Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 284(48): 33642-53, 2009 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-19801676

RESUMO

Zebrafish have the remarkable ability to regenerate body parts including the heart and fins by a process referred to as epimorphic regeneration. Recent studies have illustrated that similar to adult zebrafish, early life stage larvae also possess the ability to regenerate the caudal fin. A comparative microarray analysis was used to determine the degree of conservation in gene expression among the regenerating adult caudal fin, adult heart, and larval fin. Results indicate that these tissues respond to amputation/injury with strikingly similar genomic responses. Comparative analysis revealed raldh2, a rate-limiting enzyme for the synthesis of retinoic acid, as one of the most highly induced genes across the three regeneration platforms. In situ localization and functional studies indicate that raldh2 expression is critical for the formation of wound epithelium and blastema. Patterning during regenerative outgrowth was considered to be the primary function of retinoic acid signaling; however, our results suggest that it is also required for early stages of tissue regeneration. Expression of raldh2 is regulated by Wnt and fibroblast growth factor/ERK signaling.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Retinal Desidrogenase/genética , Proteínas de Peixe-Zebra/genética , Animais , Butadienos/farmacologia , Análise por Conglomerados , Embrião não Mamífero/embriologia , Embrião não Mamífero/lesões , Embrião não Mamífero/metabolismo , Extremidades/embriologia , Extremidades/crescimento & desenvolvimento , Extremidades/fisiologia , Feminino , Hibridização In Situ , Larva/genética , Larva/crescimento & desenvolvimento , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Pirróis/farmacologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Regeneração/efeitos dos fármacos , Regeneração/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteínas Wnt/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
2.
FASEB J ; 22(8): 3087-96, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18495758

RESUMO

Exposure to dioxins, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), causes a wide array of toxicities in vertebrates, which are mostly considered to be mediated through the inappropriate activation of the aryl hydrocarbon receptor (AHR) signaling pathway. Although transcriptional regulation by AHR is widely studied, the molecular mechanisms responsible for the adverse outcomes after AHR activation are largely unknown. To identify the important downstream events of AHR activation, we employed the zebrafish caudal fin regeneration model, where AHR activation blocks the regenerative process. Comparative toxicogenomic analysis revealed that both adult and larval fins respond to TCDD during regeneration with misexpression of Wnt signaling pathway members and Wnt target genes. R-Spondin1, a novel ligand for the Wnt coreceptor, was highly induced, and we hypothesized that misexpression of R-Spondin1 is necessary for AHR activation to block regeneration. Partial antisense repression of R-Spondin1 reversed the inhibitory effect of TCDD, and tissue regeneration was restored. This finding demonstrates that inhibition of regeneration by TCDD is mediated by misinduction of R-Spondin1. Because R-Spondin1 signals through the Wnt coreceptor LRP6, we further demonstrated that the TCDD-mediated block in regeneration is also LRP6 dependent. Collectively, these results indicate that inappropriate regulation of R-Spondin/LRP6 is absolutely required for TCDD to inhibit fin regeneration.


Assuntos
Receptores de Hidrocarboneto Arílico/fisiologia , Regeneração/fisiologia , Trombospondinas/fisiologia , Proteínas Wnt/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Sequência de Bases , DNA/genética , Poluentes Ambientais/toxicidade , Expressão Gênica/efeitos dos fármacos , Marcação de Genes , Proteínas HMGB/deficiência , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Dibenzodioxinas Policloradas/toxicidade , Receptor Cross-Talk , Receptores de LDL/antagonistas & inibidores , Receptores de LDL/genética , Receptores de LDL/fisiologia , Regeneração/efeitos dos fármacos , Fatores de Transcrição SOX9 , Trombospondinas/genética , Toxicogenética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
3.
J Biol Chem ; 282(48): 35202-10, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17848559

RESUMO

Identifying the molecular pathways that are required for regeneration remains one of the great challenges of regenerative medicine. Although genetic mutations have been useful for identifying some molecular pathways, small molecule probes of regenerative pathways might offer some advantages, including the ability to disrupt pathway function with precise temporal control. However, a vertebrate regeneration model amenable to rapid throughput small molecule screening is not currently available. We report here the development of a zebrafish early life stage fin regeneration model and its use in screening for small molecules that modulate tissue regeneration. By screening 2000 biologically active small molecules, we identified 17 that specifically inhibited regeneration. These compounds include a cluster of glucocorticoids, and we demonstrate that transient activation of the glucocorticoid receptor is sufficient to block regeneration, but only if activation occurs during wound healing/blastema formation. In addition, knockdown of the glucocorticoid receptor restores regenerative capability to nonregenerative, glucocorticoid-exposed zebrafish. To test whether the classical anti-inflammatory action of glucocorticoids is responsible for blocking regeneration, we prevented acute inflammation following amputation by antisense repression of the Pu.1 gene. Although loss of Pu.1 prevents the inflammatory response, regeneration is not affected. Collectively, these results indicate that signaling from exogenous glucocorticoids impairs blastema formation and limits regenerative capacity through an acute inflammation-independent mechanism. These studies also demonstrate the feasibility of exploiting chemical genetics to define the pathways that govern vertebrate regeneration.


Assuntos
Técnicas Genéticas , Regeneração , Animais , Anti-Inflamatórios/farmacologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Relação Dose-Resposta a Droga , Extremidades/embriologia , Glucocorticoides/metabolismo , Macrófagos/citologia , Masculino , Modelos Anatômicos , Modelos Biológicos , Neutrófilos/metabolismo , Transdução de Sinais , Cicatrização , Peixe-Zebra
4.
Toxicol Sci ; 95(1): 215-26, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17003102

RESUMO

Adult zebra fish completely regenerate their caudal (tail) fin following partial amputation. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) inhibits this regenerative process. Proper regulation of transcription, innervation, vascularization, and extracellular matrix (ECM) composition is essential for complete fin regeneration. Previous microarray studies suggest that genes involved in ECM regulation are misexpressed following activation of the aryl hydrocarbon receptor. To investigate whether TCDD blocks regeneration by impairing ECM remodeling, male zebra fish were i.p. injected with 50 ng/g TCDD or vehicle, and caudal fins were amputated. By 3 days postamputation (dpa), the vascular network in the regenerating fin of TCDD-exposed fish was disorganized compared to vehicle-exposed animals. Furthermore, immunohistochemical staining revealed that axonal outgrowth was impacted by TCDD as early as 3 dpa. Histological analysis demonstrated that TCDD exposure leads to an accumulation of collagen at the end of the fin ray just distal to the amputation site by 3 dpa. Mature lepidotrichial-forming cells (fin ray-forming cells) were not observed in the fins of TCDD-treated fish. The capacity to metabolize ECM was also altered by TCDD exposure. Quantitative real-time PCR studies revealed that the aryl hydrocarbon pathway is active and that matrix-remodeling genes are expressed in the regenerate following TCDD exposure.


Assuntos
Carcinógenos Ambientais/toxicidade , Matriz Extracelular/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/agonistas , Regeneração/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia , Animais , Colágeno/metabolismo , Citocromo P-450 CYP1A1/biossíntese , Eletroforese em Gel de Poliacrilamida , Indução Enzimática/efeitos dos fármacos , Extremidades/anatomia & histologia , Extremidades/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Metaloproteinases da Matriz/metabolismo , Reação em Cadeia da Polimerase , Proteoglicanas/metabolismo , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Fatores de Tempo , Ativador de Plasminogênio Tecidual/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Peixe-Zebra/metabolismo
5.
Toxicol Sci ; 92(1): 254-69, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16443690

RESUMO

Adult zebrafish can completely regenerate their caudal fin following amputation. This complex process is initiated by the formation of an epithelial wound cap over the amputation site by 12 h post amputation (hpa). Once the cap is formed, mesenchymal cells proliferate and migrate from sites distal to the wound plane and accumulate under the epithelial cap forming the blastemal structure within 48 hpa. Blastemal cells proliferate and differentiate, replacing the amputated tissues, which are populated with angiogenic vessels and innervating nerves during the regenerative outgrowth phase which is completed around 14 days post amputation (dpa). Regenerative outgrowth does not occur in TCDD-exposed zebrafish. To identify the molecular pathways that are perturbed by TCDD exposure, male zebrafish were ip injected with 50 ng/g TCDD or vehicle and caudal fins were amputated. Regenerating fin tissue was collected at 1, 3, and 5 dpa for mRNA abundance analysis. Microarray analysis and quantitative real time PCR revealed that wound healing and regeneration alone altered the expression of nearly 900 genes by at least two-fold between 1 and 5 dpa. TCDD altered the abundance of 370 genes at least two-fold. Among these, several known aryl hydrocarbon responsive genes were identified in addition to several genes involved in extracellular matrix composition and metabolism. The profile of misexpressed genes is suggestive of impaired cellular differentiation and extracellular matrix composition potentially regulated by Sox9b.


Assuntos
Matriz Extracelular/efeitos dos fármacos , Perfilação da Expressão Gênica , Dibenzodioxinas Policloradas/toxicidade , Regeneração/efeitos dos fármacos , Animais , Sequência de Bases , Primers do DNA , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra
6.
Mol Pharmacol ; 69(1): 257-65, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16214955

RESUMO

There is considerable literature supporting the conclusion that inappropriate activation of the aryl hydrocarbon receptor (AHR) alters cellular signaling. We have established previously that fin regeneration is specifically inhibited by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in adult zebrafish and have used this in vivo endpoint to evaluate interactions between AHR and growth-controlling pathways. Because there are experimental limitations in studying regeneration in adult animals, we have developed a larval model to evaluate the effect of AHR activation on tissue regeneration. Two-day-old zebrafish regenerate their amputated caudal fins within 3 days. Here, we demonstrate that TCDD specifically blocks regenerative growth in larvae. The AHR pathway in zebrafish is considerably more complex than in mammals, with at least three zebrafish AHR genes (zfAHR1a, zfAHR1b, and zfAHR2) and two ARNT genes (zfARNT1 and zfARNT2). Although it was presumed that the block in regeneration was mediated by AHR activation, it had not been experimentally demonstrated. Using antisense morpholinos and mutant fish lines, we report that zfAHR2 and zfARNT1 are the in vivo dimerization partners that are required for inhibition of regeneration by TCDD. Several pathways including fibroblast growth factor (FGF) signaling are essential for fin regeneration. Even though impaired FGF signaling and TCDD exposure both inhibit fin regeneration, their morphometric response is distinct, suggesting that the mechanisms of impairment are different. With the plethora of molecular and genetic techniques that can be applied to larval-stage embryos, this in vivo regeneration system can be further exploited to understand cross-talk between AHR and other signaling pathways.


Assuntos
Receptores de Hidrocarboneto Arílico/agonistas , Regeneração/efeitos dos fármacos , Peixe-Zebra/fisiologia , Animais , Sequência de Bases , Primers do DNA , Dibenzodioxinas Policloradas/toxicidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra/crescimento & desenvolvimento
7.
Pharmacogenet Genomics ; 15(4): 245-56, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15864117

RESUMO

OBJECTIVES: Previous work defined two flavin-containing monooxygenase 2 (FMO2) alleles. The major allele, FMO2*2 (g.23,238C>T), encodes truncated inactive protein (p.X472) whereas the minor allele, FMO2*1, present in African- and Hispanic-American populations, encodes active protein (p.Q472). Recently, four common (27 to 51% incidence) FMO2 single nucleotide polymorphisms (SNPs) were detected in African-Americans (N=50); they encode the following protein variants: p.71Ddup, p.V113fs, p.S195L and p.N413 K. Our objectives were to: (1) determine the incidence of these SNPs in 29 Hispanic individuals previously genotyped as g.23,238C (p.Q472) and 124 previously genotyped as homozygous g.23,238 T (p.X472); (2) determine FMO2 haplotypes in this population; and (3) assess the functional impact of SNPs in expressed proteins. METHODS: SNPs were detected via allele-specific oligonucleotide amplification coupled with real-time or electrophoretic product detection, or single strand conformation polymorphism. RESULTS: The g.7,700_7,702dupGAC SNP (p.71Ddup) was absent. The remaining SNPs were present but, except for g.13,732C>T (p.S195L), were less common in the current Hispanic study population versus the previously described African-Americans. Only expressed p.N413 K was as active as p.Q472, as determined by methimazole- and ethylenethiourea-dependent oxidation. Haplotype determination demonstrated that the g.10,951delG (p.V113fs), g.13,732C>T (p.S195L) and g.22,060T>G (p.N413 K) variants segregated with g.23,238C>T (p.X472). CONCLUSIONS: SNPs would not alter FMO2 activity in individuals possessing at least one FMO2*1 allele. It is likely that these SNPs will segregate similarly in African-American populations. Therefore, estimates that 26% of African-Americans and 2-7% of Hispanic-Americans have at least one FMO2*1 allele should closely reflect the percentages producing active FMO2 protein.


Assuntos
Haplótipos , Oxigenases/genética , Alelos , Antitireóideos/farmacologia , Primers do DNA/química , DNA Complementar/metabolismo , Etilenotioureia/farmacologia , Vetores Genéticos , Genótipo , Hispânico ou Latino , Homozigoto , Humanos , Metimazol/farmacologia , Mutagênese Sítio-Dirigida , Mutação , Farmacogenética , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único , Polimorfismo Conformacional de Fita Simples , Temperatura
8.
Toxicol Sci ; 78(1): 68-77, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14691204

RESUMO

The aryl hydrocarbon receptor (AHR) interacts with the aryl hydrocarbon receptor nuclear translocator (ARNT) to form a heterodimer that binds to promoters in target genes to alter transcription in response to xenobiotics such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The ARNT protein also forms heterodimers with other proteins such as HIF-1alpha and HIF-2alpha to alter gene expression in response to low oxygen conditions. Because ARNT is shared between multiple signaling pathways it is possible that activation of one ARNT-requiring pathway could inhibit the activation of other pathways that depend on ARNT. One hypothesis to explain TCDD toxicity in early life stage fish is that TCDD activation of zfAHR2 sequesters zfARNT2 away from the hypoxia signaling pathway. To test this hypothesis we measured the ability of TCDD to prevent induction of heme oxygenase by hypoxia (40% saturation), as well as the ability of hypoxia to increase the sensitivity of zebrafish to the effects of TCDD during the first week of life. As a further test of the model we examined mutant zebrafish that lack zfARNT2 for phenotypes that resemble the effects of TCDD exposure. Our results demonstrate that sequestration of zfARNT2 is not causing TCDD toxicity. TCDD did not inhibit hypoxia induction of heme oxygenase, hypoxia and TCDD exposures were not additive in causing developmental toxicity, and mutant embryos that lack zfARNT2 do not develop defects mimicking TCDD toxicity. However, our results demonstrate some level of cross talk between the two pathways in the zebrafish embryo. Hypoxia decreased TCDD induction of zfCYP1A mRNA, and decreased the potency of TCDD in causing edema. It is not clear whether this is mediated through competition for zfARNT2, or through other mechanisms.


Assuntos
Embrião não Mamífero/fisiologia , Poluentes Ambientais/farmacologia , Hipóxia/fisiopatologia , Dibenzodioxinas Policloradas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/fisiologia , Animais , Northern Blotting , Sistema Enzimático do Citocromo P-450/biossíntese , Sistema Enzimático do Citocromo P-450/genética , Relação Dose-Resposta a Droga , Fertilização , Cinética , Larva , Receptor Cross-Talk/fisiologia , Receptores de Hidrocarboneto Arílico/fisiologia , Teratogênicos/toxicidade , Água
9.
Toxicol Sci ; 68(2): 403-19, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12151636

RESUMO

To better understand the role of the aryl hydrocarbon receptor (AHR) signaling pathway in causing tissue-specific signs of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) toxicity in zebrafish, the temporal and spatial expression of the zebrafish aryl hydrocarbon receptor 2 (zfAHR2), aryl hydrocarbon receptor nuclear translocator 2 (zfARNT2), and an AHR regulated gene, cytochrome P4501A (zfCYP1A), were assessed in larvae exposed to vehicle or TCDD (1.55 nM) from 3-4 h postfertilization (hpf). Coexpression of a transcriptionally active AHR pathway was apparent by the expression of zfCYP1A mRNA and protein in certain larval tissues. zfCYP1A protein was first detected in the skin and vasculature of TCDD-exposed larvae at 36 hpf. Vascular-specific zfCYP1A protein expression continued from 36 to120 hpf at which time it was also detected in the heart, kidney, and liver. zfCYP1A mRNA was observed in TCDD treated larvae as early as 24 hpf in the developing vascular system. Vascular specific zfCYP1A mRNA expression in the head, trunk, and tail by 36 hpf in TCDD-exposed larvae, confirmed immunohistochemical localization. The expression of zfAHR2 and zfARNT2 mRNAs was generally similar in control and TCDD-exposed larvae. Coexpression of zfAHR2, zfARNT2, and zfCYP1A mRNAs was evident in TCDD-exposed larvae by 36 hpf and in the vasculature, heart, and trunk kidney by 48 hpf, well before the first signs of overt developmental toxicity are observed. In addition to their function in response to AHR agonists, zfAHR2 and zfARNT2 may be involved in development and function of the nervous system. zfAHR2 and zfARNT2 were detected in the brain, spinal cord, and sensory organs. However, TCDD-induced zfCYP1A expression was not detected in these tissues. Taken together, these results are consistent with the notion that the cardiovascular system is a primary target of TCDD developmental toxicity in zebrafish.


Assuntos
Sistema Enzimático do Citocromo P-450/biossíntese , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/biossíntese , Teratogênicos/toxicidade , Fatores de Transcrição/biossíntese , Peixe-Zebra , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto , Sistema Cardiovascular/embriologia , Sistema Cardiovascular/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Larva/efeitos dos fármacos , Larva/metabolismo , RNA Mensageiro/biossíntese , Receptores de Hidrocarboneto Arílico/genética , Fatores de Transcrição/genética , Peixe-Zebra/embriologia , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/genética
10.
Mol Pharmacol ; 62(2): 234-49, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12130674

RESUMO

Fish are known to have two distinct classes of aryl hydrocarbon receptors, and their roles in mediating xenobiotic toxicity remain unclear. In this study, we have identified and characterized a cDNA tentatively named zebrafish AHR1 (zfAHR1). Analysis of the deduced amino acid sequence reveals that the protein is distinct from zfAHR2 and is more closely related to the mammalian aryl hydrocarbon receptor (AHR). zfAHR1 and zfAHR2 share 40% amino acid identity overall and 58% in the N-terminal half. The zfAHR1 gene maps to linkage group 16 in a region that shares conserved synteny with human chromosome 7 containing the human AHR, suggesting that the zfAHR1 is the ortholog of the human AHR. zfAHR2 maps to a separate linkage group (LG22). Both zfAHR mRNAs are expressed in early development, but they are differentially expressed in adult tissues. zfAHR2 can dimerize with zfARNT2b and binds with specificity to dioxin-responsive elements (DREs). Under identical conditions, zfAHR1/zfARNT2b/DRE complexes are formed; however, the interactions are considerably weaker. In COS-7 cells expressing zfARNT2b and zfAHR2, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure leads to a significant induction of dioxin-responsive reporter genes. In identical experiments, TCDD exposure fails to induce the reporter gene in zfAHR1-expressing cells. Ligand-binding experiments suggested that the differential zfAHR activities are attributable to differences in TCDD binding because only zfAHR2 exhibits high-affinity binding to [(3)H]TCDD or beta-naphthoflavone. Finally, using chimeric zfAHR1/zfAHR2 constructs, the lack of TCDD-mediated transcriptional activity was localized to the ligand-binding and C-terminal domains of zfAHR1.


Assuntos
Receptores de Hidrocarboneto Arílico/genética , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Ligação Competitiva , Células COS , DNA Complementar/isolamento & purificação , Dioxinas/farmacologia , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Ensaio Radioligante , Receptores de Hidrocarboneto Arílico/classificação , Receptores de Hidrocarboneto Arílico/metabolismo , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Ativação Transcricional
11.
J Biol Chem ; 277(15): 13210-8, 2002 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-11823471

RESUMO

Two aryl hydrocarbon receptors (rtAHR2alpha and rtAHR2beta) have been identified in the rainbow trout (Oncorhynchus mykiss). These receptors share 98% amino acid identity, yet their functional properties differ. Both rtAHR2alpha and rtAHR2beta bind 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), dimerize with rainbow trout ARNTb (rtARNTb), and recognize dioxin response elements in vitro. However, in a transient transfection assay the two proteins show differential ability to recognize enhancers, produce transactivation, and respond to TCDD. To identify the sequence differences that confer the functional differences between rtAHR2alpha and rtAHR2beta, we constructed chimeric rtAHRs, in which segments of one receptor form was replaced with the corresponding part from the other isoform. This approach progressively narrowed the region being examined to a single residue, corresponding to position 111 in rtAHR2beta. Altering this residue in rtAHR2beta from the lysine to glutamate found in rtAHR2alpha produced an rtAHR2beta with the properties of rtAHR2alpha. All other known AHRs resemble rtAHR2alpha and carry glutamate at this position, located at the N terminus of the PAS-A domain. We tested the effect of altering this glutamate in the human and zebrafish AHRs to lysine. This lysine substitution produced AHRs with transactivation properties that were similar to rtAHR2beta. These results identify a critical residue in AHR proteins that has an important impact on transactivation, enhancer site recognition, and regulation by ligand.


Assuntos
Aminoácidos/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Células COS , Primers do DNA , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oncorhynchus mykiss , Conformação Proteica , Receptores de Hidrocarboneto Arílico/química , Receptores de Hidrocarboneto Arílico/genética , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA