Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nucleic Acid Ther ; 27(4): 183-196, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28609186

RESUMEN

The use of lipid formulations has greatly improved the ability to effectively deliver oligonucleotides and has been instrumental in the rapid expansion of therapeutic development programs using oligonucleotide drugs. However, the development of such complex multicomponent therapeutics requires the implementation of unique, scientifically sound approaches to the nonclinical development of these drugs, based upon a hybrid of knowledge and experiences drawn from small molecule, protein, and oligonucleotide therapeutic drug development. The relative paucity of directly applicable regulatory guidance documents for oligonucleotide therapeutics in general has resulted in the generation of multiple white papers from oligonucleotide drug development experts and members of the Oligonucleotide Safety Working Group (OSWG). The members of the Formulated Oligonucleotide Subcommittee of the OSWG have utilized their collective experience working with a variety of formulations and their associated oligonucleotide payloads, as well as their insights into regulatory considerations and expectations, to generate a series of consensus recommendations for the pharmacokinetic characterization and nonclinical safety assessment of this unique class of therapeutics. It should be noted that the focus of Subcommittee discussions was on lipid nanoparticle and other types of particulate formulations of therapeutic oligonucleotides and not on conjugates or other types of modifications of oligonucleotide structure intended to facilitate delivery.


Asunto(s)
Oligonucleótidos/uso terapéutico , Animales , Activación de Complemento , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Excipientes/toxicidad , Humanos , Dosis Máxima Tolerada , Pruebas de Mutagenicidad , Oligonucleótidos/farmacocinética , Oligonucleótidos/toxicidad , Medición de Riesgo
2.
Toxicol Sci ; 153(1): 39-54, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27255383

RESUMEN

Histone deacetylase (HDAC) inhibitors are an emerging class of anticancer agents that modify gene expression by altering the acetylation status of lysine residues of histone proteins, thereby inducing transcription, cell cycle arrest, differentiation, and cell death or apoptosis of cancer cells. In the clinical setting, treatment with HDAC inhibitors has been associated with delayed cardiac repolarization and in rare instances a lethal ventricular tachyarrhythmia known as torsades de pointes. The mechanism(s) of HDAC inhibitor-induced effects on cardiac repolarization is unknown. We demonstrate that administration of structurally diverse HDAC inhibitors to dogs causes delayed but persistent increases in the heart rate corrected QT interval (QTc), an in vivo measure of cardiac repolarization, at timepoints far removed from the Tmax for parent drug and metabolites. Transcriptional profiling of ventricular myocardium from dogs treated with various HDAC inhibitors demonstrated effects on genes involved in protein trafficking, scaffolding and insertion of various ion channels into the cell membrane as well as genes for specific ion channel subunits involved in cardiac repolarization. Extensive in vitro ion channel profiling of various structural classes of HDAC inhibitors (and their major metabolites) by binding and acute patch clamp assays failed to show any consistent correlations with direct ion channel blockade. Drug-induced rescue of an intracellular trafficking-deficient mutant potassium ion channel, hERG (G601S), and decreased maturation (glycosylation) of wild-type hERG expressed by CHO cells in vitro correlated with prolongation of QTc intervals observed in vivo The results suggest that HDAC inhibitor-induced prolongation of cardiac repolarization may be mediated in part by transcriptional changes of genes required for ion channel trafficking and localization to the sarcolemma. These data have broad implications for the development of these drug classes and suggest that the optimal time to assess potentially transcriptionally mediated physiologic effects will be delayed relative to an epigenetic drug's Tmax/Cmax.


Asunto(s)
Corazón/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Transcripción Genética , Animales , Transporte Biológico , Perros , Corazón/fisiología , Inhibidores de Histona Desacetilasas/farmacocinética , Masculino
3.
Environ Mol Mutagen ; 56(3): 277-85, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25482136

RESUMEN

Genetic toxicity tests currently used to identify and characterize potential human mutagens and carcinogens rely on measurements of primary DNA damage, gene mutation, and chromosome damage in vitro and in rodents. The International Life Sciences Institute Health and Environmental Sciences Institute (ILSI-HESI) Committee on the Relevance and Follow-up of Positive Results in In Vitro Genetic Toxicity Testing held an April 2012 Workshop in Washington, DC, to consider the impact of new understanding of biology and new technologies on the identification and characterization of genotoxic substances, and to identify new approaches to inform more accurate human risk assessment for genetic and carcinogenic effects. Workshop organizers and speakers were from industry, academe, and government. The Workshop focused on biological effects and technologies that would potentially yield the most useful information for evaluating human risk of genetic damage. Also addressed was the impact that improved understanding of biology and availability of new techniques might have on genetic toxicology practices. Workshop topics included (1) alternative experimental models to improve genetic toxicity testing, (2) Biomarkers of epigenetic changes and their applicability to genetic toxicology, and (3) new technologies and approaches. The ability of these new tests and technologies to be developed into tests to identify and characterize genotoxic agents; to serve as a bridge between in vitro and in vivo rodent, or preferably human, data; or to be used to provide dose response information for quantitative risk assessment was also addressed. A summary of the workshop and links to the scientific presentations are provided.


Asunto(s)
Pruebas de Mutagenicidad/métodos , Mutágenos/toxicidad , Animales , District of Columbia , Epigénesis Genética/efectos de los fármacos , Genómica/métodos , Humanos , Medición de Riesgo
4.
Toxicol Sci ; 131(2): 375-86, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23091169

RESUMEN

The molecular events during nongenotoxic carcinogenesis and their temporal order are poorly understood but thought to include long-lasting perturbations of gene expression. Here, we have investigated the temporal sequence of molecular and pathological perturbations at early stages of phenobarbital (PB) mediated liver tumor promotion in vivo. Molecular profiling (mRNA, microRNA [miRNA], DNA methylation, and proteins) of mouse liver during 13 weeks of PB treatment revealed progressive increases in hepatic expression of long noncoding RNAs and miRNAs originating from the Dlk1-Dio3 imprinted gene cluster, a locus that has recently been associated with stem cell pluripotency in mice and various neoplasms in humans. PB induction of the Dlk1-Dio3 cluster noncoding RNA (ncRNA) Meg3 was localized to glutamine synthetase-positive hypertrophic perivenous hepatocytes, suggesting a role for ß-catenin signaling in the dysregulation of Dlk1-Dio3 ncRNAs. The carcinogenic relevance of Dlk1-Dio3 locus ncRNA induction was further supported by in vivo genetic dependence on constitutive androstane receptor and ß-catenin pathways. Our data identify Dlk1-Dio3 ncRNAs as novel candidate early biomarkers for mouse liver tumor promotion and provide new opportunities for assessing the carcinogenic potential of novel compounds.


Asunto(s)
Biomarcadores de Tumor/genética , Impresión Genómica , Péptidos y Proteínas de Señalización Intercelular/genética , Yoduro Peroxidasa/genética , Neoplasias Hepáticas Experimentales/genética , Familia de Multigenes , ARN no Traducido/genética , Animales , Proteínas de Unión al Calcio , Receptor de Androstano Constitutivo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Reacción en Cadena de la Polimerasa , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Transcriptoma , beta Catenina/metabolismo
5.
PLoS One ; 6(3): e18216, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21455306

RESUMEN

Evidence suggests that epigenetic perturbations are involved in the adverse effects associated with some drugs and toxicants, including certain classes of non-genotoxic carcinogens. Such epigenetic changes (altered DNA methylation and covalent histone modifications) may take place at the earliest stages of carcinogenesis and their identification holds great promise for biomedical research. Here, we evaluate the sensitivity and specificity of genome-wide epigenomic and transcriptomic profiling in phenobarbital (PB)-treated B6C3F1 mice, a well-characterized rodent model of non-genotoxic liver carcinogenesis. Methylated DNA Immunoprecipitation (MeDIP)-coupled microarray profiling of 17,967 promoter regions and 4,566 intergenic CpG islands was combined with genome-wide mRNA expression profiling to identify liver tissue-specific PB-mediated DNA methylation and transcriptional alterations. Only a limited number of significant anti-correlations were observed between PB-induced transcriptional and promoter-based DNA methylation perturbations. However, the constitutive androstane receptor (CAR) target gene Cyp2b10 was found to be concomitantly hypomethylated and transcriptionally activated in a liver tissue-specific manner following PB treatment. Furthermore, analysis of active and repressive histone modifications using chromatin immunoprecipitation revealed a strong PB-mediated epigenetic switch at the Cyp2b10 promoter. Our data reveal that PB-induced transcriptional perturbations are not generally associated with broad changes in the DNA methylation status at proximal promoters and suggest that the drug-inducible CAR pathway regulates an epigenetic switch from repressive to active chromatin at the target gene Cyp2b10. This study demonstrates the utility of integrated epigenomic and transcriptomic profiling for elucidating early mechanisms and biomarkers of non-genotoxic carcinogenesis.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/genética , Epigenómica/métodos , Fenobarbital/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Esteroide Hidroxilasas/genética , Animales , Inmunoprecipitación de Cromatina , Receptor de Androstano Constitutivo , Islas de CpG/efectos de los fármacos , Islas de CpG/genética , Familia 2 del Citocromo P450 , Metilación de ADN/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
6.
Environ Health Perspect ; 117(7): 1139-46, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19654925

RESUMEN

BACKGROUND: The vertebrate aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates cellular responses to environmental polycyclic and halogenated compounds. The naive receptor is believed to reside in an inactive cytosolic complex that translocates to the nucleus and induces transcription of xenobiotic detoxification genes after activation by ligand. OBJECTIVES: We conducted an integrative genomewide analysis of AHR gene targets in mouse hepatoma cells and determined whether AHR regulatory functions may take place in the absence of an exogenous ligand. METHODS: The network of AHR-binding targets in the mouse genome was mapped through a multipronged approach involving chromatin immunoprecipitation/chip and global gene expression signatures. The findings were integrated into a prior functional knowledge base from Gene Ontology, interaction networks, Kyoto Encyclopedia of Genes and Genomes pathways, sequence motif analysis, and literature molecular concepts. RESULTS: We found the naive receptor in unstimulated cells bound to an extensive array of gene clusters with functions in regulation of gene expression, differentiation, and pattern specification, connecting multiple morphogenetic and developmental programs. Activation by the ligand displaced the receptor from some of these targets toward sites in the promoters of xenobiotic metabolism genes. CONCLUSIONS: The vertebrate AHR appears to possess unsuspected regulatory functions that may be potential targets of environmental injury.


Asunto(s)
Familia de Multigenes/genética , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Sitios de Unión/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Familia de Multigenes/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Unión Proteica/genética , Unión Proteica/fisiología
7.
EXS ; 99: 231-57, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19157064

RESUMEN

The aryl hydrocarbon receptor (AHR) has long been recognized as a ligand-activated transcription factor responsible for the induction of drug-metabolizing enzymes. Its role in the combinatorial matrix of cell functions was established long before the first report of an AHR cDNA sequence was published. It is only recently that other functions of this protein have begun to be recognized, and it is now clear that the AHR also functions in pathways outside of its well-characterized role in xenobiotic enzyme induction. Perturbation of these pathways by xenobiotic ligands may ultimately explain much of the toxicity of these compounds. This chapter focuses on the interactions of the AHR in pathways critical to cell cycle regulation, mitogen-activated protein kinase cascades, differentiation and apoptosis. Ultimately, the effect of a particular AHR ligand on the biology of the organism will depend on the milieu of critical pathways and proteins expressed in specific cells and tissues with which the AHR itself interacts.


Asunto(s)
Receptores de Hidrocarburo de Aril/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/genética , Apoptosis/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/genética
8.
EXS ; 99: 259-88, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19157065

RESUMEN

Recent advances in technological approaches for mapping and characterizing the epigenome are generating a wealth of new opportunities for exploring the relationship between epigenetic modifications, human disease and the therapeutic potential of pharmaceutical drugs. While the best examples for xenobiotic-induced epigenetic perturbations come from the field of non-genotoxic carcinogenesis, there is growing evidence for the relevance of epigenetic mechanisms associated with a wide range of disease areas and drug targets. The application of epigenomic profiling technologies to drug safety sciences has great potential for providing novel insights into the molecular basis of long-lasting cellular perturbations including increased susceptibility to disease and/or toxicity, memory of prior immune stimulation and/or drug exposure, and transgenerational effects.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Epigénesis Genética , Toxicología/métodos , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Humanos , Toxicología/tendencias
9.
Biochem Pharmacol ; 77(4): 713-22, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18817753

RESUMEN

Exposure to toxic polycyclic aromatic hydrocarbons raises a number of toxic and carcinogenic responses in experimental animals and humans mediated for the most part by the aryl hydrocarbon -- or dioxin -- receptor (AHR). The AHR is a ligand-activated transcription factor whose central role in the induction of drug-metabolizing enzymes has long been recognized. For quite some time now, it has become clear that the AHR also functions in pathways outside of its role in detoxification and that perturbation of these pathways by xenobiotic ligands may be an important part of the toxicity of these compounds. AHR activation by some of its ligands participates among others in pathways critical to cell cycle regulation, mitogen-activated protein kinase cascades, immediate-early gene induction, cross-talk within the RB/E2F axis and mobilization of crucial calcium stores. Ultimately, the effect of a particular AHR ligand may depend as much on the adaptive interactions that it established with pathways and proteins expressed in a specific cell or tissue as on the toxic responses that it raises.


Asunto(s)
Receptor Cross-Talk/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Contaminantes Ambientales/toxicidad , Humanos , Dibenzodioxinas Policloradas/toxicidad , Receptor Cross-Talk/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Mol Biol Cell ; 19(8): 3263-71, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18524851

RESUMEN

Cellular stress by DNA damage induces checkpoint kinase-2 (CHK2)-mediated phosphorylation and stabilization of the E2F1 transcription factor, leading to induction of apoptosis by activation of a subset of proapoptotic E2F1 target genes, including Apaf1 and p73. This report characterizes an interaction between the aryl hydrocarbon (Ah) receptor (AHR), a ligand-activated transcription factor, and E2F1 that results in the attenuation of E2F1-mediated apoptosis. In Ahr(-/-) fibroblasts stably transfected with a doxycycline-regulated AHR expression vector, inhibition of AHR expression causes a significant elevation of oxidative stress, gammaH2A.X histone phosphorylation, and E2F1-dependent apoptosis, which can be blocked by small interfering RNA-mediated knockdown of E2F1 expression. In contrast, ligand-dependent AHR activation protects these cells from etoposide-induced cell death. In cells expressing both proteins, AHR and E2F1 interact independently of the retinoblastoma protein (RB), because AHR and E2F1 coimmunoprecipitate from extracts of RB-negative cells. Additionally, chromatin immunoprecipitation assays indicate that AHR and E2F1 bind to the Apaf1 promoter at a region containing a consensus E2F1 binding site but no AHR binding sites. AHR activation represses Apaf1 and TAp73 mRNA induction by a constitutively active CHK2 expression vector. Furthermore, AHR overexpression blocks the transcriptional induction of Apaf1 and p73 and the accumulation of sub-G(0)/G(1) cells resulting from ectopic overexpression of E2F1. These results point to a proproliferative, antiapoptotic function of the Ah receptor that likely plays a role in tumor progression.


Asunto(s)
Apoptosis , Factor de Transcripción E2F1/metabolismo , Estrés Oxidativo , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Ciclo Celular , Línea Celular Tumoral , Fibroblastos/metabolismo , Histonas/metabolismo , Humanos , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas
11.
J Mol Biol ; 380(1): 1-16, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18508077

RESUMEN

Latent TGFbeta-binding protein 1 (LTBP-1) is a key regulator of TGFbeta targeting and activation in the extracellular matrix. LTBP-1 is recognized as a major docking molecule to localize, and possibly to activate, TGFbeta in the extracellular matrix. Despite this relevant function, the molecular mechanisms regulating Ltbp-1 transcription remain largely unknown. Previous results from our laboratory revealed that mouse embryonic fibroblasts (MEF) lacking dioxin receptor (AhR) had increased Ltbp-1 mRNA expression and elevated TGFbeta activity, suggesting that AhR repressed Ltbp-1 transcription. Here, we have cloned the mouse Ltbp-1 gene promoter and analysed its mechanism of transcriptional repression by AhR. Reporter gene assays, AhR over-expression and site-directed mutagenesis showed that basal Ltbp-1 transcription is AhR-dependent. Chromatin immunoprecipitation (ChIP) and RNA interference (RNAi) revealed that AhR regulates Ltbp-1 transcription by a mechanism involving recruitment of co-activators such as CREB1 and co-repressors such as HDAC2 to the Ltbp-1 promoter. In AhR-expressing (AhR+/+) MEF cells, the recruitment of HDAC1, 2 and 4 correlated with decreased K8H4 acetylation and impaired binding of pCREB(Ser133) to the Ltbp-1 promoter, likely maintaining a constitutive repressed state. AhR-/- MEF cells had the opposite pattern of HDACs and pCREB1(Ser133) binding to Ltbp-1 promoter, and therefore, over-expressed Ltbp-1 mRNA. In agreement, siRNA for HDAC2 increased Ltbp-1 expression and K8H4 acetylation in AhR+/+ but not in AhR-/- MEF cells. We suggest that HDAC2 binding keeps Ltbp-1 promoter repressed in AhR+/+ MEF cells, whereas in AhR-null MEF cells the absence of HDAC2 and the binding of pCREB(Ser133) allow Ltbp-1 transcription. Thus, epigenetics can contribute to constitutive Ltbp-1 repression by a mechanism requiring AhR activity.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Regiones Promotoras Genéticas/genética , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Represoras/metabolismo , Acetilación , Animales , Secuencia de Bases , Clonación Molecular , Metilación de ADN , Genotipo , Histona Desacetilasa 2 , Histona Desacetilasas/genética , Histonas/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Interferencia de ARN , Proteínas Represoras/genética , Elementos de Respuesta/genética
12.
Toxicol Sci ; 103(1): 28-34, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18281259

RESUMEN

The Critical Path Institute recently established the Predictive Safety Testing Consortium, a collaboration between several companies and the U.S. Food and Drug Administration, aimed at evaluating and qualifying biomarkers for a variety of toxicological endpoints. The Carcinogenicity Working Group of the Predictive Safety Testing Consortium has concentrated on sharing data to test the predictivity of two published hepatic gene expression signatures, including the signature by Fielden et al. (2007, Toxicol. Sci. 99, 90-100) for predicting nongenotoxic hepatocarcinogens, and the signature by Nie et al. (2006, Mol. Carcinog. 45, 914-933) for predicting nongenotoxic carcinogens. Although not a rigorous prospective validation exercise, the consortium approach created an opportunity to perform a meta-analysis to evaluate microarray data from short-term rat studies on over 150 compounds. Despite significant differences in study designs and microarray platforms between laboratories, the signatures proved to be relatively robust and more accurate than expected by chance. The accuracy of the Fielden et al. signature was between 63 and 69%, whereas the accuracy of the Nie et al. signature was between 55 and 64%. As expected, the predictivity was reduced relative to internal validation estimates reported under identical test conditions. Although the signatures were not deemed suitable for use in regulatory decision making, they were deemed worthwhile in the early assessment of drugs to aid decision making in drug development. These results have prompted additional efforts to rederive and evaluate a QPCR-based signature using these samples. When combined with a standardized test procedure and prospective interlaboratory validation, the accuracy and potential utility in preclinical applications can be ascertained.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Genómica , Animales , Perfilación de la Expresión Génica , Masculino , Ratas , Ratas Sprague-Dawley
13.
Biochem Biophys Res Commun ; 348(1): 120-3, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16876762

RESUMEN

Naphthalene (NP) has been designated a "reasonably anticipated human carcinogen" because of positive responses in carcinogenicity bioassays in rodents. Whereas CYP2F enzymes are widely regarded as responsible for NP bioactivation, other metabolic enzymes--including CYP1A1 and CYP1A2--produce NP-1,2-oxide in vitro. We investigated the role of these aryl hydrocarbon receptor (AHR)-mediated enzymes in NP toxicity in two ways. First, NP was assessed for the ability to activate transcription via the AHR in an in vitro luciferase reporter assay and was found to have no activity. Second, mice deficient in AHR, CYP1A1 or CYP1A2 were dosed with NP alone, or following pretreatment with the CYP2F inhibitor 5-phenyl-1-pentyne. None of the knockout mice were protected from olfactory toxicity of NP. In contrast, CYP1A1- and CYP1A2-null mice pretreated with 5-phenyl-1-pentyne exhibited no NP olfactory toxicity. These results suggest that AHR-mediated enzymes do not contribute significantly to NP bioactivation in the intact animal.


Asunto(s)
Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Naftalenos/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Alquinos/farmacología , Animales , Derivados del Benceno/farmacología , Citocromo P-450 CYP1A1/efectos de los fármacos , Citocromo P-450 CYP1A2/efectos de los fármacos , Inhibidores Enzimáticos del Citocromo P-450 , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Noqueados , Naftalenos/metabolismo , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Cornetes Nasales/efectos de los fármacos , Cornetes Nasales/patología
14.
J Cell Biochem ; 96(6): 1174-84, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16211578

RESUMEN

Most effects of exposure to halogenated and polycyclic aromatic hydrocarbons are mediated by the aryl hydrocarbon receptor (AHR). It has long been recognized that the AHR is a ligand-activated transcription factor that plays a central role in the induction of drug-metabolizing enzymes and hence in xenobiotic detoxification. Of late, it has become evident that outside this well-characterized role, the AHR also functions as a modulator of cellular signaling pathways. In this Prospect, we discuss the involvement of the AHR in pathways critical to cell cycle regulation, mitogen-activated protein kinase cascades, immediate-early gene induction, and the functions of the RB protein. Ultimately, the toxicity of AHR xenobiotic ligands may be intrinsically connected with the perturbation of these pathways and depend on the many critical signaling pathways and effectors with which the AHR itself interacts.


Asunto(s)
Ciclo Celular/fisiología , Genes Supresores de Tumor/efectos de los fármacos , Receptores de Hidrocarburo de Aril/fisiología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Humanos , Ligandos , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Xenobióticos/farmacología
15.
J Biol Chem ; 279(28): 29013-22, 2004 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-15123621

RESUMEN

The environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes a wide range of toxic, teratogenic, and carcinogenic effects. TCDD is a ligand for the aromatic hydrocarbon receptor (AHR), a ligand-activated transcription factor believed to be the primary mediator of these effects. Activation of the AHR by TCDD also elicits a variety of effects on cell cycle progression, ranging from proliferation to arrest. In this report, we have characterized further the role of the activated AHR in cell cycle regulation. In human mammary carcinoma MCF-7 and mouse hepatoma Hepa-1 cells, TCDD treatment decreased the number of cells in S phase and caused the accumulation of cells in G(1). In Hepa-1 cells, this effect correlated with the transcriptional repression of several E2F-regulated genes required for S phase progression. AHR-mediated gene repression was dependent on its interaction with retinoblastoma protein but was independent of its transactivation function because AHR mutants lacking DNA binding or transactivation domains repressed E2F-dependent expression as effectively as wild type AHR. Overexpression of p300 suppressed retinoblastoma protein-dependent gene repression, and this effect was reversed by TCDD. Chromatin immunoprecipitation assays showed that TCDD treatment caused the recruitment of AHR to E2F-dependent promoters and the concurrent displacement of p300. These results delineate a novel mechanism whereby the AHR, a known transcriptional activator, also mediates gene repression by pathways involving combinatorial interactions at E2F-responsive promoters, leading to the repression of E2F-dependent, S phase-specific genes. The AHR seems to act as an environmental checkpoint that senses exposure to environmental toxicants and responds by signaling cell cycle inhibition.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/metabolismo , Fase S/fisiología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proteína p300 Asociada a E1A , Factores de Transcripción E2F , Genes Reporteros , Humanos , Ligandos , Ratones , Dibenzodioxinas Policloradas/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/fisiología , Transcripción Genética
16.
J Invertebr Pathol ; 83(1): 37-45, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12725810

RESUMEN

We report on the effect of single and mixed infections with two gut symbionts, trypanosomatids and the intracellular fungus Coccidiascus legeri, on the life history of their host, Drosophila melanogaster. We also provide the first report on the prevalence of C. legeri in natural populations of Drosophila. Prevalence overall was low (3.4%), and differed with host species, but persisted from the first to the second year of our survey. We documented delayed pupation in flies exposed to trypanosomatids, but larvae exposed to the fungus eclosed more quickly than controls. Larvae exposed to mixed infections pupated more slowly, but eclosed more quickly than controls.


Asunto(s)
Ascomicetos/fisiología , Drosophila/parasitología , Micosis/fisiopatología , Infecciones por Protozoos/fisiopatología , Trypanosomatina/fisiología , Animales , Drosophila/crecimiento & desarrollo , Femenino , Interacciones Huésped-Parásitos/fisiología , Intestinos/parasitología , Masculino , Micosis/epidemiología , Prevalencia , Infecciones por Protozoos/epidemiología , Pupa , Factores Sexuales , Especificidad de la Especie , Simbiosis
17.
Toxicology ; 181-182: 171-7, 2002 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-12505305

RESUMEN

One of the most puzzling aspects of the biological impact of polycyclic aromatic hydrocarbon compounds is that they elicit an apparently unrelated variety of toxic, teratogenic, and carcinogenic responses in exposed animals and in humans. At the cellular level, these environmental toxicants affect cell cycle regulatory mechanisms and signal transduction pathways in ways that are equally diverse and often contradictory. For example, depending on the particular cell lines studied, exposure to these compounds may lead to cell proliferation, to terminal differentiation, or to apoptosis. These effects are mediated by the aryl hydrocarbon receptor, a ligand-activated transcription factor well known for its regulatory activity on the expression of several phase I detoxification cytochrome P450 genes. Research into the molecular mechanisms of aryl hydrocarbon receptor function has uncovered a novel role for this protein during cell cycle progression. The activated receptor acts as an environmental sensor and cell cycle checkpoint that commits cells exposed to adverse environmental stimuli to arrest before the onset of DNA replication.


Asunto(s)
Ciclo Celular/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/genética , Grupo Citocromo a/metabolismo , Replicación del ADN/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Humanos , Ligandos , Plásmidos/genética , Hidrocarburos Policíclicos Aromáticos/toxicidad , Receptores de Hidrocarburo de Aril/genética , Proteína de Retinoblastoma/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...