Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Toxicol Appl Pharmacol ; 296: 54-60, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26908176

RESUMO

Numerous metals are well-known human bladder carcinogens. Despite the significant occupational and public health concern of metals and bladder cancer, the carcinogenic mechanisms remain largely unknown. Chromium, in particular, is a metal of concern as incidences of bladder cancer have been found elevated in chromate workers, and there is an increasing concern for patients with metal hip implants. However, the impact of hexavalent chromium (Cr(VI)) on bladder cells has not been studied. We compared chromate toxicity in two bladder cell lines; primary human urothelial cells and hTERT-immortalized human urothelial cells. Cr(VI) induced a concentration- and time-dependent increase in chromosome damage in both cell lines, with the hTERT-immortalized cells exhibiting more chromosome damage than the primary cells. Chronic exposure to Cr(VI) also induced a concentration-dependent increase in aneuploid metaphases in both cell lines which was not observed after a 24h exposure. Aneuploidy induction was higher in the hTERT-immortalized cells. When we correct for uptake, Cr(VI) induces a similar amount of chromosome damage and aneuploidy suggesting that the differences in Cr(VI) sensitivity between the two cells lines were due to differences in uptake. The increase in chromosome instability after chronic chromate treatment suggests this may be a mechanism for chromate-induced bladder cancer, specifically, and may be a mechanism for metal-induced bladder cancer, in general.


Assuntos
Cromo/toxicidade , Instabilidade Cromossômica/efeitos dos fármacos , Instabilidade Cromossômica/fisiologia , Urotélio/efeitos dos fármacos , Urotélio/fisiologia , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Humanos , Urotélio/patologia
2.
Toxicol Appl Pharmacol ; 278(3): 259-65, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24823294

RESUMO

Cobalt exposure is increasing as cobalt demand rises worldwide due to its use in enhancing rechargeable battery efficiency, super-alloys, and magnetic products. Cobalt is considered a possible human carcinogen with the lung being a primary target. However, few studies have considered cobalt-induced toxicity in human lung cells. Therefore, in this study, we sought to determine the cytotoxicity and genotoxicity of particulate and soluble cobalt in human lung cells. Cobalt oxide and cobalt chloride were used as representative particulate and soluble cobalt compounds, respectively. Exposure to both particulate and soluble cobalt induced a concentration-dependent increase in cytotoxicity, genotoxicity, and intracellular cobalt ion levels. Based on intracellular cobalt ion levels, we found that soluble cobalt was more cytotoxic than particulate cobalt while particulate and soluble cobalt induced similar levels of genotoxicity. However, soluble cobalt induced cell cycle arrest indicated by the lack of metaphases at much lower intracellular cobalt concentrations compared to cobalt oxide. Accordingly, we investigated the role of particle internalization in cobalt oxide-induced toxicity and found that particle-cell contact was necessary to induce cytotoxicity and genotoxicity after cobalt exposure. These data indicate that cobalt compounds are cytotoxic and genotoxic to human lung fibroblasts, and solubility plays a key role in cobalt-induced lung toxicity.


Assuntos
Carcinógenos Ambientais/toxicidade , Cobalto/toxicidade , Pulmão/efeitos dos fármacos , Mutagênicos/toxicidade , Transporte Biológico , Carcinógenos Ambientais/análise , Carcinógenos Ambientais/química , Carcinógenos Ambientais/metabolismo , Ciclo Celular , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Clonais , Cobalto/análise , Cobalto/química , Cobalto/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Humanos , Pulmão/química , Pulmão/metabolismo , Mutagênicos/análise , Mutagênicos/química , Mutagênicos/metabolismo , Concentração Osmolar , Óxidos/análise , Óxidos/química , Óxidos/metabolismo , Óxidos/toxicidade , Tamanho da Partícula , Fagocitose/efeitos dos fármacos , Solubilidade
3.
Mutat Res ; 762: 1-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24561002

RESUMO

Depleted uranium (DU) is extensively used in both industry and military applications. The potential for civilian and military personnel exposure to DU is rising, but there are limited data on the potential health hazards of DU exposure. Previous laboratory research indicates DU is a potential carcinogen, but epidemiological studies remain inconclusive. DU is genotoxic, inducing DNA double strand breaks, chromosome damage and mutations, but the mechanisms of genotoxicity or repair pathways involved in protecting cells against DU-induced damage remain unknown. The purpose of this study was to investigate the effects of homologous recombination repair deficiency on DU-induced genotoxicity using RAD51D and XRCC3-deficient Chinese hamster ovary (CHO) cell lines. Cells deficient in XRCC3 (irs1SF) exhibited similar cytotoxicity after DU exposure compared to wild-type (AA8) and XRCC3-complemented (1SFwt8) cells, but DU induced more break-type and fusion-type lesions in XRCC3-deficient cells compared to wild-type and XRCC3-complemented cells. Surprisingly, loss of RAD51D did not affect DU-induced cytotoxicity or genotoxicity. DU induced selective X-chromosome fragmentation irrespective of RAD51D status, but loss of XRCC3 nearly eliminated fragmentation observed after DU exposure in wild-type and XRCC3-complemented cells. Thus, XRCC3, but not RAD51D, protects cells from DU-induced breaks and fusions and also plays a role in DU-induced chromosome fragmentation.


Assuntos
Aberrações Cromossômicas/efeitos da radiação , Cromossomos de Mamíferos/efeitos da radiação , Mutagênicos/farmacologia , Reparo de DNA por Recombinação , Urânio/farmacologia , Animais , Células CHO , Cromossomos de Mamíferos/química , Cricetulus , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Teste de Complementação Genética
4.
Mutat Res ; 735(1-2): 51-5, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22583656

RESUMO

Inappropriate survival signaling after DNA damage may facilitate clonal expansion of genetically compromised cells, and it is known that protein tyrosine phosphatase (PTP) inhibitors activate key survival pathways. In this study we employed the genotoxicant, hexavalent chromium [Cr(VI)], which is a well-documented carcinogen of occupational and environmental concern. Cr(VI) induces a complex array of DNA damage, including DNA double strand breaks (DSBs). We recently reported that PTP inhibition bypassed cell cycle arrest and abrogated Cr(VI)-induced clonogenic lethality. Notably, PTP inhibition resulted in an increase in forward mutations at the HPRT locus, supporting the hypothesis that PTP inhibition in the presence of DNA damage may lead to genomic instability (GIN), via cell cycle checkpoint bypass. The aim of the present study was to determine the effect of PTP inhibition on DNA DSB formation and chromosomal integrity after Cr(VI) exposure. Diploid human lung fibroblasts were treated with Cr(VI) in the presence or absence of the PTP inhibitor, sodium orthovanadate, for up to 24h, and cells were analyzed for DNA DSBs and chromosomal damage. Cr(VI) treatment induced a rapid increase in DNA DSBs, and a significant increase in total chromosomal damage (chromatid breaks and gaps) after 24h. In sharp contrast, PTP inhibition abrogated both DNA DSBs and chromosomal damage after Cr(VI) treatment. In summary, PTP inhibition in the face of Cr(VI) genotoxic stress decreases chromosomal instability (CIN) but increases mutagenesis, which we postulate to be a result of error-prone DNA repair.


Assuntos
Instabilidade Cromossômica/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Cromo/toxicidade , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Fibroblastos , Humanos , Pulmão/citologia
5.
Neuron ; 110(10): 1671-1688.e6, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35294901

RESUMO

Amyotrophic lateral sclerosis (ALS) is characterized by motor neuron degeneration accompanied by aberrant accumulation and loss of function of the RNA-binding protein TDP43. Thus far, it remains unresolved to what extent TDP43 loss of function directly contributes to motor system dysfunction. Here, we employed gene editing to find whether the mouse ortholog of the TDP43-regulated gene STMN2 has an important function in maintaining the motor system. Both mosaic founders and homozygous loss-of-function Stmn2 mice exhibited neuromuscular junction denervation and fragmentation, resulting in muscle atrophy and impaired motor behavior, accompanied by an imbalance in neuronal microtubule dynamics in the spinal cord. The introduction of human STMN2 through BAC transgenesis was sufficient to rescue the motor phenotypes observed in Stmn2 mutant mice. Collectively, our results demonstrate that disrupting the ortholog of a single TDP43-regulated RNA is sufficient to cause substantial motor dysfunction, indicating that disruption of TDP43 function is likely a contributor to ALS.


Assuntos
Esclerose Lateral Amiotrófica , Estatmina , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Animais , Modelos Animais de Doenças , Homozigoto , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Junção Neuromuscular/metabolismo , Estatmina/genética , Estatmina/metabolismo
6.
JCI Insight ; 6(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33351783

RESUMO

The cohesin complex plays an essential role in chromosome maintenance and transcriptional regulation. Recurrent somatic mutations in the cohesin complex are frequent genetic drivers in cancer, including myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Here, using genetic dependency screens of stromal antigen 2-mutant (STAG2-mutant) AML, we identified DNA damage repair and replication as genetic dependencies in cohesin-mutant cells. We demonstrated increased levels of DNA damage and sensitivity of cohesin-mutant cells to poly(ADP-ribose) polymerase (PARP) inhibition. We developed a mouse model of MDS in which Stag2 mutations arose as clonal secondary lesions in the background of clonal hematopoiesis driven by tet methylcytosine dioxygenase 2 (Tet2) mutations and demonstrated selective depletion of cohesin-mutant cells with PARP inhibition in vivo. Finally, we demonstrated a shift from STAG2- to STAG1-containing cohesin complexes in cohesin-mutant cells, which was associated with longer DNA loop extrusion, more intermixing of chromatin compartments, and increased interaction with PARP and replication protein A complex. Our findings inform the biology and therapeutic opportunities for cohesin-mutant malignancies.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Reparo do DNA/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutação , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Animais , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Feminino , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos SCID , Camundongos Transgênicos , Síndromes Mielodisplásicas/tratamento farmacológico , Proteínas Nucleares/genética , Ftalazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Células U937 , Ensaios Antitumorais Modelo de Xenoenxerto , Coesinas
7.
Biochem Soc Trans ; 38(6): 1687-90, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21118148

RESUMO

Exposure to toxic and carcinogenic metals is widespread; however, their mechanisms of action remain largely unknown. One potential mechanism for metal-induced carcinogenicity and toxicity is centrosome amplification. Here we review the mechanisms for metal-induced centrosome amplification, including arsenic, chromium, mercury and nano-titanium dioxide.


Assuntos
Arsênio/toxicidade , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , Cromo/toxicidade , Mercúrio/toxicidade , Titânio/toxicidade , Animais , Carcinógenos/toxicidade , Linhagem Celular , Exposição Ambiental , Humanos , Nanoestruturas/toxicidade , Titânio/química
8.
Chem Res Toxicol ; 23(2): 365-72, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20000473

RESUMO

Hexavalent chromium (Cr(VI)) compounds are well-established human lung carcinogens. Solubility plays an important role in their carcinogenicity with the particulate Cr(VI) compounds being the most carcinogenic. Epidemiology and animal studies suggest that zinc chromate is the most potent particulate Cr(VI) compound; however, there are few comparative data to support these observations. The purpose of this study was to compare the genotoxicity of zinc chromate with two other particulate Cr(VI) compounds, barium chromate and lead chromate, and one soluble Cr(VI) compound, sodium chromate. The clastogenic effects of barium chromate and zinc chromate were similar, but lead chromate induced significantly less damage. The levels of DNA damage measured by gamma-H2A.X foci formation were similar for the three particulate chromium compounds. Corrected for chromium uptake differences, we found that zinc chromate and barium chromate were the most cytotoxic, and lead chromate and sodium chromate were less cytotoxic. Zinc chromate was more clastogenic than all other chromium compounds, and lead chromate was the least clastogenic. There was no significant difference between any of the compounds for the induction of DNA double strand breaks. All together, these data suggest that the difference in the carcinogenic potency of zinc chromate over the other chromium compounds is not due solely to a difference in chromium ion uptake and that the zinc cation may in fact have an important role in its carcinogenicity.


Assuntos
Brônquios/citologia , Cromo/toxicidade , Testes de Mutagenicidade , Carcinógenos Ambientais/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos
9.
Chem Res Toxicol ; 23(2): 386-95, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20030412

RESUMO

Hexavalent chromium (Cr(VI)) compounds are known human lung carcinogens. Solubility plays an important role in its carcinogenicity with the particulate or insoluble form being the most potent. Of the particulate Cr(VI) compounds, zinc chromate appears to be the most potent carcinogen; however, very few studies have investigated its carcinogenic mechanism. In this study, we investigated the ability of chronic exposure to zinc chromate to induce numerical chromosome instability. We found no increase in aneuploidy after a 24 h exposure to zinc chromate, but with more chronic exposures, zinc chromate induced concentration- and time-dependent increases in aneuploidy in the form of hypodiploidy, hyperdiploidy, and tetraploidy. Zinc chromate also induced centrosome amplification in a concentration- and time-dependent manner in both interphase and mitotic cells after chronic exposure, producing cells with centriolar defects. Furthermore, chronic exposure to zinc chromate induced concentration- and time-dependent increases in spindle assembly checkpoint bypass with increases in centromere spreading, premature centromere division, and premature anaphase. Last, we found that chronic exposure to zinc chromate induced a G2 arrest. All together, these data indicate that zinc chromate can induce chromosome instability after prolonged exposures.


Assuntos
Centrossomo/efeitos dos fármacos , Cromatos/toxicidade , Instabilidade Cromossômica/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Fuso Acromático/efeitos dos fármacos , Compostos de Zinco/toxicidade , Aneuploidia , Linhagem Celular , Humanos , Pulmão/citologia , Tamanho da Partícula , Solubilidade
10.
Toxicol Appl Pharmacol ; 234(3): 293-9, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19027772

RESUMO

Hexavalent chromium Cr(VI) is a respiratory toxicant and carcinogen, with solubility playing an important role in its carcinogenic potential. Zinc chromate, a water insoluble or 'particulate' Cr(VI) compound, has been shown to be carcinogenic in epidemiology studies and to induce tumors in experimental animals, but its genotoxicity is poorly understood. Our study shows that zinc chromate induced concentration-dependent increases in cytotoxicity, chromosome damage and DNA double strand breaks in human lung cells. In response to zinc chromate-induced breaks, MRE11 expression was increased and ATM and ATR were phosphorylated, indicating that the DNA double strand break repair system was initiated in the cells. In addition, our data show that zinc chromate-induced double strand breaks were only observed in the G2/M phase population, with no significant amount of double strand breaks observed in G1 and S phase cells. These data will aid in understanding the mechanisms of zinc chromate toxicity and carcinogenesis.


Assuntos
Cromatos/toxicidade , Instabilidade Cromossômica , Quebras de DNA de Cadeia Dupla , DNA/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Mutagênicos/toxicidade , Compostos de Zinco/toxicidade , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Pulmão/metabolismo , Pulmão/patologia , Proteína Homóloga a MRE11 , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima
11.
Rev Environ Health ; 23(1): 39-57, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18557597

RESUMO

Hexavalent chromium is a commonly used industrial metal that has been shown to induce lung cancer in workers having long term exposure. In the particulate form, Cr(VI) dissolves slowly in vivo, leading to an extended exposure of lung cells. Hexavalent chromium is taken into the cell and rapidly reduced to Cr(V), Cr(IV), Cr(III), and reactive oxygen species. Cells treated with Cr(VI) are subject to several types of DNA damage resulting from this reduction, including base modification, single-strand breaks, double-strand breaks, Cr-DNA adducts, DNA-Cr-DNA adducts, and protein-Cr-DNA adducts. These types of damage, if left unrepaired or are misrepaired, can lead to growth arrest, cytotoxicity, and apoptosis, as well as mutations leading to neoplastic transformation and ultimately tumorigenesis. Here we review the current literature on Cr-induced DNA damage and its repair.


Assuntos
Cromo/toxicidade , Dano ao DNA , Reparo do DNA , Neoplasias Pulmonares/induzido quimicamente , Poluentes Ocupacionais do Ar/toxicidade , Humanos , Indústrias , Neoplasias Pulmonares/patologia , Doenças Profissionais/etiologia , Exposição Ocupacional/efeitos adversos , Estresse Oxidativo , Material Particulado/toxicidade
12.
Mar Pollut Bull ; 56(8): 1416-21, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18599091

RESUMO

The endangered Western population of the Steller sea lion declined for three decades for uncertain reasons. We present baseline data of metal concentrations in pups as a first step towards investigating the potential threat of developmental exposures to contaminants. Seven metals were investigated: arsenic, cadmium, silver, aluminum, mercury, lead and vanadium. Vanadium was detected in only a single blubber sample. Mercury appears to be the most toxicologically significant metal with concentrations in the liver well above the current action level for mercury in fish. The concentrations of aluminum, arsenic, silver, cadmium and lead were present in one-fourth to two-thirds of all samples and were at either comparable or below concentrations previously reported. Neither gender nor region had a significant effect on metal burdens. Future work should consider metal concentrations in juveniles and adults and toxicological studies need to be performed to begin to assess the toxicity of these metals.


Assuntos
Metais Pesados/metabolismo , Leões-Marinhos/metabolismo , Poluentes Químicos da Água/metabolismo , Animais , Feminino , Masculino , Distribuição Tecidual
13.
Cancer Res ; 66(8): 4041-8, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16618723

RESUMO

Hexavalent chromium [Cr(VI)] compounds are established human lung carcinogens. The carcinogenicity of Cr(VI) is related to its solubility, with the most potent carcinogens being the insoluble particulate Cr(VI) compounds. However, it remains unknown why particulate Cr(VI) is more carcinogenic than soluble Cr(VI). One possible explanation is that particulates may provide more chronic exposures to chromate over time. We found that aneuploid cells increased in a concentration- and time-dependent manner after chronic exposure to lead chromate. Specifically, a 24-hour lead chromate exposure induced no aneugenic effect, whereas a 120-hour exposure to 0.5 and 1 microg/cm2 lead chromate induced 55% and 60% aneuploid metaphases, respectively. We also found that many of these aneuploid cells were able to continue to grow and form colonies. Centrosome defects are known to induce aneuploidy; therefore, we investigated the effects of chronic lead chromate exposure on centrosomes. We found that centrosome amplification in interphase and mitotic cells increased in a concentration- and time-dependent manner with 0.5 and 1 microg/cm2 lead chromate for 120 hours, inducing aberrant centrosomes in 18% and 21% of interphase cells and 32% and 69% of mitotic cells, respectively; however, lead oxide did not induce centrosome amplification in interphase or mitotic cells. There was also an increase in aberrant mitosis after chronic exposure to lead chromate with the emergence of disorganized anaphase and mitotic catastrophe. These data suggest that one possible mechanism for lead chromate-induced carcinogenesis is through centrosome dysfunction, leading to the induction of aneuploidy.


Assuntos
Aneuploidia , Centrossomo/efeitos dos fármacos , Cromatos/toxicidade , Chumbo/toxicidade , Pulmão/efeitos dos fármacos , Linhagem Celular , Centrossomo/fisiologia , Humanos , Pulmão/fisiologia , Pulmão/ultraestrutura , Mitose/efeitos dos fármacos
14.
Am J Respir Cell Mol Biol ; 37(5): 544-52, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17585109

RESUMO

Particulate hexavalent chromium (Cr(VI)) is a well-established human lung carcinogen with widespread exposure among people in occupational settings and the general public. However, no studies have examined the chromate-induced malignant transformation of human lung epithelial cells, its predominant target. Human papillomavirus-immortalized human bronchial epithelial (BEP2D) cells were used to better understand the mechanisms involved in human bronchial carcinogenesis induced by particulate chromate. We found that aneuploid cells increased in a concentration-dependent manner after chronic exposure to lead chromate. Moreover, chronic exposure to lead chromate induced BEP2D cell transformation. Transformed BEP2D cells developed through a series of sequential steps, including altered cell morphology, loss of cell contact inhibition and anchorage-independent growth. Specifically, a 5-day exposure to lead chromate induced foci formation with 0, 1, 5, and 10 microg/cm2 lead chromate inducing 0, 7, 3, and 15 foci in 10 dishes. Anchorage independence was observed in cell lines derived from these foci. These foci-derived cells also showed centrosome amplification and increases in aneuploid metaphases. Our study demonstrates that particulate Cr(VI) is able to transform human bronchial epithelial cells, and that chromosome instability may play an important role in particulate Cr(VI)-induced neoplastic transformation.


Assuntos
Brônquios/efeitos dos fármacos , Carcinógenos/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Cromatos/toxicidade , Chumbo/toxicidade , Brônquios/patologia , Linhagem Celular Transformada , Transformação Celular Neoplásica/genética , Instabilidade Cromossômica/efeitos dos fármacos , Humanos
16.
Mutat Res ; 610(1-2): 2-7, 2006 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16872863

RESUMO

Particulate hexavalent chromium (Cr(VI)) is a well-established human lung carcinogen. It is currently a major public health concern, there is widespread exposure to it in occupational settings and to the general public. However, despite the potential widespread exposure and the fact that the lung is its target organ, few studies have considered the toxic effects of particulate Cr(VI) in human lung cells. Accordingly, we used lead chromate as a model particulate Cr(VI) compound and determined its cytotoxicity and genotoxicity in cultured human bronchial epithelial cells, using BEP2D cells as a model cell line. We found that lead chromate induced concentration-dependent cytotoxicity in BEP2D cells after a 24h exposure. Specifically, the relative survival was 78, 59, 53, 46 and 0% after exposure to 0.5, 1, 5, 10 and 50 microg/cm(2) lead chromate, respectively. Similarly, the amount of chromosome damage increased with concentration after 24h exposure to lead chromate. Specifically, 0.5, 1, 5 and 10 microg/cm(2) damaged 10, 13, 20 and 28% of metaphase cells with the total amount of damage reaching 11, 15, 24 and 36 aberrations per 100 metaphases, respectively. Lead chromate (50 microg/cm(2) lead chromate) induced profound cell cycle delay and no metaphases were found. In addition we investigated the effects of soluble hexavalent chromium, sodium chromate, in this cell line. We found that 1, 2.5, 5 and 10 microM sodium chromate induced 66, 35, 0 and 0% relative survival, respectively. The amount of chromosome damage increased with concentration after 24h exposure to sodium chromate. Specifically, 1, 2.5 and 5 microM damaged 25, 34 and 41% of metaphase cells with the total amount of damage reaching 33, 59 and 70 aberrations per 100 metaphases, respectively. Ten micromolar sodium chromate induced profound cell cycle delay and no metaphases were found. Overall the data clearly indicate that hexavalent Cr(VI) is cytotoxic and genotoxic to human lung epithelial cells.


Assuntos
Cromo/farmacologia , Células Epiteliais/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Carcinógenos Ambientais/química , Carcinógenos Ambientais/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatos/farmacologia , Cromo/química , Aberrações Cromossômicas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Chumbo/farmacologia , Pulmão/citologia , Pulmão/metabolismo , Metáfase/efeitos dos fármacos , Tamanho da Partícula , Compostos de Sódio/farmacologia , Solubilidade
17.
Mutat Res ; 610(1-2): 8-13, 2006 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16870495

RESUMO

Hexavalent chromium (Cr(VI)) is a well-designated human lung carcinogen, with solubility playing an important role in its carcinogenic potential. Although it is known that particulate or water-insoluble Cr(VI) compounds are more potent than the soluble species of this metal, the mechanisms of action are not fully elucidated. In this study, we investigated the hypothesis that the difference in potency between particulate and soluble Cr(VI) is due to more chronic exposures with particulate chromate because it can deposit and persist in the lungs while soluble chromate is rapidly cleared. Chronic exposure to both insoluble lead chromate and soluble sodium chromate induced a concentration and time-dependent increase in intracellular Cr ion concentrations in cultured human lung fibroblasts. Intracellular Pb levels after chronic exposure to lead chromate increased in a concentration-dependent manner but did not increase with longer exposure times up to 72 h. We also investigated the effects of chronic exposure to Cr(VI) on clastogenicity and found that chronic exposure to lead chromate induces persistent or increasing chromosome damage. Specifically, exposure to 0.5 microg/cm(2) lead chromate for 24, 48 and 72 h induced 23, 23 and 27% damaged metaphases, respectively. Contrary to lead chromate, the amount of chromosome damage after chronic exposure to sodium chromate decreased with time. For example, cells exposed to 1 microM sodium chromate for 24, 48 and 72 h induced 23, 13 and 17% damaged metaphases, respectively. Our data suggest a possible mechanism for the observed potency difference between soluble and insoluble Cr(VI) compounds is that chronic exposure to particulate Cr(VI) induces persistent chromosome damage and chromosome instability while chromosome damage is repaired with chronic exposure to soluble Cr(VI).


Assuntos
Cromo/farmacologia , Pulmão/efeitos dos fármacos , Carcinógenos Ambientais/química , Carcinógenos Ambientais/farmacologia , Linhagem Celular , Cromatos/farmacologia , Cromo/química , Aberrações Cromossômicas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Chumbo/farmacologia , Pulmão/citologia , Pulmão/metabolismo , Metáfase/efeitos dos fármacos , Tamanho da Partícula , Compostos de Sódio/farmacologia , Solubilidade , Fatores de Tempo
18.
Environ Mol Mutagen ; 57(4): 282-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27040722

RESUMO

Cobalt is a toxic metal used in various industrial applications leading to adverse lung effects by inhalation. Cobalt is considered a possible human carcinogen with the lung being a primary target. However, few studies have considered cobalt-induced toxicity in human lung cells, especially normal lung epithelial cells. Therefore, in this study, we sought to determine the cytotoxicity and genotoxicity of particulate and soluble cobalt in normal primary human lung epithelial cells. Cobalt oxide and cobalt chloride were used as representative particulate and soluble cobalt compounds, respectively. Exposure to both particulate and soluble cobalt induced a concentration-dependent increase in cytotoxicity, genotoxicity, and intracellular cobalt ion levels. Based on intracellular cobalt ion levels, we found that soluble and particulate cobalt induced similar cytotoxicity while soluble cobalt was more genotoxic than particulate cobalt. These data indicate that cobalt compounds are cytotoxic and genotoxic to human lung epithelial cells.


Assuntos
Cobalto/toxicidade , Citotoxinas/toxicidade , Células Epiteliais/efeitos dos fármacos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Mutagênicos/toxicidade , Óxidos/toxicidade , Linhagem Celular , Aberrações Cromossômicas/induzido quimicamente , Dano ao DNA/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Pulmão/metabolismo , Pulmão/patologia , Tamanho da Partícula , Solubilidade
19.
Cell Rep ; 15(11): 2488-99, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27264184

RESUMO

BRCA1/2 proteins function in homologous recombination (HR)-mediated DNA repair and cooperate with Fanconi anemia (FA) proteins to maintain genomic integrity through replication fork stabilization. Loss of BRCA1/2 proteins results in DNA repair deficiency and replicative stress, leading to genomic instability and enhanced sensitivity to DNA-damaging agents. Recent studies have shown that BRCA1/2-deficient tumors upregulate Polθ-mediated alternative end-joining (alt-EJ) repair as a survival mechanism. Whether other mechanisms maintain genomic integrity upon loss of BRCA1/2 proteins is currently unknown. Here we show that BRCA1/2-deficient tumors also upregulate FANCD2 activity. FANCD2 is required for fork protection and fork restart in BRCA1/2-deficient tumors. Moreover, FANCD2 promotes Polθ recruitment at sites of damage and alt-EJ repair. Finally, loss of FANCD2 in BRCA1/2-deficient tumors enhances cell death. These results reveal a synthetic lethal relationship between FANCD2 and BRCA1/2, and they identify FANCD2 as a central player orchestrating DNA repair pathway choice at the replication fork.


Assuntos
Proteína BRCA1/deficiência , Proteína BRCA2/deficiência , Reparo do DNA por Junção de Extremidades , Replicação do DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Neoplasias/genética , Neoplasias/patologia , Animais , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Reparo do DNA por Junção de Extremidades/genética , Replicação do DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Endodesoxirribonucleases , Instabilidade Genômica , Humanos , Camundongos Nus , Mutação/genética , Proteínas Nucleares/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Ubiquitinação , Regulação para Cima/genética , DNA Polimerase teta
20.
Mutat Res ; 586(2): 160-72, 2005 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-16112599

RESUMO

Hexavalent chromium (Cr(VI)) is a widespread environmental contaminant and a known human carcinogen, generally causing bronchial cancer. Recent studies have shown that the particulate forms of Cr(VI) are the potent carcinogens. Particulate Cr(VI) is known to induce a spectrum of DNA damage such as DNA single strand breaks, Cr-DNA adducts, DNA-protein crosslinks and chromosomal aberrations. However, particulate Cr(VI)-induced DNA double strand breaks (DSBs) have not been reported. Thus, the aim of this study was to determine if particulate Cr(VI)-induces DSBs in human bronchial cells. Using the single cell gel electrophoresis assay (comet assay), showed that lead chromate-induced concentration dependent increases in DSBs with 0.1, 0.5, 1 and 5 microg/cm2 lead chromate inducing a 20, 50, 67 and 109% relative increase in the tail integrated intensity ratio, respectively. Sodium chromate at concentrations of 1, 2.5 and 5 microM induced 38, 78 and 107% relative increase in the tail integrated intensity ratio, respectively. We also show that genotoxic concentrations of lead chromate activate the ataxia telangiectasia mutated (ATM) protein, which is thought to play a central role in the early stages of DSB detection and controls cellular responses to this damage. The H2A.X protein becomes rapidly phosphorylated on residue serine 139 in cells when DSBs are introduced into the DNA by ionizing radiation. By using immunofluorescence, we found that lead chromate-induced concentration-dependent increases in phosphorylated H2A.X (r-H2A.X) foci formation with 0.1, 0.5, 1, 5 and 10 microg/cm2 lead chromate inducing a relative increase in the number of cells with r-H2A.X foci formation of 43, 51, 115 and 129%, respectively.


Assuntos
Cromatos/toxicidade , Dano ao DNA , Chumbo/toxicidade , Pulmão/citologia , Pulmão/efeitos dos fármacos , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Ensaio Cometa , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos , Imunofluorescência , Histonas/metabolismo , Humanos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Compostos de Sódio/toxicidade , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA