Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Environ Mol Mutagen ; 57(4): 282-7, 2016 May.
Article in English | MEDLINE | ID: mdl-27040722

ABSTRACT

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.


Subject(s)
Cobalt/toxicity , Cytotoxins/toxicity , Epithelial Cells/drug effects , Lung/cytology , Lung/drug effects , Mutagens/toxicity , Oxides/toxicity , Cell Line , Chromosome Aberrations/chemically induced , DNA Damage/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Lung/metabolism , Lung/pathology , Particle Size , Solubility
2.
Toxicol Appl Pharmacol ; 296: 54-60, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26908176

ABSTRACT

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.


Subject(s)
Chromium/toxicity , Chromosomal Instability/drug effects , Chromosomal Instability/physiology , Urothelium/drug effects , Urothelium/physiology , Cells, Cultured , DNA Damage/drug effects , DNA Damage/physiology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Urothelium/pathology
3.
Toxicol Sci ; 147(2): 490-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26293554

ABSTRACT

Particulate hexavalent chromium (Cr(VI)) is a well-established human lung carcinogen. Lung tumors are characterized by structural and numerical chromosome instability. Centrosome amplification is a phenotype commonly found in solid tumors, including lung tumors, which strongly correlates with chromosome instability. Human lung cells exposed to Cr(VI) exhibit centrosome amplification but the underlying phenotypes and mechanisms remain unknown. In this study, we further characterize the phenotypes of Cr(VI)-induced centrosome abnormalities. We show that Cr(VI)-induced centrosome amplification correlates with numerical chromosome instability. We also show chronic exposure to particulate Cr(VI) induces centrosomes with supernumerary centrioles and acentriolar centrosomes in human lung cells. Moreover, chronic exposure to particulate Cr(VI) affects the timing of important centriolar events. Specifically, chronic exposure to particulate Cr(VI) causes premature centriole disengagement in S and G2 phase cells. It also induces premature centrosome separation in interphase. Altogether, our data suggest that chronic exposure to particulate Cr(VI) targets the protein linkers that hold centrioles together. These centriolar linkers are important for key events of the centrosome cycle and their premature disruption might underlie Cr(VI)-induced centrosome amplification.


Subject(s)
Centrioles/drug effects , Centrosome/drug effects , Chromium/toxicity , Lung/drug effects , Particulate Matter/toxicity , Cell Line , Chromates/administration & dosage , Chromates/toxicity , Chromium/administration & dosage , Chromosomal Instability/drug effects , Humans , Lung/cytology , Particulate Matter/administration & dosage , Zinc Compounds/administration & dosage , Zinc Compounds/toxicity
4.
Biol Trace Elem Res ; 166(1): 49-56, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25805272

ABSTRACT

Hexavalent chromium Cr(VI) is a known human lung carcinogen, with solubility playing an important role in its carcinogenic potency. Dermal exposure to Cr(VI) is common and has been associated with skin damage; however, no link between chromate exposure and skin cancer has been found. In this study, we compared the cytotoxic and clastogenic effects of Cr(VI) and its impacts on cell cycle progression in human lung and skin fibroblasts. We found human skin cells arrested earlier in their cell cycle and exhibit more cytotoxicity than human lung cells, despite taking up similar amounts of Cr. These outcomes are consistent with a hypothesis that different cellular and molecular responses underlie the differences in carcinogenic outcome in these two tissues.


Subject(s)
Cell Cycle Checkpoints/drug effects , Chromium/toxicity , Environmental Pollutants/toxicity , Fibroblasts/drug effects , Lung/drug effects , Skin/drug effects , Cell Culture Techniques , Cell Cycle Checkpoints/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Chromium/chemistry , Environmental Pollutants/chemistry , Humans , Lung/cytology , Organ Specificity , Skin/cytology , Solubility
5.
Toxicol Sci ; 142(1): 117-25, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25173789

ABSTRACT

The aim of this study was to focus on hexavalent chromium, [Cr(VI)], a chemical carcinogen and major public health concern, and consider its ability to impact DNA double strand break repair. We further focused on particulate Cr(VI), because it is the more potent carcinogenic form of Cr(VI). DNA double strand break repair serves to protect cells against the detrimental effects of DNA double strand breaks. For particulate Cr(VI), data show DNA double strand break repair must be overcome for neoplastic transformation to occur. Acute Cr(VI) exposures reveal a robust DNA double strand break repair response, however, longer exposures have not been considered. Using the comet assay, we found longer exposures to particulate zinc chromate induced concentration-dependent increases in DNA double strand breaks indicating breaks were occurring throughout the exposure time. Acute (24 h) exposure induced DNA double strand break repair signaling by inducing Mre11 foci formation, ATM phosphorylation and phosphorylated ATM foci formation, Rad51 protein levels and Rad51 foci formation. However, longer exposures reduced the Rad51 response. These data indicate a major chemical carcinogen can simultaneously induce DNA double strand breaks and alter their repair and describe a new and important aspect of the carcinogenic mechanism for Cr(VI).


Subject(s)
Carcinogenesis/drug effects , Carcinogens/toxicity , Chromates/toxicity , DNA Breaks, Double-Stranded , Lung/drug effects , Rad51 Recombinase/genetics , Recombinational DNA Repair , Zinc Compounds/toxicity , Carcinogenesis/genetics , Cell Culture Techniques , Cell Line , Chromosome Aberrations/chemically induced , Comet Assay , Humans , Lung/metabolism , Lung/pathology , Mutagens/toxicity , Time Factors
6.
Toxicol Appl Pharmacol ; 278(3): 259-65, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24823294

ABSTRACT

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.


Subject(s)
Carcinogens, Environmental/toxicity , Cobalt/toxicity , Lung/drug effects , Mutagens/toxicity , Biological Transport , Carcinogens, Environmental/analysis , Carcinogens, Environmental/chemistry , Carcinogens, Environmental/metabolism , Cell Cycle , Cell Line , Cell Survival/drug effects , Clone Cells , Cobalt/analysis , Cobalt/chemistry , Cobalt/metabolism , DNA Fragmentation/drug effects , Humans , Lung/chemistry , Lung/metabolism , Mutagens/analysis , Mutagens/chemistry , Mutagens/metabolism , Osmolar Concentration , Oxides/analysis , Oxides/chemistry , Oxides/metabolism , Oxides/toxicity , Particle Size , Phagocytosis/drug effects , Solubility
7.
Mutat Res ; 762: 1-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24561002

ABSTRACT

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.


Subject(s)
Chromosome Aberrations/radiation effects , Chromosomes, Mammalian/radiation effects , Mutagens/pharmacology , Recombinational DNA Repair , Uranium/pharmacology , Animals , CHO Cells , Chromosomes, Mammalian/chemistry , Cricetulus , DNA Breaks, Double-Stranded/drug effects , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Genetic Complementation Test
8.
Mutat Res ; 735(1-2): 51-5, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22583656

ABSTRACT

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.


Subject(s)
Chromosomal Instability/drug effects , Genomic Instability/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Chromium/toxicity , DNA Breaks, Double-Stranded , DNA Damage , Fibroblasts , Humans , Lung/cytology
9.
Biochem Soc Trans ; 38(6): 1687-90, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118148

ABSTRACT

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.


Subject(s)
Arsenic/toxicity , Centrosome/drug effects , Centrosome/metabolism , Chromium/toxicity , Mercury/toxicity , Titanium/toxicity , Animals , Carcinogens/toxicity , Cell Line , Environmental Exposure , Humans , Nanostructures/toxicity , Titanium/chemistry
10.
Article in English | MEDLINE | ID: mdl-20211760

ABSTRACT

In this study we directly compared soluble and particulate chromate cytotoxicity and genotoxicity in human (Homo sapiens) and sea lion (Eumetopias jubatus) lung fibroblasts. Our results show that hexavalent chromium induces increased cell death and chromosome damage in both human and sea lion cells with increasing intracellular chromium ion levels. The data further indicate that both sodium chromate and lead chromate are less cytotoxic and genotoxic to sea lion cells than human cells, based on an administered dose. Differences in chromium ion uptake explained some but not all of the reduced amounts of sodium chromate-induced cell death. By contrast, uptake differences could explain the differences in sodium chromate-induced chromosome damage and particulate chromate-induced toxicity. Altogether they indicate that while hexavalent chromium induces similar toxic effects in sea lion and human cells, there are different mechanisms underlying the toxic outcomes.


Subject(s)
Chromium/toxicity , Cytotoxins/toxicity , Fibroblasts/drug effects , Lung/drug effects , Sea Lions , Animals , Cell Line , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung/metabolism , Lung/pathology , Mutagenicity Tests/methods , Sea Lions/metabolism , Species Specificity
11.
Chem Res Toxicol ; 23(2): 386-95, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20030412

ABSTRACT

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.


Subject(s)
Centrosome/drug effects , Chromates/toxicity , Chromosomal Instability/drug effects , Fibroblasts/drug effects , Lung/drug effects , Spindle Apparatus/drug effects , Zinc Compounds/toxicity , Aneuploidy , Cell Line , Humans , Lung/cytology , Particle Size , Solubility
12.
Chem Res Toxicol ; 23(2): 365-72, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20000473

ABSTRACT

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.


Subject(s)
Bronchi/cytology , Chromium/toxicity , Mutagenicity Tests , Carcinogens, Environmental/toxicity , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans
13.
Toxicol Appl Pharmacol ; 234(3): 293-9, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19027772

ABSTRACT

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.


Subject(s)
Chromates/toxicity , Chromosomal Instability , DNA Breaks, Double-Stranded , DNA/drug effects , Fibroblasts/drug effects , Lung/drug effects , Mutagens/toxicity , Zinc Compounds/toxicity , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Cell Survival/drug effects , DNA/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung/metabolism , Lung/pathology , MRE11 Homologue Protein , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation
14.
Mar Pollut Bull ; 56(8): 1416-21, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18599091

ABSTRACT

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.


Subject(s)
Metals, Heavy/metabolism , Sea Lions/metabolism , Water Pollutants, Chemical/metabolism , Animals , Female , Male , Tissue Distribution
15.
Rev Environ Health ; 23(1): 39-57, 2008.
Article in English | MEDLINE | ID: mdl-18557597

ABSTRACT

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.


Subject(s)
Chromium/toxicity , DNA Damage , DNA Repair , Lung Neoplasms/chemically induced , Air Pollutants, Occupational/toxicity , Humans , Industry , Lung Neoplasms/pathology , Occupational Diseases/etiology , Occupational Exposure/adverse effects , Oxidative Stress , Particulate Matter/toxicity
16.
Am J Respir Cell Mol Biol ; 37(5): 544-52, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17585109

ABSTRACT

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.


Subject(s)
Bronchi/drug effects , Carcinogens/toxicity , Cell Transformation, Neoplastic/chemically induced , Chromates/toxicity , Lead/toxicity , Bronchi/pathology , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/drug effects , Humans
17.
Chem Res Toxicol ; 19(11): 1492-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17112237

ABSTRACT

One of the hallmarks of lung cancer is chromosome instability (CIN), particularly a tetraploid phenotype, which is normally prevented by the spindle assembly checkpoint. Hexavalent chromium Cr(VI) is an established human lung carcinogen, and Cr(VI) induces tumors at lung bifurcation sites where Cr(VI) particles impact and persist. However, the effects of Cr(VI) on the spindle assembly checkpoint are unknown and little is known about prolonged exposure to particulate Cr(VI). Accordingly, we investigated particulate Cr(VI)-induced bypass of the spindle assembly checkpoint after several days of exposure in WHTBF-6 cells. We found that lead chromate indeed induces spindle assembly checkpoint bypass in human lung cells, as 72, 96, and 120 h treatments with 0.5 or 1 microg/cm2 lead chromate induced significant increases in the percentage of cells with aberrant mitotic figures. For example, treatment with 1 microg/cm2 lead chromate for 96 h induced 11, 12.3, and 14% of cells with premature anaphase, centromere spreading and premature centromere division, respectively. In addition, we found a disruption of mitosis with more cells accumulating in anaphase; cells treated for 96 h increased from 18% in controls to 31% in cells treated with lead chromate. To confirm involvement of the spindle assembly checkpoint, Mad2 expression was used as a marker. Mad2 expression was decreased in cells exposed to chronic treatments of lead chromate, consistent with disruption of the checkpoint. We also found concentration- and time-dependent increases in tetraploid cells, which continued to grow and form colonies. When cells were treated with chronic lead alone there was no increase in aberrant mitotic cells or polyploidy; however, chronic exposure to a soluble Cr(VI) showed an increase in aberrant mitotic cells and polyploidy. These data suggest that lead chromate does induce CIN and may be one mechanism in the development of Cr(VI)-induced lung cancer.


Subject(s)
Cell Cycle/physiology , Chromates/toxicity , Chromosomal Instability/drug effects , Lead/toxicity , Lung/cytology , Particulate Matter/toxicity , Spindle Apparatus/drug effects , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Chromates/metabolism , Colchicine/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Humans , Lead/metabolism , Lung/drug effects , Mad2 Proteins , Particulate Matter/metabolism , Polyploidy , Repressor Proteins/metabolism , Solubility , Time Factors
18.
Mutat Res ; 610(1-2): 8-13, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-16870495

ABSTRACT

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).


Subject(s)
Chromium/pharmacology , Lung/drug effects , Carcinogens, Environmental/chemistry , Carcinogens, Environmental/pharmacology , Cell Line , Chromates/pharmacology , Chromium/chemistry , Chromosome Aberrations/drug effects , Dose-Response Relationship, Drug , Humans , Lead/pharmacology , Lung/cytology , Lung/metabolism , Metaphase/drug effects , Particle Size , Sodium Compounds/pharmacology , Solubility , Time Factors
19.
Mutat Res ; 610(1-2): 2-7, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-16872863

ABSTRACT

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.


Subject(s)
Chromium/pharmacology , Epithelial Cells/drug effects , Lung/drug effects , Carcinogens, Environmental/chemistry , Carcinogens, Environmental/pharmacology , Cell Cycle/drug effects , Cell Line , Cell Survival/drug effects , Chromates/pharmacology , Chromium/chemistry , Chromosome Aberrations/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Lead/pharmacology , Lung/cytology , Lung/metabolism , Metaphase/drug effects , Particle Size , Sodium Compounds/pharmacology , Solubility
20.
Cancer Res ; 66(8): 4041-8, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618723

ABSTRACT

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.


Subject(s)
Aneuploidy , Centrosome/drug effects , Chromates/toxicity , Lead/toxicity , Lung/drug effects , Cell Line , Centrosome/physiology , Humans , Lung/physiology , Lung/ultrastructure , Mitosis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...