Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Regul Toxicol Pharmacol ; 92: 213-219, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29233771

RESUMEN

ß-Galactosidase (EC 3.2.1.23) is a glycoside hydrolase that catalyzes the release of sugar monomers from ß-galactoside oligosaccharides via hydrolysis of glycosidic bonds, and has potential uses in the food industry. The safety of this enzyme and its production organism, Papiliotrema terrestris (P. terrestris), are described herein. P. terrestris was non-pathogenic upon intravenous (IV) administration of 2.4 × 108 cfu and oral administration of 1.3 × 109 cfu. Genotoxic results for the enzyme concentrate were negative in a bacterial reverse mutation test (Ames test) and chromosome aberration test in cultured Chinese hamster lung fibroblast (CHL/IU) cells. In a 13-week oral gavage study in Sprague-Dawley rats, no adverse effects were observed in any of the tested group and a No Observed Adverse Effects Level (NOAEL) of 2000 mg/kg bw/day [total organic solids (TOS) 1800 mg/kg bw/day)] was established, which was the highest dosage tested. Allergenicity sequence analysis revealed no evidence suggesting that ß-galactosidase is an allergen. The data presented in this study support the conclusion that ß-galactosidase produced from P. terrestris is safe for use in food production.


Asunto(s)
Levaduras/metabolismo , beta-Galactosidasa/toxicidad , Administración Oral , Animales , Células Cultivadas , Cricetulus , Femenino , Masculino , Ratones Endogámicos ICR , Pruebas de Mutagenicidad/métodos , Nivel sin Efectos Adversos Observados , Ratas , Ratas Sprague-Dawley
2.
Food Chem Toxicol ; 59: 541-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23831195

RESUMEN

Thermolysin is a zinc metalloprotease that has potential uses in the food industry. The safety of thermolysin has not been demonstrated before, and therefore a series of standard toxicological tests to assess its potential toxicity was undertaken. The thermolysin used in this study was derived from the thermophilic bacterium Geobacillus stearothermophilus, which had undergone chemical mutagenesis to generate strains with increased thermolysin production. Acute toxicity studies in rats and mice showed that thermolysin powder is not acutely toxic with an oral LD50 of more than 18,000 mg/kg (2520 mg/kg thermolysin protein) in rats and more than 24,000 mg/kg (3360 mg/kg protein) in mice. Subchronic feeding studies in rats for 91 days at doses up to 1000 mg/kg (390 mg/kg protein) revealed no significant differences between treated and non-treated groups and a No Observed Effect Level (NOEL) of 1000 mg/kg (390 mg/kg protein) per day was established. Results from genotoxicity tests such as in vitro chromosomal aberration assay and in vivo mouse micronucleus were negative. Allergenicity sequence analysis revealed no evidence suggesting that thermolysin is an allergen. The data presented in this study support the conclusion that thermolysin is safe for use in food production.


Asunto(s)
Proteínas Bacterianas/efectos adversos , Aditivos Alimentarios/efectos adversos , Geobacillus stearothermophilus/enzimología , Termolisina/efectos adversos , Alérgenos/administración & dosificación , Alérgenos/efectos adversos , Animales , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/efectos adversos , Proteínas Bacterianas/administración & dosificación , Línea Celular , Cricetinae , Cricetulus , Femenino , Aditivos Alimentarios/administración & dosificación , Geobacillus stearothermophilus/inmunología , Masculino , Ratones , Ratones Endogámicos , Pruebas de Micronúcleos , Pruebas de Mutagenicidad , Nivel sin Efectos Adversos Observados , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Termolisina/administración & dosificación , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Subcrónica
3.
Biochim Biophys Acta ; 1833(9): 2083-91, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23639288

RESUMEN

p27 is a cyclin-dependent kinase (CDK) inhibitor that suppresses a cell's transition from G0 to S phase, therefore acting as a tumor suppressor. Our most recent studies demonstrate that upon arsenite exposure, p27 suppresses Hsp27 and Hsp70 expressions through the JNK2/c-Jun- and HSF-1-dependent pathways, suggesting a novel molecular mechanism underlying the tumor suppressive function of p27 in a CDK-independent manner. We found that p27-deficiency (p27-/-) resulted in the elevation of cyclooxygenase-2 (COX-2) expression at transcriptional level, whereas the introduction of p27 brought back COX-2 expression to a level similar to that of p27+/+ cells, suggesting that p27 exhibits an inhibitory effect on COX-2 expression. Further studies identified that p27 inhibition of COX-2 expression was specifically due to phosphorylation of transcription factor cAMP response element binding (CREB) phosphorylation mediated by p38ß and p38δ. These results demonstrate a novel mechanism underlying tumor suppression effect of p27 and will contribute to the understanding of the overall mechanism of p27 tumor suppression in a CDK-independent manner.


Asunto(s)
Arsenitos/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclooxigenasa 2/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Teratógenos/farmacología , Animales , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Ciclooxigenasa 2/genética , Regulación Enzimológica de la Expresión Génica/genética , Ratones , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
4.
Carcinogenesis ; 30(7): 1243-51, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19376846

RESUMEN

In the present study, we examined the effects of CoCl(2) on multiple histone modifications at the global level. We found that in both human lung carcinoma A549 cells and human bronchial epithelial Beas-2B cells, exposure to CoCl(2) (>/=200 muM) for 24 h increased H3K4me3, H3K9me2, H3K9me3, H3K27me3, H3K36me3, uH2A and uH2B but decreased acetylation at histone H4 (AcH4). Further investigation demonstrated that in A549 cells, the increase in H3K4me3 and H3K27me3 by cobalt ions exposure was probably through enhancing histone methylation processes, as methionine-deficient medium blocked the induction of H3K4me3 and H3K27me3 by cobalt ions, whereas cobalt ions increased H3K9me3 and H3K36me3 by directly inhibiting JMJD2A demethylase activity in vitro, which was probably due to the competition of cobalt ions with iron for binding to the active site of JMJD2A. Furthermore, in vitro ubiquitination and deubiquitination assays revealed that the cobalt-induced histone H2A and H2B ubiquitination is the result of inhibition of deubiquitinating enzyme activity. Microarray data showed that exposed to 200 microM of CoCl(2) for 24 h, A549 cells not only increased but also decreased expression of hundreds of genes involved in different cellular functions, including tumorigenesis. This study is the first to demonstrate that cobalt ions altered epigenetic homeostasis in cells. It also sheds light on the possible mechanisms involved in cobalt-induced alteration of histone modifications, which may lead to altered programs of gene expression and carcinogenesis since cobalt at higher concentrations is a known carcinogen.


Asunto(s)
Cobalto/farmacología , Histonas/metabolismo , Acetilación , Línea Celular Tumoral , Epigénesis Genética , Humanos , Hierro/metabolismo , Histona Demetilasas con Dominio de Jumonji , Metilación , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidorreductasas N-Desmetilantes/metabolismo , Ubiquitinación
5.
Carcinogenesis ; 29(6): 1276-81, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18375956

RESUMEN

Nickel (Ni) is a known carcinogen, although the mechanism of its carcinogenicity is not clear. Here, we provide evidence that Ni can induce phosphorylation of histone H3 at its serine 10 residue in a c-jun N-terminal kinase (JNK)/stress-activated protein kinase (SAPK)-dependent manner. Ni induces the phosphorylation of JNK, with no effect on the phosphorylation states of the extracellular signal-regulated kinase (ERK) or p38 mitogen-activated protein kinases. An inhibitor of JNK eliminated the Ni-initiated JNK-mediated induction of histone H3 phosphorylation at serine 10, whereas inhibitors specific for ERK or p38 kinases had no effect on the phosphorylation levels of histone H3 at serine 10 (P-H3S10) in Ni-treated cells. A complete loss of Ni ion-induced phosphorylation of H3S10 was observed when JNK was specifically knocked down with RNAi. These results are the first to show the specific JNK-mediated phosphorylation of histone H3 at its serine 10 residue. We show that addition of Ni to an in vitro P-H3S10 dephosphorylation reaction does not change the loss of phosphorylation in the reaction, supporting the notion that Ni causes H3S10 phosphorylation via the JNK/SAPK pathway. It is likely that modification of H3S10 is one of a growing number of epigenetic changes believed to be involved in the carcinogenesis caused by Ni.


Asunto(s)
Histonas/efectos de los fármacos , MAP Quinasa Quinasa 4/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Níquel/toxicidad , Serina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Histonas/genética , Humanos , MAP Quinasa Quinasa 4/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , ARN Interferente Pequeño , Serina/genética , Transfección
6.
Toxicol Appl Pharmacol ; 228(2): 190-9, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18279901

RESUMEN

Nickel (Ni) compounds are known carcinogens but underlying mechanisms are not clear. Epigenetic changes are likely to play an important role in nickel ion carcinogenesis. Previous studies have shown epigenetic effects of nickel ions, including the loss of histone acetylation and a pronounced increase in dimethylated H3K9 in nickel-exposed cells. In this study, we demonstrated that both water-soluble and insoluble nickel compounds induce histone ubiquitination (uH2A and uH2B) in a variety of cell lines. Investigations of the mechanism by which nickel increases histone ubiquitination in cells reveal that nickel does not affect cellular levels of the substrates of this modification, i.e., ubiquitin, histones, and other non-histone ubiquitinated proteins. In vitro ubiquitination and deubiquitination assays have been developed to further investigate possible effects of nickel on enzymes responsible for histone ubiquitination. Results from the in vitro assays demonstrate that the presence of nickel did not affect the levels of ubiquitinated histones in the ubiquitinating assay. Instead, the addition of nickel significantly prevents loss of uH2A and uH2B in the deubiquitinating assay, suggesting that nickel-induced histone ubiquitination is the result of inhibition of (a) putative deubiquitinating enzyme(s). Additional supporting evidence comes from the comparison of the response to nickel ions with a known deubiquitinating enzyme inhibitor, iodoacetamide (IAA). This study is the first to demonstrate such effects of nickel ions on histone ubiquitination. It also sheds light on the possible mechanisms involved in altering the steady state of this modification. The study provides further evidence that supports the notion that nickel ions alter epigenetic homeostasis in cells, which may lead to altered programs of gene expression and carcinogenesis.


Asunto(s)
Histonas/metabolismo , Leupeptinas/farmacología , Níquel/farmacología , Ubiquitinación/efectos de los fármacos , Adenosina Trifosfato/farmacología , Western Blotting , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/farmacología , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Yodoacetamida/farmacología , Modelos Biológicos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Factores de Tiempo , Ubiquitinas/metabolismo
7.
Carcinogenesis ; 29(1): 2-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17916902

RESUMEN

N-myc downstream-regulated gene 1 (NDRG1) is an intracellular protein that is induced under a wide variety of stress and cell growth-regulatory conditions. NDRG1 is up-regulated by cell differentiation signals in various cancer cell lines and suppresses tumor metastasis. Despite its specific role in the molecular cause of Charcot-Marie-Tooth type 4D disease, there has been more interest in the gene as a marker of tumor progression and enhancer of cellular differentiation. Because it is strongly up-regulated under hypoxic conditions, and this condition is prevalent in solid tumors, its regulation is somewhat complex, governed by hypoxia-inducible factor 1 alpha (HIF-1alpha)- and p53-dependent pathways, as well as its namesake, neuroblastoma-derived myelocytomatosis, and probably many other factors, at the transcriptional and translational levels, and through mRNA stability. We survey the data for clues to the NDRG1 gene's mechanism and for indications that the NDRG1 gene may be an efficient diagnostic tool and therapy in many types of cancers.


Asunto(s)
Proteínas de Ciclo Celular/genética , División Celular/genética , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias/patología , Progresión de la Enfermedad , Humanos , Neoplasias/genética
8.
Toxicol Appl Pharmacol ; 219(1): 18-23, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17239912

RESUMEN

The carcinogenic activity of various nickel (Ni) compounds is likely dependent upon their ability to enter cells and elevate intracellular levels of Ni ions. Water-insoluble Ni compounds such as NiS and Ni(3)S(2) were shown in vitro to enter cells by phagocytosis and potently induce tumors in experimental animals at the site of exposure. These water-insoluble nickel compounds are generally considered to be more potent carcinogens than the water-soluble forms. However, recent in vitro studies have shown similar effects for insoluble and soluble Ni compounds. Using a dye that fluoresces when intracellular Ni ion binds to it, we showed that both soluble and insoluble Ni compounds were able to elevate the levels of Ni ions in the cytoplasmic and nuclear compartments. However, when the source of Ni ions was removed from the culture dish, the intracellular Ni ions derived from soluble Ni compound were lost from the cells at a significantly faster rate than those derived from the insoluble Ni compound. Within 10 h after NiCl(2) removal from the culture medium, Ni ions disappeared from the nucleus and were not detected in the cells by 16 h, while insoluble Ni(3)S(2) yielded Ni ions that persisted in the nucleus after 16 h and were detected in the cytoplasm even after 24 h following Ni removal. These effects are discussed in terms of whole body exposure to water-soluble and -insoluble Ni compounds and consistency with animal carcinogenicity studies.


Asunto(s)
Níquel/metabolismo , Bronquios/citología , Bronquios/metabolismo , Línea Celular Tumoral , Células Cultivadas , Células Epiteliales/metabolismo , Colorantes Fluorescentes , Humanos , Pulmón/citología , Pulmón/metabolismo
9.
Mol Pharmacol ; 70(5): 1469-80, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16887934

RESUMEN

Adaptation to low oxygen tension (hypoxia) in cells and tissues leads to the transcriptional induction of a series of genes that participate in angiogenesis, iron metabolism, glucose metabolism, and cell proliferation/survival. The primary factor mediating this response is the hypoxia-inducible factor-1 (HIF-1), an oxygen-sensitive transcriptional activator. HIF-1 consists of a constitutively expressed subunit HIF-1beta and an oxygen-regulated subunit HIF-1alpha (or its paralogs HIF-2alpha and HIF-3alpha). The stability and activity of the alpha subunit of HIF are regulated by its post-translational modifications such as hydroxylation, ubiquitination, acetylation, and phosphorylation. In normoxia, hydroxylation of two proline residues and acetylation of a lysine residue at the oxygen-dependent degradation domain (ODDD) of HIF-1alpha trigger its association with pVHL E3 ligase complex, leading to HIF-1alpha degradation via ubiquitin-proteasome pathway. In hypoxia, the HIF-1alpha subunit becomes stable and interacts with coactivators such as cAMP response element-binding protein binding protein/p300 and regulates the expression of target genes. Overexpression of HIF-1 has been found in various cancers, and targeting HIF-1 could represent a novel approach to cancer therapy.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/química , Factor 1 Inducible por Hipoxia/genética
10.
Mol Cell Biol ; 26(10): 3728-37, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648469

RESUMEN

We have previously reported that carcinogenic nickel compounds decreased global histone H4 acetylation and silenced the gpt transgene in G12 Chinese hamster cells. However, the nature of this silencing is still not clear. Here, we report that nickel ion exposure increases global H3K9 mono- and dimethylation, both of which are critical marks for DNA methylation and long-term gene silencing. In contrast to the up-regulation of global H3K9 dimethylation, nickel ions decreased the expression and activity of histone H3K9 specific methyltransferase G9a. Further investigation demonstrated that nickel ions interfered with the removal of histone methylation in vivo and directly decreased the activity of a Fe(II)-2-oxoglutarate-dependent histone H3K9 demethylase in nuclear extract in vitro. These results are the first to show a histone H3K9 demethylase activity dependent on both iron and 2-oxoglutarate. Exposure to nickel ions also increased H3K9 dimethylation at the gpt locus in G12 cells and repressed the expression of the gpt transgene. An extended nickel ion exposure led to increased frequency of the gpt transgene silencing, which was readily reversed by treatment with DNA-demethylating agent 5-aza-2'-deoxycytidine. Collectively, our data strongly indicate that nickel ions induce transgene silencing by increasing histone H3K9 dimethylation, and this effect is mediated by the inhibition of H3K9 demethylation.


Asunto(s)
Silenciador del Gen/efectos de los fármacos , Histonas/química , Histonas/metabolismo , Lisina/química , Níquel/farmacología , Transgenes/efectos de los fármacos , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Western Blotting , Carcinoma/patología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Metilación de ADN/efectos de los fármacos , Decitabina , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Cinética , Neoplasias Pulmonares/patología , Metiltransferasas/metabolismo , Ratones , Oligoelementos/farmacología
11.
Carcinogenesis ; 27(7): 1481-8, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16522665

RESUMEN

Although it has been well established that insoluble nickel compounds are potent carcinogens and soluble nickel compounds are less potent, the mechanisms remain unclear. Nickel compounds are weakly mutagenic, but cause epigenetic effects in cells. Previous studies have shown that insoluble nickel compounds enter cells by phagocytosis and silence gene expression, but the entry of soluble nickel compounds and their effects on gene silencing have not been well studied. Here, we have demonstrated, using a dye that fluoresces when nickel ions bind, that soluble nickel compounds were taken up by cells. Nickel ions localized initially in the cytoplasm, but later entered the nucleus and eventually silenced a transgene. In addition, we described three major changes in histone modification of cells exposed to soluble nickel compounds: (i) loss of acetylation of H2A, H2B, H3 and H4; (ii) increases of H3K9 dimethylation; and (iii) substantial increases of the ubiquitination of H2A and H2B. These effects were observed at nickel exposure conditions that had minimum effects on cell cytotoxicity. Moreover, we demonstrated that nickel-induced transgene silencing was associated with similar changes of histone modifications in their nuclesomes. This study is the first to show that nickel compounds increase histone ubiquitination in cells. These new findings will further our understanding of the epigenetic mechanisms of nickel-mediated carcinogenesis.


Asunto(s)
Silenciador del Gen/efectos de los fármacos , Histonas/efectos de los fármacos , Níquel/metabolismo , Níquel/farmacología , Transgenes/efectos de los fármacos , Animales , Western Blotting , Línea Celular , Cricetinae , Proteínas de Escherichia coli , Humanos , Pentosiltransferasa , Reacción en Cadena de la Polimerasa , Proteínas/genética
12.
Am J Physiol Lung Cell Mol Physiol ; 291(2): L257-64, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16489119

RESUMEN

Occupational exposure to crystalline silica has been associated with progressive pulmonary silicosis and lung cancer, but the underlying molecular mechanisms are not well understood. Previous studies have shown that crystalline silica exposure can generate reactive oxygen species (ROS) and induce the expression of the inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) in cells. TNF-alpha is believed to be critical in the development of silica-related diseases. Thus it will be of significance to understand the mechanisms of TNF-alpha induction by silica exposure. Given the fact that the transcription factor nuclear factor of activated T cells (NFAT) plays an important role in the regulation of TNF-alpha and can also be activated by ROS, in this study we investigated the potential role of ROS in silica-induced NFAT activity as well as TNF-alpha expression in Cl41 cells. The results showed that exposure of cells to silica led to NFAT transactivation and TNF-alpha induction, where superoxide anion radical (O(2)(-).), but not H(2)O(2), was involved. The knockdown of NFAT3 by its specific small interfering RNA significantly attenuated the silica-induced TNF-alpha transcription. This study demonstrated that silica was able to activate NFAT in an O(2)(-).-dependent manner, which was required for TNF-alpha induction.


Asunto(s)
Factores de Transcripción NFATC/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Dióxido de Silicio/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Humanos , Ratones , Factores de Transcripción NFATC/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Dióxido de Silicio/química , Activación Transcripcional
13.
Mutat Res ; 592(1-2): 79-88, 2005 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-16009382

RESUMEN

Both water soluble and insoluble nickel compounds have been implicated in the etiology of human lung and nasal cancers. Water insoluble nickel compounds have been shown to enter cells by phagocytosis and are contained in cytoplasmic vacuoles, which are acidified thus accelerating the dissolution of soluble nickel from the particles. Using Newport Green, a dye that fluoresces when ionic nickel is bound, we have shown that following exposure (48-72 h) of human lung (A549) cells to NiS particles, most of the nickel is contained in the nucleus, while cells exposed to soluble NiCl2 exhibit most of the ions localized in the cytoplasm. This effect is consistent with previously published reports showing that short-term exposure of cells to crystalline nickel particles (1-3 days) is able to epigenetically silence target genes placed near heterochromatin, while similar short-term exposure to soluble nickel compounds are not able to induce silencing of genes placed near heterochromatin. However, a 3 week exposure of cells to soluble NiCl2 is also able to induce gene silencing. A similar effect was found in yeast cells where nickel was able to silence the URA-3 gene placed near (1.3 kb) a telomere silencing element, but not when the gene was placed farther away from the silencing element (2.0 kb). In addition to epigenetic effects, nickel compounds activate hypoxia signaling pathways. The mechanism of this effect involves the ability of either soluble or insoluble nickel compounds to block iron uptake leading to cellular iron depletion, directly affect iron containing enzymes, or both. This results in the inhibition of a variety of iron-dependent enzymes, such as aconitase and the HIF proline hydroxylases (PHD1-3). The inhibition of the HIF proline hydroxylases stabilizes the HIF protein and activates hypoxic signaling. Additional studies have shown that nickel and hypoxia decrease histone acetylation and increase the methylation of H3 lysine 9. These events are involved in gene silencing and hypoxia can also cause these effects in human cells. It is hypothesised that the state of hypoxia either by low oxygen tension or as a result of agents that signal hypoxia under normal oxygen tension (iron chelation, nickel and cobalt) results in low levels of acetyl CoA, which is a substrate for histone and other protein acetylation. This effect may in part be responsible for the gene silencing following nickel exposure and during hypoxia.


Asunto(s)
Carcinógenos , Hipoxia de la Célula/efectos de los fármacos , Níquel/toxicidad , Línea Celular , Daño del ADN , Silenciador del Gen , Humanos , Níquel/farmacocinética , Distribución Tisular
14.
Int J Environ Res Public Health ; 2(1): 10-3, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16705796

RESUMEN

Although nickel and cobalt compounds have been known to cause induction of the transcription factor hypoxia-inducible factor 1 (HIF-1) and activation of a battery of hypoxia-inducible genes in the cell, the molecular mechanisms of this induction remain unclear. The post-translational modification of HIF-1a, the oxygen-sensitive subunit of HIF-1, regulates stabilization, nuclear translocation, DNA binding activity, and transcriptional activity of the protein. Among the enzymes regulating the post-translational modification of HIF-la, the factor inhibiting HIF-1 (FIH-1) hydroxylates the protein at asparagine 803, suppressing the interaction of HIF-1a with transcription coactivators p300/CBP and reducing the transcriptional activity of the protein. ARD-1, the acetyltransferase, acetylates HIF-1a at lysine 532, which enhances the interaction of HIF-1a with pVHL. Therefore, FIH-1 and ARD-1 negatively regulate the transcriptional activity and the stability of HIF-1a. We examined the mRNA levels of FIH-l and ARD-1 genes after exposure nickel (II) or cobalt (II) to the cell and found that both genes were down-regulated by the chemical treatment, which may lead to reduced levels of both proteins and result in increased level of HIF-1 a and its transcriptional activity.


Asunto(s)
Acetiltransferasas/metabolismo , Cobalto/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Níquel/toxicidad , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Acetiltransferasas/genética , Adenocarcinoma , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares , Oxigenasas de Función Mixta , Acetiltransferasa A N-Terminal , Acetiltransferasa E N-Terminal , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética
15.
Mol Carcinog ; 40(2): 104-15, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15170815

RESUMEN

Polycyclic aromatic hydrocarbons (PAHs) and their derivatives, such as benzo[a]pyrene (B[a]P), (+/-)-anti-benzo[a]pyrene-7,8-diol-9,10-epoxide (B[a]PDE), and 5-methylchrysene-1,2-diol-3,4-epoxide (5-MCDE), are complete carcinogens. However, the tumor promotion effects of PAHs remain unclear. We therefore investigated the possible activation of activator protein-1 (AP-1) and nuclear factor-kappaB (NFkappaB) in mouse epidermal Cl41 cells after different PAHs treatments, including B[a]P, B[a]PDE, chrysene-1,2-diol-3,4-epoxid (CDE), and 5-MCDE. The results showed that B[a]PDE and 5-MCDE were able to activate AP-1 and NF-kappaB, whereas B[a]P showed only marginal effect on AP-1 activation, and B[a]P and CDE had no effect on NF-kappaB activation. Treatment with either B[a]PDE or 5-MCDE also resulted in mitogen-activated protein kinases (MAPKs) activation as well as inhibitory subunit kappa-B (IkappaBalpha) phosphorylation and degradation, whereas B[a]P and CDE had no effect. Pretreatment with PD98059, a specific inhibitor for extracellular signal-regulated protein kinases (ERKs) upstream kinase MEK1/2, or SB202190, a p38 kinase inhibitor, resulted in a dramatic inhibition of B[a]PDE-induced AP-1 transactivation. In addition, B[a]PDE-induced AP-1 activation was also inhibited by overexpressing a dominant negative mutant of JNK1 in the cells. All these suggest ERKs, c-jun N-terminal kinases (JNKs), and p38 kinase signal transduction pathways are required for AP-1 induction by B[a]PDE. Taken together, B[a]PDE and 5-MCDE are the active compounds of PAHs to initiate signaling pathways. Considering the important roles of AP-1 and NF-kappaB in tumor promotion, we speculated the activation of AP-1 and NF-kappaB by B[a]PDE and 5-MCDE may involve in their or their parent compounds' tumor promotion effects. This study may help in better understanding the tumor promotion effects of PAHs.


Asunto(s)
Carcinógenos/toxicidad , Epidermis/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Factor de Transcripción AP-1/efectos de los fármacos , 7,8-Dihidro-7,8-dihidroxibenzo(a)pireno 9,10-óxido/toxicidad , Animales , Benzo(a)pireno/toxicidad , Células Cultivadas , Crisenos/toxicidad , Inhibidores Enzimáticos/farmacología , Células Epidérmicas , Epidermis/metabolismo , Flavonoides/farmacología , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Piridinas/farmacología , Transducción de Señal , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional/efectos de los fármacos
16.
Mol Cell Biochem ; 255(1-2): 57-66, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14971646

RESUMEN

Arsenic is a metalloid compound that is widely distributed in the environment. Human exposure of this compound has been associated with increased cancer incidence. Although the exact mechanisms remain to be investigated, numerous carcinogenic pathways have been proposed. Potential carcinogenic actions for arsenic include oxidative stress, genotoxic damage, DNA repair inhibition, epigenetic events, and activation of certain signal transduction pathways leading to abberrant gene expression. In this article, we summarize current knowledge on the molecular mechanisms of arsenic carcinogenesis with an emphasis on ROS and signal transduction pathways.


Asunto(s)
Arsénico/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Contaminantes Ambientales/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Arsénico/metabolismo , Cricetinae , Citocinas/metabolismo , Daño del ADN/fisiología , Reparación del ADN/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Mutágenos/metabolismo , Oncogenes/fisiología , Estrés Oxidativo/fisiología , Proteínas Quinasas/metabolismo , Conejos , Ratas , Factores de Transcripción/metabolismo
17.
Cancer Res ; 62(20): 5689-97, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12384526

RESUMEN

The p70 S6 kinase (p70(S6k)) is a Ser/Thr kinase that plays an important role in cell growth, transformation, and the transition of the cell cycle in mammalian cells. Because UV radiation has been reported to induce activation of p70(S6k), which is believed to play some role in the carcinogenic effects of sun exposure, the present study investigated the signaling pathways involved in this activation induced by UV radiation in mouse epidermal JB6 Cl41 cells. Exposure of cells to UV radiation led to marked increases in p70(S6k) activity and phosphorylation at Thr(389) and Thr(421)/Ser(424). UV radiation also generated reactive oxygen species as measured by electron spin resonance and by H(2)O(2) and O( minus sign, dot below )(2) fluorescence staining assays in JB6 Cl 41 cells. The scavenging of UV-generated H(2)O(2) by N-acety-L-cyteine (a general antioxidant) or catalase (a specific H(2)O(2) scavenger) inhibited p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424), whereas pretreatment of cells with sodium formate (an.OH radical scavenger) or superoxide dismutase (an O( minus sign, dot below )(2) radical scavenger) did not show any inhibitory effects. Importantly, UV-induced increases in p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424) were dramatically inhibited by pretreatment of cells with rapamycin, LY294002, or PD98059, whereas overexpression of dominant-negative mutants of PKClambda/iota and Akt1 did not inhibit p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424). These results demonstrated that H(2)O(2), phosphatidylinositol 3-kinase, and mammalian target of rapamycin were important players for UV-induced p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424), whereas Akt and atypical protein kinase C were not involved in this activation. The role of H(2)O(2) in p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424) was further supported by the findings that treatment of cells with H(2)O(2) also caused p70(S6k) phosphorylation at Thr(389) and Thr(421)/Ser(424).


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Células Cultivadas , Espectroscopía de Resonancia por Spin del Electrón , Inducción Enzimática/efectos de la radiación , Ratones , Fosforilación/efectos de la radiación , Proteínas Proto-Oncogénicas c-akt , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/biosíntesis , Serina/metabolismo , Transducción de Señal/efectos de la radiación , Piel/citología , Piel/metabolismo , Piel/efectos de la radiación , Serina-Treonina Quinasas TOR , Treonina/metabolismo , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...