Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Biochem Biophys Res Commun ; 551: 14-20, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33714754

RESUMO

The blood-brain barrier (BBB) is the most critical obstacle in the treatment of central nervous system disorders, such as glioma, the most typical type of brain tumor. To overcome the BBB and enhance drug-penetration abilities, we used angiopep-2-modified liposomes to deliver arsenic trioxide (ATO) across the BBB, targeting the glioma. Angiopep-2-modified calcium arsenite-loaded liposomes (A2-PEG-LP@CaAs), with uniformly distributed hydrodynamic diameter (96.75 ± 0.57 nm), were prepared using the acetate gradient method with high drug-loading capacity (7.13 ± 0.72%) and entrapment efficiency (54.30 ± 9.81%). In the acid tumor microenvironment, arsenic was responsively released, thereby exerting an anti-glioma effect. The anti-glioma effect of A2-PEG-LP@CaAs was investigated both in vitro and in vivo. As a result, A2-PEG-LP@CaAs exhibited a potent, targeted anti-glioma effect mediated by the lipoprotein receptor-related (LRP) receptor, which is overexpressed in both the BBB and glioma. Therefore, A2-PEG-LP@CaAs could dramatically promote the anti-glioma effect of ATO, as a promising strategy for glioma therapy.


Assuntos
Arsenitos/química , Cálcio/química , Sistemas de Liberação de Medicamentos , Glioma/tratamento farmacológico , Lipossomos/química , Lipossomos/farmacocinética , Peptídeos/química , Animais , Trióxido de Arsênio/química , Trióxido de Arsênio/farmacocinética , Arsenitos/farmacocinética , Barreira Hematoencefálica/metabolismo , Cálcio/farmacocinética , Ciclo Celular , Linhagem Celular Tumoral , Glioma/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Nus , Peptídeos/farmacocinética , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Environ Toxicol ; 36(2): 204-212, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32930475

RESUMO

Long-term arsenic exposure can promote cancer through epigenetic mechanisms, and arsenite methyltransferase (AS3MT) plays an important role in this process. However, the expression patterns and mechanisms of AS3MT in arsenic carcinogenesis remain unclear. In this study, we found that the AS3MT was overexpressed in arsenic exposed population, non-small cell lung cancer (NSCLC) tissues, and A549 cells with sodium arsenite (NaAsO2 ) treatment for 48 hours. Besides, the level of AS3MT expression was positively correlated with the concentrations of urinary total arsenic (tAs), inorganic arsenic (iAs), methanearsonic acid (MMA), and dimethylarsinic acid (DMA) in all subjects. Functional experiments demonstrated that siRNA-mediated knockdown of AS3MT significantly inhibited proliferation of A549 cells. Mechanism investigation revealed that silencing of AS3MT inhibited proliferation by increasing mRNA expression levels of p21 and E2F1, and inhibiting CDK1, CDK2, CDK4, CDK6, Cyclin A2, Cyclin E1, Cyclin E2, and PCNA mRNA expression. Therefore, arsenic increased AS3MT expression in vivo and in vitro, which could directly act on the cell and affect the progression of NSCLC by regulating cell cycle genes.


Assuntos
Arsenitos/toxicidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes cdc , Neoplasias Pulmonares/patologia , Metiltransferases/genética , Células A549 , Arsenitos/farmacocinética , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células/genética , Feminino , Humanos , Neoplasias Pulmonares/genética , Metiltransferases/metabolismo , Regulação para Cima
3.
Expert Opin Investig Drugs ; 29(9): 1059-1067, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32735765

RESUMO

BACKGROUND: We evaluated the tolerability, pharmacokinetics (PK) and preliminary efficacy of KML001, an oral trivalent arsenical, as a monotherapy in patients with advanced solid tumors. RESEARCH DESIGN AND METHODS: With a standard 3 + 3 design for dose-escalation stage, the planned dose levels of KML001 were 5, 7.5, 10, 12.5, and 15 mg/day for 28 days. Once the maximum tolerated dose was determined, 22 subjects were additionally enrolled for dose-expansion stage. PK analysis was performed in the 5, 10, and 15 mg/day cohort at the dose-escalation stage and also at the dose-expansion stage. Moreover, response was assessed using the standard RECIST 1.1. RESULTS: A total of 45 Korean subjects were enrolled. No DLT was reported at the dose-escalation stage. Three DLTs, two cases of prolonged QTc interval and one of neutropenia, were reported in the 12.5 mg/day cohort at the dose-expansion stage. Higher total daily doses up to 12.5 mg/day of KML001 resulted in higher trough plasma concentrations. Among the 18 subjects who completed 2 cycles of therapy, 15 had progressive disease and 3 had stable disease. CONCLUSIONS: Doses equal to or greater than 10 mg/day KML001 alone were tolerable and produced plasma concentrations higher than biologically relevant targets.


Assuntos
Antineoplásicos/administração & dosagem , Arsenitos/administração & dosagem , Neoplasias/tratamento farmacológico , Compostos de Sódio/administração & dosagem , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Arsenitos/efeitos adversos , Arsenitos/farmacocinética , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/patologia , Compostos de Sódio/efeitos adversos , Compostos de Sódio/farmacocinética , Resultado do Tratamento
4.
J Hazard Mater ; 391: 122196, 2020 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-32062345

RESUMO

The toxicity of arsenic (As) has been reported to be different depending on their chemical forms. However, its toxicity mechanisms largely remain unknown. In this study, to investigate toxicity mechanism of As in marine zooplanktons, namely, the rotifer Brachionus plicatilis and the copepod Paracyclopina nana, metabolites of As were analyzed by using a high-performance liquid chromatography coupled with inductively coupled plasma mass spectrometry with in vivo toxicity and antioxidant responses in response to inorganic As, including arsenate (AsV) and arsenite (AsIII). While AsIII was more toxic than AsV in both organisms, the rotifer B. plicatilis exhibited stronger tolerance, compared to the copepod P. nana. The As speciation analysis revealed differences in biotransformation processes in two species with B. plicatilis having a more simplified process than P. nana, contributing to a better tolerance against As in the rotifer B. plicatilis compared to P. nana. Moreover, the levels of GSH content and the regulation of omega class glutathione S-transferases were different in response to oxidative stress between B. plicatilis and P. nana. These results suggest that the rotifer B. plicatilis has a unique survival strategy with more efficient biotransformation and antioxidant responses, compared to P. nana, conferring higher tolerance to As.


Assuntos
Arseniatos/farmacocinética , Arseniatos/toxicidade , Arsenitos/farmacocinética , Arsenitos/toxicidade , Copépodes/efeitos dos fármacos , Rotíferos/efeitos dos fármacos , Poluentes Químicos da Água/farmacocinética , Poluentes Químicos da Água/toxicidade , Animais , Biotransformação , Catalase/metabolismo , Copépodes/metabolismo , Copépodes/fisiologia , Feminino , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Reprodução/efeitos dos fármacos , Rotíferos/metabolismo , Superóxido Dismutase/metabolismo
5.
Food Chem Toxicol ; 133: 110760, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31421213

RESUMO

Arsenic is a common toxic contaminant in food and drinking water. Metabolic activation of arsenic species produces reactive trivalent intermediates that can disrupt cellular regulatory systems by covalent binding to thiol groups. Arsenic exposures have been associated with human diseases including cancer, diabetes, lung and cardiovascular disorders and there is accumulating evidence that early life exposures are important in the etiology. Previous toxicokinetic studies of arsenite ingestion in neonatal CD-1 mice showed consistent evidence for metabolic and physiologic immaturity that led to elevated internal exposures to trivalent arsenic species in the youngest mice, relative to adults. The current study in rhesus monkeys showed that metabolism and binding of trivalent intermediates after arsenite ingestion were similar between adult monkeys and CD-1 mice. Unlike neonatal mice, monkeys from the age of 5-70 days showed similar metabolism and binding profiles, which were also similar to those in adults. The absence of evidence for metabolic immaturity in monkeys suggests that toxicological effects observed in mice from early postnatal exposures to arsenic could over-predict those possible in primates, based on significantly higher internal exposures.


Assuntos
Arsenitos/farmacocinética , Compostos de Sódio/farmacocinética , Poluentes Químicos da Água/farmacocinética , Administração Oral , Fatores Etários , Animais , Animais Recém-Nascidos/metabolismo , Arsenitos/administração & dosagem , Arsenitos/metabolismo , Eritrócitos/metabolismo , Feminino , Macaca mulatta/metabolismo , Masculino , Camundongos , Compostos de Sódio/administração & dosagem , Compostos de Sódio/metabolismo , Poluentes Químicos da Água/administração & dosagem , Poluentes Químicos da Água/metabolismo
6.
Nanotechnology ; 30(17): 175101, 2019 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-30654348

RESUMO

Arsenic trioxide (ATO), an FDA-approved drug for acute promyelocytic leukemia, also has great potential for treatment of solid tumors. Drug delivery powered by recent advances in nanotechnology has boosted the efficacy of many drugs, which is enlightening for applications of ATO in treating solid tumors. Herein, we reported arsenite-loaded multifunctional nanoparticles that are capable of pH-responsive ATO release for treating hepatocellular carcinoma (HCC) and real-time monitoring via magnetic resonance imaging. We fabricated these nanoparticles (designated as magnetic large-pore mesoporous silica nanoparticle (M-LPMSN)-NiAsO x ) by loading nanoparticulate ATO prodrugs (NiAsO x ) into the pores of large-pore mesoporous silica nanoparticles (LPMSNs) that contain magnetic iron oxide nanoparticles in the center. The surface of these nanodrugs was modified with a targeting ligand folic acid (FA) to further enhance the drug efficacy. Releasing profiles manifest the responsive discharging of arsenite in acidic environment. In vitro experiments with SMMC-7721 cells reveal that M-LPMSN-NiAsO x -FA nanodrugs have significantly higher cytotoxicity than traditional free ATO and induce more cell apoptosis. In vivo experiments with mice bearing H22 tumors further confirm the superior antitumor efficacy of M-LPMSN-NiAsO x -FA over traditional free ATO and demonstrate the outstanding imaging ability of M-LPMSN-NiAsO x -FA for real-time tumor monitoring. These targeted arsenite-loaded magnetic mesoporous silica nanoparticles integrating imaging and therapy hold great promise for treatment of HCC, indicating the auspicious potential of LPMSN-based nanoplatforms.


Assuntos
Antineoplásicos/administração & dosagem , Arsenitos/administração & dosagem , Carcinoma Hepatocelular/tratamento farmacológico , Portadores de Fármacos , Neoplasias Hepáticas/tratamento farmacológico , Nanopartículas de Magnetita/química , Animais , Antineoplásicos/farmacocinética , Arsenitos/química , Arsenitos/farmacocinética , Linhagem Celular Tumoral , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Feminino , Humanos , Nanopartículas de Magnetita/administração & dosagem , Camundongos Endogâmicos BALB C
7.
Environ Toxicol ; 34(2): 188-202, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30511785

RESUMO

Human exposure to inorganic arsenic (iAs) is a global health issue. Although there is strong evidence for iAs-induced toxicity at higher levels of exposure, many epidemiological studies evaluating its effects at low exposure levels have reported mixed results. We comprehensively reviewed the literature and evaluated the scientific knowledge on human exposure to arsenic, mechanisms of action, systemic and carcinogenic effects, risk characterization, and regulatory guidelines. We identified areas where additional research is needed. These priority areas include: (1) further development of animal models of iAs carcinogenicity to identify molecular events involved in iAs carcinogenicity; (2) characterization of underlying mechanisms of iAs toxicity; (3) assessment of gender-specific susceptibilities and other factors that modulate arsenic metabolism; (4) sufficiently powered epidemiological studies to ascertain relationship between iAs exposure and reproductive/developmental effects; (5) evaluation of genetic/epigenetic determinants of iAs effects in children; and (6) epidemiological studies of people chronically exposed to low iAs concentrations.


Assuntos
Arseniatos/toxicidade , Arsenitos/toxicidade , Pesquisa Biomédica , Carcinógenos Ambientais/toxicidade , Poluentes Ambientais/toxicidade , Mutagênicos/toxicidade , Animais , Arseniatos/farmacocinética , Arsenitos/farmacocinética , Pesquisa Biomédica/tendências , Biotransformação , Carcinógenos Ambientais/farmacocinética , Poluentes Ambientais/farmacocinética , Humanos , Mutagênicos/farmacocinética
8.
Toxicol Sci ; 165(2): 431-446, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29982692

RESUMO

Arsenite exposure is known to increase the risk of neurological disorders via alteration of dopamine content, but the detailed molecular mechanisms remain largely unknown. In this study, using both dopaminergic neurons of the PC-12 cell line and C57BL/6J mice as in vitro and in vivo models, our results demonstrated that 6 months of arsenite exposure via drinking water caused significant learning and memory impairment, anxiety-like behavior and alterations in conditioned avoidance and escape responses in male adult mice. We also were the first to reveal that the reduction in dopamine content induced by arsenite mainly resulted from deficits in dopaminergic neurotransmission in the synaptic cleft. The reversible N6- methyladenosine (m6A) modification is a novel epigenetic marker with broad roles in fundamental biological processes. We further evaluated the effect of arsenite on the m6A modification and tested if regulation of the m6A modification by demethylase fat mass and obesity-associated (FTO) could affect dopaminergic neurotransmission. Our data demonstrated for the first time that arsenite remarkably increased m6A modification, and FTO possessed the ability to alleviate the deficits in dopaminergic neurotransmission in response to arsenite exposure. Our findings not only provide valuable insight into the molecular neurotoxic pathogenesis of arsenite exposure, but are also the first evidence that regulation of FTO may be considered as a novel strategy for the prevention of arsenite-associated neurological disorders.


Assuntos
Adenosina/análogos & derivados , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Arsenitos/toxicidade , Comportamento Animal/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Adenosina/genética , Adenosina/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Arsenitos/farmacocinética , Aprendizagem da Esquiva/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Relação Dose-Resposta a Droga , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Células PC12 , Modificação Traducional de Proteínas , Ratos
9.
J Biochem Mol Toxicol ; 32(2)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29214690

RESUMO

The present study evaluates the protective effect of α-lipoic acid (LA) against arsenic-induced testicular and epididymal oxidative damage in rats. Arsenic caused significant reduction in the reproductive organ weights, serum testosterone levels, testicular daily sperm count, epididymal sperm count, sperm motility, sperm viability, and sperm membrane integrity. Significant reduction in the activity levels of superoxide dismutase, catalase, and glutathione levels with a concomitant increase in the lipid peroxidation and protein carbonyl content in the testis and the cauda epididymis of arsenic-exposed rats. Arsenic intoxication also enhanced the testicular caspase-3 mRNA levels, disorganization of testicular and cauda epididymal architecture as well as increased arsenic content in the testis and the cauda epididymis of rats. Arsenic exposure also deteriorated fertility ability in male rats over controls. Conversely, α-LA negated the testicular and cauda epididymal oxidative stress and restored the male reproductive health in arsenic-exposed rats.


Assuntos
Antioxidantes/uso terapêutico , Intoxicação por Arsênico/fisiopatologia , Epididimo/efeitos dos fármacos , Infertilidade Masculina/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Testículo/efeitos dos fármacos , Ácido Tióctico/uso terapêutico , Animais , Antioxidantes/administração & dosagem , Arsenitos/metabolismo , Arsenitos/farmacocinética , Arsenitos/toxicidade , Biomarcadores/sangue , Biomarcadores/metabolismo , Epididimo/metabolismo , Epididimo/patologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glutationa/metabolismo , Infertilidade Masculina/etiologia , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Injeções Intraperitoneais , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Tamanho do Órgão/efeitos dos fármacos , Oxirredutases/metabolismo , Carbonilação Proteica/efeitos dos fármacos , Distribuição Aleatória , Ratos Wistar , Compostos de Sódio/metabolismo , Compostos de Sódio/farmacocinética , Compostos de Sódio/toxicidade , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia , Testosterona/sangue , Ácido Tióctico/administração & dosagem , Distribuição Tecidual
10.
Sci Rep ; 7: 44424, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28303940

RESUMO

Arsenic is a common environmental and occupational toxicant with dramatic species differences in its susceptibility and metabolism. Mouse strain variability may provide a better understanding of the arsenic pathological profile but is largely unknown. Here we investigated oxidative lesion induced by acute arsenic exposure in the two frequently used mouse strains C57BL/6J and 129X1/SvJ in classical gene targeting technique. A dose of 5 mg/kg body weight arsenic led to a significant alteration of blood glutathione towards oxidized redox potential and increased hepatic malondialdehyde content in C57BL/6J mice, but not in 129X1/SvJ mice. Hepatic antioxidant enzymes were induced by arsenic in transcription in both strains and many were higher in C57BL/6J than 129X1/SvJ mice. Arsenic profiles in the liver, blood and urine and transcription of genes encoding enzymes involved in arsenic biomethylation all indicate a higher arsenic methylation capacity, which contributes to a faster hepatic arsenic excretion, in 129X1/SvJ mice than C57BL/6J mice. Taken together, C57BL/6J mice are more susceptible to oxidative hepatic injury compared with 129X1/SvJ mice after acute arsenic exposure, which is closely associated with arsenic methylation pattern of the two strains.


Assuntos
Intoxicação por Arsênico/sangue , Arsenitos/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Compostos de Sódio/toxicidade , Alanina Transaminase/genética , Alanina Transaminase/metabolismo , Animais , Intoxicação por Arsênico/patologia , Intoxicação por Arsênico/urina , Arsenitos/farmacocinética , Aspartato Aminotransferases/genética , Aspartato Aminotransferases/metabolismo , Biotransformação , Glutamato-Cisteína Ligase/genética , Glutamato-Cisteína Ligase/metabolismo , Glutationa/sangue , Fígado/metabolismo , Fígado/patologia , Masculino , Malondialdeído/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Compostos de Sódio/farmacocinética , Especificidade da Espécie , Transcrição Gênica
11.
Biochem Pharmacol ; 126: 79-86, 2017 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-27889539

RESUMO

Inorganic arsenicals are well-known carcinogens, whereas arsenite (iAsIII) compounds are now recognized as potent therapeutic agents for several leukemias, and arsenic trioxide has been used for the treatment of recurrent acute promyelocytic leukemia (APL). However, recent clinical trials revealed that arsenite is not always effective for non-APL malignancies. Another arsenical, S-dimethylarsino-glutathione ([DMAIII(GS)]), which is a putative metabolic intermediate in the hepatic metabolism of iAsIII, shows promise for treating several types of lymphoma. However, the metabolism of [DMAIII(GS)] has not been well investigated, probably because [DMAIII(GS)] is not stable in biological fluids where the concentration of glutathione is low. In the present study, we injected [DMAIII(GS)] intravenously into mice and compared the tissue distribution and metabolic dynamics of [DMAIII(GS)] with those of sodium arsenite (NaAsO2). We found a unique organ preference for the distribution of [DMAIII(GS)] to the lung and brain in comparison to NaAsO2. Furthermore, [DMAIII(GS)] appeared to bind to serum albumin by exchanging its glutathione moiety quickly after administration, providing novel insights into the longer retention of [DMAIII(GS)] in plasma.


Assuntos
Antineoplásicos/farmacocinética , Arsenicais/farmacocinética , Arsenitos/farmacocinética , Glutationa/análogos & derivados , Compostos de Sódio/farmacocinética , Animais , Antineoplásicos/sangue , Arsenicais/sangue , Glutationa/sangue , Glutationa/farmacocinética , Injeções Intravenosas , Masculino , Camundongos Endogâmicos C57BL , Ligação Proteica , Albumina Sérica/metabolismo , Distribuição Tecidual
12.
New Phytol ; 209(2): 746-61, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26372374

RESUMO

The fern Pteris vittata is an arsenic hyperaccumulator. The genes involved in arsenite (As(III)) transport are not yet clear. Here, we describe the isolation and characterization of a new P. vittata aquaporin gene, PvTIP4;1, which may mediate As(III) uptake. PvTIP4;1 was identified from yeast functional complement cDNA library of P. vittata. Arsenic toxicity and accumulating activities of PvTIP4;1 were analyzed in Saccharomyces cerevisiae and Arabidopsis. Subcellular localization of PvTIP4;1-GFP fusion protein in P. vittata protoplast and callus was conducted. The tissue expression of PvTIP4;1 was investigated by quantitative real-time PCR. Site-directed mutagenesis of the PvTIP4;1 aromatic/arginine (Ar/R) domain was studied. Heterologous expression in yeast demonstrates that PvTIP4;1 was able to facilitate As(III) diffusion. Transgenic Arabidopsis showed that PvTIP4;1 increases arsenic accumulation and induces arsenic sensitivity. Images and FM4-64 staining suggest that PvTIP4;1 localizes to the plasma membrane in P. vittata cells. A tissue location study shows that PvTIP4;1 transcripts are mainly expressed in roots. Site-directed mutation in yeast further proved that the cysteine at the LE1 position of PvTIP4;1 Ar/R domain is a functional site. PvTIP4;1 is a new represented tonoplast intrinsic protein (TIP) aquaporin from P. vittata and the function and location results imply that PvTIP4;1 may be involved in As(III) uptake.


Assuntos
Aquaporinas/genética , Aquaporinas/metabolismo , Arsenitos/farmacocinética , Proteínas de Plantas/metabolismo , Pteris/metabolismo , Aquaporinas/química , Arabidopsis/efeitos dos fármacos , Arabidopsis/genética , Arabidopsis/metabolismo , Arsênio/toxicidade , Arsenitos/metabolismo , Transporte Biológico , Cisteína , Regulação da Expressão Gênica de Plantas , Proteínas de Membrana/genética , Proteínas de Plantas/química , Proteínas de Plantas/genética , Plantas Geneticamente Modificadas , Pteris/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
13.
Arch Toxicol ; 89(10): 1751-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25092181

RESUMO

Arsenic-induced toxicity appears to be dependent on the tissue- or cell-specific accumulation of this metalloid. An early study showed that arsenic was retained in the esophagus as well as the liver, kidney cortex and skin of marmosets after intraperitoneal administration of (74)As-arsenite. However, there is little available information regarding the distribution of arsenic in the esophagus. Here, we compared the retention of arsenic in the esophagus, liver, lung, kidney and heart in mice intraperitoneally administered 1 or 5 mg/kg sodium arsenite (As(III)) daily for 3 or 7 days. The results showed that the arsenic concentration was highest in the esophagus. We compared the mRNA levels of aquaglyceroporin (AQP) 3, AQP7 and AQP9, which are responsible for arsenic influx, and those of multidrug-resistance protein (MRP) 1 and MRP2, which are responsible for arsenic efflux. The levels of AQP3 mRNA in the esophagus were much higher than those in liver, lung and heart, while the mRNA levels of MRP2 were very low in the esophagus. In addition, we found extremely low expression of Nrf2 in the esophagus at the basal and under the activated conditions, which might have resulted in low levels of glutamyl-cysteine ligase catalytic and modulatory subunits, and subsequently in the low levels of glutathione. Thus, the highest retention of arsenic was detected in the esophagus after intraperitoneal administration of As(III) to mice, and this appeared to result from multiple factors, including high expression of AQP3, low expression of MRP2, low capacity of glutathione synthesis and low activation of Nrf2.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Aquaporina 3/genética , Arsenitos/farmacocinética , Esôfago/metabolismo , Compostos de Sódio/farmacocinética , Animais , Arsenitos/administração & dosagem , Arsenitos/toxicidade , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , RNA Mensageiro/metabolismo , Compostos de Sódio/administração & dosagem , Compostos de Sódio/toxicidade , Fatores de Tempo , Distribuição Tecidual , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
14.
Toxicol Sci ; 134(1): 83-91, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23570992

RESUMO

The zebrafish (Danio rerio) was used to investigate protein expression in the liver following arsenic exposure. Several disorders have been linked to arsenic exposure, including cancer, diabetes, and cardiovascular disease. The mechanisms of arsenic toxicity are poorly understood. Prior studies have described altered gene expression, inflammation, and mitogenic signaling in acute or chronic exposure models. A proteomic approach was employed to investigate arsenic-induced alteration in the zebrafish liver proteome following a 7-day exposure to 50 ppb sodium arsenite. Over 740 unique proteins were identified, with fewer than 2% showing differential expression. Molecular pathway analysis software identified lipid metabolism and transport as potential molecular targets. Immunoblots were used to confirm protein expression changes, whereas qPCR was employed to investigate gene expression changes. Overall, 25 proteins were differentially expressed in a gender-specific manner, 11 in males and 14 in females. Of these 25, a single protein, hydroxysteroid dehydrogenase like 2, showed decreased expression in both males and females following arsenic exposure. These findings indicate that protein expression is altered following arsenic exposure. The changes presented here seem to be most prevalent in lipid transport and metabolic pathways, suggesting a potential increase in fibrosis in males and decreased lipid accumulation and uptake in females.


Assuntos
Arsenitos/toxicidade , Metabolismo dos Lipídeos/efeitos dos fármacos , Cirrose Hepática/induzido quimicamente , Proteoma/genética , Caracteres Sexuais , Compostos de Sódio/toxicidade , Poluentes Químicos da Água/toxicidade , Peixe-Zebra/metabolismo , Animais , Arsenitos/farmacocinética , Relação Dose-Resposta a Droga , Feminino , Perfilação da Expressão Gênica , Immunoblotting , Metabolismo dos Lipídeos/genética , Cirrose Hepática/metabolismo , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Compostos de Sódio/farmacocinética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transcriptoma , Poluentes Químicos da Água/farmacocinética
15.
Hum Exp Toxicol ; 32(9): 972-82, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23475432

RESUMO

Humans are commonly exposed to nicotine, one of the most important lifestyle chemicals. The occurrence of high levels of arsenic in the groundwater of the southeast region of Asia has received much attention in the past decade and has become a global health concern. Predominant occurrence of both these chemicals and ease of their human exposure led us to investigate the effect of nicotine, a major tobacco alkaloid, on arsenic toxicity. Adult male rats were pre-exposed to two different doses of nicotine (0.75 and 3 mg/kg, intraperitoneally) for 7 days followed by 30 days of arsenic exposure (50 ppm sodium arsenite in drinking water). Nicotine pre-exposure resulted in an increased brain arsenic accumulation and a decreased liver arsenic concentration. Arsenic also caused a significant oxidative stress in the blood, brain and liver of the exposed rats. Glutathione-S-transferase, a phase II enzyme, was inhibited by both arsenic and nicotine but no such inhibition was noted in arsenic-treated animals pre-exposed to nicotine. Upon nicotine pre-exposure, brain acetylcholinesterase increased, while monoamine oxidase (MAO) decreased. The toxic effects of MAO significantly attenuated with nicotine pre-exposure. The present study suggests that nicotine may not be the major contributing factor for the previously reported synergistic toxic interaction between tobacco and arsenic. Nicotine pre-exposure in arsenic-exposed animals revealed interesting toxicokinetics and oxidative stress modulating interactions in the brain and liver of rats, which requires further exploration.


Assuntos
Arsenitos/toxicidade , Encéfalo/efeitos dos fármacos , Fígado/efeitos dos fármacos , Nicotina/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Compostos de Sódio/toxicidade , Poluentes Químicos da Água/toxicidade , Animais , Arsenitos/administração & dosagem , Arsenitos/farmacocinética , Encéfalo/metabolismo , Interações Medicamentosas , Fígado/metabolismo , Masculino , Nicotina/administração & dosagem , Nicotina/farmacologia , Ratos , Ratos Wistar , Compostos de Sódio/administração & dosagem , Compostos de Sódio/farmacocinética , Poluentes Químicos da Água/administração & dosagem , Poluentes Químicos da Água/farmacocinética
16.
Toxicology ; 300(3): 149-57, 2012 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-22750630

RESUMO

The protective effect of telmisartan, the angiotensin II-receptor antagonist, against liver toxicity induced by sodium arsenite (5 mg/kg/day, p.o., for 30 days) was investigated in mice. Telmisartan treatment (10 mg/kg/day, p.o.) was applied for 30 days, starting on the same day of arsenic administration. Telmisartan significantly reduced serum alanine aminotransferase level which was increased by sodium arsenite. Telmisartan significantly suppressed lipid peroxidation, and prevented the reduced glutathione depletion and nitric oxide elevation in the liver tissue resulted from arsenic administration. Also, the increase of arsenic ion, and the reductions of selenium and zinc ions in liver tissue were attenuated by telmisartan. Histopathological examination showed that liver tissue injury mediated by arsenic was ameliorated by telmisartan treatment. Immunohistochemical analysis revealed that telmisartan significantly decreased the arsenic-induced expression of inducible nitric oxide synthase, tumor necrosis factor-α, cyclooxygenase-2, nuclear factor-κB and caspase-3 in liver tissue. It was concluded that telmisartan may represent a potential option to protect the liver tissue from the detrimental effects of arsenic toxicity.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Arsenitos/toxicidade , Benzimidazóis/uso terapêutico , Benzoatos/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Poluentes Ambientais/toxicidade , Fígado/efeitos dos fármacos , Compostos de Sódio/toxicidade , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Animais , Arsenitos/farmacocinética , Benzimidazóis/administração & dosagem , Benzoatos/administração & dosagem , Caspase 3/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Poluentes Ambientais/farmacocinética , Glutationa/metabolismo , Imuno-Histoquímica , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Testes de Função Hepática , Masculino , Camundongos , NF-kappa B/metabolismo , Selênio/metabolismo , Compostos de Sódio/farmacocinética , Telmisartan , Zinco/metabolismo
17.
Toxicol Appl Pharmacol ; 262(2): 185-93, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22575231

RESUMO

Arsenic methylation is an important cellular metabolic process that modulates arsenic toxicity and carcinogenicity. Biomethylation of arsenic produces a series of mono-, di- and tri-methylated arsenic metabolites that can be detected in tissues and excretions. Here we report that zebrafish exposed to arsenite (As(III)) produces organic arsenicals, including MMA(III), MMA(V) and DMA(V) with characteristic tissue ratios, demonstrating that an arsenic methylation pathway exists in zebrafish. In mammals, cellular inorganic arsenic is methylated by a SAM-dependent arsenic methyltransferase, AS3MT. A zebrafish arsenic methyltransferase homolog, As3mt, was identified by sequence alignment. Western blotting analysis showed that As3mt was universally expressed in zebrafish tissues. Prominent expression in liver and intestine correlated with methylated arsenic metabolites detected in those tissues. As3mt was expressed in and purified from Escherichia coli for in vitro functional studies. Our results demonstrated that As3mt methylated As(III) to DMA(V) as an end product and produced MMA(III) and MMA(V) as intermediates. The activity of As3mt was inhibited by elevated concentrations of the substrate As(III) as well as the metalloid selenite, which is a well-known antagonistic micronutrient of arsenic toxicity. The activity As3mt was abolished by substitution of either Cys160 or Cys210, which corresponds to conserved cysteine residues in AS3MT homologs, suggesting that they are involved in catalysis. Expression in zebrafish of an enzyme that has a similar function to human and rodent orthologs in catalyzing intracellular arsenic biomethylation validates the applicability of zebrafish as a valuable vertebrate model for understanding arsenic-associated diseases in humans.


Assuntos
Arsenitos/farmacocinética , Arsenitos/toxicidade , Metiltransferases/metabolismo , S-Adenosilmetionina/metabolismo , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Arsenicais/farmacocinética , Sequência de Bases , Clonagem Molecular/métodos , Feminino , Masculino , Metilação , Metiltransferases/antagonistas & inibidores , Metiltransferases/genética , Modelos Moleculares , Dados de Sequência Molecular , RNA Mensageiro/química , RNA Mensageiro/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Selenito de Sódio/farmacologia
18.
Mutat Res ; 735(1-2): 32-8, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22564430

RESUMO

Calcineurin is a Ca(2+)-dependent serine/threonine phosphatase and the target of the immunosuppressive drugs cyclosporin and tacrolimus, which are used in transplant recipients to prevent rejection. Unfortunately, the therapeutic use of this drugs is complicated by a high incidence of skin malignancy, which has set off a number of studies into the role of calcineurin signaling in skin, particularly with respect to cell cycle control and DNA repair. Both UVA1 radiation and arsenic species are known to promote skin cancer development via production of reactive oxygen species. In light of the well-documented sensitivity of calcineurin to oxidative stress, we examined and compared the effects of UVA1 and arsenite on calcineurin signaling. In this paper, we show that physiologically relevant doses of UVA1 radiation and low micromolar concentrations of arsenite strongly inhibit calcineurin phosphatase activity in Jurkat and skin cells and decrease NFAT nuclear translocation in Jurkat cells. The effects on calcineurin signaling could be partly prevented by inhibition of NADPH oxidase in Jurkat cells or increased dismutation of superoxide in Jurkat and skin cells. In addition, both UVA1 and arsenite decreased NF-κB activity, although at lower concentrations, arsenite enhanced NF-κB activity. These data indicate that UVA1 and arsenite affect a signal transduction route of growingly acknowledged importance in skin and that calcineurin may serve as a potential link between ROS exposure and impaired tumor suppression.


Assuntos
Arsenitos/farmacocinética , Calcineurina/metabolismo , Calcineurina/farmacologia , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Raios Ultravioleta , Células Cultivadas , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Humanos , Células Jurkat , Fatores de Transcrição NFATC/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Pele/metabolismo , Superóxidos/metabolismo , Fator de Transcrição RelA/metabolismo
19.
Exp Toxicol Pathol ; 64(1-2): 25-30, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20580540

RESUMO

UNLABELLED: The (As) arsenic exposure is a risk factor for causing disturbances in the endocrine organs. OBJECTIVE: To evaluate if sub-chronic As exposure during the pre- and postnatal development causes disturbances in the puberty. Moreover, determine adverse effects of As on the ovarian follicle and adrenocortical cell maturation. METHODS: Females adult Wistar rats were exposed to sodium arsenite at 3 ppm calculated as As in drinking water from mating, gestation. Following the birth, the female offspring continued exposured to As via lactation. Weaned pups received the same As treatment as mothers, until they were 1-4 months (mo) old. At these ages, blood sampling and tissue harvest were done. The tissues were fixed in situ with 4% paraformaldehyde in phosphate buffer. After the perfusion the ovaries, uterus, adrenal glands were harvested, dissected out, weighted. The ovaries and the adrenal glands were processed to paraffin and sectioned at 5 µM and stained with hematoxylin and eosin for light microscopy. STATISTICAL ANALYSIS: Comparisons between groups were made by unpaired t-test or nonparametric Mann-Whitney test as appropriate. RESULTS: 100% As treated rats at 1 mo of age were at diestrous stage, with low estradiol E2. As treatment caused disturbances in the morphology of the ovarian cell consisting in DNA damage evidenced by picknotic chromatin, cariorexis, significant cytoplasmic vacuolization and also vasculature damaged. Arrest in follicle maturation was also present. CONCLUSIONS: We found that the onset of puberty in the As treated rats was 1 mo delayed since vagina was still closed, the vaginal smear showed that they were at diestrus stage with plasma low E2 levels.


Assuntos
Envelhecimento/efeitos dos fármacos , Arsenitos/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Maturidade Sexual/efeitos dos fármacos , Compostos de Sódio/toxicidade , Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/crescimento & desenvolvimento , Envelhecimento/sangue , Animais , Arsenitos/farmacocinética , Estradiol/sangue , Ciclo Estral/sangue , Ciclo Estral/efeitos dos fármacos , Feminino , Lactação , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/sangue , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Progesterona/sangue , Ratos , Ratos Wistar , Compostos de Sódio/farmacocinética , Vagina/efeitos dos fármacos , Vagina/embriologia , Vagina/crescimento & desenvolvimento
20.
Toxicol Lett ; 204(2-3): 127-33, 2011 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-21554932

RESUMO

Arsenic is a pollutant widely distributed in the environment. There are numerous studies on the toxicity of trivalent arsenic forms As(III), MMA(III), and DMA(III), but few data are available on the processes of digestion and absorption of these arsenic species and the mechanisms involved are unknown. The present study evaluated the processes involved in intestinal absorption of trivalent arsenic species, using the Caco-2 cell model as system. The apparent permeability values obtained for As(III) in apical-basolateral direction (4.6±0.3)×10(-6)cm/s, showing moderate intestinal absorption. Transport of MMA(III) [P(app)=(7.0±0.9)×10(-6)cm/s] and DMA(III) [P(app)=(10.6±1.4)×10(-6)cm/s] is greater than that of As(III). The cellular retention of As(III) (0.9-2.4%) was less than that observed for MMA(III) (30%) and DMA(III) (35%). A substantial paracellular component was observed in transport of As(III) and MMA(III), whereas DMA(III) does not use this pathway for its absorption. For all the trivalent species, transport depends on temperature, with an active transcellular component for MMA(III) and DMA(III). Variations in pH do not affect transport of these species. The presence of GSH and green tea extract significantly alters transport of As(III) across Caco-2 cells.


Assuntos
Arsenitos/farmacocinética , Ácido Cacodílico/análogos & derivados , Mucosa Intestinal/metabolismo , Compostos Organometálicos/farmacocinética , Transporte Biológico , Células CACO-2 , Ácido Cacodílico/farmacocinética , Glutationa/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Permeabilidade , Extratos Vegetais/farmacologia , Chá , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA