Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-37711997

RESUMO

One of the challenges of teaching scientific courses is helping students understand research methods, biological models, and data analysis, which can be especially difficult in classes without a laboratory component. Within the field of toxicology, it is also important for students to understand how living organisms are affected by exposure to toxicants and how these toxicants can impact the ecosystem. Resources focusing on active learning pedagogy are scarce in the field of toxicology compared to other disciplines. In this activity, upper-level students in an introductory toxicology course learn to interpret data from primary literature, draw conclusions about how toxicants, specifically metals, can impact susceptible populations, and understand the One Environmental Health approach. Students work in small groups to answer questions concerning data from a paper and then share their responses with the entire class building their communication skills. The instructor serves as a moderator, allowing the students to work through concepts, intervening only when necessary. This approach enables a deeper level of understanding of content and allows the students to engage actively in the learning process. As such, students think critically through relevant problems and find connections to the real world. This lesson can be adapted for several levels of students and could be modified depending on the objectives of the course.

2.
Sci Total Environ ; 678: 761-767, 2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31085492

RESUMO

Cadmium (Cd) is a toxic heavy metal that accumulates in living systems. Exposure can occur occupationally or environmentally. Workers within the electroplating, battery production, and pigment industries are at the highest risk for exposure and have been reported to have increased levels of Cd in their blood and urine. Environmental exposure can be the result of anthropogenic activities or smoking. Cd has a long half-life and bio accumulates in plants, invertebrates, and vertebrates. The toxic effects following exposure include growth retardation and organ system toxicity, with kidney and liver toxicity most reported with in higher organisms. At the molecular level, Cd leads to the production of reactive oxygen species, DNA damage, and inhibition of DNA repair. This article gives a brief overview of the correlations between exposure to cadmium occupationally and environmentally and levels measured in blood and urine. It also examines the bioaccumulation of cadmium in aquatic invertebrates and vertebrates indicating that accumulation varies not only by location but also within and between various species.


Assuntos
Organismos Aquáticos/efeitos dos fármacos , Cádmio/efeitos adversos , Exposição Ambiental/efeitos adversos , Poluentes Ambientais/efeitos adversos , Mamíferos , Plantas/efeitos dos fármacos , Animais , Humanos , Exposição Ocupacional/efeitos adversos
3.
Mutat Res Genet Toxicol Environ Mutagen ; 816-817: 12-17, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28464991

RESUMO

Cadmium is a toxic heavy metal with many industrial and commercial uses resulting in widespread environmental pollution. Exposure to cadmium occurs occupationally, through tainted food, tobacco leaves, and the environment. Cadmium exposure has been linked to neurological and respiratory problems and cancer. Past studies have examined cytotoxicity, DNA damage, micronuclei formation, or mutagenesis individually, but no study has examined these endpoints together and correlated them with intracellular cadmium concentrations. This study examines the cytotoxic, genotoxic, and mutagenic effects of cadmium exposure in V79 cells providing a correlation between the induction of DNA damage, the generation of mutations, and intracellular cadmium concentration. Cells exposed to 2.5-40µM cadmium exhibited a concentration-dependent decrease in survival which correlated with increases in PARP cleavage and caspase activity. Cytotoxicity was detected at intracellular cadmium concentrations of 12.9ppb. Exposure to cadmium also resulted in the production of single and double strand breaks and the induction micronuclei and mutations in the Hprt gene. Collectively, this study demonstrates that exposure of V79 cells to cadmium results in the induction of apoptosis which is related to the formation of DNA strand breaks and genotoxic damage.


Assuntos
Cloreto de Cádmio/toxicidade , Dano ao DNA/efeitos dos fármacos , Animais , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Cricetinae , Intoxicação por Metais Pesados , Hipoxantina Fosforribosiltransferase/genética , Metais Pesados/toxicidade , Mutagênicos/toxicidade , Intoxicação
4.
Toxicol Lett ; 222(3): 265-72, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23948120

RESUMO

Environmental toxicants such as methylmercury play a critical role in the pathogenesis of many neurodevelopmental disorders. Environmental exposure to methylmercury frequently occurs at low doses, most frequently through fish consumption. Although the general population is at risk for exposure, pregnant women and young children are the most vulnerable. A common symptom of perinatal exposure to methylmercury is increased sensory (visual) deficits, motor impairment, and an overall cognitive decline. Research has indicated that the developing cerebellum, specifically the cerebellar granular layer, is particularly vulnerable to methylmercury neurotoxicity. This review examines the effects of low-level methylmercury exposure on motor coordination. We specifically focus on the role of cerebellar granule cells in methylmercury neurotoxicity. We suggest that methylmercury induces oxidative stress in cerebellar granule cells, which subsequently results in apoptotic cell death. Understanding the mechanism by which methylmercury induces toxicity within the developing brain will allow for enhanced treatments and potential reversal of the detrimental effects.


Assuntos
Cerebelo/efeitos dos fármacos , Desenvolvimento Infantil/efeitos dos fármacos , Compostos de Metilmercúrio/toxicidade , Movimento/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Cerebelo/citologia , Criança , Feminino , Humanos , Transtornos dos Movimentos/etiologia , Gravidez , Espécies Reativas de Oxigênio/metabolismo
5.
Mutagenesis ; 27(4): 437-43, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22241526

RESUMO

Cultured human cells are invaluable biological models for mechanistic studies of genotoxic chemicals and drugs. Continuing replacement of animals in toxicity testing will further increase the importance of in vitro cell systems, which should accurately reproduce key in vivo characteristics of toxicants such as their profiles of metabolites and DNA lesions. In this work, we examined how a common severe deficiency of cultured cells in ascorbate (Asc) impacts the formation of oxidative DNA damage by hexavalent chromium (chromate). Cr(VI) is reductively activated inside the cells by both Asc and small thiols but with different rates and spectra of intermediates and DNA adducts. We found that Cr(VI) exposure of H460 human lung epithelial cells in standard culture (<0.01 mM cellular Asc) induced biologically significant amounts of oxidative DNA damage. Inhibition of oxidative damage repair in these cells by stable XRCC1 knockdown strongly enhanced cytotoxic effects of Cr(VI) and led to depletion of cells from G(1) and accumulation in S and G(2) phases. However, restoration of physiological levels of Asc (≈ 1 mM) completely eliminated Cr(VI) hypersensitivity of XRCC1 knockdown. The induction of chromosomal breaks assayed by the micronucleus test in Asc-restored H460, primary human lung fibroblasts, and CHO cells was also unaffected by the XRCC1 status. Centromere-negative (clastogenic) micronuclei accounted for 80-90% of all Cr(VI)-induced micronuclei. Consistent with the micronuclei results, Asc-restored cells also showed no increase in the levels of poly(ADP-ribose), which is a biochemical marker of single-stranded breaks. Asc had no effect on cytotoxicity of O(6)-methylguanine, a lesion produced by direct DNA alkylation. Overall, our results indicate that the presence of physiological levels of Asc strongly suppresses pro-oxidant pathways in Cr(VI) metabolism and that the use of standard cell cultures creates a distorted profile of its genotoxic properties.


Assuntos
Ácido Ascórbico/farmacologia , Carcinógenos Ambientais/efeitos adversos , Ciclo Celular/efeitos dos fármacos , Cromo/efeitos adversos , Dano ao DNA/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Western Blotting , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Cricetinae , Cricetulus , Adutos de DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Glutationa/metabolismo , Guanina/análogos & derivados , Guanina/farmacologia , Humanos , Pulmão/citologia , Testes para Micronúcleos , Oxirredução , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
6.
Toxicol Appl Pharmacol ; 258(3): 367-75, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22172632

RESUMO

Nickel and cobalt are heavy metals found in land, water, and air that can enter the body primarily through the respiratory tract and accumulate to toxic levels. Nickel compounds are known to be carcinogenic to humans and animals, while cobalt compounds produce tumors in animals and are probably carcinogenic to humans. People working in industrial and manufacturing settings have an increased risk of exposure to these metals. The cytotoxicity of nickel and cobalt has individually been demonstrated; however, the underlying mechanisms of co-exposure to these heavy metals have not been explored. In this study, we investigated the effect of exposure of H460 human lung epithelial cells to nickel and cobalt, both alone and in combination, on cell survival, apoptotic mechanisms, and the generation of reactive oxygen species and double strand breaks. For simultaneous exposure, cells were exposed to a constant dose of 150 µM cobalt or nickel, which was found to be relatively nontoxic in single exposure experiments. We demonstrated that cells exposed simultaneously to cobalt and nickel exhibit a dose-dependent decrease in survival compared to the cells exposed to a single metal. The decrease in survival was the result of enhanced caspase 3 and 7 activation and cleavage of poly (ADP-ribose) polymerase. Co-exposure increased the production of ROS and the formation of double strand breaks. Pretreatment with N-acetyl cysteine alleviated the toxic responses. Collectively, this study demonstrates that co-exposure to cobalt and nickel is significantly more toxic than single exposure and that toxicity is related to the formation of ROS and DSB.


Assuntos
Cobalto/toxicidade , Pulmão/efeitos dos fármacos , Níquel/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cobalto/administração & dosagem , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Humanos , Pulmão/citologia , Pulmão/patologia , Níquel/administração & dosagem , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo
7.
Mutagenesis ; 25(4): 381-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410141

RESUMO

DNA-protein cross-links (DPCs) are caused by a large number of human carcinogens and anti-cancer drugs. However, cellular processes involved in decreasing a burden of these genotoxic lesions remain poorly understood. Here, we examined the impact of nucleotide excision repair (NER), which is a principal repair pathway for bulky DNA adducts, and the main cellular reducers on removal of chromium(VI)-induced DPC. We found that standard and ascorbate-restored cultures of isogenic XPA-null (NER deficient) and XPA-complemented human fibroblasts had very similar repair of Cr-DPC (60-65% average DPC removal after 24 h). However, XPA absence caused depletion of G1 and accumulation of G2 cells at low Cr(VI) doses, suggesting that Cr-DPC were not a significant cause of cell cycle perturbations. Interestingly, although pro-oxidant metabolism of Cr(VI) in glutathione-depleted cells generated significantly fewer DPC, they were repair resistant irrespective of the NER status of cells. Inhibition of proteasome activity by MG132 abolished DPC repair in both XPA-null and XPA-complemented cells. XPA loss caused two to three times higher initial DPC formation, demonstrating the importance of NER in removal of the precursor lesions. Our results indicate that human NER is not involved in removal of Cr-DPC containing non-histone proteins but it acts as a defence mechanism against these large lesions by preventing their formation. Therefore, individual differences in NER activity are expected to alter sensitivity but not persistence of DPC as a biomarker of hexavalent Cr.


Assuntos
Cromatos/toxicidade , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Mutagênicos/toxicidade , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo , Adutos de DNA/metabolismo , Dano ao DNA , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos
8.
Cancer Res ; 69(3): 1071-9, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19141647

RESUMO

Mismatch repair (MMR) strongly enhances cyto- and genotoxicity of several chemotherapeutic agents and environmental carcinogens. DNA double-strand breaks (DSB) formed after two replication cycles play a major role in MMR-dependent cell death by DNA alkylating drugs. Here, we examined DNA damage detection and the mechanisms of the unusually rapid induction of DSB by MMR proteins in response to carcinogenic chromium(VI). We found that MSH2-MSH6 (MutSalpha) dimer effectively bound DNA probes containing ascorbate-Cr-DNA and cysteine-Cr-DNA cross-links. Binary Cr-DNA adducts, the most abundant form of Cr-DNA damage, were poor substrates for MSH2-MSH6, and their toxicity in cells was weak and MMR independent. Although not involved in the initial recognition of Cr-DNA damage, MSH2-MSH3 (MutSbeta) complex was essential for the induction of DSB, micronuclei, and apoptosis in human cells by chromate. In situ fractionation of Cr-treated cells revealed MSH6 and MSH3 chromatin foci that originated in late S phase and did not require replication of damaged DNA. Formation of MSH3 foci was MSH6 and MLH1 dependent, whereas MSH6 foci were unaffected by MSH3 status. DSB production was associated with progression of cells from S into G(2) phase and was completely blocked by the DNA synthesis inhibitor aphidicolin. Interestingly, chromosome 3 transfer into MSH3-null HCT116 cells activated an alternative, MSH3-like activity that restored dinucleotide repeat stability and sensitivity to chromate. Thus, sequential recruitment and unprecedented cooperation of MutSalpha and MutSbeta branches of MMR in processing of Cr-DNA cross-links is the main cause of DSB and chromosomal breakage at low and moderate Cr(VI) doses.


Assuntos
Cromatos/farmacologia , Adutos de DNA/metabolismo , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Proteína MutS de Ligação de DNA com Erro de Pareamento/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Compostos de Potássio/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Cromatos/química , Cromatos/metabolismo , Cromossomos Humanos Par 3 , Adutos de DNA/química , DNA de Neoplasias/química , DNA de Neoplasias/metabolismo , Dimerização , Células HCT116 , Células HeLa , Humanos , Proteína 1 Homóloga a MutL , Proteínas Nucleares/biossíntese , Compostos de Potássio/química , Compostos de Potássio/metabolismo
9.
Infect Immun ; 76(7): 3164-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18426875

RESUMO

Despite effective chemotherapy, schistosomiasis remains a major public health problem in the developing world, with at least 200 million active infections resulting in significant morbidity. Rapid reinfection after treatment, accompanied by extensive residual morbidity, mandates alternative control strategies, including vaccine development. Paramyosin, a myofibrillar protein found only in invertebrates, has been widely studied as a vaccine candidate for both Schistosoma mansoni and Schistosoma japonicum. Recently, we demonstrated that Th2-biased immune responses to paramyosin are associated with resistance to reinfection with S. japonicum in humans; however, challenges in the pilot-scale production of schistosome paramyosin have hampered further studies of this promising vaccine candidate. Here we report a method for the pilot-scale expression and purification of recombinant S. japonicum paramyosin (rSj97). rSj97 was extracted from Escherichia coli inclusion bodies and purified with sequential anion-exchange, hydroxyapatite, and size exclusion chromatography. The purified rSj97 was >95% pure as judged by sodium dodecyl sulfate-polyacrylamide gel electrophoretic analysis and was free of significant endotoxin contamination. We demonstrate that, like native paramyosin, rSj97 adopts an alpha-helical coiled-coil tertiary structure and binds immunoglobulin and collagen. Naïve mice infected with S. japonicum produce anti-rSj97 immunoglobulin G (IgG) antibodies as early as 4 weeks postinfection, while sera collected from S. japonicum-infected individuals contain anti-rSj97 IgE antibodies. Our method for pilot-scale production of recombinant full-length paramyosin will facilitate preclinical evaluation of paramyosin as a vaccine for schistosomiasis.


Assuntos
Anticorpos Anti-Helmínticos/imunologia , Proteínas Recombinantes/metabolismo , Schistosoma japonicum/imunologia , Esquistossomose Japônica/prevenção & controle , Tropomiosina/metabolismo , Vacinas , Animais , Biotecnologia/métodos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Corpos de Inclusão/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Projetos Piloto , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Schistosoma japonicum/genética , Schistosoma japonicum/metabolismo , Esquistossomose Japônica/parasitologia , Tropomiosina/genética , Tropomiosina/imunologia , Tropomiosina/isolamento & purificação
10.
Carcinogenesis ; 28(7): 1613-20, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17301063

RESUMO

Ascorbate (Asc) plays a key role in reductive activation of carcinogenic chromium(VI) in vivo. In addition to much higher rates (t(1/2) = 1 min for 1 mM Asc), its reactions at physiological conditions differ from other reducers by low yields of Cr(V) intermediates. Human cells in culture are severely Asc deficient, which results in distorted metabolism and potentially abnormal responses to Cr(VI). We found that restoration of physiological Asc levels in human lung cells (primary IMR90 fibroblasts and epithelial H460 cells) increased clonogenic lethality and apoptosis by Cr(VI). Enhanced cytotoxicity in mass cultures was more evident after normalization for lower Cr uptake caused by leakage of Asc into media. Asc did not change uptake-adjusted yields of Cr-DNA adducts and had no effect on cytotoxicity when delivered shortly after Cr(VI) exposure. Protein and Ser-15 phosphorylation levels of p53 did not show any association with the presence of Asc and there were no increases in p53-driven reporter activity in Cr-treated cells. Stable silencing of p53 expression by short hairpin RNA (shRNA) had no effect on toxicity of Cr(VI) in both -Asc and +Asc IMR90 and H460 cells. In contrast, shRNA-mediated depletion of essential components of MutS or MutL mismatch repair complexes greatly improved survival of all Cr-treated cells and eliminated Asc-potentiated effects on cell death. Thus, mismatch repair-mediated enhancement of Cr(VI) cytotoxicity by Asc should promote the selection of MSI+/wt-p53 phenotype found among chromate-induced human lung cancers. Our findings also indicate that Asc plays a dual role in Cr(VI) toxicity: protective outside and potentiating inside the cell.


Assuntos
Apoptose , Ácido Ascórbico/metabolismo , Cromatos/toxicidade , Adutos de DNA/metabolismo , Reparo de Erro de Pareamento de DNA , Compostos de Potássio/toxicidade , Proteína Supressora de Tumor p53/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ácido Ascórbico/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Pulmão/citologia , Pulmão/metabolismo , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação , Proteína Supressora de Tumor p53/genética
11.
Nucleic Acids Res ; 35(2): 465-76, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17169990

RESUMO

Here we examined the role of cellular vitamin C in genotoxicity of carcinogenic chromium(VI) that requires reduction to induce DNA damage. In the presence of ascorbate (Asc), low 0.2-2 microM doses of Cr(VI) caused 10-15 times more chromosomal breakage in primary human bronchial epithelial cells or lung fibroblasts. DNA double-strand breaks (DSB) were preferentially generated in G2 phase as detected by colocalization of H2AX and 53BP1 foci in cyclin B1-expressing cells. Asc dramatically increased the formation of centromere-negative micronuclei, demonstrating that induced DSB were inefficiently repaired. DSB in G2 cells were caused by aberrant mismatch repair of Cr damage in replicated DNA, as DNA polymerase inhibitor aphidicolin and silencing of MSH2 or MLH1 by shRNA suppressed induction of H2AX and micronuclei. Cr(VI) was also up to 10 times more mutagenic in cells containing Asc. Increasing Asc concentrations generated progressively more mutations and DSB, revealing the genotoxic potential of otherwise nontoxic Cr(VI) doses. Asc amplified genotoxicity of Cr(VI) by altering the spectrum of DNA damage, as total Cr-DNA binding was unchanged and post-Cr loading of Asc exhibited no effects. Collectively, these studies demonstrated that Asc-dependent metabolism is the main source of genotoxic and mutagenic damage in Cr(VI)-exposed cells.


Assuntos
Ácido Ascórbico/fisiologia , Carcinógenos Ambientais/toxicidade , Cromo/toxicidade , Quebras de DNA de Cadeia Dupla , Reparo de Erro de Pareamento de DNA , Mutagênicos/toxicidade , Carcinógenos Ambientais/metabolismo , Células Cultivadas , Cromo/metabolismo , Fase G2 , Histonas/análise , Humanos , Proteína 2 Homóloga a MutS/metabolismo , Mutagênese , Mutagênicos/metabolismo , Proteínas Nucleares/metabolismo , Fase S
12.
Free Radic Biol Med ; 40(11): 1981-92, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16716899

RESUMO

Carcinogenic chromates induce DNA single-strand breaks (SSB) that are detectable by conventional alkali-based assays. However, the extent of direct breakage has been uncertain because excision repair and hydrolysis of Cr-DNA adducts at alkaline pH also generate SSB. We examined mechanisms of SSB production during chromate reduction by glutathione (GSH) and assessed the significance of these lesions in cells using genetic approaches. Cr(VI) reduction was biphasic and the formation of SSB occurred exclusively during the slow reaction phase. Catalase or iron chelators completely blocked DNA breakage, as did the use of GSH purified by a modified Chelex procedure. Thus, the direct intermediates of GSH-chromate reactions were unable to cause SSB unless activated by H2O2. SSB repair-deficient XRCC1(-/-) and proficient XRCC1+ EM9 cells had identical survival at doses causing up to 60% clonogenic death and accumulation of 1 mM Cr(VI). However, XRCC1(-/-) cells displayed higher lethality in the more toxic range and the depletion of GSH made them hypersensitive even to moderate doses. Elevation of cellular catalase or GSH levels eliminated survival differences between XRCC1(-/-) and XRCC1+ cells. In summary, formation of toxic SSB in cells occurs at relatively high chromate doses, requires H2O2, and is suppressed by high GSH concentrations.


Assuntos
Cromatos/metabolismo , Dano ao DNA , DNA de Cadeia Simples/efeitos dos fármacos , Glutationa/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetinae , Peróxido de Hidrogênio/farmacologia , Técnicas In Vitro , Ferro/farmacologia
13.
J Cell Biochem ; 97(5): 1025-35, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16288478

RESUMO

Exposure of human lung cells to carcinogenic nickel compounds in the presence of oxygen up-regulated carbonic anhydrase IX (CA IX) and NDRG1/Cap43, both known as intrinsic hypoxia markers and cancer-associated genes. This suggests that factors other than a shortage of oxygen may be involved in this induction. Both proteins can also be induced in the presence of oxygen by culturing these cells to a high density without medium change. The intracellular ascorbate measurements revealed its rapid depletion in both metal- and density-exposed cells. Nickel exposure caused strong activation of HIF-1alpha and HIF-2alpha proteins, underscoring activation of HIF-1-dependent transcription. In contrast, cell density-dependent transcription was characterized by minor induction of HIF-1alpha or HIF-2alpha. Moreover, the up-regulation of NDRG1/Cap43 in HIF-1alpha deficient fibroblasts suggested the involvement of different transcription factor(s). The repletion of intracellular ascorbate reversed the induction of CA IX and NDRG1/Cap43 caused by cell density or nickel exposure. Thus, the loss of intracellular ascorbate triggered the induction of both tumor markers by two different conditions in the presence of oxygen. Ascorbate is delivered to lung cells via the SVCT2 ascorbate transporter, which was found to be sensitive to nickel or cell density. Collectively these findings establish the importance of intracellular ascorbate levels for the regulation of expression of CA IX and NDRG1/Cap43. We suggest, that, in addition to low oxygenation, insufficient supply of ascorbate or its excessive oxidation in tumors, can contribute to the induction of hypoxia-associated proteins via both HIF-dependent and independent mechanisms.


Assuntos
Ácido Ascórbico/farmacologia , Contagem de Células , Níquel/farmacologia , Fatores de Transcrição/metabolismo , Regulação para Cima , Antígenos de Neoplasias/metabolismo , Anidrase Carbônica IX , Anidrases Carbônicas/metabolismo , Proteínas de Ciclo Celular , Hipóxia Celular , Relação Dose-Resposta a Droga , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Ferro/farmacologia , Ácido Láctico/farmacologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Proteínas/metabolismo , Transportadores de Sódio Acoplados à Vitamina C , Simportadores/metabolismo , Transcrição Gênica , Transfecção
14.
Cell Cycle ; 4(8): 1050-2, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15970712

RESUMO

Hexavalent chromium compounds are widespread environmental contaminants that are well recognized as human carcinogens and potent respiratory toxicants. Intracellular metabolism of chromium(VI) leads to the production of numerous chromium-DNA adducts that are primarily formed at the phosphate groups. The mechanism of toxicity of these DNA modifications in human cells has been uncertain for a long time because chromium and other phosphate-based adducts did not block DNA replication with purified polymerases. Our recent studies identified mismatch repair proteins as activators of toxic responses to chromium-DNA damage, which resolved an apparent discrepancy in genotoxic activity of chromium adducts in cells and in vitro. The discovered mechanism of toxicity provided the basis for a novel model of chromium carcinogenesis based on the selection of resistant clones that lack mismatch repair and progress to cancer due to high levels of spontaneous mutagenesis.


Assuntos
Pareamento Incorreto de Bases , Cromo/toxicidade , Reparo do DNA , Neoplasias/genética , Linhagem Celular Tumoral , Adutos de DNA , Dano ao DNA , Replicação do DNA , Fase G2 , Humanos , Técnicas In Vitro , Repetições de Microssatélites , Mutagênese , Mutação , Neoplasias/patologia , Fosfatos/química , Plasmídeos/metabolismo
15.
Mol Cell Biol ; 25(9): 3596-607, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15831465

RESUMO

Chromium(VI) is a toxic and carcinogenic metal that causes the formation of DNA phosphate-based adducts. Cr-DNA adducts are genotoxic in human cells, although they do not block replication in vitro. Here, we report that induction of cytotoxicity in Cr(VI)-treated human colon cells and mouse embryonic fibroblasts requires the presence of all major mismatch repair (MMR) proteins. Cr-DNA adducts lost their ability to block replication of Cr-modified plasmids in human colon cells lacking MLH1 protein. The presence of functional mismatch repair caused induction of p53-independent apoptosis associated with activation of caspases 2 and 7. Processing of Cr-DNA damage by mismatch repair resulted in the extensive formation of gamma-H2AX foci in G(2) phase, indicating generation of double-stranded breaks as secondary toxic lesions. Induction of gamma-H2AX foci was observed at 6 to 12 h postexposure, which was followed by activation of apoptosis in the absence of significant G(2) arrest. Our results demonstrate that mismatch repair system triggers toxic responses to Cr-DNA backbone modifications through stress mechanisms that are significantly different from those for other forms of DNA damage. Selection for Cr(VI) resistant, MMR-deficient cells may explain the very high frequency of lung cancers with microsatellite instability among chromate workers.


Assuntos
Pareamento Incorreto de Bases/fisiologia , Cromo/toxicidade , Dano ao DNA , Proteínas de Ligação a DNA/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose , Pareamento Incorreto de Bases/genética , Proteínas de Transporte , Caspase 2 , Caspase 7 , Caspases/metabolismo , Células Cultivadas , Colo/citologia , Colo/efeitos dos fármacos , Adutos de DNA/metabolismo , Reparo do DNA/genética , Reparo do DNA/fisiologia , Replicação do DNA/genética , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/genética , Fibroblastos/efeitos dos fármacos , Fase G2/fisiologia , Histonas/análise , Histonas/metabolismo , Humanos , Camundongos , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/fisiologia
16.
J Biol Chem ; 279(29): 30419-24, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15087443

RESUMO

Intracellular reduction of carcinogenic Cr(VI) leads to the extensive formation of Cr(III)-DNA phosphate adducts. Repair mechanisms for chromium and other DNA phosphate-based adducts are currently unknown in human cells. We found that nucleotide excision repair (NER)-proficient human cells rapidly removed chromium-DNA adducts, with an average t((1/2)) of 7.1 h, whereas NER-deficient XP-A, XP-C, and XP-F cells were severely compromised in their ability to repair chromium-DNA lesions. Activation of NER in Cr(VI)-treated human fibroblasts or lung epithelial H460 cells was manifested by XPC-dependent binding of the XPA protein to the nuclear matrix, which was also observed in UV light-treated (but not oxidant-stressed) cells. Intracellular replication of chromium-modified plasmids demonstrated increased mutagenicity of binary Cr(III)-DNA and ternary cysteine-Cr(III)-DNA adducts in cells with inactive NER. NER deficiency created by the loss of XPA in fibroblasts or by knockdown of this protein by stable expression of small interfering RNA in H460 cells increased apoptosis and clonogenic death by Cr(VI), providing genetic evidence for the role of monofunctional chromium-DNA adducts in the toxic effects of this metal. The rate of NER of chromium-DNA adducts under saturating conditions was calculated to be approximately 50,000 lesions/min/cell. Because chromium-DNA adducts cause only small changes in the DNA helix, rapid repair of these modifications in human cells indicates that the presence of major structural distortions in DNA is not required for the efficient detection of the damaged sites by NER proteins in vivo.


Assuntos
Cromatos/toxicidade , Cromo/química , Reparo do DNA , DNA/química , Linhagem Celular , Núcleo Celular/metabolismo , Cromo/farmacologia , Adutos de DNA , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Vetores Genéticos , Humanos , Peróxido de Hidrogênio/farmacologia , Microscopia de Fluorescência , Oxidantes/farmacologia , Fosfatos , Plasmídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Frações Subcelulares/metabolismo , Fatores de Tempo , Raios Ultravioleta , Proteína de Xeroderma Pigmentoso Grupo A
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...