Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Med Rep ; 27(2)2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36524366

RESUMO

Although arsenic is an environmental toxicant, arsenic trioxide (ATO) is used to treat acute promyelocytic leukemia (APL) with anticancer effects. Studies have demonstrated oral cancer is in the top 10 cancers in Taiwan. High rate of oral cancers is linked to various behaviors, such as excessive alcohol consumption and tobacco use. Similarly, betel chewing is a strong risk factor in oral cancer. In the present study, oral squamous carcinoma OC3 cells were investigated with the treatments of sodium arsenite (NaAsO2) and dimethylarsenic acid (DMA), respectively, to examine if arsenic compounds have anti­cancer efforts. It was found that 1 µM NaAsO2 and 1 mM DMA for 24 h induced rounded contours with membrane blebbing phenomena in OC3 cells, revealing cell apoptotic characteristics. In addition, NaAsO2 (10­100 µM) and DMA (1­100 mM) significantly decreased OC3 cell survival. In cell cycle regulation detected by flow cytometry, NaAsO2 and DMA significantly augmented percentage of subG1 and G2/M phases in OC3 cells, respectively. Annexin V/PI double staining assay was further used to confirm NaAsO2 and DMA did induce OC3 cell apoptosis. In mechanism investigation, western blotting assay was applied and the results showed that NaAsO2 and DMA significantly induced phosphorylation of JNK, ERK1/2 and p38 and then the cleavages of caspase­8, ­9, ­3 and poly ADP­ribose polymerase (PARP) in OC3 cells, dynamically. In conclusion, NaAsO2 and DMA activated MAPK pathways and then apoptotic pathways to induce OC3 oral cancer cell apoptosis.


Assuntos
Arsenicais , Neoplasias Bucais , Humanos , Ácido Cacodílico/farmacologia , Linhagem Celular Tumoral , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/patologia , Apoptose , Arsenicais/farmacologia
2.
Artigo em Chinês | MEDLINE | ID: mdl-36229210

RESUMO

Objective: To investigate the effect of arsenic and its main metabolites on the apoptosis of human lung adenocarcinoma cell line A549 and the expression of pro-apoptotic genes Bad and Bik. Methods: In October 2020, A549 cells were recovered and cultured, and the cell viability was detected by the cell counting reagent CCK-8 to determine the concentration and time of sodium arsenite exposure to A549. The study was divided into NaAsO(2) exposure groups and metobol: le expoure groups: the metabolite comparison groups were subdivided into the control group, the monomethylarsinic acid exposure group (60 µmol/L) , and the dimethylarsinic acid exposure group (60 µmol/L) ; sodium arsenite dose groups were subdivided into 4 groups: control group (0) , 20, 40, 60 µmol/L sodium arsenite NaAsO(2). Hoechst 33342/propidium iodide double staining (Ho/PI) was used to observe cell apoptosis and real-time quantitative polymerase chain reaction (qRT-PCR) was used to detect the expression levels of Bad and Bik mRNA in cells after exposure. Western blotting was used to detect the protein expressions of Bad, P-Bad-S112, Bik, cleaved Bik and downstream proteins poly ADP-ribose polymerase PARP1 and cytochrome C (Cyt-C) , using spectrophotometry to detect the activity changes of caspase 3, 6, 8, 9. Results: Compared with the control group, the proportion of apoptotic cells in the 20, 40, and 60 µmol/L NaAsO(2) dose groups increased significantly (P<0.01) , and the expression levels of Bad, Bik mRNA, the protein expression levels of Bad, P-Bad-S112, Bik, cleaved Bik, PARP1, Cyt-C were increased (all P<0.05) , and the activities of Caspase 3, 6, 8, and 9 were significantly increased with significantly differences (P<0.05) . Compared with the control group, the expression level of Bad mRNA in the DMA exposure group (1.439±0.173) was increased with a significant difference (P=0.024) , but there was no significant difference in the expression level of Bik mRNA (P=0.788) . There was no significant differences in the expression levels of Bad and Bik mRNA in the poison groups (P=0.085, 0.063) . Compared with the control group, the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to MMA were 0.696±0.023, 0.707±0.014, 0.907±0.031, 1.032±0.016, and there was no significant difference between the two groups (P=0.469, 0.669, 0.859, 0.771) ; the gray values of proteins Bad, Bik, PARP1 and Cyt-C exposed to DMA were 0.698±0.030, 0.705±0.022, 0.908±0.015, 1.029±0.010, and there was no difference between the two groups (P=0.479, 0.636, 0.803, 0.984) . Conclusion: Sodium arsenite induces the overexpression of Bad and Bik proteins, initiates the negative feedback regulation of phosphorylated Bad and the degradation of Bik, activates the downstream proteins PARP1, Cyt-C and Caspase pathways, and mediates the apoptosis of A549 cells.


Assuntos
Arsênio , Venenos , Células A549 , Adenosina Difosfato Ribose/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose , Arsenitos , Ácido Cacodílico/farmacologia , Caspase 3 , Caspases/farmacologia , Citocromos c/farmacologia , Humanos , Proteínas Mitocondriais/farmacologia , Propídio/farmacologia , RNA Mensageiro , Sincalida/farmacologia , Compostos de Sódio , Proteína de Morte Celular Associada a bcl/metabolismo
3.
Xenobiotica ; 48(2): 124-134, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28134025

RESUMO

1. Dimethylarsinic acid (DMA(V)) is the major metabolite of inorganic arsenic in human body. Thus we investigated the effect of DMA(V) on the alteration of phase I (typified by Cyp1a) and phase II (typified by Nqo1) AhR-regulated genes in vivo. C57BL/6 mice received DMA(V) (13.3 mg/kg, i.p.) with or without TCDD (15 µg/kg, i.p.), thereafter the liver, lung, and kidney were harvested at 6 and 24 h post-treatment. 2. Results demonstrated that DMA(V) has no significant effect on Cyp1a mRNA and protein expression or catalytic activity in the liver. On the other hand, DMA(V) significantly potentiated the TCDD-mediated induction of Cyp1a mRNA and protein expression, with a subsequent potentiation of catalytic activity in the lung. Moreover, DMA(V) significantly inhibited the TCDD-mediated induction of Cyp1a mRNA and protein expression with subsequent inhibition of catalytic activity in the kidney. 3. Regarding to phase II AhR-regulated genes, DMA(V) has no significant effect on Nqo1 mRNA and protein expression, or activity neither in the liver, lung, or kidney. 4. In conclusion, the present work demonstrates for the first time that DMA(V) modulates AhR-regulated genes in a tissue- and enzyme-specific manner. This modulation may play a crucial role in arsenic-induced toxicity and carcinogenicity.


Assuntos
Ácido Cacodílico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Herbicidas/farmacologia , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , RNA Mensageiro/metabolismo
4.
Mediators Inflamm ; 2015: 790652, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26617437

RESUMO

Dimethylarsinic acid (DMA(V)) is the main product of arsenic methylation metabolism in vivo and is rat bladder carcinogen and tumor promoting agent. In this study, we measured the expressions of mRNA and proteins of NF-κB pathway members, IKKα, IKKß, p65, and p50 in rat bladder epithelium by qRT-PCR and immunohistochemical analysis after rats received drinking water containing 100 and 200 ppm DMA(V) for 10 weeks. Transforming growth factor-ß (TGF-ß) immunoexpression in rat bladder epithelium and urine level of IL-1ß also were determined. We found that DMA(V) dramatically increased the mRNA levels of NF-κB p50 and IKKα in the bladder epithelium of rats compared to the control group. Immunohistochemical examinations showed that DMA(V) increased immunoreactivities of IKKα, IKKß, and phospho-NF-κB p50 in the cytoplasm and phospho-NF-κB p50 and p65 in nucleus of rat urothelial cells. In addition, DMA(V) treated rats exhibited significantly increased inflammatory factor TGF-ß immunoreactivity in bladder epithelium and IL-1ß secretion in urine. These data suggest that DMA(V) could activate NF-κB signal pathway and increase TGF-ß and IL-1ß expressions in bladder epithelial cells of rats.


Assuntos
Ácido Cacodílico/farmacologia , Água Potável/química , Interleucina-1beta/análise , NF-kappa B/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/análise , Bexiga Urinária/efeitos dos fármacos , Animais , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Feminino , Imuno-Histoquímica , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Bexiga Urinária/imunologia
5.
Oncotarget ; 6(28): 25646-59, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26213848

RESUMO

Arsenic trioxide (As2O3) is one of the most effective therapeutic agents used for patients with acute promyelocytic leukemia (APL). The probable explanation for As2O3-induced cell differentiation is the direct targeting of PML-RARα oncoprotein by As2O3, which results in initiation of PML-RARα degradation. However, after injection, As2O3 is rapidly methylated in body to different intermediate metabolites such as trivalent monomethylarsonous acid (MMA(III)) and dimethylarsinous acid (DMA(III)), therefore, it remains unknown that which arsenic specie is actually responsible for the therapeutic effects against APL. Here we have shown the role of As2O3 (as iAs(III)) and its intermediate metabolites (i.e., MMA(III)/DMA(III)) in NB4 cells. Inorganic iAs(III) predominantly showed induction of cell differentiation, while MMA(III) and DMA(III) specifically showed to induce mitochondria and endoplasmic reticulum-mediated apoptosis, respectively. On the other hand, in contrast to iAs(III), MMA(III) showed stronger binding affinity for ring domain of PML recombinant protein, however, could not induce PML protein SUMOylation and ubiquitin/proteasome degradation. In summary, our results suggest that the binding of arsenicals to the ring domain of PML proteins is not associated with the degradation of PML-RARα fusion protein. Moreover, methylated arsenicals can efficiently lead to cellular apoptosis, however, they are incapable of inducing NB4 cell differentiation.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Arsenicais/metabolismo , Arsenicais/farmacologia , Diferenciação Celular/efeitos dos fármacos , Leucemia Promielocítica Aguda/tratamento farmacológico , Proteínas Nucleares/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Óxidos/metabolismo , Óxidos/farmacologia , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Apoptose/efeitos dos fármacos , Trióxido de Arsênio , Biotransformação , Ácido Cacodílico/análogos & derivados , Ácido Cacodílico/metabolismo , Ácido Cacodílico/farmacologia , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HEK293 , Células HeLa , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Metilação , Compostos Organometálicos/metabolismo , Compostos Organometálicos/farmacologia , Proteína da Leucemia Promielocítica , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteólise , Sumoilação , Fatores de Tempo
6.
Rev. bras. enferm ; 67(6): 979-986, Nov-Dec/2014. graf
Artigo em Português | LILACS, BDENF | ID: lil-732807

RESUMO

Este artigo apresenta uma compreensão sobre o autocuidado do cuidador familiar segundo a teoria de Dorothea Orem. Resulta de uma pesquisa qualitativa com aporte da Teoria Fundamentada nos Dados, utilizando-se das técnicas de visita domiciliar, registro de notas de campo e entrevista semiestruturada com 11 cuidadores, após a internação de familiar em um Hospital Universitário de Minas Gerais. Foram obtidas quatro categorias, destacando uma categoria central, em torno da qual se analisaram as facilidades, dificuldades e estratégias para o autocuidado do cuidador. Entre as dificuldades, foram evidenciadas: tempo insuficiente para os cuidados com a saúde e, entre as facilidades, o apoio de outros familiares. As principais estratégias foram: apoio na fé; revezamento nos cuidados e recursos na comunidade. Concluiu-se que orientações no momento da alta e o acompanhamento de enfermagem após a alta contribuem para o autocuidado do cuidador, atuando sobre suas dificuldades e estimulando suas potencialidades.


This article presents an understanding concerning self-care in family caregivers according to Dorothea Orem's theory. Resulting from a qualitative research based on Grounded Theory, this work uses the techniques of home visiting, field notes and semistructured interviews with 11 caregivers after the hospitalization of a family member in a teaching Hospital located in Minas Gerais. Four categories were found and among them a central category is highlighted from which some facilities, difficulties and strategies for selfcare in caregiver were analyzed. Considering the difficulties, insufficient time for healthcare was noticed whereas the support from other family members appeared as a facility. The main strategies were: faith as a support; shift work in healthcare and community resources. This study demonstrated that hospital discharge guidelines and nursing follow-up after discharge were responsible for positive contributions to self-care in caregivers helping them to overcome their difficulties and enhancing their potentialities.


El artículo presenta una comprensión sobre el autocuidado del cuidador familiar, según la teoría de Dorothea Orem. Resulta de investigación cualitativa con aporte de la Teoría Fundamentada en los Datos, utilizándo se de las técnicas de visitas a domicilio; registro de apuntes de campo y entrevista semiestructurada, tras la internación de un familiar en un hospital universitario de Minas Gerais. Se llegó a cuatro categorías, señalando una categoría central, alrededor de la cual se analizaron las facilidades, dificultades y estrategias para el autocuidado del cuidador. Se evidenció, entre las dificultades, tiempo insuficiente para los cuidados con la salud y, entre las facilidades, el apoyo de otros familiares. Las principales estrategias fueron: apoyo en la fe, revezo en los cuidados y recursos en la comunidad. Se concluyó que orientaciones el momento del alta y el acompañamiento de enfermería tras el alta contribuyen para el autocuidado del cuidador, actuando sobre sus dificultades y estimulando sus potencialidades.


Assuntos
Animais , Masculino , Ratos , Arsenicais/farmacologia , Ácido Cacodílico/farmacologia , Compostos Férricos/farmacocinética , Absorção Intestinal/fisiologia , Ânions/farmacocinética , Coloides , Cátions/farmacocinética , Histocitoquímica , Ratos Endogâmicos
7.
Metallomics ; 6(8): 1502-12, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24870979

RESUMO

Arsenic trioxide (As2O3) has remarkable therapeutic efficacy against leukemia. However, after As2O3 biotransformation, the role of arsenic metabolites in the clinical efficacy against leukemia still needs to be elucidated. Therefore, to explore the contribution of trivalent methylated arsenicals in the therapeutic effects, we investigated and compared the effects of arsenite (iAs(III)), monomethylarsonous acid (MMA(III)) and dimethylarsinous acid (DMA(III)) on HL-60 cells. Methylated arsenic species MMA(III) and DMA(III) showed potentially reduced cell survival with IC50 values of 3 and 2 µM, respectively. We found that methylated metabolites caused apoptosis through oxidative stress and loss of mitochondrial membrane potential. Furthermore, we found that the caspase-9 and -3 were markedly activated by exposure to methylated metabolites, with cleavage of poly-ADP ribose polymerase (PARP). Conversely, cellular apoptosis, generation of ROS, activation of caspase-3, -9 as well as PARP cleavage were significantly attenuated by pretreatment with an antioxidant, N-acetylcysteine (NAC). DNA damage was also markedly observed in HL-60 cells exposed to either MMA(III) or DMA(III), while iAs(III) did not show any relevant effects in HL-60 cells. Likewise, phosphorylation of the histone H2A variant (γ-H2AX), a biomarker of DNA damage, significantly occurred in cellular nuclei following exposure to two methylated species, which was reduced in the presence of NAC, suggesting that the induction of DNA damage was predominantly caused by the two metabolites via oxidative stress. In conclusion, we suggest that arsenic intermediate metabolites; MMA(III) and DMA(III) might prove to be of clinical relevance in future as such approaches may help in the treatment of leukemia and other types of cancers.


Assuntos
Arsênio/farmacologia , Apoptose/efeitos dos fármacos , Trióxido de Arsênio , Arsenicais/farmacologia , Arsenitos/farmacologia , Ácido Cacodílico/análogos & derivados , Ácido Cacodílico/farmacologia , Células HL-60 , Humanos , Óxidos/farmacologia , Espécies Reativas de Oxigênio/metabolismo
8.
Toxicol Sci ; 128(1): 137-46, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22539622

RESUMO

Arsenic is known to be a human carcinogen as well as one of the most effective drugs for treatment of patients with acute promyelocytic leukemia. The intermediate metabolites of arsenic, monomethylarsonous acid (MMA(III)) and dimethylarsinous acid (DMA(III)), are formed by methylation reactions, and they are more reactive and toxic than the inorganic precursor arsenite (iAs(III)); however, the detailed mechanism of toxicity is poorly understood. Here, we studied the effects of three arsenic compounds (i.e., iAs(III), MMA(III), and DMA(III)) on mitochondrial permeability transition pore (mPTP) and release of apoptotic cytochrome c (Cyt c) after incubating with rat liver mitochondria. Inorganic iAs(III) had no effect on mitochondrial swelling even at higher concentrations ranging up to 100 µM, but swelling was significantly induced in the presence of Ca(2+). Additionally, mitochondrial swelling was strongly induced by exposure to the methylated forms of MMA(III) and DMA(III) in a dose-dependent manner in the absence of Ca(2+), suggesting that the methylated forms may have potent effects on cellular mitochondria. Although mitochondrial swelling was completely inhibited in the presence of cyclosporin-A (an inhibitor of mitochondrial permeability transition) or ruthenium red (an inhibitor of Ca(2+) uniporter) following exposure to methylated arsenicals, the release of apoptotic Cyt c from mitochondria was not inhibited, indicating that release of Cyt c is probably not dependent on mPTP opening. In addition, inhibitors of Bax (e.g., Bax-inhibiting peptide) did not reduce the release of Cyt c from the mitochondria by formation of Bax-voltage-dependent anion channel (VDAC) complex, whereas the recombinant Bcl-x(L )proteins significantly reduced the release of Cyt c after exposure to DMA(III), suggesting that dimethylated DMA(III) directly interacted with the VDAC in mitochondria and caused the release of Cyt c from mitochondria.


Assuntos
Apoptose , Ácido Cacodílico/análogos & derivados , Citocromos c/metabolismo , Canais Iônicos/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Animais , Ácido Cacodílico/farmacologia , Linhagem Celular , Mitocôndrias Hepáticas/enzimologia , Ratos
9.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 5): 421-33, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19390147

RESUMO

The signal recognition particle (SRP) Alu domain has been implicated in translation elongation arrest in yeasts and mammals. Fission yeast SRP RNA is similar to that of mammals, but has a minimal Alu-domain RNA lacking two stem-loops. The mammalian Alu-domain proteins SRP9 and SRP14 bind their cognate Alu RNA as a heterodimer. However, in yeasts, notably Saccharomyces cerevisiae, SRP14 is thought to bind Alu RNA as a homodimer, the SRP9 protein being replaced by SRP21, the function of which is not yet clear. Structural characterization of the Schizosaccharomyces pombe Alu domain may thus help to identify the critical features required for elongation arrest. Here, the crystal structure of the SRP14 subunit of S. pombe SRP (SpSRP14) which crystallizes as a homodimer, is presented. Comparison of the SpSRP14 homodimer with the known structure of human SRP9/14 in complex with Alu RNA suggests that many of the protein-RNA contacts centred on the conserved U-turn motif are likely to be conserved in fission yeast. Initial attempts to solve the structure using traditional selenomethionine SAD labelling failed. However, two As atoms originating from the cacodylate buffer were found to make cysteine adducts and strongly contributed to the anomalous substructure. These adducts were highly radiation-sensitive and this property was exploited using the RIP (radiation-damage-induced phasing) method. The combination of SAD and RIP phases yielded an interpretable electron-density map. This example will be of general interest to crystallographers attempting de novo phasing from crystals grown in cacodylate buffer.


Assuntos
Cristalografia por Raios X/métodos , Proteínas de Ligação a RNA/química , Proteínas de Schizosaccharomyces pombe/química , Schizosaccharomyces/química , Partícula de Reconhecimento de Sinal/química , Elementos Alu , Sequência de Aminoácidos , Sítios de Ligação , Ácido Cacodílico/farmacologia , Sequência Conservada , Cristalização , Dimerização , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Conformação Proteica , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/isolamento & purificação , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/isolamento & purificação , Proteínas de Schizosaccharomyces pombe/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
10.
Mutat Res ; 675(1-2): 66-70, 2009 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-19386250

RESUMO

A short-term cell transformation assay has recently been developed, using Bhas 42 cells which were established from BALB/c 3T3 cells transfected by v-Ha-ras gene and postulated to be initiated in the two-stage carcinogenesis theory. The Bhas 42 cell transformation assay has been reported to be capable of detecting initiating and promoting activities of chemical carcinogens, according to the different protocols, initiation assay and promotion assay, respectively. The assay is superior to classical transformation assays in cost and labor performance. The present study was carried out to compare its sensitivity with that of a classical BALB/c 3T3 cell system. We performed the Bhas 42 cell transformation assay with inorganic arsenic compounds which are potent environmental carcinogens in human but not mutagens in bacteria or weak mutagens in mammalian cells in vitro. Sodium arsenite, disodium arsenate, and their metabolites, monomethylarsonic acid and dimethylarsinic acid (DMAA) were included in the study. Sodium arsenite was positive in the initiation assay and all compounds except for DMAA were positive in the promotion assay. These results were compared with reported data in a two-stage BALB/c 3T3 cell transformation assay. The sensitivity of Bhas 42 cell transformation assay was found to be similar to that of the conventional BALB/c 3T3 cell transformation assay for the detection of initiating activities of arsenic compounds. For the detection of promoting activities, its sensitivity was equivalent to that of the two-stage BALB/c 3T3 cell transformation assay where the target cells were initiated with sub-threshold dose of 3-methylcholanthrene, confirming that Bhas 42 cells behave as initiated cells in the transformation assay.


Assuntos
Arsenicais/farmacologia , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Animais , Arseniatos/farmacologia , Arsenitos/farmacologia , Células 3T3 BALB , Ácido Cacodílico/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Camundongos
11.
Chem Res Toxicol ; 22(2): 382-90, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19146383

RESUMO

Recently, inorganic arsenite (iAs(III)) and its mono- and dimethylated metabolites have been examined for their interference with the formation and repair of benzo[a]pyrene diol epoxide (BPDE)-induced DNA adducts in human cells (Schwerdtle, ., Walter, I., and Hartwig, A. (2003) DNA Repair 2, 1449 - 1463). iAs(III) and monomethylarsonous acid (MMA(III)) were found to be able to enhance the formation of BPDE-DNA adducts, whereas dimethylarsinous acid (DMA(III)) had no enhancing effect at all. The anomaly manifested by DMA(III) prompted us to further investigate the effects of the three trivalent arsenic species on the formation of BPDE-DNA adducts. Use of a nucleotide excision repair (NER)-deficient Xeroderma pigmentosum complementation group A cell line (GM04312C) allowed us to dissect DNA damage induction from DNA repair and to examine the effects of arsenic on the formation of BPDE-DNA adducts only. At concentrations comparable to those used in the study by Schwerdtle et al., we found that each of the three trivalent arsenic species was able to enhance the formation of BPDE-DNA adducts with the potency in a descending order of MMA(III) > DMA(III) > iAs(III), which correlates well with their cytotoxicities. Similar to iAs(III), DMA(III) modulation of reduced glutathione (GSH) or total glutathione S-transferase (GST) activity could not account for its enhancing effect on DNA adduct formation. Additionally, the enhancing effects elicited by the trivalent arsenic species were demonstrated to be highly time-dependent. Thus, although our study made use of short-term assays with relatively high doses, our data may have meaningful implications for carcinogenesis induced by chronic exposure to arsenic at low doses encountered environmentally.


Assuntos
7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/metabolismo , Arsenitos/toxicidade , Ácido Cacodílico/análogos & derivados , Adutos de DNA/metabolismo , Compostos Organometálicos/toxicidade , 7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/química , Arsenitos/farmacologia , Ácido Cacodílico/farmacologia , Ácido Cacodílico/toxicidade , Linhagem Celular , Linhagem Celular Transformada , Dano ao DNA , Reparo do DNA , Eletroforese Capilar , Fibroblastos/efeitos dos fármacos , Glutationa Transferase/metabolismo , Humanos , Compostos Organometálicos/farmacologia , Fatores de Tempo , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo
12.
Chem Biol ; 15(3): 274-86, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18355727

RESUMO

Eukaryotic cells produce a family of diverse inositol polyphosphates (IPs) containing pyrophosphate bonds. Inositol pyrophosphates have been linked to a wide range of cellular functions, and there is growing evidence that they act as second messengers. Inositol hexakisphosphate kinase (IP6K) is able to convert the natural substrates inositol pentakisphosphate (IP 5) and inositol hexakisphosphate (IP 6) to several products with an increasing number of phospho-anhydride bonds. In this study, we structurally analyzed IPs synthesized by three mammalian isoforms of IP6K from IP 5 and IP 6. The NMR and mass analyses showed a number of products with diverse, yet specific, stereochemistry, defined by the architecture of IP6K's active site. We now report that IP6K synthesizes both pyrophosphate (diphospho) as well as triphospho groups on the inositol ring. All three IP6K isoforms share the same activities both in vitro and in vivo.


Assuntos
Difosfatos/análise , Difosfatos/química , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Polifosfatos/análise , Polifosfatos/química , Ácido Cacodílico/farmacologia , Cromatografia Líquida de Alta Pressão , Fosfatos de Inositol/química , Fosfatos de Inositol/metabolismo , Isoenzimas/metabolismo , Espectrometria de Massas , Fosfotransferases (Aceptor do Grupo Fosfato)/antagonistas & inibidores , Ácido Fítico/química , Ácido Fítico/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Estereoisomerismo , Especificidade por Substrato
13.
Toxicol Appl Pharmacol ; 220(3): 262-70, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17321558

RESUMO

In a poisonous incident in Kamisu, Japan, it is understood that diphenylarsinic acid (DPAA) was a critical contaminant of ground water. Most patients showed dysfunction of the central nervous system. To understand the overall mechanism of DPAA toxicity and to gain some insight into the application of a remedy specific for intoxication, the molecular target must be clarified. As an approach, a high throughput analysis of cell proteins in cultured human hepatocarcinoma HpG2 exposed to DPAA was performed by two-dimensional electrophoresis (2-DE). Four proteins, which were up- and down-regulated by exposure of cultured HepG2 cells to DPAA, were identified. They were chaperonin containing TCP-1 (CCT) beta subunit, aldehyde dehydrogenase 1 (ALDH1), ribosomal protein P0 and glutaminase C (GAC). Of these, GAC was the only protein that was down-regulated by DPAA exposure, and cellular expression levels were reduced by DPAA in a concentration- and time-dependent manner. Decrease in cellular GAC levels was accompanied by decreased activity of the enzyme, phosphate-activated glutaminase (PAG). Decreased expression of GAC by DPAA was also observed in human cervical carcinoma HeLa and neuroblastoma SH-SY5Y cells. By contrast, no significant changes in GAC protein expression were observed when cells were incubated with arsenite [iAs (III)] and trivalent dimethylarsinous acid [DMA (III)]. In the central nervous system, GAC plays a role in the production of the neurotransmitter glutamic acid. Selective inhibition of GAC expression by DPAA may be a cause of dysfunction of glutamatergic neuronal transmission and the resultant neurological impairments.


Assuntos
Arsenicais/farmacologia , Regulação para Baixo/efeitos dos fármacos , Glutaminase/metabolismo , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Arsenicais/metabolismo , Arsenitos/farmacologia , Ácido Cacodílico/análogos & derivados , Ácido Cacodílico/farmacologia , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Chaperonina com TCP-1 , Chaperoninas/farmacologia , Substâncias para a Guerra Química/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel Bidimensional , Imunofluorescência , Células HeLa , Humanos , Immunoblotting , Isoenzimas/metabolismo , Retinal Desidrogenase , Proteínas Ribossômicas/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fatores de Tempo
14.
Carcinogenesis ; 24(5): 967-74, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12771042

RESUMO

Even though a well-known human carcinogen the underlying mechanisms of arsenic carcinogenicity are still not fully understood. For arsenite, proposed mechanisms are the interference with DNA repair processes and an increase in reactive oxygen species. Even less is known about the genotoxic potentials of its methylated metabolites monomethylarsonous [MMA(III)] and dimethylarsinous [DMA(III)] acid, monomethylarsonic [MMA(V)] and dimethylarsinic [DMA(V)] acid. Within the present study we compared the induction of oxidative DNA damage by arsenite and its methylated metabolites in cultured human cells and in isolated PM2 DNA, by frequencies of DNA strand breaks and of lesions recognized by the bacterial formamidopyrimidine-DNA glycosylase (Fpg). Only DMA(III) (> or =10 micro M) generated DNA strand breaks in isolated PM2 DNA. In HeLa S3 cells, short-term incubations (0.5-3 h) with doses as low as 10 nM arsenite induced high frequencies of Fpg-sensitive sites, whereas the induction of oxidative DNA damage after 18 h incubation was rather low. With respect to the methylated metabolites, both trivalent and pentavalent metabolites showed a pronounced induction of Fpg-sensitive sites in the nanomolar or micromolar concentration range, respectively, which was present after both short-term and long-term incubations. Furthermore MMA(III) and DMA(V) generated DNA strand breaks in a concentration-dependent manner. Taken together our results show that very low physiologically relevant doses of arsenite and the methylated metabolites induce high levels of oxidative DNA damage in cultured human cells. Thus, biomethylation of inorganic arsenic may be involved in inorganic arsenic-induced genotoxicity/carcinogenicity.


Assuntos
Arsenicais/farmacologia , Dano ao DNA , DNA/efeitos dos fármacos , Bacteriófagos/genética , Ácido Cacodílico/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ensaio Cometa , Adutos de DNA/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , DNA Bacteriano/efeitos dos fármacos , DNA-Formamidopirimidina Glicosilase , Relação Dose-Resposta a Droga , Células HeLa/efeitos dos fármacos , Humanos , Cinética , N-Glicosil Hidrolases/farmacologia , Compostos Organometálicos/farmacologia , Oxirredução
15.
Toxicol Lett ; 143(2): 145-53, 2003 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-12749818

RESUMO

The purpose of the present study was to elucidate the genotoxic mechanism of trivalent dimethylated arsenic, particularly the induction mechanism of oxidative stress in nuclear bases. Cis-thymine glycol was used as a biomarker of DNA oxidation damage. The treatment of thymine with dimethylarsinous iodide (DMI), a model compound of dimethylarsinous acid, induced the formation of cis-thymine glycol. This oxidative damage was induced via the production of dimethylated arsenic peroxide, but not via the production of superoxide anion or hydrogen peroxide. Trivalent dimethylated arsenic may thus play an important role in arsenic carcinogenesis through the induction of oxidative base damage.


Assuntos
Ácido Cacodílico/análogos & derivados , Ácido Cacodílico/farmacologia , Dano ao DNA/efeitos dos fármacos , Timina/análogos & derivados , Timina/química , Ácido Cacodílico/química , Catalase/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Estrutura Molecular , Oxirredução/efeitos dos fármacos , Superóxido Dismutase/metabolismo
16.
Cancer Chemother Pharmacol ; 51(5): 427-32, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12736761

RESUMO

PURPOSE: Arsenic trioxide (As(2)O(3)), an inorganic arsenic compound, has recently been approved for the treatment of relapsed or refractory acute promyelocytic leukemia. However, systemic toxicity associated with As(2)O(3) treatment remains a problem. Inorganic arsenic is detoxified in vivo by methylation reactions into organic arsenic compounds that are less toxic. METHODS AND RESULTS: We investigated the antiproliferative and cytotoxic activity of dimethylarsinic acid (DMAA), an organic arsenic derivative and major metabolic by-product of As(2)O(3), against a panel of eight leukemia and multiple myeloma cell lines. As(2)O(3) was tested in comparison. In clonogenic assay, the average concentration of DMAA that suppressed cell colony growth by 50% was 0.5-1 m M, while for As(2)O(3) it was on average 1-2 microM. At those concentrations DMAA and As(2)O(3) had significantly less effect on colony growth of normal progenitor cells. Cytotoxic doses of DMAA and As(2)O(3) in 3-day trypan blue dye exclusion assay experiments were similar to doses effective in clonogenic assay. Assessment of apoptosis by annexin V assay revealed a high rate of apoptosis in all cell lines treated with DMAA and As(2)O(3), but significantly less effect on normal progenitor cells. DMAA, unlike As(2)O(3), had no effect on the maturation of leukemic cells. CONCLUSIONS: DMAA exerts differential antiproliferative and cytotoxic activity against leukemia and multiple myeloma cells, with no significant effect on normal progenitor cells. However, concentrations of DMAA needed to achieve such efficacy are up to 1000 times those of As(2)O(3). Evaluation of novel organic arsenic that would combine the high efficacy of As(2)O(3) and the low toxicity of DMAA is warranted.


Assuntos
Ácido Cacodílico/farmacologia , Herbicidas/farmacologia , Leucemia/patologia , Mieloma Múltiplo/patologia , Morte Celular , Divisão Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Células Tumorais Cultivadas
17.
Carcinogenesis ; 23(8): 1387-97, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12151359

RESUMO

Arsenic is a well-documented human carcinogen, and contamination with this heavy metal is of global concern, presenting a major issue in environmental health. However, the mechanism by which arsenic induces cancer is unknown, in large part due to the lack of an appropriate animal model. In the present set of experiments, we focused on dimethylarsinic acid (DMA), a major metabolite of arsenic in most mammals including humans. We provide, for the first time, the full data, including detailed pathology, of the carcinogenicity of DMA in male F344 rats in a 2-year bioassay, along with the first assessment of the genetic alteration patterns in the induced rat urinary bladder tumors. Additionally, to test the hypothesis that reactive oxygen species (ROS) may play a role in DMA carcinogenesis, 8-hydroxy-2'-deoxyguanosine (8-OHdG) formation in urinary bladder was examined. In experiment 1, a total of 144 male F344 rats at 10 weeks of age were randomly divided into four groups that received DMA at concentrations of 0, 12.5, 50 and 200 p.p.m. in the drinking water, respectively, for 104 weeks. From weeks 97-104, urinary bladder tumors were observed in 8 of 31 and 12 of 31 rats in groups treated with 50 and 200 p.p.m. DMA, respectively, and the preneoplastic lesion, papillary or nodular hyperplasias (PN hyperplasia), was noted in 12 and 14 rats, respectively. DMA treatment did not cause tumors in other organs and no urinary bladder tumors or preneoplastic lesions were evident in the 0 and 12.5 p.p.m.-treated groups. Urinary levels of arsenicals increased significantly in a dose-responsive manner except for arsenobetaine (AsBe). DMA and trimethylarsine oxide (TMAO) were the major compounds detected in the urine, with small amounts of monomethylarsonic acid (MMA) and tetramethylarsonium (TeMa) also detected. Significantly increased 5-bromo-2'-deoxyuridine (BrdU) labeling indices were observed in the morphologically normal epithelium of the groups treated with 50 and 200 p.p.m. DMA. Mutation analysis showed that DMA-induced rat urinary bladder tumors had a low rate of H-ras mutations (2 of 20, 10%). No alterations of the p53, K-ras or beta-catenin genes were detected. Only one TCC (6%) demonstrated nuclear accumulation of p53 protein by immunohistochemistry. In 16 of 18 (89%) of the TTCs and 3 of 4 (75%) of the papillomas, decreased p27(kip1) expression could be demonstrated. Cyclin D1 overexpression was observed in 26 of 47 (55%) PN hyperplasias, 3 of 4 (75%) papillomas, and 10 of 18 (56%) TCCs. As a molecular marker of oxidative stress, increased COX-2 expression was noted in 17 of 18 (94%) TCCs, 4 of 4 (100%) papillomas, and 39 of 47 (83%) PN hyperplasias. In experiment 2, 8-OHdG formation in urinary bladder was significantly increased after treatment with 200 p.p.m. DMA in the drinking water for 2 weeks compared with the controls. The studies demonstrated DMA to be a carcinogen for the rat urinary bladder and suggested that DMA exposure may be relevant to the carcinogenic risk of inorganic arsenic in humans. Diverse genetic alterations observed in DMA-induced urinary bladder tumors imply that multiple genes are involved in stages of DMA-induced tumor development. Furthermore, generation of ROS is likely to play an important role in the early stages of DMA carcinogenesis.


Assuntos
Ácido Cacodílico/farmacologia , Carcinógenos/toxicidade , Desoxiguanosina/análogos & derivados , Mutação , Neoplasias da Bexiga Urinária/induzido quimicamente , 8-Hidroxi-2'-Desoxiguanosina , Animais , Proteínas do Citoesqueleto/genética , Desoxiguanosina/metabolismo , Genes p53 , Genes ras , Imuno-Histoquímica , Masculino , Ratos , Ratos Endogâmicos F344 , Transativadores/genética , Urinálise , Neoplasias da Bexiga Urinária/genética , beta Catenina
18.
Int J Cancer ; 100(2): 136-9, 2002 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-12115560

RESUMO

Dimethylarsinic acid (DMA) is a major metabolite of inorganic arsenicals, which are epidemiologically significant chemicals in relation to liver cancer in mammals. The present study was conducted to determine the promoting effects of organic arsenicals related to DMA [monomethylarsonic acid (MMA) and trimethylarsine oxide (TMAO)] on rat liver carcinogenesis using a liver medium-term bioassay (the Ito test). Male, 10-week-old, F344 rats were given a single i.p. injection of diethylnitrosamine at a dose of 200 mg/kg b.w. as an initiator. Starting 2 weeks thereafter they received 100 ppm of MMA, DMA or TMAO in their drinking water, or no supplement as a control, for 6 weeks. All animals underwent 2/3 partial hepatectomy in week 3 after initiation. Quantification of glutathione S-transferase placental form (GST-P)-positive foci as preneoplastic lesions in liver sections revealed significantly increased numbers and areas in all 3 treated groups compared with controls. Hepatic microsome cytochrome P-450 content was markedly increased with all 3 arsenic treatments. Markedly elevated CYP 2B1 protein levels and CYP 2B1/2 mRNA levels were thus observed in all cases. The potency of promotion was similar for MMA, DMA and TMAO. Since hydroxyradicals were found to be generated in the relatively early phase while methylated arsenicals were metabolized in liver, the resultant oxidative stress might have promoted lesion development.


Assuntos
Arsenicais/farmacologia , Hidrocarboneto de Aril Hidroxilases , Ácido Cacodílico/farmacologia , Carcinógenos/farmacologia , Desoxiguanosina/análogos & derivados , Glutationa Transferase/biossíntese , Isoenzimas/biossíntese , Neoplasias Hepáticas Experimentais/enzimologia , Fígado/efeitos dos fármacos , Lesões Pré-Cancerosas/enzimologia , Espécies Reativas de Oxigênio/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Animais , Western Blotting , Citocromo P-450 CYP2B1/genética , Citocromo P-450 CYP2B1/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Desoxiguanosina/metabolismo , Glutationa S-Transferase pi , Técnicas Imunoenzimáticas , Fígado/enzimologia , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/patologia , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Estresse Oxidativo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo
19.
Mutat Res ; 499(1): 73-84, 2002 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-11804606

RESUMO

The role of microtubule-based motors in the induction of abnormal centrosome integrity by dimethylarsinic acid (DMAA) was investigated with the use of monastrol, a specific inhibitor of mitotic kinesin, and vanadate, an inhibitor of dynein ATPase. Cytoplasmic dynein co-localized with multiple foci of gamma-tubulin in mitotic cells arrested by DMAA. Disruption of microtubules caused dispersion of dynein while multiple foci of gamma-tubulin were coalesced to a single dot. Vanadate also caused dispersion of dynein, which had been co-localized with multiple foci of gamma-tubulin by DMAA, without affecting spindle organization. However, the dispersion of dynein did not prohibit the induction of abnormal centrosome integrity by DMAA. Inhibition of mitotic kinesin by monastrol resulted in monoastral cells with non-migrated centrosomes in the cell center. Monastrol, when applied to mitotic cells with abnormal centrosome integrity, rapidly reduced the incidence of cells with the centrosome abnormality. Moreover, monastrol completely inhibited reorganization of abnormal centrosomes that had been coalesced to a single dot by microtubule disruption. These results suggest that abnormal centrosome integrity caused by DMAA is not simply due to dispersion of fragments of microtubule-organizing centers, but is dependent on the action of kinesin. In addition, the results suggest that kinesin plays a role not only in the induction of mitotic centrosome abnormality, but also in maintenance.


Assuntos
Ácido Cacodílico/farmacologia , Centrossomo/efeitos dos fármacos , Dineínas/metabolismo , Cinesinas/metabolismo , Fuso Acromático/fisiologia , Animais , Células Cultivadas , Cricetinae , Cricetulus , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Dineínas/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Cinesinas/antagonistas & inibidores , Cinesinas/efeitos dos fármacos , Masculino , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Pirimidinas/farmacologia , Fuso Acromático/efeitos dos fármacos , Tionas/farmacologia , Tubulina (Proteína)/metabolismo , Vanadatos/farmacologia
20.
Mutat Res ; 493(1-2): 31-8, 2001 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-11516713

RESUMO

Numerous epidemiological studies suggest that arsenic (As) compounds are carcinogens, however, recent data have renewed the interest in their anticarcinogenic properties. The cytotoxic effects of three arsenic compounds were assessed: sodium arsenite, sodium arsenate and sodium cacodylate, representing the trivalent and pentavalent species of arsenic, along with a dimethylated pentavalent arsenic species. HeLa cells and Salmonella typhimurium (strains TA98 and TA100) were exposed to As compounds and the cytotoxic effects were evaluated. Alterations on RNA and DNA synthesis in HeLa cells were also examined. All arsenic compounds produced a dose-dependent inhibition on colony formation and DNA synthesis in HeLa cells, yet any of them significantly influenced RNA synthesis in these cells. No evidence of arsenic-induced mutagenicity or antimutagenicity was observed using the Ames assay. In bacterial cells, only sodium arsenite caused a dose-dependent inhibition of colony formation.Collectively, these results indicate that in both, HeLa and S. typhimurium cell systems, only trivalent sodium arsenite can act as an effective inhibitor of cell growth. The possible mechanism(s) of the cytotoxic effect of arsenite in these two different cell systems might be due to its reactivity with intracellular sulfhydryl groups.


Assuntos
Antineoplásicos/farmacologia , Arseniatos/farmacologia , Arseniatos/toxicidade , Arsenitos/farmacologia , Arsenitos/toxicidade , Ácido Cacodílico/farmacologia , Ácido Cacodílico/toxicidade , Salmonella typhimurium/efeitos dos fármacos , Compostos de Sódio/farmacologia , Compostos de Sódio/toxicidade , Antimutagênicos/farmacologia , Divisão Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , DNA/biossíntese , Células HeLa , Humanos , Testes de Mutagenicidade , Mutagênicos/toxicidade , RNA/biossíntese , Salmonella typhimurium/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA