Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arch Toxicol ; 82(8): 493-512, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18496671

RESUMO

Mechanisms of carcinogenicity are discussed for metals and their compounds, classified as carcinogenic to humans or considered to be carcinogenic to humans: arsenic, antimony, beryllium, cadmium, chromium, cobalt, lead, nickel and vanadium. Physicochemical properties govern uptake, intracellular distribution and binding of metal compounds. Interactions with proteins (e.g., with zinc finger structures) appear to be more relevant for metal carcinogenicity than binding to DNA. In general, metal genotoxicity is caused by indirect mechanisms. In spite of diverse physicochemical properties of metal compounds, three predominant mechanisms emerge: (1) interference with cellular redox regulation and induction of oxidative stress, which may cause oxidative DNA damage or trigger signaling cascades leading to stimulation of cell growth; (2) inhibition of major DNA repair systems resulting in genomic instability and accumulation of critical mutations; (3) deregulation of cell proliferation by induction of signaling pathways or inactivation of growth controls such as tumor suppressor genes. In addition, specific metal compounds exhibit unique mechanisms such as interruption of cell-cell adhesion by cadmium, direct DNA binding of trivalent chromium, and interaction of vanadate with phosphate binding sites of protein phosphatases.


Assuntos
Arsenicais/efeitos adversos , Carcinógenos/toxicidade , Metais Pesados/toxicidade , Metais Leves/toxicidade , Animais , Arsenicais/química , Arsenicais/farmacocinética , Disponibilidade Biológica , Carcinógenos/química , Carcinógenos/farmacocinética , DNA/efeitos dos fármacos , Dano ao DNA , Humanos , Metais Pesados/química , Metais Pesados/farmacocinética , Metais Leves/química , Metais Leves/farmacocinética , Mutagênicos/toxicidade
2.
Biol Trace Elem Res ; 108(1-3): 87-104, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16327063

RESUMO

Zinc homeostasis in mammalian cells is precisely regulated by cellular signal transduction mechanisms. The main result of this study is the finding that modulators of phospholipase C (PLC) activity affect cellular zinc export. Two different PLC inhibitors caused an increase of the total cellular zinc level whereas two different PLC activators caused a decrease. Furthermore, both the inhibition of cyclic nucleotide phosphodiesterases as well as the administration of 8-bromo-cAMP evoked a drop in the intracellular zinc level, indicating the involvement of cAMP in the control of cellular zinc export. It is concluded that the activity of PLC controls cellular zinc transport and that the effect of elevated zinc concentrations on PLC activity might be mediated by cAMP. However, modulation of other major signaling enzymes did not affect the cellular zinc homeostasis. These include activation and inhibition of guanylate cyclase, activation of protein kinase G, activation of protein kinase A, and activation or inhibition of protein kinase C. Furthermore there was no evidence for the existence of a zinc-sensing receptor in C6 glioma cells, which would stimulate PLC activity and evoke a mobilization of intracellular free-calcium levels.


Assuntos
Glioma/metabolismo , Fosfolipases Tipo C/metabolismo , Zinco/metabolismo , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Cálcio/metabolismo , Células Cultivadas , AMP Cíclico/química , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Citosol/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Homeostase , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Proteína Quinase C/metabolismo , Ratos , Transdução de Sinais , Células Tumorais Cultivadas , Zinco/farmacologia
3.
Anal Biochem ; 332(1): 145-52, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15301959

RESUMO

The intracellular homeostasis of zinc is postulated to be controlled by signaling through nitric oxide (NO). Administration of the NO donor S-nitrosocysteine (SNOC) caused a rapid drop in the fluorescence of the zinc-specific fluorescence of the zinc probe zinquin in C6 glioma cells. Tentatively, a strong effect of NO on the level of mobile intracellular zinc ions was concluded. However, zinc analysis with atomic absorption spectrometry demonstrated that the total cellular zinc level was not changed under these conditions. Sodium nitrite or an NO donor devoid of sulfhydryl groups (diethylamine NONOate) exerted no degrading effect on the Zn/zinquin fluorescence, but cysteine alone evoked a similar decline as SNOC. Hence, the sulfhydryl groups of cysteine seem to compete for zinc from the Zn/zinquin complex. Analysis of the reaction products by mass spectrometry demonstrated that cysteine caused a depletion of zinc from the Zn/zinquin complex, whereas an NO donor without sulfhydryl groups (diethylamine NONOate) did not. It is concluded that great caution should be employed when using S-nitroso compounds together with zinquin in investigations of intracellular zinc homeostasis.


Assuntos
Cisteína/análogos & derivados , Cisteína/metabolismo , Sondas Moleculares , Quinolonas/metabolismo , S-Nitrosotióis/metabolismo , Compostos de Tosil/metabolismo , Zinco/metabolismo , Animais , Fluorescência , Óxido Nítrico/metabolismo , Ratos , Espectrometria de Fluorescência , Espectrometria de Massas por Ionização por Electrospray , Fatores de Tempo , Zinco/análise
5.
Biometals ; 17(1): 65-78, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14977363

RESUMO

Cadmium has recently been shown to induce apoptosis in C6 glioma cells via disruption of the mitochondrial membrane potential and subsequent caspase 9-activation. Here we show that both H2O2 and CdCl2 induced apoptotic DNA fragmentation in C6 cells. The employment of glutathione as an antioxidant prevented the induction of apoptotic DNA fragmentation by cadmium completely and catalase strongly reduced cadmium-induced DNA fragmentation suggesting that cadmium exerts its apoptotic effects at least partly via the production of H2O2. Apoptosis may be induced by cadmium indirectly through formation of oxidative stress, e.g., by inhibition of antioxidant enzymes. After incubation of C6 cells with cadmium for short times (up to 4 h), we analyzed the formation of intracellular reactive oxygen species and cellular lipid peroxidation. After 1 h of incubation with inreasing concentrations of CdCl2 (1-500 microM), no increase in dichlorofluorescein fluorescence was found. At variance, lipid peroxidation was slightly elevated after 2 h incubation with cadmium (50-100 microM). Furthermore, we analyzed the modulation of markers for oxidative stress after prolonged (24 h) exposure to cadmium. The intracellular glutathione content as measured using the fluorescent probe monobromobimane was decreased after incubation with CdCl2 (0.5-10 microM) for 24 h. Furthermore, we measured the effect of cadmium on the level of oxidized DNA lesions (predominantly 8-hydroxyguanine) using the bacterial Fpg-DNA-repair protein. After 24 h of incubation with 5 microM CdCl2 we found a sixfold increase in Fpg-sensitive DNA-lesions. We conclude that short time incubations with cadmium (up to 4 h) caused only slight or insignificant effects on the generation of reactive oxygen species (formation of thiobarbituric acid reactive substances, fluorescence of dichlorofluorescein), whereas incubation with this heavy metal for 24 h lead to a decrease in intracellular glutathione concentration and an increase in oxidative DNA-lesions. Our data demonstrate that cadmium as similar to H2O2 is a potent inducer of apoptosis in C6 cells. Even if cadmium unlike Fenton-type metals can not produce reactive oxygen species directly, the apoptotic effects of cadmium at least in part are mediated via induction of oxidative stress. Because both apoptosis and oxidative stress are thought to play important roles in neurodegenerative diseases, low concentrations of cadmium that initiate programmed cell death may lead to a selective cell death in distinct brain regions via generation of oxidative stress.


Assuntos
Apoptose/efeitos dos fármacos , Cádmio/farmacologia , Glioma/patologia , Estresse Oxidativo , Animais , Antioxidantes/farmacologia , Cloreto de Cádmio/farmacologia , Catalase/metabolismo , Linhagem Celular Tumoral , DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , Glioma/tratamento farmacológico , Glioma/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Concentração Inibidora 50 , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Oxirredução/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Enxofre/metabolismo
6.
Toxicology ; 192(2-3): 95-117, 2003 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-14580780

RESUMO

Cadmium is a heavy metal, which is widely used in industry, affecting human health through occupational and environmental exposure. In mammals, it exerts multiple toxic effects and has been classified as a human carcinogen by the International Agency for Research on Cancer. Cadmium affects cell proliferation, differentiation, apoptosis and other cellular activities. Cd2+ does not catalyze Fenton-type reactions because it does not accept or donate electrons under physiological conditions, and it is only weakly genotoxic. Hence, indirect mechanisms are implicated in the carcinogenicity of cadmium. In this review multiple mechanisms are discussed, such as modulation of gene expression and signal transduction, interference with enzymes of the cellular antioxidant system and generation of reactive oxygen species (ROS), inhibition of DNA repair and DNA methylation, role in apoptosis and disruption of E-cadherin-mediated cell-cell adhesion. Cadmium affects both gene transcription and translation. The major mechanisms of gene induction by cadmium known so far are modulation of cellular signal transduction pathways by enhancement of protein phosphorylation and activation of transcription and translation factors. Cadmium interferes with antioxidant defense mechanisms and stimulates the production of reactive oxygen species, which may act as signaling molecules in the induction of gene expression and apoptosis. The inhibition of DNA repair processes by cadmium represents a mechanism by which cadmium enhances the genotoxicity of other agents and may contribute to the tumor initiation by this metal. The disruption of E-cadherin-mediated cell-cell adhesion by cadmium probably further stimulates the development of tumors. It becomes clear that there exist multiple mechanisms which contribute to the carcinogenicity of cadmium, although the relative weights of these contributions are difficult to estimate.


Assuntos
Cádmio/toxicidade , Carcinógenos Ambientais/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Neoplasias/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Humanos , Ativação Transcricional
7.
Biol Chem ; 384(1): 161-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12674510

RESUMO

The transcription factor E2F1 plays a decisive role in the G1/S and G2/M checkpoint transitions of proliferating cells. Because cells are arrested at these checkpoints after heat shock it was of interest to test heat shock effects on E2F1 activity. In human A549 cells, heat shock (44 degrees C, 30 min) caused an immediate reduction of E2F1-DNA binding as determined by electrophoretic mobility shift assay (EMSA). The complex of E2F1-DNA with the retinoblastoma protein (pRB) was also reduced after heat shock. This indicates that the former effect is not caused by a lower phosphorylation and therefore a higher binding capacity of pRB. Western blot analyses showed that the lower E2F1-DNA binding is probably due to a decrease of the E2F1 level (40% of the controls) induced by heat shock. This result was confirmed by an experiment with HeLa cells in which heat shock decreased the level to 60% of the controls. In order to test whether this decrease resulted from inhibition of transcription, RT-PCR measurements were conducted and showed only a slight reduction of the E2F1 mRNA (89% of controls). This indicates that the heat shock effect is not predominantly caused by transcriptional inhibition. Six hours after heat shock the E2F1-DNA binding capacity recovered to control levels. These results provide evidence for E2F1 involvement in heat shock-induced cell cycle arrests at the G1/S and G2/M checkpoints, which also may be relevant for hyperthermic cancer therapy.


Assuntos
Proteínas de Transporte/metabolismo , DNA/metabolismo , Temperatura Alta , Proteínas Adaptadoras de Transdução de Sinal , Western Blotting , Ciclo Celular/fisiologia , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , DNA/análise , Sondas de DNA , DNA Complementar/biossíntese , Eletroforese em Gel de Ágar , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Indicadores e Reagentes , Cinética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/isolamento & purificação , Ligação Proteica , RNA/biossíntese , RNA/isolamento & purificação , Proteínas de Ligação a RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Biol Trace Elem Res ; 96(1-3): 133-42, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14716092

RESUMO

Zinc ions are essential, but at elevated concentrations, they also have toxic effects on mammalian cells. Zinc plays a crucial role in cell proliferation and differentiation and it even protects cells against apoptosis caused by various reagents. On the other hand, zinc at high concentrations causes cell death that was characterized as apoptotic by internucleosomal DNA fragmentation, formation of apoptotic bodies, and breakdown of the mitochondrial membrane potential. In the present work, a clone of rat C6 glioma cells that was resistant to toxic effects of ZnCl2 up to 250 microM was employed to study the effect of the ionophore A23187 on zinc-induced apoptosis. Neither 150 microM Zn2+ nor 100 nM A23187 alone caused apoptosis as measured by internucleosomal DNA fragmentation. However, combined exposure of C6 cells to 100 nM A23187 and 150 microM Zn2+ for 48 h was effective in inducing apoptosis. Because the so-called calcium ionophore A23187 is not specific for Ca2+ ions but also transports Zn2+ with high selectivity over Ca2+, we investigated whether this substance promoted the uptake of Zn2+ ions into C6 cells. Employing the zinc-specific fluorescence probe Zinquin, we observed that the very low concentration of 1.9 nM A23187 significantly and rapidly raised the intracellular mobile Zn2+ content. Analysis by atomic absorption spectroscopy revealed that incubation with 1.9 nM A23187 caused a doubling of the total intracellular zinc level within 60 min. We conclude that the apoptosis evoked by the combined action of Zn2+ and A23187 was the result of enhanced Zn2+ influx evoked by the ionophore, resulting in higher intracellular zinc levels.


Assuntos
Apoptose/efeitos dos fármacos , Calcimicina/farmacologia , Cálcio/antagonistas & inibidores , Glioma/patologia , Ionóforos/farmacologia , Zinco/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Líquido Extracelular , Glioma/metabolismo , Peróxido de Hidrogênio/farmacologia , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Microscopia de Fluorescência , Ratos , Espectrofotometria Atômica
9.
Environ Health Perspect ; 110 Suppl 5: 865-7, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12426148

RESUMO

In various mammalian cells, two group IIb metals, cadmium and zinc, induce several morphological and biochemical effects that are salient features of programmed cell death. In C6 rat glioma cells, cadmium caused externalization of phosphatidylserine, breakdown of the mitochondrial membrane potential, activation of caspase-9, internucleosomal DNA fragmentation, chromatin condensation, and nuclear fragmentation. In NIH3T3 murine fibroblasts, cadmium-induced apoptosis was inhibited by overexpression of the antiapoptotic protein Bcl-2. Cadmium-induced DNA fragmentation in C6 cells was independent of inhibition of protein kinase A (PKA), protein kinase C (PKC), mitogen-activated protein kinase (MAPK), phosphatidylinositol-3-kinase, Ca-calmodulin-dependent protein kinase, and protein kinase G. Zinc at moderate concentrations (10-50 microM) protected against programmed cell death induced by cadmium, whereas deprivation of zinc by the membrane-permeable chelator N,N,N',N-terakis-(2-pyridylmethyl)ethylenediamine (TPEN) caused cell death with features characteristic of apoptosis. On the other hand, at elevated extracellular levels (150-200 microM), zinc alone caused programmed cell death in C6 cells. Zinc-induced apoptosis was independent of inhibition of PKA, PKC, guanylate cyclase and MAPK, but it was suppressed in the presence of 100 microM lanthanum chloride.


Assuntos
Apoptose/efeitos dos fármacos , Cádmio/efeitos adversos , Zinco/efeitos adversos , Animais , Cádmio/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/farmacologia , Fibroblastos , Glioma/patologia , Guanilato Ciclase/efeitos dos fármacos , Guanilato Ciclase/farmacologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/farmacologia , Ratos , Células Tumorais Cultivadas , Zinco/farmacologia
10.
Biochem Biophys Res Commun ; 296(4): 923-8, 2002 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-12200136

RESUMO

In mammalian cells, the intracellular availability of zinc influences numerous crucial processes. Its distribution has previously been visualized with several fluorescent probes, but it was unclear how these probes are compartmentalized within the cell. Here, we show that in C6 cells the zinc-specific probe Zinquin is evenly distributed. Thus, the significantly lower level of fluorescence in the nucleus and a punctuate vesicular staining are real differences in the concentrations of zinc. Chemical perturbation of the steady state by releasing intracellular protein-bound zinc with the sulfhydryl-reactive N-ethylmaleimide (NEM) resulted in a vanadate sensitive transport of zinc out of the nucleus and into zincosomes. If the zinc-release was performed with the histidine-reactive diethylpyrocarbonate, sequestration was reduced compared to treatment with NEM, indicating the importance of histidine within membrane zinc transporters. Another major factor regulating the zinc homeostasis is ion export. As determined by atomic absorption spectroscopy, up to 50% of the cellular zinc was exported by a mechanism sensitive to lanthanum ions. We conclude that different concentrations of labile zinc exist in different cellular compartments, which are maintained by export and intracellular transport of zinc.


Assuntos
Zinco/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Etilmaleimida/farmacologia , Corantes Fluorescentes/farmacologia , Histidina/metabolismo , Íons , Microscopia de Fluorescência , Transporte Proteico , Quinolonas/farmacologia , Ratos , Espectrofotometria Atômica , Fatores de Tempo , Compostos de Tosil/farmacologia , Células Tumorais Cultivadas , Vanadatos/farmacologia
11.
Toxicol Lett ; 127(1-3): 63-8, 2002 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-12052642

RESUMO

Six metals and/or their compounds have been recognized as carcinogens: arsenic, beryllium, cadmium, chromium, cobalt and nickel. With the exception of arsenic, the main rote of exposure is inhalation and the main target organ is the lung. Arsenic is exceptional because it also produces tumors of skin and lung after oral uptake. With the exception of hexavalent chromium, carcinogenic metals are weak mutagens, if at all, and their mechanisms of carcinogenicity are still far from clear. A general feature of arsenic, cadmium, cobalt and nickel is their property to enhance the mutagenicity and carcinogenicity of directly acting genotoxic agents. These properties can be interpreted in terms of the ability of these metals to inhibit the repair of damaged DNA. However, because carcinogenic metals cause tumor development in experimental animals even under exclusion of further carcinogens, other mechanisms have to be envisaged, too. Evidence will be discussed that carcinogenic metal compounds alter patterns of gene expression leading to stimulated cell proliferation, either by activation of early genes (proto-oncogenes) or by interference with genes downregulating cell growth. Special reference will be devoted to the effects of cadmium and arsenic on gene expression, which have been studied extensively. Possible implications for occupational safety and health will be discussed.


Assuntos
Carcinógenos/efeitos adversos , Dano ao DNA , Metais/efeitos adversos , Animais , Arsênio/efeitos adversos , Arsênio/toxicidade , Berílio/efeitos adversos , Berílio/toxicidade , Cádmio/efeitos adversos , Cádmio/toxicidade , Carcinógenos/toxicidade , Cromo/efeitos adversos , Cromo/toxicidade , Cobalto/efeitos adversos , Cobalto/toxicidade , DNA/efeitos dos fármacos , DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metais/toxicidade , Modelos Biológicos , Neoplasias/induzido quimicamente , Neoplasias/genética , Níquel/efeitos adversos , Níquel/toxicidade
12.
Biometals ; 15(1): 15-25, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11860019

RESUMO

The induction of apoptotic cell death by cadmium was investigated in eight mammalian cell lines. Great differences in the cytotoxicity of cadmium were found with different cell lines: Rat C6 glioma cells turned out to be most sensitive with an IC50-value of 0.7 microM, while human A549 adenocarcinoma cells were relatively resistant with an IC50-value of 164 microM CdCl2. The mode of cadmium-induced cellular death was identified to involve apoptotic DNA fragmentation in three cell lines, i.e., in C6 glioma cells, E367 neuroblastoma cells and NIH3T3 fibroblasts. In C6 glioma cells, this process was investigated in detail. Internucleosomal DNA-fragmentation occurred 40 h after application of CdCl2 and was concentration-dependent between 1-100 microM CdCl2, followed by a decrease at higher concentrations due to necrotic processes. Apoptotic chromatin-condensation and nuclear fragmentation was observed 48 h after application of 2.5 microM CdCl2. Furthermore, cadmium (1 microM, 48 h) caused a breakdown of the mitochondrial membrane potential as shown by the decline in mitochondrial uptake of rhodamine 123. Also, we found an activation of caspase 9, a protease known to be activated in apoptotic processes following mitochondrial damage. Besides Cd2+, other toxic heavy metal ions (Hg2+, Pb2+, Ni2+, Fe2+, CrO4(2-), Cu2+ or Co2+) did not induce apoptotic DNA fragmentation in C6 cells. The only exception was Zn2+ which caused apotosis at high concentrations (>150 microM) whereas it protected against cadmium-induced apoptosis at low concentrations (10-50 microM).


Assuntos
Apoptose/efeitos dos fármacos , Cádmio/toxicidade , Caspases/metabolismo , Células 3T3 , Animais , Caspase 9 , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Resistência a Medicamentos , Ativação Enzimática/efeitos dos fármacos , Glioma/metabolismo , Glioma/patologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Metais Pesados/toxicidade , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células PC12 , Ratos , Células Tumorais Cultivadas , Zinco/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...