Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(1): 105525, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38043800

RESUMEN

The innate antiviral response to RNA viruses is initiated by sensing of viral RNAs by RIG-I-like receptors and elicits type I interferon (IFN) production, which stimulates the expression of IFN-stimulated genes that orchestrate the antiviral response to prevent systemic infection. Negative regulation of type I IFN and its master regulator, transcription factor IRF7, is essential to maintain immune homeostasis. We previously demonstrated that AIP (aryl hydrocarbon receptor interacting protein) functions as a negative regulator of the innate antiviral immune response by binding to and sequestering IRF7 in the cytoplasm, thereby preventing IRF7 transcriptional activation and type I IFN production. However, it remains unknown how AIP inhibition of IRF7 is regulated. We show here that the kinase TBK1 phosphorylates AIP and Thr40 serves as the primary target for TBK1 phosphorylation. AIP Thr40 plays critical roles in regulating AIP stability and mediating its interaction with IRF7. The AIP phosphomimetic T40E exhibited increased proteasomal degradation and enhanced interaction with IRF7 compared with wildtype AIP. AIP T40E also blocked IRF7 nuclear translocation, which resulted in reduced type I IFN production and increased viral replication. In sharp contrast, AIP phosphonull mutant T40A had impaired IRF7 binding, and stable expression of AIP T40A in AIP-deficient mouse embryonic fibroblasts elicited a heightened type I IFN response and diminished RNA virus replication. Taken together, these results demonstrate that TBK1-mediated phosphorylation of AIP at Thr40 functions as a molecular switch that enables AIP to interact with and inhibit IRF7, thus preventing overactivation of type I IFN genes by IRF7.


Asunto(s)
Inmunidad Innata , Factor 7 Regulador del Interferón , Interferón Tipo I , Proteínas Serina-Treonina Quinasas , Infecciones por Virus ARN , Virus ARN , Receptores de Hidrocarburo de Aril , Animales , Ratones , Fibroblastos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Virus ARN/inmunología , Infecciones por Virus ARN/inmunología , Humanos , Células HEK293
2.
Toxicol Sci ; 187(1): 150-161, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35172007

RESUMEN

Frog metamorphosis, the development of an air-breathing froglet from an aquatic tadpole, is controlled by thyroid hormone (TH) and glucocorticoids (GC). Metamorphosis is susceptible to disruption by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an aryl hydrocarbon receptor (AHR) agonist. Krüppel-like factor 9 (klf9), an immediate early gene in the endocrine-controlled cascade of expression changes governing metamorphosis, can be synergistically induced by both hormones. This process is mediated by an upstream enhancer cluster, the klf9 synergy module (KSM). klf9 is also an AHR target. We measured klf9 mRNA following exposures to triiodothyronine (T3), corticosterone (CORT), and TCDD in the Xenopus laevis cell line XLK-WG. klf9 was induced 6-fold by 50 nM T3, 4-fold by 100 nM CORT, and 3-fold by 175 nM TCDD. Cotreatments of CORT and TCDD or T3 and TCDD induced klf9 7- and 11-fold, respectively, whereas treatment with all 3 agents induced a 15-fold increase. Transactivation assays examined enhancers from the Xenopus tropicalis klf9 upstream region. KSM-containing segments mediated a strong T3 response and a larger T3/CORT response, whereas induction by TCDD was mediated by a region ∼1 kb farther upstream containing 5 AHR response elements (AHREs). This region also supported a CORT response in the absence of readily identifiable GC responsive elements, suggesting mediation by protein-protein interactions. A functional AHRE cluster is positionally conserved in the human genome, and klf9 was induced by TCDD and TH in HepG2 cells. These results indicate that AHR binding to upstream AHREs represents an early key event in TCDD's disruption of endocrine-regulated klf9 expression and metamorphosis.


Asunto(s)
Dioxinas , Dibenzodioxinas Policloradas , Animales , Corticosterona/farmacología , Glucocorticoides/farmacología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Metamorfosis Biológica , Dibenzodioxinas Policloradas/toxicidad , Hormonas Tiroideas/metabolismo
3.
Gen Comp Endocrinol ; 299: 113592, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32858041

RESUMEN

The aryl hydrocarbon receptor (AHR) plays pleiotropic roles in the development and physiology of vertebrates in conjunction with xenobiotic and endogenous ligands. It is best known for mediating the toxic effects of dioxin-like pollutants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). While most vertebrates possess at least one AHR that binds TCDD tightly, amphibian AHRs bind TCDD with very low affinity. Previous analyses of AHRs from Xenopus laevis (a frog; order Anura) and Ambystoma mexicanum (a salamander; order Caudata) identified three amino acid residues in the ligand-binding domain (LBD) that underlie low-affinity binding. In X. laevis AHR1ß, these are A354, A370, and N325. Here we extend the analysis of amphibian AHRs to the caecilian Gymnopis multiplicata, representing the remaining extant amphibian order, Gymnophiona. G. multiplicata AHR groups with the monophyletic vertebrate AHR/AHR1 clade. The LBD includes all three signature residues of low TCDD affinity, and a structural homology model suggests that its architecture closely resembles those of other amphibians. In transactivation assays, the EC50 for reporter gene induction by TCDD was 17.17 nM, comparable to X. laevis AhR1ß (26.23 nM) and Ambystoma AHR (34.09 nM) and dramatically higher than mouse AhR (0.13 nM), a trend generally reflected in direct measures of TCDD binding. These shared properties distinguish amphibian AHRs from the high-affinity proteins typical of both vertebrate groups that diverged earlier (teleost fish) and those that appeared more recently (other tetrapods). These findings suggest the hypothesis that AHRs with low TCDD affinity represent a characteristic that evolved in a common ancestor of all three extant amphibian groups.


Asunto(s)
Ambystoma mexicanum/metabolismo , Dibenzodioxinas Policloradas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Xenopus laevis/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Ligandos , Filogenia , Dibenzodioxinas Policloradas/química , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/genética , Homología de Secuencia
4.
Toxicol Sci ; 161(1): 196-206, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29294139

RESUMEN

Amphibian metamorphosis is driven by thyroid hormone (TH). We used prometamorphic tadpoles and a cell line of the African clawed frog (Xenopus laevis) to examine immediate effects of dioxin exposure on TH. Gene expression patterns suggest cross-talk between the thyroid hormone receptor (TR) and aryl hydrocarbon receptor (AHR) signaling pathways. In XLK-WG cells, expression of Cytochrome P450 1A6 (cyp1A6), an AHR target, was induced 1000-fold by 100 nM TCDD (2, 3, 7, 8 tetrachlorodibenzo-p-dioxin). Krüppel-Like Factor 9 (klf9), the first gene induced in a cascade of TH responses tied to metamorphosis, was upregulated over 5-fold by 50 nM triiodothyronine (T3) and 2-fold by dioxin. Co-exposure to T3 and TCDD boosted both responses, further inducing cyp1A6 by 75% and klf9 about 60%. Additional canonical targets of each receptor, including trßa and trßb (TR) and udpgt1a (AHR) responded similarly. Induction of TH targets by TCDD in XLK-WG cells predicts that exposure could speed metamorphosis. We tested this hypothesis in two remodeling events: tail resorption and hind limb growth. Resorption of ex vivo cultured tails was accelerated by 10 nM T3, while a modest increase in resorption by 100 nM TCDD lacked statistical significance. Hind limbs doubled in length over four days following 1 nM T3 treatment, but limb length was unaffected by 100 nM TCDD. TCDD co-exposure reduced the T3 effect by nearly 40%, despite TCDD induction of klf9 in whole tadpoles, alone or with T3. These results suggest that tissue-specific TCDD effects limit or reverse the increased metamorphosis rate predicted by klf9 induction.


Asunto(s)
Disruptores Endocrinos/toxicidad , Larva/efectos de los fármacos , Metamorfosis Biológica/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Hormonas Tiroideas/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Larva/metabolismo , Metamorfosis Biológica/genética , Receptor Cross-Talk/efectos de los fármacos , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal/efectos de los fármacos , Hormonas Tiroideas/farmacología , Triyodotironina/metabolismo , Triyodotironina/farmacología , Xenopus laevis
5.
Toxicol Sci ; 155(2): 337-347, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27994169

RESUMEN

Gene duplication confers genetic redundancy that can facilitate subfunctionalization, the partitioning of ancestral functions between paralogs. We capitalize on a recent genome duplication in Xenopus laevis (African clawed frog) to interrogate possible functional differentiation between alloalleles of the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor that mediates toxicity of dioxin-like compounds and plays a role in the physiology and development of the cardiovascular, hepatic, and immune systems in vertebrates. X. laevis has 2 AHR genes, AHR1α and AHR1ß To test the hypothesis that the encoded proteins exhibit different molecular functions, we used TALENs in XLK-WG cells, generating mutant lines lacking functional versions of each AHR and measuring the transcriptional responsiveness of several target genes to the toxic xenobiotic 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and the candidate endogenous ligand 6-formylindolo[3,2-b]carbazole (FICZ). Mutation of either AHR1α or AHR1ß reduced TCDD induction of the canonical AHR target, Cytochrome P4501A6, by 75%, despite the much lower abundance of AHR1ß in wild-type cells. More modestly induced target genes, encoding aryl hydrocarbon receptor repressor (AHRR), spectrin repeat-containing nuclear envelope protein 1 (SYNE-1), and gap junction protein gamma 1 (GJC1), were regulated solely by AHR1α. AHR1ß was responsible for CYP1A6 induction by FICZ, while AHR1α mediated FICZ induction of AHRR We conclude that AHR1α and AHR1ß have distinct transcriptional functions in response to specific agonists, even within a single cell type. Functional analysis of frog AHR paralogs advances the understanding of AHR evolution and as well as the use of frog models of developmental toxicology such as FETAX.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Carbazoles/toxicidad , Expresión Génica/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/fisiología , Proteínas de Xenopus/fisiología , Xenopus laevis/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular , Sistema Enzimático del Citocromo P-450/biosíntesis , Duplicación de Gen , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/genética , Proteínas de Xenopus/agonistas , Proteínas de Xenopus/genética
6.
Environ Sci Technol ; 49(11): 6993-7001, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25941739

RESUMEN

Structural features of the aryl hydrocarbon receptor (AHR) can underlie species- and population-specific differences in its affinity for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). These differences often explain variations in TCDD toxicity. Frogs are relatively insensitive to dioxin, and Xenopus AHRs bind TCDD with low affinity. Weak TCDD binding results from the combination of three residues in the ligand-binding domain: A354 and A370, and N325. Here we sought to determine whether this mechanism of weak TCDD binding is shared by other amphibian AHRs. We isolated an AHR cDNA from the Mexican axolotl (Ambystoma mexicanum). The encoded polypeptide contains identical residues at positions that confer low TCDD affinity to X. laevis AHRs (A364, A380, and N335), and homology modeling predicts they protrude into the binding cavity. Axolotl AHR bound one-tenth the TCDD of mouse AHR in velocity sedimentation analysis, and in transactivation assays, the EC50 for TCDD was 23 nM, similar to X. laevis AHR1ß (27 nM) and greater than AHR containing the mouse ligand-binding domain (0.08 nM). Sequence, modeled structure, and function indicate that axolotl AHR binds TCDD weakly, predicting that A. mexicanum lacks sensitivity toTCDD toxicity. We hypothesize that this characteristic of axolotl and Xenopus AHRs arose in a common ancestor of the Caudata and Anura.


Asunto(s)
Ambystoma mexicanum/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Secuencia de Aminoácidos , Animales , Carbazoles/farmacología , Genes Reporteros , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Dibenzodioxinas Policloradas/química , Receptores de Hidrocarburo de Aril/química , Activación Transcripcional/efectos de los fármacos , Xenopus laevis/genética
7.
Biochemistry ; 52(10): 1746-54, 2013 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23394719

RESUMEN

The aryl hydrocarbon receptor (AHR) is a Per-ARNT-Sim (PAS) family protein that mediates the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in vertebrates. Frogs are remarkably insensitive to TCDD, and AHRs from Xenopus laevis bind TCDD with low affinity. We sought to identify structural features of X. laevis AHR1ß associated with low TCDD sensitivity. Substitution of the entire ligand binding domain (LBD) with the corresponding sequence from mouse AHR(b-1) dramatically increased TCDD responsiveness in transactivation assays. To identify the amino acid residues responsible, we constructed a comparative model of the AHR1ß LBD using homologous domains of PAS proteins HIF2α and ARNT. The model revealed an internal cavity with dimensions similar to those of the putative binding cavity of mouse AHR(b-1), suggesting the importance of side chain interactions over cavity size. Of residues with side chains clearly pointing into the cavity, only two differed from the mouse sequence. When A354, located within a conserved ß-strand, was changed to serine, the corresponding mouse residue, the EC50 for TCDD decreased more than 15-fold. When N325 was changed to serine, the EC50 decreased 3-fold. When the mutations were combined, the EC50 decreased from 18.6 to 0.8 nM, the value nearly matching the TCDD sensitivity of mouse AHR. Velocity sedimentation analysis confirmed that mutant frog AHRs exhibited correspondingly increased levels of TCDD binding. We also assayed mutant AHRs for responsiveness to a candidate endogenous ligand, 6-formylindolo[3,2-b]carbazole (FICZ). Mutations that increased sensitivity to TCDD also increased sensitivity to FICZ. This comparative study represents a novel approach to discerning fundamental information about the structure of AHR and its interactions with biologically important agonists.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/química , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sitios de Unión , Carbazoles/metabolismo , Ligandos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Dibenzodioxinas Policloradas/metabolismo , Estructura Terciaria de Proteína , Receptores de Hidrocarburo de Aril/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Activación Transcripcional , Proteínas de Xenopus/genética , Xenopus laevis/genética , Xenopus laevis/metabolismo
8.
Aquat Toxicol ; 114-115: 165-72, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22446828

RESUMEN

Cytochrome P450 family 1 (CYP1) includes four subfamilies of enzymes: CYP1A, CYP1B, CYP1C, and CYP1D. In many vertebrates, CYP1A, 1B, and 1C expression is induced by agonists of the aryl hydrocarbon receptor, including toxic contaminants such as chlorinated dioxins, coplanar chlorinated biphenyls, and polynuclear aromatic hydrocarbons. Assessed at the level of mRNA, protein, or enzyme activity, CYP1s (especially CYP1As) represent potent and popular biomarkers of contaminant exposure in aquatic vertebrates. Alkylated resorufins are synthetic substrates used to detect, quantify, and describe catalytic activities of cytochrome P450s. The ability to oxidize specific resorufin-based substrates can distinguish the catalytic activities of individual CYP1s. Xenopus laevis, the African clawed frog, is the most widely employed amphibian model in aquatic toxicology, yet the number, inducibility, and activities of CYP1s have not been systematically characterized in this species. Here we report the cloning of cDNAs encoding two new CYP1 family members, X. laevis CYP1B and CYP1C, along with an integrated assessment of the induction of alkyloxyuresorufin-O-dealkylase (AROD) activities and mRNA expression of four known X. laevis CYP1s: CYP1A6, CYP1A7, CYP1B, and CYP1C. Using XLK-WG, an X. laevis kidney epithelial cell line, we determined that EROD (ethoxyresorufin substrate) and MROD (methoxyresorufin) were both induced 3000- to 5000-fold following 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) exposure up to 250 nM, while BROD (benzyloxyresorufin) and PROD (pentyloxyresorufin) activity was not detectable regardless of TCDD treatment. TCDD induced CYP1A6 and CYP1A7 mRNAs by 2-3 orders of magnitude, while CYP1B and CYP1C were unchanged. The more potent AHR agonist, FICZ (6-formylindolo[3,2-b]carbazole), induced CYP1B up to 10-fold at concentrations between 0.1 and 250 nM, while CYP1C induction was less than 3-fold. CYP1B mRNA showed the highest constitutive mRNA expression, 5- to 75-fold greater than the other CYP1 transcripts. Taken together, these results suggest that CYP1A6 and CYP1A7 perform the bulk of EROD and MROD activities we observed in these cells. The ability of each X. laevis CYP1 to catalyze oxidation of individual resorufin substrates remains to be determined. Correlating CYP1 mRNA and induced AROD activity is a significant step toward clarifying the biochemical meaning of these biomarkers and the roles of CYP1 enzymes in X. laevis. The cell culture approach represents an important complement to the long standing use of frog embryos and tadpoles in toxicological studies, providing a well suited model system for determining the molecular mechanisms underlying the regulation of these important biomarkers of contaminant exposure.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Dibenzodioxinas Policloradas/toxicidad , ARN Mensajero/metabolismo , Secuencia de Aminoácidos , Animales , Biomarcadores , Línea Celular , Clonación Molecular , Sistema Enzimático del Citocromo P-450/genética , Filogenia , ARN Mensajero/genética , Xenopus laevis
9.
Chem Biol Interact ; 183(1): 202-11, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19799885

RESUMEN

The aryl hydrocarbon receptor (AHR) mediates the toxic effects of environmental contaminants, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Frogs are very insensitive to TCDD toxicity, and AHRs from Xenopus laevis (African clawed frog) bind TCDD with >20-fold lower affinity than mouse AHR(b-1). Frog AHRs may nonetheless be highly responsive to structurally distinct compounds, especially putative endogenous ligands. We sought to determine the responsiveness of an X. laevis cell line, XLK-WG, to the candidate endogenous AHR ligand 6-formylindolo[3,2-b]carbazole (FICZ), a tryptophan photoproduct that exhibits high potency in mammalian systems. FICZ readily induced mRNAs for CYP1A6 and CYP1A7. Cells exposed to FICZ for 3h expressed up to 5-fold greater quantities of CYP1A6/7 mRNAs than those exposed for 24h, suggesting FICZ is metabolized following rapid enzyme induction. FICZ appeared more potent than TCDD. Following a 3-h exposure, the EC(50) for CYP1A6 mRNA induction by FICZ was approximately 6nM, while the TCDD response was greater than 174nM. These potencies were lower than those determined for mouse hepatoma cells (Hepa1c1c7; EC(50)= approximately 0.06nM each). The difference in ligand potency between cell lines was confirmed by induction of ethoxyresorufin-O-deethylase (EROD) activity. mRNA from XLK-WG cells treated with 100nM FICZ, 100nM TCDD, or vehicle was also analyzed on expression microarrays. FICZ altered the expression of 105 more transcripts than TCDD, and common targets were altered more dramatically by FICZ. Overall, these studies demonstrate that although FICZ is a less potent CYP1A inducer in frog cells than in mouse cells, the reduction is much less than for TCDD. Relative conservation of the FICZ response in a TCDD-insensitive species suggests its physiological importance as an AHR ligand.


Asunto(s)
Carbazoles/toxicidad , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Carbazoles/química , Línea Celular , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Perfilación de la Expresión Génica , Ligandos , Ratones , Dibenzodioxinas Policloradas/química , Xenopus laevis
10.
Toxicol Sci ; 104(1): 124-34, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18385208

RESUMEN

Xenopus laevis and other frogs are extremely insensitive to the toxicity of xenobiotic ligands of the aryl hydrocarbon receptor (AHR), including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Premetamorphic life stages are especially insensitive, and they are reported to be refractory to induction of Cytochrome P4501As, which are readily induced in older animals. The AHR repressor (AHRR) is a member of the AHR gene family. AHRR expression is induced by TCDD; it then represses AHR in an apparent negative feedback loop. In this study, we sought to test the hypothesis that constitutive AHRR expression underlies the lack of TCDD responsiveness in frog early life stages. We determined the sequence of an AHRR complimentary DNA encoding an 85.3-kDa protein sharing 52-55% identity with the bHLH/PAS domains of other AHRRs. In transient transfection assays, X. laevis AHRR inhibited TCDD-induced reporter gene expression mediated by either X. laevis AHR paralog, AHR1alpha or AHR1beta. AHRR messenger RNA was expressed at low levels in embryos (Nieuwkoop-Faber stage 33-38; approximately 52 h.p.f.) and was induced approximately twofold following TCDD exposure (42 ng/g wet weight). In contrast, AHRR exhibited higher constitutive expression and was induced more than threefold in tadpoles at stage 52-55 (prometamorphic; approximately 4 weeks postfertilization) and in isolated viscera of stage 62 tadpoles (in the metamorphic climax; approximately 7 weeks postfertilization). Although the magnitude of induction was smaller, the temporal pattern of AHRR expression and inducibility resembled that of CYP1A6. Thus, attenuated transcriptional activation of AHR target genes and low TCDD toxicity in X. laevis embryos cannot be explained by constitutive, high-level expression of AHRR.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Represoras/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células COS , Chlorocebus aethiops , Citocromo P-450 CYP1A1/genética , ADN Complementario/genética , Embrión no Mamífero/metabolismo , Datos de Secuencia Molecular , Filogenia , ARN Mensajero/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN
11.
Mar Environ Res ; 62 Suppl: S34-7, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16697456

RESUMEN

Although 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) is a potent developmental toxicant in most vertebrates, several frog species are insensitive to TCDD, especially during early life stages. Previous experiments with ranid frogs suggest that TCDD insensitivity results largely from rapid elimination. Recent studies in the African clawed frog (Xenopus laevis; family Pipidae) link low TCDD toxicity with the low binding affinity of aryl hydrocarbon receptors, which mediate the toxic effects of dioxin-like compounds. The present study sought to examine TCDD elimination in X. laevis embryos and tadpoles, enabling an integrated assessment of the relative roles of TCDD elimination and AHR-related mechanisms in TCDD insensitivity within a single frog species. Using tadpoles (stage 52-55; approximately 1 month old) exposed to [3H]TCDD, we observed that TCDD has a relatively short half life of 102.6 h, consistent with other frogs and much faster than reported clearance rates in developing fish. In contrast, TCDD elimination is much slower during early development. Embryos exposed during primary organogenesis (from stage 31-41, beginning approximately 36 h after fertilization) exhibited little TCDD elimination during the subsequent 96 h. Enhanced TCDD clearance in later developmental stages may follow the appearance of a functional digestive tract and the onset of feeding. These results suggest that rapid elimination is unlikely to contribute mechanistically to TCDD insensitivity during development of the cardiovascular system, which is significantly perturbed by TCDD in fish embryos.


Asunto(s)
Dibenzodioxinas Policloradas/farmacocinética , Contaminantes Químicos del Agua/farmacocinética , Xenopus laevis/metabolismo , Animales , Embrión no Mamífero/metabolismo , Semivida , Corazón/efectos de los fármacos , Corazón/embriología , Larva/metabolismo , Tasa de Depuración Metabólica , Dibenzodioxinas Policloradas/análisis , Factores de Tiempo , Tritio/análisis , Contaminantes Químicos del Agua/análisis , Xenopus laevis/crecimiento & desarrollo
12.
Toxicol Sci ; 88(1): 60-72, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15958654

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent developmental toxicant in most vertebrates. However, frogs are relatively insensitive to TCDD toxicity, especially during early life stages. Toxicity of TCDD and related halogenated aromatic hydrocarbons is mediated by the aryl hydrocarbon receptor (AhR), and specific differences in properties of the AhR signaling pathway can underlie in TCDD toxicity in different species. This study investigated the role of AhR in frog TCDD insensitivity, using Xenopus laevis as a model system. X. laevis, a pseudotetraploid species, expresses two distinct AhR1 genes, AhR1alpha and AhR1beta. Sharing 86% amino acid identity, these likely represent distinct genes, both orthologous to mammalian AhR and paralogous to the AhR2 gene(s) in most fish. Both AhR1alpha and AhR1beta exhibit TCDD-dependent binding of cognate DNA sequences, but they bind TCDD with at least 20-fold lower affinity than the mouse AhR(b-1) protein, and they are similarly less responsive in TCDD-induced reporter gene induction in conjunction with the mouse CYP1A1 promoter. Furthermore, CYP1A6 and CYP1A7 induction by TCDD in cultured X. laevis A6 cells appears much less responsive than CYP1A induction in cell lines derived from more sensitive animals. Taken together, these data suggest that low affinity binding by X. laevis AhRs plays an important mechanistic role in the insensitivity of frogs to TCDD. An understanding of these molecular mechanisms should aid amphibian ecotoxicology and refine the use of frog embryos as a model [e.g. in FETAX (Frog Embryo Teratogenesis Assay-Xenopus)] for determining developmental toxicity of samples containing dioxin-like compounds.


Asunto(s)
Embrión no Mamífero/metabolismo , Dibenzodioxinas Policloradas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Teratógenos/metabolismo , Pruebas de Toxicidad/métodos , Xenopus laevis , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Embrión no Mamífero/anomalías , Embrión no Mamífero/efectos de los fármacos , Datos de Secuencia Molecular , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/genética , Análisis de Secuencia de Proteína , Teratógenos/toxicidad , Xenopus laevis/embriología , Xenopus laevis/genética
13.
Mar Environ Res ; 58(2-5): 119-24, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15178023

RESUMEN

Enzymes in the cytochrome P450 gene family 1 (CYP1) catalyze the metabolic activation of numerous hydrocarbon carcinogens and various natural compounds. CYP1 family members have been identified in several vertebrates, including fish, amphibians, birds, and mammals, and are inducible by aromatic hydrocarbons acting through the aryl hydrocarbon receptor (AHR). Together with its heterodimeric partner ARNT, the ligand-bound AHR binds conserved xenobiotic response elements (XREs) near the promoter of CYP1A and other genes. However, some populations of the Atlantic killifish Fundulus heteroclitus inhabiting highly contaminated sites are refractory to CYP1A induction by aromatic hydrocarbons. To better understand the mechanisms underlying this phenomenon, we are characterizing the AHR-CYP1A signaling pathway in this species. We report here the characterization of a genomic clone containing the 5(') end of the wild-type F. heteroclitus CYP1A gene. The 5(') coding sequence matches that of the F. heteroclitus CYP1A cDNA reported earlier [Comp. Biochem. Physiol. 121C (1998) 231]. Consistent with its inducibility by AHR agonists, the CYP1A gene contains three consensus XREs (5(')CACGC3(')) within 1.6 kb of the putative transcriptional start site. When oligonucleotides containing each of these sites were analyzed in an electrophoretic mobility shift assay, one of these showed a strong, TCDD-inducible mobility shift in the presence of in vitro expressed mouse AHR protein. These sequence data and initial functional characterization provide a valuable tool for the study of genetic variations in CYP1A expression and activity in sensitive and resistant populations. These studies may ultimately shed light on the importance of P4501A activity in xenobiotic toxicity.


Asunto(s)
Citocromo P-450 CYP1A1/genética , Fundulidae/genética , Regiones Promotoras Genéticas/genética , Elementos de Respuesta/genética , Animales , Secuencia de Bases , Ensayo de Cambio de Movilidad Electroforética , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Dibenzodioxinas Policloradas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Elementos de Respuesta/fisiología , Análisis de Secuencia de ADN
14.
J Exp Zool B Mol Dev Evol ; 300(1): 48-57, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14984034

RESUMEN

The aryl hydrocarbon receptor nuclear translocator (ARNT) is a member of the Per-ARNT-Sim (PAS) protein superfamily, transcription factors that mediate the cellular responses to various developmental signals and environmental conditions. A beta-class ("partner") PAS protein, ARNT exhibits the capacity to form transcriptionally active heterodimers with several alpha-class ("sensor") proteins, including the aryl hydrocarbon receptors (AHRs), the hypoxia-inducible factors (HIFs), and the Single minded (Sim) proteins. Two genes encode different forms of ARNT in mammals: ARNT1, which is widely expressed, and ARNT2, which is limited to the brain and kidneys of adults and specific neural and branchial tissues of embryos. In contrast, fish apparently express only a single ARNT gene, although in different species, this may be either ARNT1 or ARNT2. In efforts to understand the evolution of ARNT proteins throughout the vertebrate lineage, we isolated an ARNT cDNA from early life stages of the amphibian Xenopus laevis. The encoded protein binds cognate DNA sequences in concert with mouse AHR. Phylogenetic analysis reveals that this sequence is orthologous to mammalian ARNT2 and paralogous to the recently reported X. laevis ARNT1. ARNT2 mRNA expression begins later than ARNT1 (stage 22 vs. stage 8), suggesting the two proteins play distinct roles during development. Hence, in the expression of two well-conserved ARNT paralogs with distinct expression patterns, X. laevis resembles mammals rather than fish. Diversity in the number and function of PAS proteins, including ARNT, may underlie significant species differences in developmental programming and biochemical response to environmental conditions. The identification of multiple amphibian ARNT paralogs represents an important step in the understanding of evolution and functional variation of ARNT in vertebrates.


Asunto(s)
Expresión Génica , Filogenia , Receptores de Hidrocarburo de Aril/genética , Factores de Transcripción/genética , Proteínas de Xenopus , Xenopus laevis/genética , Secuencia de Aminoácidos , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo , Western Blotting , Cartilla de ADN , ADN Complementario/genética , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Análisis de Secuencia de ADN
15.
J Exp Zool ; 294(1): 17-29, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11932946

RESUMEN

The hypoxia-inducible factors (HIFs) are dimeric transcription factors that mediate changes in gene expression during adaptation of animals to oxygen stress. Both alpha (HIFalpha) and beta (ARNT) subunits are members of the basic helix-loop-helix/Per-ARNT-Sim family of proteins. Mammals have at least three different HIF-alpha subunits, paralogous proteins expressed in tissue-specific fashion (HIF-1alpha, HIF-2alpha, and HIF-3alpha). However, the diversity and functional properties of teleost HIFs are poorly understood. In efforts to characterize mechanisms of hypoxia adaptation in estuarine fish, we have isolated cDNAs encoding HIF subunits from Fundulus heteroclitus (Atlantic killifish or mummichog), including a HIF-2alpha homolog and ARNT2alt, a splice variant of ARNT2 that contains an additional exon encoding 16 amino acids near the amino terminus. HIF-2alpha protein synthesized in vitro binds cognate DNA elements in concert with either Fundulus ARNT2 splice variant or murine ARNT1. HIF-2alpha, ARNT2, and ARNT2alt mRNAs are expressed in all organs examined. The HIF-2alpha cDNA encodes a protein of 96.4 kDa, sharing 53-54% identity with mammalian and avian orthologs. The oxygen-dependent degradation domain, however, exhibits substantial divergence from well-conserved mammalian sequences, suggesting the possibility of important functional differences, perhaps in the sensitivity to induction of activity by hypoxia. Hypoxia-tolerant fishes such as F. heteroclitus represent a unique opportunity for the study of functional and evolutionary aspects of adaptation to hypoxia at the molecular, cellular, and organismal levels. This study extends the understanding of hypoxia signaling in fish, the evolution and diversity of HIF function, and the evolution of the PAS family of proteins.


Asunto(s)
Empalme Alternativo/genética , Fundulidae/genética , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Clonación Molecular , Secuencia Conservada , Evolución Molecular , Datos de Secuencia Molecular , Familia de Multigenes/genética , Filogenia , Subunidades de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores/química , Factores de Transcripción/química
16.
J Biol Chem ; 277(9): 6949-59, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11742002

RESUMEN

The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds occur via the aryl hydrocarbon receptor (AHR), a member of the basic helix-loop-helix-Per-ARNT-Sim homology (bHLH-PAS) protein superfamily. A single AHR gene has been identified in mammals, whereas many fish species, including the Atlantic killifish (Fundulus heteroclitus) possess two distinct AHR genes (AHR1 and a novel form, AHR2). A mouse bHLH-PAS protein closely related to AHR and designated AHR repressor (AHRR) is induced by 3-methylcholanthrene and represses the transcriptional activity of the AHR. To determine whether AHRR is the mammalian ortholog of fish AHR2 and to investigate the mechanisms by which AHRR regulates AHR function, we cloned an AHRR ortholog in F. heteroclitus with high sequence identity to the mouse and human AHRRs. Killifish AHRR encodes a 680-residue protein with a predicted molecular mass of 75.2 kDa. We show that in vitro expressed AHRR proteins from human, mouse, and killifish all fail to bind [(3)H]TCDD or [(3)H]beta-naphthoflavone. In transient transfection experiments using a luciferase reporter gene under control of AHR response elements, killifish AHRR inhibited the TCDD-dependent transactivation function of both AHR1 and AHR2. AHRR mRNA is widely expressed in killifish tissues and is inducible by TCDD or polychlorinated biphenyls, but its expression is not altered in a population of fish exhibiting genetic resistance to these compounds. The F. heteroclitus AHRR promoter contains three putative AHR response elements. Both AHR1 and AHR2 activated transcription of luciferase driven by the AHRR promoter, and AHRR could repress its own promoter. Thus, AHRR is an evolutionarily conserved, TCDD-inducible repressor of AHR1 and AHR2 function. Phylogenetic analysis shows that AHRR, AHR1, and AHR2 are distinct genes, members of an AHR gene family; these three vertebrate AHR-like genes descended from a single invertebrate AHR.


Asunto(s)
Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células COS , Mapeo Cromosómico , Cartilla de ADN/farmacología , ADN Complementario/metabolismo , Peces , Genoma , Humanos , Ligandos , Luciferasas/metabolismo , Ratones , Datos de Secuencia Molecular , Filogenia , Regiones Promotoras Genéticas , Unión Proteica , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Distribución Tisular , Transcripción Genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...