Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 60(22): 6346-52, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103796

ABSTRACT

Arsenic compounds are potent human carcinogens. Accumulated evidence has shown that arsenite-induced cytogenetic alterations are associated with the carcinogenicity of arsenic. Because p53 plays a guarding role in maintaining genome integrity and accuracy of chromosome segregation, the mechanistic effects of arsenite on p53 activation were analyzed. In the present study, arsenite-induced DNA strand breaks were confirmed by alkaline single-cell gel electrophoresis (comet assay) in human fibroblast (HFW) cells. Accompanying the appearance of DNA strand breaks was a significant accumulation of p53 in arsenite-treated HFW cells, as demonstrated by immunoblotting and immunofluorescence techniques. p53 downstream proteins, such as p21 and the human homologue of murine double minute-2, were also significantly induced by arsenite treatment. Cell cycle retardation and G2-M arrest were observed in 5-bromo-2'-deoxyuridine pulse-labeled HFW cells by flow cytometry. Wortmannin, an inhibitor of phosphatidylinositol 3-kinases, inhibited arsenite- or X-ray irradiation-induced p53 accumulation but did not alter UV irradiation- or N-acetyl-Leu-Leu-norleucinal-induced p53 accumulation. p53 phosphorylation on serine 15 was also confirmed by immunoblotting technique in arsenite- and X-ray-treated HFW cells but was not observed in UV- or N-acetyl-Leu-Leu-norleucinal-treated HFW cells. These results suggest the involvement of a phosphatidylinositol 3-kinase-related protein kinase in arsenite-induced p53 accumulation. For confirmation, we demonstrated that arsenite treatment, similar to X-ray irradiation, did not induce p53 accumulation in GM3395 fibroblasts derived from a patient with ataxia telangiectasia. In contrast, UV irradiation did cause p53 accumulation in these cells. Together, these findings infer that arsenite-induced DNA strand breaks may lead to p53 phosphorylation and accumulation through an ataxia telangiectasia mutated-dependent pathway in HFW cells.


Subject(s)
Arsenites/toxicity , Carcinogens/toxicity , Fibroblasts/drug effects , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/metabolism , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/drug effects , Cell Cycle Proteins , Cell Death/drug effects , DNA Damage , DNA-Binding Proteins , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Tumor Suppressor Proteins
2.
Mutat Res ; 452(1): 41-50, 2000 Jul 20.
Article in English | MEDLINE | ID: mdl-10894889

ABSTRACT

Numerous reports have shown that oxidative stress is involved in arsenite-induced genetic damage. Arsenite is also a potent inducer of heme oxygenase (HO)-1. To understand whether HO-1 could function as a cellular antioxidant and protect cells from arsenite injury, the effects of tin-protoporphyrin (SnPP), a competitive inhibitor of HO-1, on arsenite-induced genetic damage were examined in human skin fibroblasts (HFW). In the present study, we found that SnPP at 100 microM significantly potentiated arsenite-induced cytotoxicity, DNA strand breaks (assayed by alkaline single cell gel electrophoresis(SCGE)), and chromatid breaks. Although arsenite alone mainly induced kinetochore-plus micronuclei (K(+)-MN), SnPP only synergistically enhanced kinetochore-negative micronuclei (K(-)-MN). The increase in K(-)-MN by SnPP cotreatment was consistent with the increase in DNA strand breaks and chromatid breaks caused by SnPP. However, at higher arsenite doses, K(+)-MN was significantly reduced by SnPP. Pretreatment of HFW cells with hemin, an inducer of HO-1, significantly attenuated the cytotoxicity of arsenite. Therefore, the present results suggest that HO-1 induction by arsenite plays certain roles in protecting cells from arsenite-induced injury.


Subject(s)
Arsenites/pharmacology , Chromosome Aberrations , DNA/drug effects , Fibroblasts/drug effects , Metalloporphyrins/pharmacology , Micronuclei, Chromosome-Defective/drug effects , Protoporphyrins/pharmacology , Cell Survival/drug effects , Cells, Cultured , Comet Assay , DNA/genetics , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Humans , Infant, Newborn , Kinetochores , Male , Tumor Cells, Cultured
3.
Mutat Res ; 440(1): 75-82, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10095130

ABSTRACT

Arsenic, widely distributed in the environment, is a potent human carcinogen. Arsenite genotoxicity has been observed in a variety of cells and animal systems. However, the underlying mechanism is not completely clear. In this study, human fibroblasts (HFW) were treated with 1.25-10 microM arsenite for 24 h (low dose and long exposure) and 5-80 microM for 4 h (high dose and short exposure), and the arsenite accumulation, cytotoxicity, and micronucleus (MN) induction were examined. By these two different protocols, HFW cells showed equivalent levels of arsenite accumulation, but exhibited different kinetics of cell killing and different types of MN generation. Arsenite induced mainly kinetochore-positive MN (K+-MN) in HFW cells by low dose exposure whereas mainly kinetochore-negative MN (K--MN) was induced by high dose exposure. Catalase reduced both K+- and K--MN induced by these two exposure protocols. Except for the case of K+-MN induction by the high dose exposure protocol, N-acetyl-cysteine (NAC) in both low and high dose protocols was also shown to effectively reduce arsenite-induced MN. The present results imply that oxidative stress is involved in arsenite-induced MN in diploid human fibroblasts. However, different protocols for arsenite exposure may result in different cellular damage.


Subject(s)
Arsenites/toxicity , Carcinogens/toxicity , Kinetochores/drug effects , Micronuclei, Chromosome-Defective/drug effects , Sodium Compounds/toxicity , Acetylcysteine/pharmacology , Arsenites/administration & dosage , Arsenites/metabolism , Carcinogens/administration & dosage , Carcinogens/metabolism , Catalase/pharmacology , Cell Survival/drug effects , Cells, Cultured , Diploidy , Dose-Response Relationship, Drug , Drug Administration Schedule , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Micronucleus Tests , Oxidative Stress , Sodium Compounds/administration & dosage , Sodium Compounds/metabolism
4.
Cancer Res ; 57(22): 5051-9, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9371502

ABSTRACT

Arsenite, a unique human carcinogen, induces many types of cytogenetic alterations, such as sister chromatid exchanges, chromosome aberrations, and endoreduplication in a variety of in vivo and in vitro systems. Cytogenetic alterations are frequently associated with cancer development. The purpose of this study was to explore how arsenite induces cytogenetic alterations in human skin fibroblasts (HFW). The present results show that treatment of G2-enriched HFW cells with 5 microM arsenite results in significant delay of cell cycle progression, accumulation of mitotic cells, and prolongation of mitosis. Arsenite-induced G2 and mitotic delay are accompanied by accumulation of cyclin B1 and hyperphosphorylation of cdc2 and Mos proteins. In addition to mitotic delay and prolongation, arsenite treatment also induced out-of-phase centromere separation and alterations of chromosome segregation, such as the appearance of c-metaphase, ball-metaphase, and lagged chromosomes. Unlike spindle poisons, arsenite at the dose range used did not inhibit the spindle fiber formation but conceivably deranges the spindle apparatus. By analyzing the karyotype of established subclones surviving arsenite injury, 18% (8 of 44) showed one chromosome loss, whereas all 26 subclones derived from the untreated cultures were diploid. Furthermore, most arsenite-treated clones manifest prolonged life span (86 +/- 18 population doublings) as compared to those derived from the untreated cultures (44 +/- 11 population doublings). Unfortunately, none became immortal. Collectively, treatment of the G2-enriched HFW cells with arsenite can disturb the mitotic events and subsequently induce chromosome loss.


Subject(s)
Arsenites/pharmacology , Chromosome Deletion , Mitosis/drug effects , Sodium Compounds/pharmacology , CDC2 Protein Kinase/metabolism , Cell Survival/drug effects , Chromosome Aberrations , Cyclin B/metabolism , Cyclin B1 , Fibroblasts/drug effects , G2 Phase/drug effects , G2 Phase/genetics , Humans , Karyotyping , Mitosis/genetics , Oncogene Proteins v-mos/metabolism , Phosphorylation , Spindle Apparatus/drug effects
5.
Environ Mol Mutagen ; 25(3): 188-96, 1995.
Article in English | MEDLINE | ID: mdl-7737136

ABSTRACT

Arsenic, strongly associated with increased risks of human cancers, is a potent clastogen in a variety of mammalian cell systems. The effect of sodium arsenite (a trivalent arsenic compound) on chromatid separation was studied in human skin fibroblasts (HFW). Human fibroblasts were arrested in S phase by the aid of serum starvation and aphidicolin blocking and then these cells were allowed to synchronously progress into G2 phase. Treatment of the G2-enriched HFW cells with sodium arsenite (0-200 microM) resulted in arrest of cells in the G2 phase, interference with mitotic division, inhibition of spindle assembly, and induction of chromosome endoreduplication in their second mitosis. Sodium arsenite treatment also inhibited the activities of serine/threonine protein phosphatases and enhanced phosphorylation levels of a small heat shock protein (HSP27). These results suggest that sodium arsenite may mimic okadaic acid to induce chromosome endoreduplication through its inhibitory effect on protein phosphatase activity.


Subject(s)
Arsenites/toxicity , Chromosome Aberrations , Fibroblasts/drug effects , Phosphoprotein Phosphatases/metabolism , Sodium Compounds/toxicity , Sulfhydryl Reagents/toxicity , Cell Division/drug effects , Cell Division/genetics , Cell Survival/drug effects , Cell Survival/genetics , Electrophoresis, Polyacrylamide Gel , Fibroblasts/enzymology , Flow Cytometry , G2 Phase/drug effects , Heat-Shock Proteins/metabolism , Histones/drug effects , Histones/metabolism , Humans , Immunohistochemistry , Infant, Newborn , Mitosis/drug effects , Mitosis/genetics , Multigene Family , Phosphorylation/drug effects , Sister Chromatid Exchange/drug effects , Sister Chromatid Exchange/genetics , Skin/cytology , Skin/drug effects
6.
Biochem Biophys Res Commun ; 202(2): 1015-22, 1994 Jul 29.
Article in English | MEDLINE | ID: mdl-8048913

ABSTRACT

Treatment of Chinese hamster ovary (CHO-K1) cells with 10 microM sodium arsenite for 24 h resulted in enhancement of a proteolytic activity toward the chromogenic substrate CBZ-Phe-Arg-AMC. Presence of dithiothreitol and a pH between 4 and 6 were required for displaying its full hydrolytic activity. According to its substrate- and inhibitor-specificity, this arsenite-induced proteolytic activity was very similar to lysosomal cathepsin B. Arsenite cytotoxicity was further shown to be partially prevented by inhibitors that inhibited the arsenite-induced protease, such as antipain and chymostatin, but not by protease inhibitors without inhibitory effects on the arsenite-induced protease. Our present results suggest that the arsenite-induced protease activity may be involved in arsenite's killing effects.


Subject(s)
Arsenites/pharmacology , CHO Cells/enzymology , Cell Death/drug effects , Endopeptidases/metabolism , Sodium Compounds/pharmacology , Animals , CHO Cells/drug effects , Coumarins/metabolism , Cricetinae , Dipeptides/metabolism , Dithiothreitol/pharmacology , Fluorescent Dyes , Hydrogen-Ion Concentration , Molecular Weight , Protease Inhibitors/pharmacology , Substrate Specificity
7.
Cell Biol Int Rep ; 15(3): 253-64, 1991 Mar.
Article in English | MEDLINE | ID: mdl-2032293

ABSTRACT

Our present results show that treatment with sodium arsenite apparently decreases cellular ATP levels in a dose- and time-dependent manner in HeLa S-3 cells. The reduction in ATP induced by sodium arsenite was possibly through mitochondrial damage, since treatment with sodium arsenite resulted in reduction of rhodamine 123 accumulation and disruption of the structure of the cristae in mitochondria. However, all of these changes could be reversed by removing sodium arsenite from the culture medium. The levels of ATP depletion were correlated with the killing effects of sodium arsenite in HeLa S-3 cells.


Subject(s)
Adenosine Triphosphate/metabolism , Arsenic/pharmacology , Arsenites , Mitochondria/drug effects , Sodium Compounds , HeLa Cells , Humans , Kinetics , Microscopy, Fluorescence , Mitochondria/pathology
8.
Arch Toxicol ; 65(8): 640-5, 1991.
Article in English | MEDLINE | ID: mdl-1747062

ABSTRACT

Post-treatment with sodium arsenite synergistically increased the cytotoxicity of ultraviolet (UV) light. The potentiation of UV cytotoxicity by sodium arsenite was apparently suppressed by cycloheximide (CHM), a protein synthesis inhibitor. The protective effect of CHM against sodium arsenite-potentiated UV cytotoxicity was well correlated to its activity in inhibiting the synthesis of stress proteins, particularly a small polypeptide with a molecular weight of 8500 dalton. This small stress protein was demonstrated as ubiquitin by immunoprecipitation. Our results also showed that neither ubiquitin induction nor potentiation of UV cytotoxicity by post-treatment with sodium arsenite was observed in the stationary cells. Thus, we suggested that ubiquitin is possibly involved in the action of arsenite in potentiating UV-induced cell killing.


Subject(s)
Arsenic/toxicity , Arsenites , Cycloheximide/pharmacology , Sodium Compounds , Ultraviolet Rays/adverse effects , Animals , CHO Cells , Cell Survival/drug effects , Cell Survival/radiation effects , Chromosome Aberrations , Cricetinae , DNA Repair , Heat-Shock Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...