Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Growth Differ ; 12(10): 487-96, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11682460

RESUMO

The cellular responses to carcinogen exposure influence cellular fate, which in turn modulates the neoplastic response. Certain hexavalent chromium [Cr(VI)] compounds are implicated as occupational respiratory carcinogens at doses that are both genotoxic and cytotoxic. We examined the mechanism of Cr(VI)-induced apoptosis in normal human fibroblasts (BJ) immortalized by human telomerase gene transfection (BJ-hTERT), and we assessed the spectrum of cumulative cellular fates [(a) regaining of replicative potential; (b) terminal growth arrest; or (c) apoptosis] for a narrow range of increasingly genotoxic doses of Cr(VI). Exposure of BJ-hTERT cells to Cr(VI) resulted in a dose-dependent increase in apoptosis that involved mitochondrial disruption as evidenced by mitochondrial membrane depolarization and cytochrome c release. The initial response to Cr(VI) exposure was inhibition of cell cycle progression. At the lowest dose tested (1 microM; 32% clonogenic survival), the cell cycle inhibition led to terminal growth arrest but no apoptosis. The fraction of terminally growth arrested cells increased as the dose was increased to 3 microM but then decreased at 4, 5, and 6 microM as apoptosis became the predominant cell fate. Our results suggest that cell populations exposed to Cr(VI) have a different spectrum of responses, depending on the extent of DNA damage, and that the regaining of replicative potential after relatively higher genotoxic exposures may be attributable to either escape from, or resistance to, terminal growth arrest or apoptosis.


Assuntos
Apoptose , Cromatos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Compostos de Sódio/farmacologia , Telomerase/metabolismo , Caspase 3 , Caspases/metabolismo , Ciclo Celular , Divisão Celular/efeitos dos fármacos , Linhagem da Célula , Células Cultivadas , Proteínas de Ligação a DNA , Relação Dose-Resposta a Droga , Humanos , Imuno-Histoquímica , Membranas Intracelulares , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Fenótipo , Estresse Fisiológico , Telomerase/genética , Fatores de Tempo , Transfecção
2.
Mol Cell Biochem ; 222(1-2): 173-82, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11678599

RESUMO

Hexavalent chromium (Cr (VI)) is reduced intracellularly to Cr (V), Cr (IV) and Cr (III) by ascorbate (Asc), cysteine and glutathione (GSH). These metabolites induce a spectrum of genomic DNA damage resulting in the inhibition of DNA replication. Our previous studies have shown that treatment of DNA with Cr (III) or Cr (VI) plus Asc results in the formation of DNA-Cr-DNA crosslinks (Cr-DDC) and guanine-specific arrests of both prokaryotic and mammalian DNA polymerases. GSH not only acts as a reductant of Cr (VI) but also becomes crosslinked to DNA by Cr, thus, the focus of the present study was to examine the role of GSH in Cr-induced DNA damage and polymerase arrests. Co-incubation of Cr (III) with plasmid DNA in the presence of GSH led to the crosslinking of GSH to DNA. GSH co-treatment with Cr (III) also led to a decrease in the degree of Cr-induced DNA interstrand crosslinks relative to Cr (III) alone, without affecting total Cr DNA binding. DNA polymerase arrests were observed following treatment of DNA with Cr (III) alone, but were markedly reduced when GSH was added to the reaction mixture. Pre-formed polymerase-arresting lesions (Cr-DDC) were not removed by subsequent addition of GSH. Treatment of DNA with Cr (VI), in the presence of GSH, resulted in crosslinking of GSH to DNA, but failed to produce detectable DNA interstrand crosslinks or polymerase arrests. The inhibitory effect of GSH on Cr-induced polymerase arrest was further confirmed in human genomic DNA using quantitative PCR (QPCR) analysis. Treatment of genomic DNA with Cr (III) resulted in a marked inhibition of the amplification of a 1.6 kb target fragment of the p53 gene by Taq polymerase. This was almost completely prevented by co-treatment with GSH and Cr (III). These results indicate that Cr-induced DNA interstrand crosslinks, and not DNA-Cr-GSH crosslinks, are the principal lesions responsible for blocking DNA replication. Moreover, the formation of DNA-Cr-GSH crosslinks may actually preclude the formation of the polymerase arresting lesions.


Assuntos
Cromo/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , DNA Polimerase Dirigida por DNA/metabolismo , DNA/efeitos dos fármacos , Glutationa/farmacologia , Sítios de Ligação , Linhagem Celular , DNA/metabolismo , Adutos de DNA , Dano ao DNA , Replicação do DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/efeitos dos fármacos , Interações Medicamentosas/fisiologia , Fibroblastos , Genoma , Humanos , Pulmão/citologia , Oxirredução , Plasmídeos
3.
Mol Pharmacol ; 60(3): 559-67, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11502888

RESUMO

Physiological stress conditions associated with the tumor microenvironment play a role in resistance to anticancer therapy. In this study, treatment of EMT6 mouse mammary tumor cells with hypoxia or the chemical stress agents brefeldin A (BFA) or okadaic acid (OA) causes the development of resistance to the topoisomerase II inhibitor etoposide. The mechanism of physiological stress-induced drug resistance may involve the activation of stress-responsive proteins and transcription factors. Our previous work shows that treatment with BFA or OA causes activation of the nuclear transcription factor NF-kappa B. Pretreatment with the proteasome inhibitor carbobenzyoxyl-leucinyl-leucinyl-leucinal inhibits stress-induced NF-kappa B activation and reverses BFA-induced drug resistance. To test whether NF-kappa B specifically mediates stress-induced drug resistance, an inducible phosphorylation site-deficient mutant of I kappa B alpha (I kappa B alpha M, S32/36A) was introduced into EMT6 cells. In this study, we show that I kappa B alpha M expression inhibits stress-induced NF-kappa B activation and prevents BFA-, hypoxia-, and OA-induced resistance to etoposide. These results indicate that NF-kappa B activation mediates both chemical and physiological drug resistance to etoposide. Furthermore, they imply that coadministration of agents that inhibit NF-kappa B may enhance the efficacy of topoisomerase II inhibitors in clinical cancer chemotherapy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Proteínas de Ligação a DNA/biossíntese , Etoposídeo/farmacologia , Proteínas I-kappa B , NF-kappa B/metabolismo , Oxigênio/metabolismo , Animais , Brefeldina A/farmacologia , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Interações Medicamentosas , Resistencia a Medicamentos Antineoplásicos/genética , Camundongos , Mutação , Inibidor de NF-kappaB alfa , Ácido Okadáico/farmacologia , Fosforilação , Células Tumorais Cultivadas
4.
Carcinogenesis ; 21(11): 2027-33, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11062164

RESUMO

A variety of key events in the molecular apoptotic pathway involve the mitochondria. Cyclosporin A (csA) affects the mitochondria by inhibiting the mitochondrial permeability transition (MPT), thereby preventing disruption of the transmembrane potential. The role of the MPT in apoptosis is not fully understood, but inhibition of the MPT may prevent the release of mitochondrial caspase activators, such as cytochrome c (cyt c), into the cytosol. Certain hexavalent chromium [Cr(VI)] compounds are known occupational respiratory tract toxins and carcinogens. We have previously shown that these compounds induce apoptosis as a predominant mode of cell death and that this effect can be observed in cell culture using soluble Cr(VI). We show here that Cr(VI)-induced apoptosis in Chinese hamster ovary (CHO) cells involves disruption of mitochondrial stability. Using a cyt c-specific monoclonal antibody, we observed a dose-dependent release of mitochondrial cyt c in cytosolic extracts of CHO cells exposed to apoptogenic doses of sodium chromate. Co-treatment of these cells with csA inhibited the release of cyt c and abrogated Cr(VI)-induced apoptosis as determined by a reduction in internucleosomal DNA fragmentation. Co-treatment with csA also markedly increased clonogenic survival of Cr(VI)-treated CHO cells. In contrast, the general caspase inhibitor Z-VAD-FMK markedly inhibited most of the morphological and biochemical parameters of apoptosis but did not prevent cyt c release and did not increase clonogenic survival. These results suggest that the MPT plays an important role in the regulation of mitochondrial cyt c release and that this may be a critical point in the apoptotic pathway in which cells are irreversibly committed to death.


Assuntos
Apoptose/efeitos dos fármacos , Cromatos/antagonistas & inibidores , Cromatos/toxicidade , Ciclosporina/farmacologia , Grupo dos Citocromos c/metabolismo , Mitocôndrias/efeitos dos fármacos , Compostos de Sódio/antagonistas & inibidores , Compostos de Sódio/toxicidade , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/fisiologia , Células CHO/efeitos dos fármacos , Inibidores de Caspase , Caspases/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Inibidores de Cisteína Proteinase/farmacologia , Citosol/efeitos dos fármacos , Citosol/enzimologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
Cancer Res ; 60(16): 4629-37, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10969817

RESUMO

Eicosanoids modulate the interaction of tumor cells with various host components in cancer metastasis. Their synthesis involves the release of arachidonic acid (AA) from cellular phospholipids by phospholipase A2 (PLA2), followed by metabolism by cyclooxygenases (COXs) and lipooxygenases (LOXs). This study aimed to identify the pathway(s) of AA metabolism that are required for the invasion of prostate tumor cells. DU-145 and PC-3 human prostate cancer cell lines were used to test the effect of inhibitors of PLA2, COX, or LOX on the invasion of prostate tumor cells through Matrigel in vitro using the Boyden chamber assay and fibroblast-conditioned medium as the chemoattractant. We used nontoxic doses that did not inhibit simple cell motility and did not decrease clonogenic survival. All of the inhibitors caused a significant reduction in AA release from treated cells compared with control cells, which indicated that the treatments were biochemically active. Invasion through Matrigel was inhibited by the PLA2 inhibitor 4-bromophenacyl bromide (4-BPB), the general COX inhibitor ibuprofen (IB), and the highly selective COX-2 inhibitor NS398. Inhibition of cell invasiveness by 4-BPB (1.0 microM), IB (10.0 microM), and NS398 (10.0 microM) was reversed by the addition of prostaglandin E2 (PGE2). PGE2 alone, however, did not stimulate invasiveness, which suggests that its production is necessary for rendering the cells invasive-permissive but not sufficient for inducing invasiveness. In contrast, we found no significant inhibition of invasion of prostate tumor cells treated with esculetin (1.0 microM) or nordihydroguiaretic acid (1.0 microM), which are specific inhibitors of LOX. We also tested the effect of 4-BPB, IB, NS398, and esculetin on the secretion of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs), as key enzymes in the proteolysis of Matrigel during invasion, using gelatin zymograms and Western blots. Cells that received 4-BPB, IB, or NS398, but not esculetin showed a significant reduction in the levels of proMMP-2, MMP-9, and proMMP-9 in the culture medium. DU-145 cells did not secrete TIMP-1, and the drugs did not alter the secretion of TIMP-2. This work highlights the role played by COX in disturbing the balance between MMPs and TIMPs in prostate cancer cells, and it points to the potential use of COX inibitors, especially COX-2 selective inhibitors, in the prevention and therapy of prostate cancer invasion.


Assuntos
Eicosanoides/fisiologia , Inibidores Enzimáticos/farmacologia , Metaloproteinases da Matriz/metabolismo , Prostaglandinas/biossíntese , Neoplasias da Próstata/patologia , Células 3T3 , Acetofenonas/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Ácido Araquidônico/metabolismo , Ácido Araquidônico/fisiologia , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/farmacologia , Eicosanoides/metabolismo , Humanos , Ibuprofeno/farmacologia , Isoenzimas/antagonistas & inibidores , Lipoxigenase/metabolismo , Inibidores de Lipoxigenase/farmacologia , Masculino , Inibidores de Metaloproteinases de Matriz , Proteínas de Membrana , Camundongos , Invasividade Neoplásica , Nitrobenzenos/farmacologia , Fosfolipases A/antagonistas & inibidores , Fosfolipases A2 , Prostaglandina-Endoperóxido Sintases , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/metabolismo , Sulfonamidas/farmacologia , Inibidores Teciduais de Metaloproteinases/metabolismo , Células Tumorais Cultivadas , Umbeliferonas/farmacologia
7.
Mol Carcinog ; 28(2): 111-8, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10900468

RESUMO

Some forms of hexavalent chromium [Cr(VI)] are known to cause damage to respiratory-tract tissue and DNA and are thought to be human lung carcinogens. In general, Cr(VI) is mutagenic and carcinogenic at doses that also evoke some cell death, and we previously showed that the predominant mode of death is apoptosis. Because p53 has been shown to initiate apoptosis after genotoxic insults, the objective of these experiments was to determine whether p53 is activated in and necessary for apoptosis of normal diploid human lung fibroblasts (HLF cells) after chromium exposure. By using annexin(V) staining and fluorescent microscopy, we found that Cr(VI) caused up to 14% of HLF cells to undergo apoptosis within 24 h after exposure. In addition, by using western blotting, we found that p53 protein levels increased fourfold to sixfold after exposure to sodium chromate. Because the major function of p53 is as a transcription factor, it must be translocated from the cytoplasm to the nucleus after chromate exposure to be active. Immunofluorescence studies using an antibody against p53 showed that, after chromate exposure, p53 was located in the nucleus of the treated HLF cells. The necessity of p53 for chromium-induced apoptosis was examined in two ways. One approach used dermal fibroblasts from p53 wild-type, heterozygous, and null mice, and the other approach used HLF cells that were transiently transfected with the human papilloma virus E6 gene, which targets p53 for degradation and creates a functional p53-null cell. These studies showed that chromium-induced apoptosis was p53 dependent. Mol. Carcinog. 28:111-118, 2000.


Assuntos
Apoptose/efeitos dos fármacos , Cromo/farmacologia , Pulmão/efeitos dos fármacos , Pele/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose/fisiologia , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Humanos , Pulmão/citologia , Camundongos , Camundongos Knockout , Pele/citologia
8.
Toxicol Sci ; 55(1): 60-8, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10788560

RESUMO

Some forms of hexavalent chromium [Cr(VI)] are known to cause damage to respiratory tract tissue, and are thought to be human lung carcinogens. Because Cr(VI) is mutagenic and carcinogenic at doses that evoke cell toxicity, the objective of these experiments was to examine the effect of Cr(VI) on the growth, survival, and mode of cell death in normal human lung fibroblasts (HLF cells). DNA adduct formation was monitored as a marker for bioavailability of genotoxic chromium. We also examined the modulation of these endpoints by vitamins C and E. Long-term Cr(VI) exposures were employed, which decreased clonogenic cell survival by 25% to 95% in a dose-dependent manner. The predominant cellular response to Cr(VI) was growth arrest. We found that Cr(VI) caused up to 20% of HLF cells to undergo apoptosis, and documented apoptotic morphology and the phagocytosis of apoptotic bodies by neighboring cells. P53 levels increased 4- to 6-fold in chromium-treated cells. In contrast with previous studies using CHO cells, the present study using HLFs found that pretreatment with either vitamin C or E did not exhibit a significant effect on Cr-induced apoptosis or clonogenic survival. In addition, pretreatment with vitamin C did not affect the p53 induction observed after chromium treatment. Neither vitamin had any effect on Cr-DNA adduct formation. These data indicate that although pretreatment with vitamin C or E alters the spectrum of cellular and/or genetic lesions induced by chromium(VI), neither vitamin altered the initiation or progression of apoptosis in diploid human lung cells.


Assuntos
Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Ácido Ascórbico/farmacologia , Cromo/toxicidade , Pulmão/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Vitamina E/farmacologia , Western Blotting , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cromatos/antagonistas & inibidores , Cromatos/toxicidade , Cromo/antagonistas & inibidores , Células Clonais/efeitos dos fármacos , Adutos de DNA/efeitos dos fármacos , Fibroblastos , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Microscopia Eletrônica , Fosfatidilserinas/metabolismo , Compostos de Sódio/antagonistas & inibidores , Compostos de Sódio/toxicidade
9.
Breast Cancer Res Treat ; 59(1): 15-26, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10752676

RESUMO

The 78 kDa glucose-regulated stress protein GRP78 is induced by physiological stress conditions such as hypoxia, low pH, and glucose deprivation which often exist in the microenvironments of solid tumors. Activation of this stress pathway occurs in response to several pro-apoptotic stimuli. In vitro studies have demonstrated a correlation between induced expression of GRP78 and resistance to apoptotic death induced by topoisomerase II-directed drugs. We were interested in characterizing this protein in human breast lesions for potential implications in chemotherapeutic intervention. Surgical specimens of human breast lesions and paired normal tissues from the same patients were flash frozen for these studies. Total RNA and/or protein were extracted from these tissues and used in northern and/or western blot analyses, respectively, to quantify the relative expression of GRP78. Northern blot analysis indicated that 0/5 benign breast lesions, 3/5 estrogen receptor positive (ER+) breast tumors, and 6/9 estrogen receptor negative (ER-) breast tumors exhibited overexpression of GRP78 mRNA compared to paired normal tissues, with fold overexpressions ranging from 1.8 to 20. Western blot analyses correlated with these findings since 0/5 benign breast lesions, 4/6 ER+ breast tumors, and 3/3 ER- breast tumors overexpressed GRP78 protein with fold overexpressions ranging from 1.8 to 19. Immunohistochemical analysis of these tissues demonstrated that the expression of GRP78 was heterogeneous among the cells comprising different normal and malignant glands, but confirmed the overexpression of GRP78 in most of the more aggressive ER- tumors. These results suggest that some breast tumors exhibit adverse microenvironment conditions that induce the overexpression of specific stress genes that may play a role in resistance to apoptosis and decreased chemotherapeutic efficacy.


Assuntos
Neoplasias da Mama/genética , Proteínas de Transporte/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico , Chaperonas Moleculares/genética , Proteínas de Neoplasias/biossíntese , Adulto , Idoso , Apoptose , Doenças Mamárias/genética , Doenças Mamárias/fisiopatologia , Neoplasias da Mama/fisiopatologia , Proteínas de Transporte/biossíntese , Chaperona BiP do Retículo Endoplasmático , Feminino , Proteínas de Choque Térmico HSP70/biossíntese , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Chaperonas Moleculares/biossíntese , Proteínas de Neoplasias/genética
10.
Oncol Res ; 12(9-10): 383-95, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11697817

RESUMO

Stress conditions associated with solid tumors lead to the formation of heterogeneous tumor cell subpopulations and insensitivity to cancer chemotherapeutics. In this report, we show that EMT6 mouse mammary tumor cells treated with the chemical stress, brefeldin A (BFA), or the physiological stress, hypoxia, develop resistance to the topoisomerase II (topoII) inhibitors teniposide and etoposide. BFA and hypoxia treatment did not alter intracellular drug concentrations, topoll protein levels, or inhibit topoII activity. BFA and hypoxia did cause the activation of the nuclear transcription factor NF-kappaB. We demonstrate that pretreatment with the synthetic cyclopentenone prostaglandin A1 (PGA1) inhibits stress-induced NF-kappaB activation and reverses BFA- and hypoxia-induced resistance. The reversal of BFA-induced resistance can occur when PGA1 is administered either before or several hours after the induction of stress. Taken together, these data support the involvement of NF-kappaB in stress-induced drug resistance, show that pharmacologic inhibitors of NF-kappaB can disrupt the biological consequences of stress, and imply that inhibitors of NF-kappaB may be useful agents to enhance the clinical efficacy of topoII-directed chemotherapeutics.


Assuntos
Resistencia a Medicamentos Antineoplásicos , NF-kappa B/metabolismo , Prostaglandinas A/farmacologia , Inibidores da Topoisomerase II , Animais , Western Blotting , Brefeldina A/farmacologia , Relação Dose-Resposta a Droga , Etoposídeo/farmacologia , Humanos , Hipóxia , Luciferases/metabolismo , Camundongos , Teniposídeo/farmacologia , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
11.
Toxicol Appl Pharmacol ; 161(3): 240-8, 1999 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10620481

RESUMO

Occupational exposure to certain particulate hexavalent chromium [Cr(VI)] compounds, such as lead chromate, has been associated with lung cancer and respiratory tract toxicity. We have previously shown that apoptosis is a major mode of death in cultured rodent cells treated with soluble sodium chromate and particulate lead chromate. Here we report the cellular and molecular effects of lead chromate and sodium chromate in normal human lung small airway epithelial (HSAE) cells, which may be one of the targets for Cr(VI)-induced lung cancer and respiratory tract toxicity. Phagocytosed lead chromate particles and intracellular lead-inclusion bodies (LIB) were observed by transmission electron microscopy and confirmed by X-ray analysis. HSAE cells exposed to lead chromate and sodium chromate underwent dose-dependent apoptosis. The cellular uptake and genomic interactions of both Cr and lead (Pb) were examined by inductively coupled plasma mass spectrometry (ICPMS) coupled with a novel, direct-injection high-efficiency nebulizer (DIHEN). Using this approach, we have quantitated a dose-dependent formation of Cr-DNA adducts and DNA-associated Pb in lead chromate-treated HSAE cells. The formation of LIB in normal human lung cells exposed to lead chromate indicates that ionic Pb is released from the particles and thus might contribute to the cell toxicity caused by lead chromate. Internalization and dissolution of lead chromate particles and the interaction of ionic Cr and Pb with DNA, may be components of the mechanism of lead chromate carcinogenesis. Lead chromate-induced apoptosis may be a mechanism to eliminate cells with chromium- and/or lead-damaged DNA.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos/toxicidade , Cromatos/toxicidade , Células Epiteliais/efeitos dos fármacos , Corpos de Inclusão/efeitos dos fármacos , Chumbo/toxicidade , Pulmão/efeitos dos fármacos , Transporte Biológico , Carcinógenos/metabolismo , Células Cultivadas , Cromatos/metabolismo , Adutos de DNA/análise , Dano ao DNA , Relação Dose-Resposta a Droga , Microanálise por Sonda Eletrônica , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Humanos , Chumbo/metabolismo , Pulmão/metabolismo , Pulmão/ultraestrutura , Microscopia de Fluorescência , Tamanho da Partícula , Fagocitose/efeitos dos fármacos
12.
Mol Carcinog ; 23(4): 201-6, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9869448

RESUMO

We have previously shown that trivalent chromium, and hexavalent chromium in the presence of one of its primary in vivo reductants, ascorbate, can bind to DNA and form interstrand crosslinks capable of obstructing replication. This effect was demonstrated in vitro by using Sequenase Version 2.0 T7 DNA polymerase; its parent enzyme, the unmodified T7 DNA polymerase; and Escherichia coli polymerase I large (Klenow) fragment; and it was demonstrated ex vivo by using Taq polymerase and DNA from chromium-treated human lung cells as template. This study was performed to determine whether DNA-bound chromium affects mammalian DNA polymerases in the same manner. Two mammalian enzymes, DNA polymerase alpha and DNA polymerase beta, were used. DNA polymerase alpha is a processive enzyme believed to be the primary lagging-stand synthetase, whereas DNA polymerase beta is a non-processive enzyme believed to function in DNA repair by filling single stranded gaps one base at a time. DNA polymerase arrest assays were performed with each of these enzymes to replicate DNA with toxicologically relevant levels of chromium adducts produced by either trivalent chromium or hexavalent chromium and ascorbate. Both enzymes responded to chromium-DNA damage by arresting replication, and the arrests increased in a dose-dependent manner. Furthermore, the guanine-specific pattern of arrests produced when an exonuclease-free preparation of DNA polymerase beta was used corresponded exactly to the arrest patterns produced in vitro by the exonuclease-free enzyme Sequenase and ex vivo by Taq polymerase. These results suggest that replication arrest may be a common response of polymerases to DNA-chromium lesions and provide a plausible mechanism for the inhibition of DNA synthesis and S-phase cell-cycle delay that occurs in mammalian cells treated with genotoxic chromium compounds.


Assuntos
Cromo/toxicidade , Adutos de DNA/metabolismo , Dano ao DNA , DNA Polimerase I/metabolismo , DNA Polimerase beta/metabolismo , Replicação do DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/metabolismo , Ácido Ascórbico/farmacologia , Ciclo Celular/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , DNA Bacteriano/efeitos dos fármacos , Humanos , Plasmídeos/efeitos dos fármacos
13.
Toxicol Sci ; 45(1): 72-6, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9848113

RESUMO

Some compounds of hexavalent chromium are well-established carcinogens. Chromium enters mammalian cells in the hexavalent form and is reduced to chromium (III). Treatment of purified DNA with chromium (III) produces DNA-DNA interstrand crosslinks (DDC) which obstruct the progression of DNA polymerases in vitro. DDC were also detected in chromate-treated cultured normal human lung cells using the renaturing agarose gel electrophoresis (RAGE) assay and correlated with base-specific inhibition of DNA replication. Curiously, DDC have gone undetected in studies of cultured cells using the alkaline elution (AE) technique, whereas chromium-mediated DNA-protein crosslinks (DPC) were readily detected by AE. We tested the hypothesis that AE conditions [60 mM tetraethyl ammonium hydroxide (TEA), 20 mM EDTA, pH 12.6, for 16 h at room temperature] dissociate DDC but not DPC using chromium(III)-treated plasmid DNA and the RAGE assay. Dose-dependent chromium-induced DDC were unaffected by TEA (pH 11.8) alone or by more rigorous alkaline denaturation conditions (200 mM NaOH, pH 13.5, for 16 h). DDC were, however, completely disrupted by EDTA (pH 12.6) alone or the combination of TEA and EDTA (pH 12.6). In contrast, DPC remained largely intact under these conditions. Therefore, past AE-based studies which have failed to detect chromium-induced DDC do not prove the absence of this lesion. AE may not be suitable for detecting DDC induced by EDTA-chelatable agents such as metals.


Assuntos
Cáusticos/farmacologia , Cloretos/farmacologia , Compostos de Cromo/farmacologia , Adutos de DNA/metabolismo , DNA/efeitos dos fármacos , Ácido Edético/farmacologia , Hidróxido de Sódio/farmacologia , Tetraetilamônio/farmacologia , DNA/metabolismo , Plasmídeos , Proteínas/metabolismo
14.
Oncol Rep ; 5(6): 1307-18, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9769362

RESUMO

The adverse health effects linked with chromium (Cr) exposure, the role of solubility and chemical speciation of Cr compounds, and the diverse cellular and molecular effects of Cr make the study of Cr carcinogenesis and toxicology very interesting and complex. Certain Cr compounds are prominent metal carcinogens in both occupational and environmental settings. Inhaled particulate forms of hexavalent Cr [Cr(VI)] cause lung cancer as well as lung toxicity. Some of the important factors in determining the biological outcome of Cr exposure include the bioavailability, chemical speciation and solubility of Cr compounds, intracellular reduction, and interaction of Cr with DNA. The stable oxidation states of Cr found in nature are Cr(III) and Cr(VI). Cr(III) is unable to enter cells but Cr(VI) enters into cells through membrane anionic transporters. Intracellular Cr(VI) is metabolically reduced to the ultimate Cr(III). Cr(VI) does not react with macromolecules such as DNA, RNA, proteins and lipids. However, both Cr(III) and the reductional intermediate Cr(V) are capable of co-ordinate covalent interactions with macromolecules. At the genomic level, Cr genotoxicity manifests as gene mutations, several types of DNA lesions and inhibition of macromolecular synthesis. At the cellular level, Cr exposure may lead to cell cycle arrest, apoptosis, premature terminal growth arrest, or neoplastic transformation. Cr-induced DNA-DNA interstrand crosslinks (DDC), the tumor suppressor gene p53 and oxidative processes are some of the major factors that may play a significant role in determining the cellular outcome in response to Cr exposure. We have utilized cellular, molecular, pharmacological, and genetic approaches to understand the interrelationship between Cr-induced genotoxicity, apoptosis and carcinogenesis. This review is based on the results and inferences of this research. We hope this review will clarify existing concepts and also introduce novel perspectives in chromium carcinogenesis research.


Assuntos
Apoptose/fisiologia , Carcinógenos/toxicidade , Transformação Celular Neoplásica , Cromo/toxicidade , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , DNA/efeitos dos fármacos , Dano ao DNA , Exposição Ambiental , Humanos , Exposição Ocupacional
15.
Cancer Res ; 58(14): 3059-65, 1998 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9679971

RESUMO

Brefeldin A, an agent that disrupts protein transport from the endoplasmic reticulum to the Golgi, induces the expression of GRP78 and the activation of nuclear factor (NF)-kappaB in cells. Treatment of cells with brefeldin A causes the development of resistance to topoisomerase II-directed agents, such as etoposide and doxorubicin. In this study, we show that treatment of EMT6 mouse mammary tumor cells with brefeldin A strongly induces GRP78 mRNA (8.5-fold) and resistance to teniposide (VM26). Treatment with okadaic acid causes a minor increase in GRP78 mRNA (2.1-fold) yet still induces resistance to VM26 as effectively as brefeldin A. In contrast, cells treated with castanospermine show a moderate increase in GRP78 mRNA (3.9-fold) but no resistance to VM26. These data imply that GRP78 induction does not mediate the development of drug resistance. An alternative mechanism of drug resistance may involve activation of the transcription factor, NF-kappaB, and we show that both brefeldin A and okadaic acid activate NF-kappaB in EMT6 cells. Furthermore, we demonstrate that treatment with the proteasome inhibitor MG-132 blocks the activation of NF-kappaB and prevents the development of resistance to VM26 induced by brefeldin A. Collectively, these results suggest that the resistance to VM26 in EMT6 cells treated with brefeldin A is mediated by the activation of NF-kappaB rather than the induction of GRP78. Our results also suggest that inhibition of NF-kappaB activation in tumor cells may increase the efficacy of topoisomerase II-directed agents in chemotherapy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Proteínas de Transporte/metabolismo , Ciclopentanos/farmacologia , Cisteína Endopeptidases/efeitos dos fármacos , Proteínas de Choque Térmico , Chaperonas Moleculares/metabolismo , Complexos Multienzimáticos/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Teniposídeo/farmacologia , Animais , Antibacterianos/farmacologia , Brefeldina A , Inibidores de Cisteína Proteinase/farmacologia , Resistencia a Medicamentos Antineoplásicos , Chaperona BiP do Retículo Endoplasmático , Leupeptinas/farmacologia , Macrolídeos , Camundongos , NF-kappa B/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma , Células Tumorais Cultivadas/efeitos dos fármacos
16.
Toxicol Sci ; 46(2): 260-5, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10048129

RESUMO

A novel method is described for the sensitive detection of chromium-DNA adducts. Chromium-DNA adducts were determined in 1 microgram of DNA from normal human lung fibroblasts exposed to sodium chromate using microscale flow injection analysis with a direct injection high-efficiency nebulizer and inductively coupled plasma mass spectrometry detection. The frequency of Cr-DNA adducts increased in a dose-dependent sigmoidal manner, indicating saturation and toxicity. The low detection limits (on the order of parts per trillion) allows the detection of as few as two Cr adducts per 10,000 bases, which, coupled with the small DNA sample requirement, makes this technique suitable for measuring metal-DNA adducts as biomarkers of exposure to toxic and carcinogenic metals such as Cr, in cultured cells, animals, and humans.


Assuntos
Cromo/metabolismo , Adutos de DNA/análise , Pulmão/citologia , Espectrometria de Massas/métodos , Adutos de DNA/isolamento & purificação , Relação Dose-Resposta a Droga , Fibroblastos , Humanos , Nebulizadores e Vaporizadores , Sensibilidade e Especificidade
17.
Toxicol Appl Pharmacol ; 146(2): 270-80, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9344895

RESUMO

Certain hexavalent chromium compounds are documented human carcinogens. Exposure of cells to particulate forms of chromium results in cell-enhanced dissolution of particles in the extracellular microenvironment and chronic production of chromium oxyanions, which are taken up by the cell through an anion transport system and are genotoxic and clastogenic. It was previously shown that apoptosis is the mode of cell death of nearly all of the Chinese hamster ovary cells (CHO-AA8 cell line), which die after high-dose, short-term treatments with soluble sodium chromate. In this report the mode of cell killing by particulate lead chromate and of low-dose continuous treaments of soluble sodium chromate designed to mimic conditions of ionic chromate uptake after lead chromate exposure was examined. CHO-AA8 cells were treated for 24 hr with doses of sodium chromate or lead chromate which cause a 50% decrease in survival in colony-forming effeciency assays. Longer treatments (up to 72 hr) at the same doses did not decrease survival further than the 24-hr exposure. Morphological changes indicative of apoptosis, as well as internucleosomal DNA fragmentation, were detectable by 24 hr after treatment with lead chromate or soluble sodium chromate. All of the cells killed by treatments with lead chromate particles underwent apoptosis as the mode of cell death and this was accurately modeled in cell culture by continuous treatments with low-dose soluble sodium chromate. Exposure of cells to hexavalent chromium compounds causes a spectrum of DNA damage which can be selectively altered by pretreatment of cells with antioxidant vitamins prior to chromium exposure. Here we show that ascorbate and alpha-tocopherol markedly inhibited the chromosomal aberrations induced by both particulate and soluble chromate compounds, even though chromium adduct levels were not decreased by either vitamin pretreatment. Cell survival assays showed that ascorbate, but not alpha-tocopherol, protected cells from apoptosis induced by sodium chromate. The results differentiate chromium-induced apoptosis from both chromosomal damage and adduct levels and suggest that other lesions sensitive to ascorbate but not tocopherol are the proximal inducing signal for chromium-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Ácido Ascórbico/farmacologia , Cromatos/toxicidade , Dano ao DNA , Chumbo/toxicidade , Vitamina E/farmacologia , Animais , Apoptose/genética , Células CHO/efeitos dos fármacos , Células CHO/patologia , Células CHO/ultraestrutura , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Corantes/toxicidade , Cricetinae , Adutos de DNA/análise , Fragmentação do DNA/efeitos dos fármacos , Microscopia Eletrônica , Microscopia de Fluorescência , Tamanho da Partícula
18.
Mol Hum Reprod ; 3(7): 563-8, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9268133

RESUMO

In order to test the hypothesis that integrin and uteroglobin (UG) expression in cultured endometrial cells are affected by hormone treatment, Ishikawa-CH endometrial cancer cells were cultured and exposed to oestradiol or oestradiol and progesterone regimens and assayed using immunohistochemistry. We evaluated the intensity of immunohistochemical staining for the integrin monomers alpha(v) and beta1, the dimers alpha(v)beta3 and alpha(v)beta6, and for the secretory protein uteroglobin under various experimental conditions. Cells grown in control media stained positively for the integrin monomers alpha(v) and beta1, the dimer alpha(v)beta3, and for UG. Oestradiol and sequential oestradiol/progesterone reversibly suppressed staining for the dimer alpha(v)beta3. Hormone treatment had no effect on the staining of the beta1 and alpha(v) monomers or UG. The alpha(v)beta6 dimer antibody did not stain under any experimental treatment conditions. These data indicate that expression of the integrin complex alpha(v)beta3 is reversibly suppressed by oestradiol in Ishikawa cells and that these cells may be a good model for studying hormone-driven molecular changes in endometrium.


Assuntos
Endométrio/metabolismo , Estradiol/farmacologia , Integrinas/biossíntese , Progesterona/farmacologia , Uteroglobina/biossíntese , Adenocarcinoma , Implantação do Embrião/fisiologia , Neoplasias do Endométrio , Feminino , Humanos , Células Tumorais Cultivadas
19.
Clin Cancer Res ; 3(12 Pt 1): 2295-300, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9815627

RESUMO

We have shown previously that the secretory protein uteroglobin (UG) is highly expressed in normal human prostate tissue but this expression is either lost or altered in human prostate cancer cell lines. Treatment of these cell lines with recombinant human UG inhibits their ability to invade human reconstituted basement membrane by up to 90%, implying that the loss of normal UG expression may be related to the invasive potential of prostate cancer. Because invasion represents a critical step in metastasis, the expression patterns of UG could provide a unique and relevant indicator of cancer progression. In this study, we present the immunohistochemical analyses of fresh frozen prostate tissues from surgical specimens taken from 50 patients. Eight slides per patient were analyzed for UG staining. Slides from 26 patients showed evidence of prostate cancer, whereas slides from the remaining 24 patients showed only benign glands. The results demonstrate UG immunoreactivity in normal prostate, benign prostatic hyperplasia, and prostatic atrophy; low but clearly positive expression in prostatic intraepithelial neoplasia; positive expression in cancerous glands of Gleason's pattern

Assuntos
Neoplasias da Próstata/patologia , Uteroglobina/análise , Adulto , Idoso , Atrofia , Carcinoma de Células de Transição/patologia , Carcinoma de Células de Transição/cirurgia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Próstata/patologia , Prostatectomia , Hiperplasia Prostática/patologia , Neoplasias da Próstata/cirurgia , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/cirurgia , Uteroglobina/genética
20.
Carcinogenesis ; 17(7): 1511-7, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8706257

RESUMO

Previous studies have shown that in vitro treatment of a synthetic double-stranded DNA template with chromium(III), or chromium(VI) in the presence of ascorbate, resulted in guanine-specific DNA polymerase arrests that correlated strongly with DNA-DNA cross-linking. In vivo chromium(VI) undergoes a more complicated intracellular cascade of reductive metabolism than is achievable in an in vitro model. Moreover, in living cells, DNA is highly packaged in the form of chromatin which may alter the accessibility of DNA to chromium. A repetitive primer-extension assay was employed to determine whether chromium forms polymerase-arresting lesions in vivo. Normal human lung fibroblasts treated with chromium(VI) exhibited adduct levels of 0.13-0.92 mmol Cr/mol DNA-nucleotides in the total genome (0.26-1.84 Cr adducts/Kbp DNA) and DNA interstrand cross-links. Genomic DNA was isolated and alphoid sequences (1-5% of the genome) were used as a substrate for repetitive primer extension using Taq polymerase. The results showed a dose-dependent, guanine-specific, replication termination, even at low doses resulting in greater than 90% survival. The same treatment resulted in dose-dependent suppression of thymidine incorporation into DNA immediately after treatment. Thymidine incorporation increased during the first 6 h after the 2-h exposure, probably related to the repair of the single strand breaks, but then returned to high suppression levels at 24 h. The chromate treatments inhibited cell growth by specific blocking of the progression of cells through S-phase of the cell cycle. The results confirmed our studies in cell-free systems and taken together they strongly indicate that guanine-guanine DNA interstrand cross-links induced by chromate in living cells is the lesion responsible for blocking DNA replication processivity.


Assuntos
Carcinógenos Ambientais/farmacologia , Ciclo Celular/efeitos dos fármacos , Cromo/farmacologia , DNA/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Inibidores da Síntese de Ácido Nucleico , Sequência de Bases , Linhagem Celular , Pré-Escolar , Cromo/metabolismo , Reagentes de Ligações Cruzadas , DNA/biossíntese , DNA/química , Dano ao DNA , Primers do DNA , Fibroblastos , Humanos , Cinética , Masculino , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Fase S
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA