Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acid Ther ; 27(4): 183-196, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28609186

ABSTRACT

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.


Subject(s)
Oligonucleotides/therapeutic use , Animals , Complement Activation , Cytokines/metabolism , Drug Evaluation, Preclinical/methods , Excipients/toxicity , Humans , Maximum Tolerated Dose , Mutagenicity Tests , Oligonucleotides/pharmacokinetics , Oligonucleotides/toxicity , Risk Assessment
2.
Environ Health Perspect ; 117(7): 1139-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19654925

ABSTRACT

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.


Subject(s)
Multigene Family/genetics , Receptors, Aryl Hydrocarbon/metabolism , Animals , Binding Sites/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Expression Profiling , Mice , Molecular Sequence Data , Multigene Family/physiology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein Binding/genetics , Protein Binding/physiology
3.
EXS ; 99: 231-57, 2009.
Article in English | MEDLINE | ID: mdl-19157064

ABSTRACT

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.


Subject(s)
Receptors, Aryl Hydrocarbon/physiology , Signal Transduction/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/genetics
4.
Biochem Pharmacol ; 77(4): 713-22, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18817753

ABSTRACT

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.


Subject(s)
Receptor Cross-Talk/physiology , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/physiology , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle/physiology , Environmental Pollutants/toxicity , Humans , Polychlorinated Dibenzodioxins/toxicity , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects
5.
Mol Biol Cell ; 19(8): 3263-71, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524851

ABSTRACT

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.


Subject(s)
Apoptosis , E2F1 Transcription Factor/metabolism , Oxidative Stress , Receptors, Aryl Hydrocarbon/metabolism , Animals , Apoptotic Protease-Activating Factor 1/metabolism , Cell Cycle , Cell Line, Tumor , Fibroblasts/metabolism , Histones/metabolism , Humans , Mice , Models, Biological , Promoter Regions, Genetic
6.
J Mol Biol ; 380(1): 1-16, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18508077

ABSTRACT

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.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Latent TGF-beta Binding Proteins/genetics , Promoter Regions, Genetic/genetics , Receptors, Aryl Hydrocarbon/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Base Sequence , Cloning, Molecular , DNA Methylation , Genotype , Histone Deacetylase 2 , Histone Deacetylases/genetics , Histones/metabolism , Latent TGF-beta Binding Proteins/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , RNA Interference , Repressor Proteins/genetics , Response Elements/genetics
7.
J Cell Biochem ; 96(6): 1174-84, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16211578

ABSTRACT

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.


Subject(s)
Cell Cycle/physiology , Genes, Tumor Suppressor/drug effects , Receptors, Aryl Hydrocarbon/physiology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Humans , Ligands , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Xenobiotics/pharmacology
8.
J Biol Chem ; 279(28): 29013-22, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15123621

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/metabolism , S Phase/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , E1A-Associated p300 Protein , E2F Transcription Factors , Genes, Reporter , Humans , Ligands , Mice , Polychlorinated Dibenzodioxins/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , Transcription, Genetic
9.
J Invertebr Pathol ; 83(1): 37-45, 2003 May.
Article in English | MEDLINE | ID: mdl-12725810

ABSTRACT

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.


Subject(s)
Ascomycota/physiology , Drosophila/parasitology , Mycoses/physiopathology , Protozoan Infections/physiopathology , Trypanosomatina/physiology , Animals , Drosophila/growth & development , Female , Host-Parasite Interactions/physiology , Intestines/parasitology , Male , Mycoses/epidemiology , Prevalence , Protozoan Infections/epidemiology , Pupa , Sex Factors , Species Specificity , Symbiosis
SELECTION OF CITATIONS
SEARCH DETAIL
...