Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Signal ; 8(370): ra32, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25829446

RESUMEN

The tumor suppressor PTEN restrains cell migration and invasion by a mechanism that is independent of inhibition of the PI3K pathway and decreased activation of the kinase AKT. PREX2, a widely distributed GEF that activates the GTPase RAC1, binds to and inhibits PTEN. We used mouse embryonic fibroblasts and breast cancer cell lines to show that PTEN suppresses cell migration and invasion by blocking PREX2 activity. In addition to metabolizing the phosphoinositide PIP3, PTEN inhibited PREX2-induced invasion by a mechanism that required the tail domain of PTEN, but not its lipid phosphatase activity. Fluorescent nucleotide exchange assays revealed that PTEN inhibited the GEF activity of PREX2 toward RAC1. PREX2 is a frequently mutated GEF in cancer, and examination of human tumor data showed that PREX2 mutation was associated with high PTEN expression. Therefore, we tested whether cancer-derived somatic PREX2 mutants, which accelerate tumor formation of immortalized melanocytes, were inhibited by PTEN. The three stably expressed, somatic PREX2 cancer mutants that we tested were resistant to PTEN-mediated inhibition of invasion but retained the ability to inhibit the lipid phosphatase activity of PTEN. In vitro analysis showed that PTEN did not block the GEF activity of two PREX2 cancer mutants and had a reduced binding affinity for the third. Thus, PTEN antagonized migration and invasion by restraining PREX2 GEF activity, and PREX2 mutants are likely selected in cancer to escape PTEN-mediated inhibition of invasion.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Invasividad Neoplásica/prevención & control , Fosfohidrolasa PTEN/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Vectores Genéticos , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Immunoblotting , Inmunoprecipitación , Lentivirus , Ratones , Fosfohidrolasa PTEN/genética , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Estadísticas no Paramétricas
2.
Trends Biochem Sci ; 39(4): 183-90, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24656806

RESUMEN

Phosphatase and tensin homolog deleted on chromosome ten (PTEN) is a phosphatase that is frequently altered in cancer. PTEN has phosphatase-dependent and -independent roles, and genetic alterations in PTEN lead to deregulation of protein synthesis, the cell cycle, migration, growth, DNA repair, and survival signaling. PTEN localization, stability, conformation, and phosphatase activity are controlled by an array of protein-protein interactions and post-translational modifications. Thus, PTEN-interacting and -modifying proteins have profound effects on the tumor suppressive functions of PTEN. Moreover, recent studies identified mechanisms by which PTEN can exit cells, via either exosomal export or secretion, and act on neighboring cells. This review focuses on modes of PTEN protein regulation and ways in which perturbations in this regulation may lead to disease.


Asunto(s)
Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Animales , Ciclo Celular , Regulación de la Expresión Génica , Inestabilidad Genómica , Humanos , Fosfohidrolasa PTEN/química , Procesamiento Proteico-Postraduccional
3.
Proc Natl Acad Sci U S A ; 111(1): 155-60, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24367090

RESUMEN

Insulin activation of phosphoinositide 3-kinase (PI3K) signaling regulates glucose homeostasis through the production of phosphatidylinositol 3,4,5-trisphosphate (PIP3). The dual-specificity phosphatase and tensin homolog deleted on chromosome 10 (PTEN) blocks PI3K signaling by dephosphorylating PIP3, and is inhibited through its interaction with phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchanger 2 (P-REX2). The mechanism of inhibition and its physiological significance are not known. Here, we report that P-REX2 interacts with PTEN via two interfaces. The pleckstrin homology (PH) domain of P-REX2 inhibits PTEN by interacting with the catalytic region of PTEN, and the inositol polyphosphate 4-phosphatase domain of P-REX2 provides high-affinity binding to the postsynaptic density-95/Discs large/zona occludens-1-binding domain of PTEN. P-REX2 inhibition of PTEN requires C-terminal phosphorylation of PTEN to release the P-REX2 PH domain from its neighboring diffuse B-cell lymphoma homology domain. Consistent with its function as a PTEN inhibitor, deletion of Prex2 in fibroblasts and mice results in increased Pten activity and decreased insulin signaling in liver and adipose tissue. Prex2 deletion also leads to reduced glucose uptake and insulin resistance. In human adipose tissue, P-REX2 protein expression is decreased and PTEN activity is increased in insulin-resistant human subjects. Taken together, these results indicate a functional role for P-REX2 PH-domain-mediated inhibition of PTEN in regulating insulin sensitivity and glucose homeostasis and suggest that loss of P-REX2 expression may cause insulin resistance.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Regulación Enzimológica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Resistencia a la Insulina , Fosfohidrolasa PTEN/antagonistas & inhibidores , Animales , Sitios de Unión , Proteínas Sanguíneas/química , Dominio Catalítico , Proliferación Celular , Fibroblastos/metabolismo , Glucosa/metabolismo , Células HEK293 , Homeostasis , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/química , Fosforilación , Unión Proteica
4.
Toxicol Appl Pharmacol ; 247(2): 83-90, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20600213

RESUMEN

Phytoestrogens are plant compounds that structurally mimic the endogenous estrogen 17beta-estradiol (E(2)). Despite intense investigation, the net effect of phytoestrogen exposure on the breast remains unclear. The objective of the current study was to examine the effects of quercetin on E(2)-induced breast cancer in vivo. Female ACI rats were given quercetin (2.5 g/kg food) for 8 months. Animals were monitored weekly for palpable tumors, and at the end of the experiment, rats were euthanized, breast tumor and different tissues excised so that they could be examined for histopathologic changes, estrogen metabolic activity and oxidant stress. Quercetin alone did not induce mammary tumors in female ACI rats. However, in rats implanted with E(2) pellets, co-exposure to quercetin did not protect rats from E(2)-induced breast tumor development with 100% of the animals developing breast tumors within 8 months of treatment. No changes in serum quercetin levels were observed in quercetin and quercetin+E(2)-treated groups at the end of the experiment. Tumor latency was significantly decreased among rats from the quercetin+E(2) group relative to those in the E(2) group. Catechol-O-methyltransferase (COMT) activity was significantly downregulated in quercetin-exposed mammary tissue. Analysis of 8-isoprostane F(2alpha) (8-iso-PGF(2alpha)) levels as a marker of oxidant stress showed that quercetin did not decrease E(2)-induced oxidant stress. These results indicate that quercetin (2.5 g/kg food) does not confer protection against breast cancer, does not inhibit E(2)-induced oxidant stress and may exacerbate breast carcinogenesis in E(2)-treated ACI rats. Inhibition of COMT activity by quercetin may expose breast cells chronically to E(2) and catechol estrogens. This would permit longer exposure times to the carcinogenic metabolites of E(2) and chronic exposure to oxidant stress as a result of metabolic redox cycling to estrogen metabolites, and thus quercetin may exacerbate E(2)-induced breast tumors in female ACI rats.


Asunto(s)
Estrógenos/toxicidad , Neoplasias Mamarias Experimentales/inducido químicamente , Fitoestrógenos/toxicidad , Quercetina/toxicidad , Animales , Catecol O-Metiltransferasa/metabolismo , Inhibidores de Catecol O-Metiltransferasa , Dinoprost/análogos & derivados , Dinoprost/biosíntesis , Sinergismo Farmacológico , Femenino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Fitoestrógenos/administración & dosificación , Quercetina/administración & dosificación , Quercetina/metabolismo , Ratas , Ratas Endogámicas ACI
5.
J Biochem Mol Toxicol ; 23(3): 202-11, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19526586

RESUMEN

Exposure to estrogens is suggested to be a risk factor in human breast cancer development. The mechanisms underlying estrogen-induced cancer have not been fully elucidated. Both estrogen receptor (ER)-mediated proliferative processes and ER-independent generation of oxidative stress are suggested to play important roles in estrogen-induced breast carcinogenesis. In the current study, we investigated the role of oxidative stress in breast carcinogenesis using the ACI rat model of mammary tumorigenesis. Female ACI rats were treated with 17beta-estradiol (E(2)), butylated hydroxyanisole (BHA), or a combination of E(2) + BHA for up to 240 days. Cotreatment of rats with E(2) + BHA reduced estrogen-induced breast tumor development with tumor incidence of 24%, a significant decrease relative to E(2) where tumor incidence was 82%. Proliferative changes in the breast tissue of E(2) + BHA-treated animals were similar to those observed in E(2)-treated animals. Tissue levels of 8-isoprostane, a marker of oxidant stress, as well as the activities of antioxidant enzymes including glutathione peroxidase, superoxide dismutase, and catalase were quantified in the breast tissues of rats treated with E(2) + BHA and compared to activity levels found in E(2)-treated animals and respective age-matched controls. Cotreatment with BHA inhibited E(2)-mediated increases in 8-isoprostane levels as well as activities of antioxidant enzymes. In summary, these data suggest that estrogen-mediated oxidant stress plays a critical role in the development of estrogen-dependent breast cancers and BHA inhibits E(2)-dependent breast carcinogenesis by decreasing oxidant stress.


Asunto(s)
Antioxidantes/metabolismo , Antioxidantes/farmacología , Hidroxianisol Butilado/farmacología , Estradiol/efectos adversos , Estrógenos/efectos adversos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Animales , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Humanos , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas ACI
6.
Carcinogenesis ; 30(7): 1202-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19406931

RESUMEN

The mechanisms underlying the pathogenesis of estrogen-induced breast carcinogenesis remain unclear. The present study investigated the roles of estrogen metabolism and oxidative stress in estrogen-mediated mammary carcinogenesis in vivo. Female August Copenhagen Irish (ACI) rats were treated with 17beta-estradiol (E(2)), the antioxidant vitamin C, the estrogen metabolic inhibitor alpha-naphthoflavone (ANF), or cotreated with E(2) + vitamin C or E(2) + ANF for up to 8 months. E(2) (3 mg) was administered as an subcutaneous implant, ANF was given via diet (0.2%) and vitamin C (1%) was added to drinking water. At necropsy, breast tumor incidence in the E(2), E(2) + vitamin C and E(2) + ANF groups was 82, 29 and 0%, respectively. Vitamin C and ANF attenuated E(2)-induced alterations in oxidative stress markers in breast tissue, including 8-iso-prostane F(2alpha) formation and changes in the activities of antioxidant enzymes superoxide dismutase and glutathione peroxidase. Quantification of 2-hydroxyestradiol (2-OHE(2)) and 4-hydroxyestradiol (4-OHE(2)) formation in breast tissue confirmed that ANF inhibited 4-hydroxylation of E(2) and decreased formation of the highly carcinogenic 4-OHE(2). These results demonstrate that antioxidant vitamin C reduces the incidence of estrogen-induced mammary tumors, increases tumor latency and decreases oxidative stress in vivo. Further, our data indicate that ANF completely abrogates breast cancer development in ACI rats. The present study is the first to demonstrate the inhibition of breast carcinogenesis by antioxidant vitamin C or the estrogen metabolic inhibitor ANF in an animal model of estrogen-induced mammary carcinogenesis. Taken together, these results suggest that E(2) metabolism and oxidant stress are critically involved in estrogen-induced breast carcinogenesis.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Benzoflavonas/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Estradiol/toxicidad , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/uso terapéutico , Ácido Ascórbico/uso terapéutico , Benzoflavonas/uso terapéutico , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Estradiol/análogos & derivados , Estradiol/metabolismo , Estrógenos de Catecol , Femenino , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Hormono-Dependientes/inducido químicamente , Neoplasias Hormono-Dependientes/prevención & control , Ratas , Ratas Endogámicas ACI
7.
Toxicol Appl Pharmacol ; 232(1): 78-85, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18640140

RESUMEN

Epidemiological evidence indicates that prolonged lifetime exposure to estrogen is associated with elevated breast cancer risk in women. Oxidative stress and estrogen receptor-associated proliferative changes are suggested to play important roles in estrogen-induced breast carcinogenesis. In the present study, we investigated changes in breast morphology and oxidative stress following estrogen exposure. Female ACI rats were treated with 17beta-estradiol (E(2), 3 mg, s.c.) for either 7, 15, 120 or 240 days. Animals were euthanized, tissues were excised, and portions of the tissues were either fixed in 10% buffered formalin or snap-frozen in liquid nitrogen. Paraffin-embedded tissues were examined for histopathologic changes. Proliferative changes appeared in the breast after 7 days of E(2) exposure. Atypical ductal proliferation and significant reduction in stromal fat were observed following 120 days of E(2) exposure. Both in situ and invasive carcinomas were observed in the majority of the mammary glands from rats treated with E(2) for 240 days. Palpable breast tumors were observed in 82% of E(2)-treated rats after 228 days, with the first palpable tumor appearing after 128 days. No morphological changes were observed in the livers, kidneys, lungs or brains of rats treated with E(2) for 240 days compared to controls. Furthermore, 8-isoprostane (8-isoPGF(2alpha)) levels as well as the activities of antioxidant enzymes, such as glutathione peroxidase, superoxide dismutase and catalase, were quantified in the breast tissues of rats treated with E(2) for 7, 15, 120 and 240 days and compared to activity levels in age-matched controls. 8-isoPGF(2alpha) levels displayed time-dependent increases upon E(2) treatment and were significantly higher than control levels at the 15, 120 and 240 day time-points. 8-isoPGF(2alpha) observed in E(2)-induced mammary tumors were significantly higher than levels found in control mammary tissue from age-matched animals. Similarly, alterations in glutathione peroxidase and superoxide dismutase activities were detected in both mammary and tumor tissue from E(2)-treated rats. Taken together, our data reveal that proliferative changes in the breast tissue of ACI rats are associated with increases in 8-isoPGF(2alpha) formation as well as changes in the activities of antioxidant enzymes. These oxidative changes appear to be a function of E(2) exposure and occur prior to tumor development.


Asunto(s)
Proliferación Celular , Glándulas Mamarias Animales , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Hormono-Dependientes/inducido químicamente , Estrés Oxidativo , Animales , Catalasa/metabolismo , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Modelos Animales de Enfermedad , Implantes de Medicamentos , Estradiol/administración & dosificación , Femenino , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Invasividad Neoplásica , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Ratas , Ratas Endogámicas ACI , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Regulación hacia Arriba
8.
J Steroid Biochem Mol Biol ; 110(1-2): 157-62, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18456490

RESUMEN

Estrogen metabolism is suggested to play an important role in estrogen-induced breast carcinogenesis. Epidemiologic studies suggest that diets rich in phytoestrogens are associated with a reduced risk of breast cancer. Phytoestrogens are biologically active plant compounds that structurally mimic 17beta-estradiol (E(2)). We hypothesize that phytoestrogens, may provide protection against breast carcinogenesis by altering the expression of estrogen-metabolizing enzymes cytochrome P450 1A1 (Cyp1A1) and 1B1 (Cyp1B1). Cyp1A1 and Cyp1B1 are responsible for the metabolism of E(2) to generate 2-hydroxyestradiol (2-OHE(2)) and 4-hydroxyestradiol (4-OHE(2)), respectively. Studies suggest that 2-OHE(2) and 2-methoxyestradiol may protect against breast carcinogenesis, while 4-OHE(2) is carcinogenic in rodent models. Thus, agents that increase the metabolism of E(2) by Cyp1A1 to produce 2-OHE(2) may have chemoprotective properties. The human immortalized non-neoplastic breast cell line MCF10F was treated with quercetin at 10 and 50muM concentrations for time points ranging from 3 to 48h. Total RNA and protein were isolated. Real-time PCR was used to measure the expression of Cyp1A1 and Cyp1B1 mRNA. Quercetin treatment produced differential regulation of Cyp1A1 and Cyp1B1 mRNA expression in a time- and dose-dependent manner. Treatment with 10 and 50 microM doses of quercetin produced 6- and 11-times greater inductions of Cyp1A1 mRNA over Cyp1B1 mRNA, respectively. Furthermore, quercetin dramatically increased Cyp1A1 protein levels and only slightly increased Cyp1B1 protein levels in MCF10F cells. Thus, our data suggest that phytoestrogens may provide protection against breast cancer by modulating expression of estrogen-metabolizing genes such that production of the highly carcinogenic estrogen metabolite 4-OHE(2) by Cyp1B1 is reduced and the production of the less genotoxic 2-OHE(2) by Cyp1A1 is increased.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Células Epiteliales/efectos de los fármacos , Quercetina/farmacología , Antioxidantes/farmacología , Hidrocarburo de Aril Hidroxilasas/genética , Western Blotting , Mama/citología , Línea Celular , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1B1 , Células Epiteliales/metabolismo , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Environ Health Perspect ; 116(4): 426-33, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18414622

RESUMEN

OBJECTIVE: Phytoestrogens display an array of pharmacologic properties, and in recent years investigation of their potential as anticancer agents has increased dramatically. In this article we review the published literature related to phytoestrogens and breast cancer as well as suggest the possible mechanisms that may underlie the relationship between phytoestrogens and breast cancer. DATA SOURCES: Electronic searches on phytoestrogens and breast cancer were performed on MEDLINE and EMBASE in June 2007. No date restriction was placed on the electronic search. DATA EXTRACTION: We focused on experimental data from published studies that examined the characteristics of phytoestrogens using in vivo or in vitro models. We also include human intervention studies in this review. DATA SYNTHESIS: We evaluated evidence regarding the possible mechanisms of phytoestrogen action. Discussions of these mechanisms were organized into those activities related to the estrogen receptor, cell growth and proliferation, tumor development, signaling pathways, and estrogen-metabolizing enzymes. CONCLUSIONS: We suggest that despite numerous investigations, the mechanisms of phytoestrogen action in breast cancer have yet to be elucidated. It remains uncertain whether these plant compounds are chemoprotective or whether they may produce adverse outcomes related to breast carcinogenesis.


Asunto(s)
Anticarcinógenos/farmacología , Neoplasias de la Mama/prevención & control , Fitoestrógenos/farmacología , Animales , Anticarcinógenos/efectos adversos , Anticarcinógenos/clasificación , Aromatasa/genética , Aromatasa/metabolismo , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/metabolismo , Aumento de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , Dieta , Estrógenos/biosíntesis , Humanos , Fitoestrógenos/efectos adversos , Fitoestrógenos/clasificación , Receptores de Estrógenos/metabolismo , Factores de Riesgo , Transducción de Señal
10.
Neurotoxicology ; 28(3): 478-89, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17175027

RESUMEN

Exposure of adult humans to manganese (Mn) has long been known to cause neurotoxicity. Recent evidence also suggests that exposure of children to Mn is associated with developmental neurotoxicity. Astrocytes are critical for the proper functioning of the nervous system, and they play active roles in neurogenesis, synaptogenesis and synaptic neurotransmission. In this report, to help elucidate the molecular events underlying Mn neurotoxicity, we systematically identified the molecular targets of Mn in primary human astrocytes at a genome-wide level, by using microarray gene expression profiling and computational data analysis algorithms. We found that Mn altered the expression of diverse genes ranging from those encoding cytokines and transporters to signal transducers and transcriptional regulators. Particularly, 28 genes encoding proinflammatory chemokines, cytokines and related functions were up-regulated, whereas 15 genes encoding functions involved in DNA replication and repair and cell cycle checkpoint control were down-regulated. Consistent with the increased expression of proinflammatory factors, analysis of common regulators revealed that 16 targets known to be positively affected by the interferon-gamma signaling pathway were up-regulated by Mn(2+). In addition, 68 genes were found to be similarly up- or down-regulated by both Mn(2+) and hypoxia. These results from genomic analysis are further supported by data from real-time RT-PCR, Western blotting, flow cytometric and toxicological analyses. Together, these analyses show that Mn(2+) selectively affects cell cycle progression, the expression of hypoxia-responsive genes, and the expression of proinflammatory factors in primary human astrocytes. These results provide important insights into the molecular mechanisms underlying Mn neurotoxicity.


Asunto(s)
Astrocitos/efectos de los fármacos , Cloruros/toxicidad , Perfilación de la Expresión Génica , Western Blotting , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Ciclo Celular/efectos de los fármacos , Hipoxia de la Célula/genética , Células Cultivadas , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Factor de Crecimiento Epidérmico/biosíntesis , Factor de Crecimiento Epidérmico/genética , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Celular/efectos de los fármacos , Inflamación/genética , Interferón gamma/biosíntesis , L-Lactato Deshidrogenasa/metabolismo , Compuestos de Manganeso , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transcripción Genética , Regulación hacia Arriba/efectos de los fármacos
11.
Cell Res ; 16(8): 681-92, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16894358

RESUMEN

Heme (iron protoporphyrin IX) is an essential molecule for numerous living organisms. Not only does it serve as a prosthetic group in enzymes, it also acts as a signaling molecule that controls diverse molecular and cellular processes ranging from signal transduction to protein complex assembly. Deficient heme synthesis or function impacts the hematopoietic, hepatic and nervous systems in humans. Recent studies have revealed a series of heme-regulated transcription factors and signal transducers including Hap1, a heme-activated transcription factor that mediates the effects of oxygen on gene transcription in the yeast Saccharomyces cerevisiae; Bach1, a transcriptional repressor that is negatively regulated by heme in mammalian cells; IRR, an iron regulatory protein that mediates the iron-dependant regulation of heme synthesis in the bacterium Bradyrhizobium japonicum; and heme-regulated inhibitor, an eucaryotic initiation factor 2alpha kinase that coordinates protein synthesis with heme availability in reticulocytes. In this review, we summarize the current knowledge about how heme controls the activity of these transcriptional regulators and signal transducers, and discuss diseases associated with defective heme synthesis, degradation and function.


Asunto(s)
Hemo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Bradyrhizobium/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación de la Expresión Génica , Hemo/genética , Hemo/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Hierro/metabolismo , Oxidación-Reducción , Oxígeno/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transactivadores/metabolismo , Proteínas ras/metabolismo
12.
Toxicol Sci ; 93(1): 125-35, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16790487

RESUMEN

Given the widespread use of insecticides in the environment, it is important to perform studies evaluating their potential effects on humans. Organophosphate insecticides, such as chlorpyrifos, are being phased out; however, the use of pyrethroids in household pest control is increasing. While chlorpyrifos is relatively well studied, much less is known about the potential neurotoxicity of cyfluthrin and other pyrethroids. To gain insights into the neurotoxicity of cyfluthrin, we compared and evaluated the toxicity profiles of chlorpyrifos and cyfluthrin in primary human fetal astrocytes. We found that at the same concentrations, cyfluthrin exerts as great as, or greater toxic effects on the growth, survival, and proper functioning of human astrocytes. By using microarray gene expression profiling, we systematically identified and compared the potential molecular targets of chlorpyrifos and cyfluthrin, at a genome-wide scale. We found that chlorpyrifos and cyfluthrin affect a similar number of transcripts. These targets include molecular chaperones, signal transducers, transcriptional regulators, transporters, and those involved in behavior and development. Further computational and biochemical analyses show that cyfluthrin and chlorpyrifos upregulate certain targets of the interferon-gamma and insulin-signaling pathways and that they increase the protein levels of activated extracellular signal-regulated kinase 1/2, a key component of insulin signaling; interleukin 6, a key inflammatory mediator; and glial fibrillary acidic protein, a marker of inflammatory astrocyte activation. These results suggest that inflammatory activation of astrocytes might be an important mechanism underlying neurotoxicity of both chlorpyrifos and cyfluthrin.


Asunto(s)
Astrocitos/efectos de los fármacos , Cloropirifos/toxicidad , Expresión Génica/efectos de los fármacos , Insecticidas/toxicidad , Nitrilos/toxicidad , Piretrinas/toxicidad , Apoptosis/efectos de los fármacos , Astrocitos/metabolismo , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ , Mediadores de Inflamación/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Transducción de Señal
13.
Proc Natl Acad Sci U S A ; 103(15): 5875-80, 2006 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-16595622

RESUMEN

Many questions remain about the process of DNA double strand break (DSB) repair by homologous recombination (HR), particularly concerning the exact function played by individual proteins and the details of specific steps in this process. Some recent studies have shown that RecQ DNA helicases have a function in HR. We studied the role of the RecQ helicase Rqh1 with HR proteins in the repair of a DSB created at a unique site within the Schizosaccharomyces pombe genome. We found that DSBs in rqh1(+) cells, are predominantly repaired by interchromosomal gene conversion, with HR between sister chromatids [sister-chromatid conversion (SCC)], occurring less frequently. In Deltarqh1 cells, repair by SCC is favored, and gene conversion rates slow significantly. When we limited the potential for SCC in Deltarqh1 cells by reducing the length of the G2 phase of the cell cycle, DSB repair continued to be predominated by SCC, whereas it was essentially eliminated in wild-type cells. These data indicate that Rqh1 acts to regulate DSB repair by blocking SCC. Interestingly, we found that this role for Rqh1 is independent of its helicase activity. In the course of these studies, we also found nonhomologous end joining to be largely faithful in S. pombe, contrary to current belief. These findings provide insight into the regulation of DSB repair by RecQ helicases.


Asunto(s)
Daño del ADN , ADN Helicasas/genética , Reparación del ADN , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/genética , Intercambio de Cromátides Hermanas , ADN Helicasas/metabolismo , Eliminación de Gen , Genoma Fúngico , Schizosaccharomyces/enzimología , Proteínas de Schizosaccharomyces pombe/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...