUMO

La exposición crónica al arsénico (As) inorgánico a través del agua de bebida da lugar al desarrollo de la enfermedad conocida como hidroarsenicismo. Esta enfermedad presenta sintomatología característica, sin embargo, para la mayoría de los efectos tóxicos que produce del As aún no se conoce en detalle el mecanismo de acción tóxica. Los mecanismos moleculares de acción del arsenito (unión a grupos sulfhidrilos) y del arseniato (sustitución del fosfato) están bien identificados, sin embargo, las consecuencias a nivel subcelular, celular, tisular y orgánico de esos mecanismos todavía presentan muchos huecos por llenar. A nivel subcelular y celular, la generación de especies reactivas de oxígeno (ERO) y de nitrógeno (ERN) son los mecanismos de acción tóxica del As más estudiados últimamente. Se los ha vinculado con la diferenciación y proliferación de queratinocitos, con la disfunción endotelial, con la resistencia a la insulina, con la inducción de peroxidación lipídica en hígado, de necrosis tubular renal y con cambios en la expresión del receptor estrogénico. Por último, la respuesta celular a proteínas no plegadas (como consecuencia del estrés del retículo endoplásmico) podría ser un mecanismo para explicar la afectación de la inmunidad humoral y la celular.


Chronic exposure to inorganic arsenic (As) through drinking water leads to the development of the disease known as hydroarsenicism. This disease presents characteristic symptomatology but the mechanisms underlying most of the toxic effects produced by As are not fully understand. The molecular mechanisms of action of arsenite (binding to sulfhydryl groups) and arsenate (phosphate substitution) are well identified, however, the consequences at the subcellular, cellular, tissue and organic levels of these mechanisms still have many gaps to fill. At the subcellular and cellular level, the generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are the most studied mechanisms of toxic action. They have been linked to the differentiation and proliferation of keratinocytes, endothelial dysfunction, insulin resistance, induction of lipid peroxidation in the liver, renal tubular necrosis and changes in the expression of estrogen receptor. Finally, the cellular response to unfolded proteins (as a consequence of the stress of the endoplasmic reticulum) could be a mechanism to explain the affectation of humoral and cellular immunity.


Assuntos
Humanos , Arsênio/toxicidade , Estresse Oxidativo , Intoxicação por Arsênico/complicações , Intoxicação por Arsênico/metabolismo , Poluentes Químicos da Água/efeitos adversos
19.
Toxicol Lett ; 295: 220-228, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-29705342

RESUMO

Arsenic, an established human carcinogen, causes genetic toxicity. However, the molecular mechanisms involved remain unknown. MicroRNAs (miRNAs) are regulators that participate in fundamental cellular processes. In the present investigation, we selected, as research subjects, patients with arsenic poisoning caused by burning of coal in Guizhou Province, China. For these patients, the plasma levels of miR-145 were up-regulated. In L-02 cells, arsenite, an active form of arsenic, induced up-regulation of miR-145 and down-regulation of ERCC1 and ERCC2, and caused DNA damage. For L-02 cells, transfection with an miR-145 inhibitor prevented arsenite-induced DNA damage and decreased ERCC2 levels. Luciferase reporter assays showed that miR-145 regulated ERCC2 expression by targeting the 3'-UTR of ERCC2, but not that for ERCC1. The present results demonstrate that arsenite induces the over-expression of miR-145 and inhibits DNA repair via targeting ERCC2, thus promoting DNA damage. The information provides a new mechanism for arsenic-induced liver injury.


Assuntos
Intoxicação por Arsênico/etiologia , Arsenitos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Dano ao DNA , Poluentes Ambientais/toxicidade , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , MicroRNAs/metabolismo , Compostos de Sódio/toxicidade , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Regiões 3' não Traduzidas , Adulto , Apoptose/efeitos dos fármacos , Intoxicação por Arsênico/genética , Intoxicação por Arsênico/metabolismo , Intoxicação por Arsênico/patologia , Sítios de Ligação , Estudos de Casos e Controles , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endonucleases/genética , Endonucleases/metabolismo , Feminino , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Transdução de Sinais/efeitos dos fármacos , Proteína Grupo D do Xeroderma Pigmentoso/genética
20.
Food Chem Toxicol ; 115: 178-184, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29530638

RESUMO

Arsenic is ubiquitous in the earth's crust, and human diseases are linked with exposures that are similar to dietary intake estimates. Metabolic methylation of inorganic arsenic facilitates excretion of pentavalent metabolites and decreases acute toxicity; however, tissue binding of trivalent arsenic intermediates is evidence for concomitant metabolic activation. Pregnant and fetal CD-1 mice comprise a key animal model for arsenic carcinogenesis since adult-only exposures have minimal effects. This study evaluated inorganic arsenic and its metabolites in pentavalent and trivalent states in blood and tissues from maternal and fetal CD-1 mice after repeated administration of arsenite through drinking water. After 8 days of exposure, DMA species were ubiquitous in dams and fetuses. Despite the presence of MMAIII in dams, none was observed in any fetal sample. This difference may be important in assessing fetal susceptibility to arsenic toxicity because MMA production has been linked with human disease. Binding of DMAIII in fetal tissues provided evidence for metabolic activation, although the role for such binding in arsenic toxicity is unclear. This study provides links between administered dose, metabolism, and internal exposures from a key animal model of arsenic toxicity to better understand risks from human exposure to environmental arsenic.


Assuntos
Intoxicação por Arsênico/metabolismo , Arsenitos/metabolismo , Água Potável/análise , Gravidez/metabolismo , Compostos de Sódio/metabolismo , Poluentes Químicos da Água/metabolismo , Animais , Animais Recém-Nascidos/sangue , Arsênio/sangue , Arsênio/química , Arsênio/metabolismo , Intoxicação por Arsênico/sangue , Intoxicação por Arsênico/embriologia , Intoxicação por Arsênico/etiologia , Arsenitos/química , Feminino , Feto/metabolismo , Humanos , Masculino , Exposição Materna/efeitos adversos , Camundongos , Gravidez/sangue , Compostos de Sódio/química , Poluentes Químicos da Água/sangue , Poluentes Químicos da Água/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA