Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arch Toxicol ; 88(2): 249-61, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24091636

RESUMO

Inorganic arsenic (iAs) and its toxic methylated metabolite, methylarsonous acid (MMA(III)), both have carcinogenic potential. Prior study shows iAs-induced malignant transformation in both arsenic methylation-proficient (liver) and methylation-deficient (prostate) cells, but only methylation-proficient cells show oxidative DNA damage (ODD) during this transformation. To further define whether arsenic methylation is necessary for transformation or ODD induction, here we chronically exposed these same liver or prostate cell lines to MMA(III) (0.25-1.0 µM) and tested for acquired malignant phenotype. Various metrics of oncogenic transformation were periodically assessed along with ODD during chronic MMA(III) exposure. Methylation-deficient and methylation-proficient cells both acquired a cancer phenotype with MMA(III) exposure at about 20 weeks, based on increased matrix metalloproteinase secretion, colony formation, and invasion. In contrast, prior work showed iAs-induced transformation took longer in biomethylation-deficient cells (~30 weeks) than in biomethylation-proficient cells (~18 weeks). In the present study, MMA(III) caused similar peak ODD levels at similar concentrations and at similar exposure times (18-22 weeks) in both cell types. At the approximate peak of ODD production, both cell types showed similar alterations in arsenic and oxidative stress adaptation factors (i.e., ABCC1, ABCC2, GST-π, SOD-1). Thus, MMA(III) causes oncogenic transformation associated with ODD in methylation-deficient cells, indicating that further methylation is not required to induce ODD. Together, these results show that MMA(III) and iAs cause an acquired malignant phenotype in methylation-deficient cells, yet iAs does not induce ODD. This indicates iAs likely has both genotoxic and non-genotoxic mechanisms dictated by the target cell's ability to methylate arsenic.


Assuntos
Arsenicais/farmacologia , Transformação Celular Neoplásica/induzido quimicamente , Dano ao DNA/efeitos dos fármacos , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Animais , Intoxicação por Arsênico/patologia , Linhagem Celular/efeitos dos fármacos , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Masculino , Metilação , Proteína 2 Associada à Farmacorresistência Múltipla , Oxirredução , PTEN Fosfo-Hidrolase/genética , Próstata/citologia , Próstata/metabolismo , Ratos , Testes de Toxicidade Crônica
2.
Toxicol Mech Methods ; 23(6): 389-95, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23301828

RESUMO

The carcinogen inorganic arsenic (iAs) undergoes biomethylation (BMT) in some cells. The methylated metabolite, monomethylarsonous (MMA(3+)), may cause oxidative DNA damage (ODD). With chronic iAs exposure, BMT-competent cells show ODD while BMT-deficient do not. To further define these events, we studied ODD produced by acute iAs or MMA(3+) in the BMT-deficient human prostate cell line, RWPE-1. ODD, measured by the immuno-spin trapping method, was assessed after exposure to iAs or MMA(3+) alone, with the arsenic BMT inhibitor selenite or after glutathione (GSH) depletion. The expression of oxidative stress-related genes (HO-1, SOD-1, SOD-2, Nrf2 and Keap-1) was also assessed. Exposure to iAs at 24 h (0-20 µM), stimulated ODD only at levels above the LC50 of a 48 h exposure (17 µM). If iAs induced ODD, it also activated oxidative stress-related genes. Selenium did not alter iAs-induced ODD. MMA(3+) at 24 h (0-0.5 µM) caused ODD at levels below the LC50 of a 48 h exposure (1.5 µM), which were greatly increased by GSH depletion but not selenite. MMA(3+) induced ODD at levels not activating oxidant stress response genes. Overall, iAs induced ODD in BMT-deficient cells only at toxic levels. MMA(3+) caused ODD at non-toxic levels, independently of cellular BMT capacity and in a fashion not requiring further BMT.


Assuntos
Arsenitos/toxicidade , Dano ao DNA , Células Epiteliais/efeitos dos fármacos , Compostos Organometálicos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Compostos de Sódio/toxicidade , Poluentes Químicos da Água/toxicidade , Arsenitos/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica , Glutationa/metabolismo , Humanos , Metilação , Compostos Organometálicos/metabolismo , Compostos de Sódio/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Poluentes Químicos da Água/metabolismo
3.
Toxicol Appl Pharmacol ; 255(3): 242-50, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21820459

RESUMO

Inorganic arsenic and UV, both human skin carcinogens, may act together as skin co-carcinogens. We find human skin keratinocytes (HaCaT cells) are malignantly transformed by low-level arsenite (100nM, 30weeks; termed As-TM cells) and with transformation concurrently undergo full adaptation to arsenic toxicity involving reduced apoptosis and oxidative stress response to high arsenite concentrations. Oxidative DNA damage (ODD) is a possible mechanism in arsenic carcinogenesis and a hallmark of UV-induced skin cancer. In the current work, inorganic arsenite exposure (100nM) did not induce ODD during the 30weeks required for malignant transformation. Although acute UV-treatment (UVA, 25J/cm(2)) increased ODD in passage-matched control cells, once transformed by arsenic to As-TM cells, acute UV actually further increased ODD (>50%). Despite enhanced ODD, As-TM cells were resistant to UV-induced apoptosis. The response of apoptotic factors and oxidative stress genes was strongly mitigated in As-TM cells after UV exposure including increased Bcl2/Bax ratio and reduced Caspase-3, Nrf2, and Keap1 expression. Several Nrf2-related genes (HO-1, GCLs, SOD) showed diminished responses in As-TM cells after UV exposure consistent with reduced oxidant stress response. UV-exposed As-TM cells showed increased expression of cyclin D1 (proliferation gene) and decreased p16 (tumor suppressor). UV exposure enhanced the malignant phenotype of As-TM cells. Thus, the co-carcinogenicity between UV and arsenic in skin cancer might involve adaptation to chronic arsenic exposure generally mitigating the oxidative stress response, allowing apoptotic by-pass after UV and enhanced cell survival even in the face of increased UV-induced oxidative stress and increased ODD.


Assuntos
Arsenitos/toxicidade , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Queratinócitos/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Oxidantes/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Apoptose/efeitos da radiação , Arsenitos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Dano ao DNA/fisiologia , Humanos , Queratinócitos/metabolismo , Oxidantes/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Estresse Oxidativo/efeitos da radiação , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos
4.
J Natl Cancer Inst ; 101(24): 1670-81, 2009 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19933942

RESUMO

BACKGROUND: Inorganic arsenic is an environmental carcinogen that may act through multiple mechanisms including formation of methylated derivatives in vivo. Sodium arsenite (up to 5.0 microM) renders arsenic methylation-competent TRL1215 rat liver epithelial cells tumorigenic in nude mice at 18 weeks of exposure and arsenic methylation-deficient RWPE-1 human prostate epithelial cells tumorigenic at 30 weeks of exposure. We assessed the role of arsenic biomethylation in oxidative DNA damage (ODD) using a recently developed immuno-spin trapping method. METHODS: Immuno-spin trapping was used to measure ODD after chronic exposure of cultured TRL1215 vs RWPE-1 cells, or of methylation-competent UROtsa/F35 vs methylation-deficient UROtsa human urothelial cells, to sodium arsenite. Secreted matrix metalloproteinase (MMP)-2 and -9 activity, as analyzed by zymography, cellular invasiveness by using a transwell assay, and colony formation by using soft agar assay were compared in cells exposed to arsenite with and without selenite, an arsenic biomethylation inhibitor, to assess the role of ODD in the transition to an in vitro cancer phenotype. RESULTS: Exposure of methylation-competent TRL1215 cells to up to 1.0 microM sodium arsenite was followed by a substantial increase in ODD at 5-18 weeks (eg, at 16 weeks with 1.0 microM arsenite, 1138% of control, 95% confidence interval [CI] = 797% to 1481%), whereas exposure of methylation-deficient RWPE-1 cells to up to 5.0 microM arsenite did not increase ODD for a 30-week period. Inhibition of arsenic biomethylation with sodium selenite abolished arsenic-induced ODD and invasiveness, colony formation, and MMP-2 and -9 hypersecretion in TRL1215 cells. Arsenic induced ODD in methylation-competent UROtsa/F35 cells (eg, at 16 weeks, with 1.0 microM arsenite 225% of control, 95% CI = 188% to 262%) but not in arsenic methylation-deficient UROtsa cells, and ODD levels corresponded to the levels of increased invasiveness, colony formation, and hypersecretion of active MMP-2 and -9 seen after transformation to an in vitro cancer phenotype. CONCLUSION: Arsenic biomethylation appears to be obligatory for arsenic-induced ODD and appears linked in some cells with the accelerated transition to an in vitro cancer phenotype.


Assuntos
Arsenitos/efeitos adversos , Carcinógenos/toxicidade , Dano ao DNA/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Inibidores Enzimáticos/efeitos adversos , Metiltransferases/genética , Neoplasias Experimentais/genética , Estresse Oxidativo/efeitos dos fármacos , Polimorfismo Genético/efeitos dos fármacos , Compostos de Sódio/efeitos adversos , Animais , Arsênio/efeitos adversos , Células Cultivadas , Células Epiteliais , Humanos , Fígado/citologia , Masculino , Inibidores de Metaloproteinases de Matriz , Metiltransferases/efeitos dos fármacos , Neoplasias Experimentais/induzido quimicamente , Fenótipo , Próstata/citologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Selenito de Sódio/farmacologia , Detecção de Spin , Fatores de Tempo , Bexiga Urinária/citologia
5.
Toxicon ; 48(8): 1002-10, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16973201

RESUMO

Plancitoxin I, the major lethal factor from the crown-of-thorns starfish Acanthaster planci venom, is quite unique not only in exhibiting potent hepatotoxicity but also in sharing high sequence homology with mammalian deoxyribonulease II. In this study, morphological and biochemical changes in rat liver epithelial cells (TRL 1215 cells) treated with the toxin were examined to understand the mechanism by which plancitoxin I displays hepatotoxicity. AlamarBlue assay established that plancitoxin I is cytolethal to TRL 1215 cells. This cytolethalithy was ascribable to apoptotic cell death. Nuclear fragmentation evidenced by either Diff-Quick or Hoechst 33258 staining, DNA fragmentation by TUNEL assay and electrophoretic analysis on agarose gel and phosphatidylserine externalization by flow cytometric analysis of annexin V-FITC stained cells were all characteristics of apoptosis. The observed apoptosis was shown to be independent of the caspase 3 cascade that is generally accepted as the effector of the apoptotic process. Very interestingly, experiments using FITC-labeled plancitoxin I proved that the toxin can enter the nucleus of TRL 1215 cells. Our results suggested that plancitoxin I induces apoptosis of TRL 1215 cells through the following procedure: binding to a specific receptor in the cytoplasmic membrane, entering the cell, entering the nucleus and degrading DNA.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Fígado/efeitos dos fármacos , Toxinas Marinhas/toxicidade , Estrelas-do-Mar/química , Peçonhas/toxicidade , Animais , Núcleo Celular/química , Núcleo Celular/metabolismo , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Fígado/citologia , Toxinas Marinhas/química , Toxinas Marinhas/metabolismo , Transporte Proteico , Ratos , Peçonhas/química
6.
Int Immunopharmacol ; 6(2): 304-15, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16459422

RESUMO

A trivalent inorganic arsenic, arsenite, has been causing chronic inflammation in humans through the consumption of contaminated well water. The total peripheral blood arsenic concentrations of chronic arsenic-exposed patients, who had inflammatory-like immune responses, are less than 1 microM, thus, nM concentrations may be very important regarding the chronic inflammatory effects by arsenite. However, there are few reports about the biological effects of low concentrations of arsenite in mammalian cells, especially in normal immune effector cells. In this study, we examined whether arsenite has any biological and/or toxicological effects on the differentiation of human peripheral blood monocytes into macrophages using the colony-stimulating factor (CSF) in vitro compared with that of other metallic compounds, and found that arsenite sensitively inhibited the CSF-induced in vitro maturation of monocytes into macrophages at nM levels, and it also induced small, nonadhesive and CD14-positive abnormal macrophage generation from monocytes with granulocyte-macrophage CSF (GM-CSF) at 50-500 nM without cell death. The addition of other metallic compounds, including chromium, selenium, mercury, cadmium, nickel, copper, zinc, cobalt, manganese and other human pentavalent arsenic metabolites, such as inorganic arsenate, monomethylarsonic acid and dimethylarsinic acid, could not induce the same abnormal cell generation from monocytes with CSFs at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at n-mM levels accompanied by cell death. This work may have implications in the arsenic-induced chronic inflammation in humans.


Assuntos
Arsenitos/toxicidade , Imunidade Celular/efeitos dos fármacos , Imunotoxinas , Macrófagos/imunologia , Monócitos/imunologia , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/biossíntese , Poluentes Ambientais/toxicidade , Glutationa/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células HL-60 , Humanos , Interleucina-12/metabolismo , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
7.
Toxicol Sci ; 91(1): 70-81, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16436460

RESUMO

Although inorganic arsenicals are toxic and carcinogenic in humans, inorganic arsenite has recently emerged as a highly effective chemotherapeutic agent for acute promyelocytic leukemia (APL). Inorganic arsenicals are enzymatically methylated to monomethylarsonic acid (MMAs(V)), dimethylarsinic acid (DMAs(V)), and trimethylarsine oxide (TMAs(V)O) in mammals. We examined the effects of chronic exposure to methylated arsenicals on arsenic tolerance by using rat normal liver TRL 1215 cells. TRL 1215 cells were exposed for 20 weeks to MMAs(V), DMAs(V), or TMAs(V)O at levels that produced submicromolar cellular concentrations of arsenic. On chronic exposure to these methylated arsenicals, the cells acquired tolerance to acute arsenic cytolethality. Cellular arsenic uptake was reduced in these cells compared to passage-matched control cells. The long-term arsenic exposure increased glutathione S-transferase (GST) activity and cellular glutathione (GSH) levels. Glutathione S-transferase, multidrug resistance-associated proteins (Mrps; efflux transporters encoded by Mrp genes), and P-glycoprotein [P-gp; efflux transporter encoded by multidrug resistance gene (MDR)] had also increased in these cells at the transcript and protein levels. The depletion of cellular GSH and the inhibition of Mrps and P-gp functions increased cellular arsenic uptake and reduced arsenic tolerance in these cells. These results indicate that chronic exposure to methylated arsenicals induces a generalized arsenic tolerance that is caused by increased arsenic excretion. Because accumulation of methylated arsenicals may occur in patients with chronic arsenic poisoning and arsenic-treated APL patients, this study may provide important information regarding chronic arsenic poisoning and the latent risk of developing multidrug resistance in APL therapy using inorganic arsenite.


Assuntos
Arsênio/toxicidade , Fígado/efeitos dos fármacos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Arsênio/administração & dosagem , Arsênio/farmacocinética , Glutationa Transferase/metabolismo , Leucemia Promielocítica Aguda/patologia , Fígado/citologia , Fígado/enzimologia , Fígado/metabolismo , Metilação , Ratos , Ratos Endogâmicos F344
8.
Biol Pharm Bull ; 28(10): 1827-32, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16204930

RESUMO

Arsenicals are known to be toxic and carcinogenic in humans. Inorganic arsenicals are enzymatically methylated to monomethylarsonic acid (MMAsV) and dimethylarsinic acid (DMAsV), which are the major pentavalent methyl arsenic metabolites. Recent reports indicate that trivalent methyl arsenicals are produced through methylation of inorganic arsenicals and participate in arsenic poisoning. Trivalent methyl arsenicals may be generated as arsenical-glutathione conjugates, such as monomethylarsonous diglutathione (MMAsIIIDG) and dimethylarsinous glutathione (DMAsIIIG), during the methylation process. It has been well known that reduced glutathione (GSH) reduces MMAsV and DMAsV in vitro, and produces MMAsIIIDG and DMAsIIIG. Some studies have shown that exogenous GSH increased cytolethality of MMAsV and DMAsV in vitro, while other studies have suggested that exogenous GSH decreased them. In this study, we examined the true effects of exogenous GSH on the cytolethality of MMAsV and DMAsV by investigating reactions between various concentrations of MMAsV or DMAsV and GSH. GSH significantly increased the cytolethality and cellular uptake of pentavalent methyl arsenicals when GSH over 25 mM was pre-incubated with mM levels of arsenicals, and this cytolethality might have been caused by arsenical-GSH conjugate generation. However, GSH at less than 25 mM did not affect the cytolethality and cellular uptake of pentavalent methyl arsenicals. These findings suggest that high concentrations of arsenicals and GSH are needed to form arsenical-GSH conjugates and to show significant cytolethality. Furthermore, we speculated that MMAsIIIDG and DMAsIIIG may separate into trivalent methyl arsenicals and glutathione, which are then transported into cells where they show significant cytolethality.


Assuntos
Arsenicais/química , Glutationa/toxicidade , Animais , Arsenicais/farmacocinética , Linhagem Celular , Cromatografia em Camada Fina , Glutationa/química , Glutationa/farmacocinética , Ratos
9.
Toxicol Appl Pharmacol ; 206(1): 54-65, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15963344

RESUMO

Human pentavalent arsenic metabolic intermediate, monomethylarsonic acid (MMAs(V)), is a major arsenic type found in the blood in chronic arsenic poisoning patients, but little information is available on its toxicity potential or mechanisms of action. In this study, we investigated the molecular mechanisms of in vitro cytolethality of MMAs(V) using rat liver TRL 1215 cells. Cellular arsenic concentrations reached the nanomolar range in TRL 1215 cells when cells were exposed to millimolar levels of MMAs(V), and most of the MMAs(V) was not metabolized during the 48-h incubation. Under these conditions, MMAs(V) showed significant cytolethality when cellular reserves of reduced glutathione (GSH) were depleted. Morphological and biochemical evidence confirmed that MMAs(V) induced both necrosis and apoptosis in the cellular GSH-depleted cells. MMAs(V) significantly enhanced cellular caspase 3 activity in the cellular GSH-depleted cells, and a caspase 3 inhibitor blocked MMAs(V)-induced apoptosis. MMAs(V) also enhanced the production of cellular reactive oxygen species (ROS) in the cellular GSH-depleted cells, and addition of a membrane-permeable radical trapping reagent completely prevented both MMAs(V)-induced cellular caspase 3 activation and cytolethality in these cells. These observations suggest that MMAs(V) typically generates harmful ROS in cells, and cellular GSH prevents cytolethality by scavenging these toxic ROS. However, when cellular GSH levels are decreased, MMAs(V) induces oxidative stress in the cells, and this leads to apoptosis and/or necrosis depending on the cellular ROS/GSH ratio.


Assuntos
Arsenicais/efeitos adversos , Glutationa/uso terapêutico , Herbicidas/toxicidade , Fígado/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Arsenicais/antagonistas & inibidores , Células Cultivadas , Herbicidas/antagonistas & inibidores , Marcação In Situ das Extremidades Cortadas , Fígado/metabolismo , Fígado/patologia , Ratos , Ratos Endogâmicos F344
10.
Int Immunopharmacol ; 4(13): 1661-73, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15454118

RESUMO

A trivalent inorganic arsenic, arsenite, has been causing chronic inflammation in humans through the consumption of contaminated well water. The total peripheral blood arsenic concentrations of chronic arsenic-exposed patients, who had inflammatory-like immune responses, are less than 1 microM, thus, nM concentrations may be very important regarding the chronic inflammatory effects by arsenite. However, there are few reports about the biological effects of low concentrations of arsenite in mammalian cells, especially in normal immune effector cells. In this study, we examined whether arsenite has any biological and/or toxicological effects on the differentiation of human peripheral blood monocytes into macrophages using the colony-stimulating factor (CSF) in vitro compared with that of other metallic compounds, and found that arsenite sensitively inhibited the CSF-induced in vitro maturation of monocytes into macrophages at nM levels, and it also induced small, nonadhesive and CD14-positive abnormal macrophage generation from monocytes with granulocyte-macrophage CSF (GM-CSF) at 50-500 nM without cell death. The addition of other metallic compounds, including chromium, selenium, mercury, cadmium, nickel, copper, zinc, cobalt, manganese and other human pentavalent arsenic metabolites, such as inorganic arsenate, monomethylarsonic acid and dimethylarsinic acid, could not induce the same abnormal cell generation from monocytes with CSFs at any concentration and any additional time schedules; they showed only simple cytolethality in monocytes and macrophages at n-mM levels accompanied by cell death. This work may have implications in the arsenic-induced chronic inflammation in humans.


Assuntos
Arsenitos/efeitos adversos , Arsenitos/imunologia , Imunotoxinas/efeitos adversos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Compostos de Sódio/efeitos adversos , Compostos de Sódio/imunologia , Arseniatos/efeitos adversos , Arseniatos/imunologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Poluentes Ambientais/efeitos adversos , Poluentes Ambientais/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células HL-60 , Humanos , Imunotoxinas/química , Imunotoxinas/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Fator Estimulador de Colônias de Macrófagos/imunologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/patologia , Macrófagos/fisiologia , Monócitos/patologia , Monócitos/fisiologia , Testes de Toxicidade/métodos
11.
Toxicol Appl Pharmacol ; 198(3): 354-65, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15276415

RESUMO

Inorganic arsenicals are clearly toxicants and carcinogens in humans. In mammals, including humans, inorganic arsenicals often undergo methylation, forming compounds such as dimethylarsinic acid (DMAs(V)). Recent evidence indicates that DMAs(V) is a complete carcinogen in rodents although evidence for inorganic arsenicals as carcinogens in rodents remains equivocal. Thus, we studied the molecular mechanisms of in vitro cytolethality of DMAs(V) using a rat liver epithelial cell line (TRL 1215). DMAs(V) selectively induced apoptosis in TRL 1215 cells; its LC(50) value after 48 h exposure was 4.5 mM. The addition of a glutathione synthase inhibitor, L-buthionine-[S,R]-sulfoximine (BSO), actually decreased DMAs(V)-induced apoptosis. DMAs(V) exposure temporarily decreased cellular reduced glutathione (GSH) levels and enhanced cellular glutathione S-transferase (GST) activity from 6 h after the exposure when the cells were still alive. Also, DMAs(V) exposure activated cellular caspase 3 activity with a peak at 18 h after the exposure when apoptosis began, and BSO treatment completely inhibited this enzyme activity. The additions of inhibitors of caspase 3, caspase 8, and caspase 9 significantly reduced DMAs(V)-induced apoptosis. Taken together, these data indicate that cellular GSH was required for DMAs(V)-induced apoptosis to occur, and activation of cellular caspases after conjugation of DMAs(V) with cellular GSH appears to be of mechanistic significance. Further research will be required to determine the role of intracellular GSH and methylation in the toxicity of arsenicals in chronic arsenic poisoning or in cases where arsenicals are used as chemotherapeutics.


Assuntos
Apoptose/efeitos dos fármacos , Butionina Sulfoximina/farmacologia , Ácido Cacodílico/toxicidade , Inibidores Enzimáticos/farmacologia , Herbicidas/toxicidade , Fígado/efeitos dos fármacos , Animais , Ácido Cacodílico/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular , Glutationa Transferase/metabolismo , Herbicidas/metabolismo , Humanos , Fígado/enzimologia , Fígado/metabolismo , Ratos , Ratos Endogâmicos F344
12.
Int Immunopharmacol ; 4(2): 179-84, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14996409

RESUMO

In this study, we observed the in vivo acute immunotoxicity of a trimethyl arsenic compound, arsenobetaine (AsBe), which is present in large quantities in various marine animals that are daily ingested as seafood in many countries. The synthetic pure AsBe was orally administered to CDF(1) mice at a dose of 1.625 g/kg mouse weight once a day on days -6, -4, -2 and 0 (four times, total 6.5 g/kg mouse weight), and its effect on the immune organs and immune effector cells were assessed until day 8. Orally administered AsBe was temporally distributed to the immune organs, such as the spleen and thymus, but was not very toxic both quantitatively and qualitatively on these immune organs and immune effector cells, splenocytes, thymocytes, Peyer's patch lymphocytes and peritoneal macrophages. This finding suggests that the ingestion of AsBe contained in marine animals is relatively safe to the health of people who often consume marine animals in their daily diet.


Assuntos
Arsênio/toxicidade , Arsenicais , Nódulos Linfáticos Agregados/efeitos dos fármacos , Alimentos Marinhos , Baço/efeitos dos fármacos , Timo/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Animais , Arsenicais/farmacocinética , Contaminação de Alimentos , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Nível de Efeito Adverso não Observado , Nódulos Linfáticos Agregados/citologia , Medição de Risco , Baço/citologia , Timo/citologia , Distribuição Tecidual , Poluentes Químicos da Água/farmacocinética
13.
Toxicol Appl Pharmacol ; 195(2): 129-41, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14998680

RESUMO

Inorganic arsenicals are clearly toxicants and carcinogens in humans. In mammals, including humans, inorganic arsenic often undergoes methylation, forming compounds such as monomethylarsonic acid (MMAs(V)) and dimethylarsinic acid (DMAs(V)). However, much less information is available on the in vitro toxic potential or mechanisms of these methylated arsenicals, especially MMAs(V). We studied the molecular mechanisms of in vitro cytolethality of MMAs(V) using a rat liver epithelial cell line (TRL 1215). MMAs(V) was not cytotoxic in TRL 1215 cells even at concentrations exceeding 10 mM, but it became weakly cytotoxic and induced both necrotic and apoptotic cell death when cellular reduced glutathione (GSH) was depleted with the glutathione synthase inhibitor, l-buthionine-[S,R]-sulfoximine (BSO), or the glutathione reductase inhibitor, carmustine. Similar results were observed in the other mammalian cells, such as human skin TIG-112 cells, chimpanzee skin CRT-1609 cells, and mouse metallothionein (MT) positive and MT negative embryonic cells. Ethacrynic acid (EA), an inhibitor of glutathione S-transferase (GST) that catalyses GSH-substrate conjugation, also enhanced the cytolethality of MMAs(V), but aminooxyacetic acid (AOAA), an inhibitor of beta-lyase that catalyses the final breakdown of GSH-substrate conjugates, had no effect. Both the cellular GSH levels and the cellular GST activity were increased by the exposure to MMAs(V) in TRL 1215 cells. On the other hand, the addition of exogenous extracellular GSH enhanced the cytolethality of MMAs(V), although cellular GSH levels actually prevented the cytolethality of combined MMAs(V) and exogenous GSH. These findings indicate that human arsenic metabolite MMAs(V) is not a highly toxic compound in mammalian cells, and the level of cellular GSH is critical to its eventual toxic effects.


Assuntos
Arsenicais/farmacologia , Glutationa/metabolismo , Ácido Amino-Oxiacético/farmacologia , Animais , Carmustina/farmacologia , Catálise , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ácido Etacrínico/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Glutationa/antagonistas & inibidores , Glutationa/farmacologia , Glutationa Transferase/antagonistas & inibidores , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pan troglodytes , Ratos , Ratos Endogâmicos F344 , Pele/citologia , Pele/efeitos dos fármacos , Pele/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...