Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicon ; 59(2): 365-72, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22206814

RESUMO

Vertebrate xenobiotic receptors are ligand-activated nuclear receptors (NRs) that bind exogenous biologically active chemicals before activating the transcription of genes involved in xenobiotic metabolism and excretion. Typically, xenobiotic receptors have ligand binding domains (LBDs) that can accommodate a structurally diverse array of molecules and in addition display high levels of inter-taxa sequence diversity suggestive of positive selection. Pursuing the idea that xenobiotic receptors may adaptively evolve to bind toxic chemicals commonly present in an organism's environment/diet, we examined ligand binding by a xenobiotic receptor orthologue of a marine filter-feeding organism. The solitary tunicate Ciona intestinalis (Phylum Chordata) genome encodes an orthologue of the vertebrate pregnane X receptor (PXR) and vitamin D receptor (VDR), here denoted CiVDR/PXRα. In a luciferase reporter assay the CiVDR/PXRα was activated, at nanomolar concentrations, by two of four natural marine microalgal biotoxins tested (okadaic acid, EC50 = 18.2 ± 0.9 nM and pectenotoxin-2, EC50 = 37.0 ± 3.5 nM) along with 1 of 11 synthetic toxicants (esfenvalerate: EC50 = 0.59 ± 0.7 µM). Two related C. intestinalis NRs, orthologous to vertebrate farnesoid X receptor and liver X receptors, respectively, along with the PXR of a freshwater fish (zebrafish, Danio rerio), were not activated by any of the 15 chemicals tested. In contrast, human PXR was activated by okadaic acid at similar concentrations to CiVDR/PXRα (EC50 = 7.2 ± 1.1 nM) but not by pectenotoxin-2. A common features pharmacophore developed for the CiVDR/PXRα ligand consisted of an off-center hydrogen bond acceptor flanked by two hydrophobic regions. The results of this study are consistent with the original hypothesis that natural toxins, present in the diet of filter-feeding marine invertebrates, may have acted as selective agents in the molecular evolution of tunicate xenobiotic receptors. Bioassays based on tunicate xenobiotic receptor activation may find application in marine environmental monitoring and bioprospecting.


Assuntos
Ciona intestinalis/metabolismo , Toxinas Marinhas/toxicidade , Praguicidas/toxicidade , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Xenobióticos/metabolismo , Animais , Evolução Molecular , Genes Reporter , Humanos , Ligantes , Luciferases/metabolismo , Microalgas/química , Modelos Moleculares , Receptor de Pregnano X , Ligação Proteica , Receptores de Calcitriol/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo
2.
BMC Biochem ; 12: 5, 2011 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-21291553

RESUMO

BACKGROUND: The farnesoid X receptor (FXR), pregnane X receptor (PXR), and vitamin D receptor (VDR) are three closely related nuclear hormone receptors in the NR1H and 1I subfamilies that share the property of being activated by bile salts. Bile salts vary significantly in structure across vertebrate species, suggesting that receptors binding these molecules may show adaptive evolutionary changes in response. We have previously shown that FXRs from the sea lamprey (Petromyzon marinus) and zebrafish (Danio rerio) are activated by planar bile alcohols found in these two species. In this report, we characterize FXR, PXR, and VDR from the green-spotted pufferfish (Tetraodon nigriviridis), an actinopterygian fish that unlike the zebrafish has a bile salt profile similar to humans. We utilize homology modelling, docking, and pharmacophore studies to understand the structural features of the Tetraodon receptors. RESULTS: Tetraodon FXR has a ligand selectivity profile very similar to human FXR, with strong activation by the synthetic ligand GW4064 and by the primary bile acid chenodeoxycholic acid. Homology modelling and docking studies suggest a ligand-binding pocket architecture more similar to human and rat FXRs than to lamprey or zebrafish FXRs. Tetraodon PXR was activated by a variety of bile acids and steroids, although not by the larger synthetic ligands that activate human PXR such as rifampicin. Homology modelling predicts a larger ligand-binding cavity than zebrafish PXR. We also demonstrate that VDRs from the pufferfish and Japanese medaka were activated by small secondary bile acids such as lithocholic acid, whereas the African clawed frog VDR was not. CONCLUSIONS: Our studies provide further evidence of the relationship between both FXR, PXR, and VDR ligand selectivity and cross-species variation in bile salt profiles. Zebrafish and green-spotted pufferfish provide a clear contrast in having markedly different primary bile salt profiles (planar bile alcohols for zebrafish and sterically bent bile acids for the pufferfish) and receptor selectivity that matches these differences in endogenous ligands. Our observations to date present an integrated picture of the co-evolution of bile salt structure and changes in the binding pockets of three nuclear hormone receptors across the species studied.


Assuntos
Receptores de Calcitriol/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/genética , Animais , Ácidos e Sais Biliares/metabolismo , Evolução Molecular , Humanos , Ligantes , Camundongos , Modelos Moleculares , Receptor de Pregnano X , Domínios e Motivos de Interação entre Proteínas , Ratos , Receptores de Calcitriol/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/metabolismo , Especificidade da Espécie , Relação Estrutura-Atividade , Tetraodontiformes , Peixe-Zebra
3.
Environ Health Perspect ; 118(10): 1412-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20558333

RESUMO

BACKGROUND: The pregnane X receptor (PXR) is a key transcriptional regulator of many genes [e.g., cytochrome P450s (CYP2C9, CYP3A4, CYP2B6), MDR1] involved in xenobiotic metabolism and excretion. OBJECTIVES: As part of an evaluation of different approaches to predict compound affinity for nuclear hormone receptors, we used the molecular docking program GOLD and a hybrid scoring scheme based on similarity weighted GoldScores to predict potential PXR agonists in the ToxCast database of pesticides and other industrial chemicals. We present some of the limitations of different in vitro systems, as well as docking and ligand-based computational models. METHODS: Each ToxCast compound was docked into the five published crystallographic structures of human PXR (hPXR), and 15 compounds were selected based on their consensus docking scores for testing. In addition, we used a Bayesian model to classify the ToxCast compounds into PXR agonists and nonagonists. hPXR activation was determined by luciferase-based reporter assays in the HepG2 and DPX-2 human liver cell lines. RESULTS: We tested 11 compounds, of which 6 were strong agonists and 2 had weak agonist activity. Docking results of additional compounds were compared with data reported in the literature. The prediction sensitivity of PXR agonists in our sample ToxCast data set (n = 28) using docking and the GoldScore was higher than with the hybrid score at 66.7%. The prediction sensitivity for PXR agonists using GoldScore for the entire ToxCast data set (n = 308) compared with data from the NIH (National Institutes of Health) Chemical Genomics Center data was 73.8%. CONCLUSIONS: Docking and the GoldScore may be useful for prioritizing large data sets prior to in vitro testing with good sensitivity across the sample and entire ToxCast data set for hPXR agonists.


Assuntos
Bases de Dados Factuais , Receptores de Esteroides/agonistas , Teorema de Bayes , Linhagem Celular , Cristalografia por Raios X , Genes Reporter , Humanos , Receptor de Pregnano X
4.
PLoS Comput Biol ; 5(12): e1000594, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20011107

RESUMO

Transcriptional regulation of some genes involved in xenobiotic detoxification and apoptosis is performed via the human pregnane X receptor (PXR) which in turn is activated by structurally diverse agonists including steroid hormones. Activation of PXR has the potential to initiate adverse effects, altering drug pharmacokinetics or perturbing physiological processes. Reliable computational prediction of PXR agonists would be valuable for pharmaceutical and toxicological research. There has been limited success with structure-based modeling approaches to predict human PXR activators. Slightly better success has been achieved with ligand-based modeling methods including quantitative structure-activity relationship (QSAR) analysis, pharmacophore modeling and machine learning. In this study, we present a comprehensive analysis focused on prediction of 115 steroids for ligand binding activity towards human PXR. Six crystal structures were used as templates for docking and ligand-based modeling approaches (two-, three-, four- and five-dimensional analyses). The best success at external prediction was achieved with 5D-QSAR. Bayesian models with FCFP_6 descriptors were validated after leaving a large percentage of the dataset out and using an external test set. Docking of ligands to the PXR structure co-crystallized with hyperforin had the best statistics for this method. Sulfated steroids (which are activators) were consistently predicted as non-activators while, poorly predicted steroids were docked in a reverse mode compared to 5alpha-androstan-3beta-ol. Modeling of human PXR represents a complex challenge by virtue of the large, flexible ligand-binding cavity. This study emphasizes this aspect, illustrating modest success using the largest quantitative data set to date and multiple modeling approaches.


Assuntos
Relação Quantitativa Estrutura-Atividade , Receptores de Esteroides/química , Biologia Computacional , Cristalografia por Raios X , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular , Receptor de Pregnano X , Estrutura Terciária de Proteína , Receptores de Esteroides/agonistas , Receptores de Esteroides/metabolismo
5.
J Proteome Res ; 7(10): 4359-72, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18651760

RESUMO

Many proteins possess intrinsic disorder (ID) and lack a rigid three-dimensional structure in at least part of their sequence. ID has been hypothesized to influence protein-protein and protein-ligand interactions. We calculated ID for nearly 400 vertebrate and invertebrate members of the biomedically important nuclear hormone receptor (NHR) superfamily, including all 48 known human NHRs. The predictions correctly identified regions in 20 of the 23 NHRs suggested as disordered based on published X-ray and NMR structures. Of the four major NHR domains (N-terminal domain, DNA-binding domain, D-domain, and ligand-binding domain), we found ID to be highest in the D-domain, a region of NHRs critical in DNA recognition and heterodimerization, coactivator/corepressor interactions and protein-protein interactions. ID in the D-domain and LBD was significantly higher in "hub" human NHRs that have 10 or more downstream proteins in their interaction networks compared to "non-hub" NHRs that interact with fewer than 10 downstream proteins. ID in the D-domain and LBD was also higher in classic, ligand-activated NHRs than in orphan, ligand-independent NHRs in human. The correlation between ID in human and mouse NHRs was high. Less correlation was found for ID between mammalian and non-mammalian vertebrate NHRs. For some invertebrate species, particularly sea squirts ( Ciona), marked differences were observed in ID between invertebrate NHRs and their vertebrate orthologs. Our results indicate that variability of ID within NHRs, particularly in the D-domain and LBD, is likely an important evolutionary force in shaping protein-protein interactions and NHR function. This information enables further understanding of these therapeutic targets.


Assuntos
Estrutura Terciária de Proteína , Receptores Citoplasmáticos e Nucleares/química , Animais , Cristalografia por Raios X , Redes Reguladoras de Genes , Humanos , Camundongos , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Receptores Citoplasmáticos e Nucleares/metabolismo
6.
Chem Res Toxicol ; 21(7): 1457-67, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18547065

RESUMO

The pregnane X receptor (PXR) regulates the expression of genes involved in xenobiotic metabolism and transport. In vitro methods to screen for PXR agonists are used widely. In the current study, computational models for human PXR activators and PXR nonactivators were developed using recursive partitioning (RP), random forest (RF), and support vector machine (SVM) algorithms with VolSurf descriptors. Following 10-fold randomization, the models correctly predicted 82.6-98.9% of activators and 62.0-88.6% of nonactivators. The models were validated using separate test sets. The overall ( n = 15) test set prediction accuracy for PXR activators with RP, RF, and SVM PXR models is 80-93.3%, representing an improvement over models previously reported. All models were tested with a second test set ( n = 145), and the prediction accuracy ranged from 63 to 67% overall. These test set molecules were found to cover the same area in a principal component analysis plot as the training set, suggesting that the predictions were within the applicability domain. The FlexX docking method combined with logistic regression performed poorly in classifying this PXR test set as compared with RP, RF, and SVM but may be useful for qualitative interpretion of interactions within the LBD. From this analysis, VolSurf descriptors and machine learning methods had good classification accuracy and made reliable predictions within the model applicability domain. These methods could be used for high throughput virtual screening to assess for PXR activation, prior to in vitro testing to predict potential drug-drug interactions.


Assuntos
Inteligência Artificial , Avaliação Pré-Clínica de Medicamentos/métodos , Mapeamento de Interação de Proteínas , Receptores de Esteroides/antagonistas & inibidores , Receptores de Esteroides/fisiologia , Algoritmos , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Simulação por Computador , Expressão Gênica , Hepatócitos/metabolismo , Humanos , Redes Neurais de Computação , Valor Preditivo dos Testes , Receptor de Pregnano X , Reprodutibilidade dos Testes
7.
BMC Evol Biol ; 8: 103, 2008 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-18384689

RESUMO

BACKGROUND: The pregnane X receptor (PXR) shows the highest degree of cross-species sequence diversity of any of the vertebrate nuclear hormone receptors. In this study, we determined the pharmacophores for activation of human, mouse, rat, rabbit, chicken, and zebrafish PXRs, using a common set of sixteen ligands. In addition, we compared in detail the selectivity of human and zebrafish PXRs for steroidal compounds and xenobiotics. The ligand activation properties of the Western clawed frog (Xenopus tropicalis) PXR and that of a putative vitamin D receptor (VDR)/PXR cloned in this study from the chordate invertebrate sea squirt (Ciona intestinalis) were also investigated. RESULTS: Using a common set of ligands, human, mouse, and rat PXRs share structurally similar pharmacophores consisting of hydrophobic features and widely spaced excluded volumes indicative of large binding pockets. Zebrafish PXR has the most sterically constrained pharmacophore of the PXRs analyzed, suggesting a smaller ligand-binding pocket than the other PXRs. Chicken PXR possesses a symmetrical pharmacophore with four hydrophobes, a hydrogen bond acceptor, as well as excluded volumes. Comparison of human and zebrafish PXRs for a wide range of possible activators revealed that zebrafish PXR is activated by a subset of human PXR agonists. The Ciona VDR/PXR showed low sequence identity to vertebrate VDRs and PXRs in the ligand-binding domain and was preferentially activated by planar xenobiotics including 6-formylindolo-[3,2-b]carbazole. Lastly, the Western clawed frog (Xenopus tropicalis) PXR was insensitive to vitamins and steroidal compounds and was activated only by benzoates. CONCLUSION: In contrast to other nuclear hormone receptors, PXRs show significant differences in ligand specificity across species. By pharmacophore analysis, certain PXRs share similar features such as human, mouse, and rat PXRs, suggesting overlap of function and perhaps common evolutionary forces. The Western clawed frog PXR, like that described for African clawed frog PXRs, has diverged considerably in ligand selectivity from fish, bird, and mammalian PXRs.


Assuntos
Evolução Molecular , Filogenia , Receptores de Esteroides/agonistas , Receptores de Esteroides/genética , Sequência de Aminoácidos , Animais , Humanos , Ligantes , Funções Verossimilhança , Receptor de Pregnano X , Receptores de Esteroides/química , Especificidade da Espécie
8.
J Steroid Biochem Mol Biol ; 110(1-2): 83-94, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18395439

RESUMO

Liver X receptors (LXRs) are key regulators of lipid and cholesterol metabolism in mammals. Little is known, however, about the function and evolution of LXRs in non-mammalian species. The present study reports the cloning of LXRs from African clawed frog (Xenopus laevis), Western clawed frog (Xenopus tropicalis), and zebrafish (Danio rerio), and their functional characterization and comparison with human and mouse LXRs. Additionally, an ortholog of LXR in the chordate invertebrate Ciona intestinalis was cloned and functionally characterized. Ligand specificities of the frog and zebrafish LXRs were very similar to LXRalpha and LXRbeta from human and mouse. All vertebrate LXRs studied were activated robustly by the synthetic ligands T-0901317 and GW3965 and by a variety of oxysterols. In contrast, Ciona LXR was not activated by T-0901317 or GW3965 but was activated by a limited number of oxysterols, as well as some androstane and pregnane steroids. Pharmacophore analysis, homology modeling, and docking studies of Ciona LXR predict a receptor with a more restricted ligand-binding pocket and less intrinsic disorder in the ligand-binding domain compared to vertebrate LXRs. The results suggest that LXRs have a long evolutionary history, with vertebrate LXRs diverging from invertebrate LXRs in ligand specificity.


Assuntos
Proteínas de Ligação a DNA/agonistas , Proteínas de Ligação a DNA/genética , Evolução Molecular , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/genética , Androstenos/química , Androstenos/farmacologia , Animais , Benzoatos/química , Benzoatos/farmacologia , Benzilaminas/química , Benzilaminas/farmacologia , Carbazóis/química , Carbazóis/farmacologia , Linhagem Celular Tumoral , Colesterol/análogos & derivados , Colesterol/química , Colesterol/farmacologia , Ciona intestinalis , Proteínas de Ligação a DNA/metabolismo , Humanos , Hidrocarbonetos Fluorados , Hidroxicolesteróis/química , Hidroxicolesteróis/farmacologia , Receptores X do Fígado , Camundongos , Estrutura Molecular , Receptores Nucleares Órfãos , Filogenia , Receptores Citoplasmáticos e Nucleares/metabolismo , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia , Xenopus laevis , Peixe-Zebra
9.
J Lipid Res ; 49(7): 1577-87, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18362391

RESUMO

Bile salts, the major end metabolites of cholesterol, vary significantly in structure across vertebrate species, suggesting that nuclear receptors binding these molecules may show adaptive evolutionary changes. We compared across species the bile salt specificity of the major transcriptional regulator of bile salt synthesis, the farnesoid X receptor (FXR). We found that FXRs have changed specificity for primary bile salts across species by altering the shape and size of the ligand binding pocket. In particular, the ligand binding pockets of sea lamprey (Petromyzon marinus) and zebrafish (Danio rerio) FXRs, as predicted by homology models, are flat and ideal for binding planar, evolutionarily early bile alcohols. In contrast, human FXR has a curved binding pocket best suited for the bent steroid ring configuration typical of evolutionarily more recent bile acids. We also found that the putative FXR from the sea squirt Ciona intestinalis, a chordate invertebrate, was completely insensitive to activation by bile salts but was activated by sulfated pregnane steroids, suggesting that the endogenous ligands of this receptor may be steroidal in nature. Our observations present an integrated picture of the coevolution of bile salt structure and of the binding pocket of their target nuclear receptor FXR.


Assuntos
Ácidos e Sais Biliares/metabolismo , Evolução Molecular , Proteínas de Ligação a RNA/metabolismo , Vertebrados/metabolismo , Animais , Ácidos e Sais Biliares/química , Bovinos , Linhagem Celular , Cristalografia por Raios X , Ligantes , Modelos Moleculares , Estrutura Molecular , Proteínas de Ligação a RNA/química , Espectrometria de Massas por Ionização por Electrospray , Vertebrados/genética
10.
BMC Evol Biol ; 7: 222, 2007 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-17997857

RESUMO

BACKGROUND: The vitamin D receptor (VDR) and pregnane X receptor (PXR) are nuclear hormone receptors of the NR1I subfamily that show contrasting patterns of cross-species variation. VDR and PXR are thought to have arisen from duplication of an ancestral gene, evident now as a single gene in the genome of the chordate invertebrate Ciona intestinalis (sea squirt). VDR genes have been detected in a wide range of vertebrates including jawless fish. To date, PXR genes have not been found in cartilaginous fish. In this study, the ligand selectivities of VDRs were compared in detail across a range of vertebrate species and compared with those of the Ciona VDR/PXR. In addition, several assays were used to search for evidence of PXR-mediated hepatic effects in three model non-mammalian species: sea lamprey (Petromyzon marinus), zebrafish (Danio rerio), and African clawed frog (Xenopus laevis). RESULTS: Human, mouse, frog, zebrafish, and lamprey VDRs were found to have similar ligand selectivities for vitamin D derivatives. In contrast, using cultured primary hepatocytes, only zebrafish showed evidence of PXR-mediated induction of enzyme expression, with increases in testosterone 6beta-hydroxylation activity (a measure of cytochrome P450 3A activity in other species) and flurbiprofen 4-hydroxylation activity (measure of cytochrome P450 2C activity) following exposure to known PXR activators. A separate assay in vivo using zebrafish demonstrated increased hepatic transcription of another PXR target, multidrug resistance gene (ABCB5), following injection of the major zebrafish bile salt, 5alpha-cyprinol 27-sulfate. The PXR target function, testosterone hydroxylation, was detected in frog and sea lamprey primary hepatocytes, but was not inducible in these two species by a wide range of PXR activators in other animals. Analysis of the sea lamprey draft genome also did not show evidence of a PXR gene. CONCLUSION: Our results show tight conservation of ligand selectivity of VDRs across vertebrate species from Agnatha to mammals. Using a functional approach, we demonstrate classic PXR-mediated effects in zebrafish, but not in sea lamprey or African clawed frog liver cells. Using a genomic approach, we failed to find evidence of a PXR gene in lamprey, suggesting that VDR may be the original NR1I gene.


Assuntos
Evolução Biológica , Receptores de Calcitriol/metabolismo , Receptores de Esteroides/metabolismo , Animais , Linhagem Celular , Ciona intestinalis , Hepatócitos/metabolismo , Humanos , Hidroxilação , Ligantes , Camundongos , Petromyzon , Receptor de Pregnano X , Receptores de Calcitriol/efeitos dos fármacos , Receptores de Calcitriol/genética , Receptores de Esteroides/efeitos dos fármacos , Receptores de Esteroides/genética , Especificidade da Espécie , Testosterona/metabolismo , Ativação Transcricional , Transfecção , Xenopus , Peixe-Zebra
11.
Mol Pharmacol ; 72(3): 592-603, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17576789

RESUMO

The pregnane X receptor (PXR) is an important transcriptional regulator of the expression of xenobiotic metabolism and transporter genes. The receptor is promiscuous, binding many structural classes of molecules that act as agonists at the ligand-binding domain, triggering up-regulation of genes, increasing the metabolism and excretion of therapeutic agents, and causing drug-drug interactions. It has been suggested that human PXR antagonists represent a means to counteract such interactions. Several azoles have been hypothesized to bind the activation function-2 (AF-2) surface on the exterior of PXR when agonists are concurrently bound in the ligand-binding domain. In the present study, we have derived novel computational models for PXR agonists using different series of imidazoles, steroids, and a set of diverse molecules with experimental PXR agonist binding data. We have additionally defined a novel pharmacophore for the steroidal agonist site. All agonist pharmacophores showed that hydrophobic features are predominant. In contrast, a qualitative comparison with the corresponding PXR antagonist pharmacophore models using azoles and biphenyls showed that they are smaller and hydrophobic with increased emphasis on hydrogen bonding features. Azole antagonists were docked into a proposed hydrophobic binding pocket on the outer surface at the AF-2 site and fitted comfortably, making interactions with key amino acids involved in charge clamping. Combining computational and experimental data for different classes of molecules provided strong evidence for agonists and antagonists binding distinct regions on PXR. These observations bear significant implications for future discovery of molecules that are more selective and potent antagonists.


Assuntos
Receptores de Esteroides/agonistas , Receptores de Esteroides/antagonistas & inibidores , Sítios de Ligação , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Simulação por Computador , Genes Reporter , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Concentração Inibidora 50 , Neoplasias Hepáticas/patologia , Luciferases/metabolismo , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Plasmídeos , Receptor de Pregnano X , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Esteroides/química , Receptores de Esteroides/metabolismo , Ativação Transcricional
12.
Blood ; 110(7): 2650-8, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17585052

RESUMO

Oncogenic Notch1 mutations are found in most T-lineage acute lymphoblastic leukemias in humans and T-cell lymphomas in mice. However, the mechanism by which Notch1 promotes transformation or maintains malignant cell survival has not been determined fully. Here, we report that expression of the transcription factor lymphoid enhancer factor 1 (Lef1) is Notch dependent in murine T-cell lymphomas in vitro and in vivo, and that the intracellular domain of Notch1 (ICN1) is present at the Lef1 promoter. Lef1 expression is not Notch dependent in primary T-cell progenitors, but Lef1 mRNA is increased by ectopic expression of ICN1 in these cells. We show that Lef1 is required for survival of T-cell lymphoma lines, and that ectopic expression of Lef1 delays lymphoma cell death in the absence of Notch signaling, indicating that Lef1 is an important Notch target in these cells. Therefore, Notch1 co-opts Lef1 during the process of transformation to maintain survival of T-cell lymphomas.


Assuntos
Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Receptor Notch1/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Sobrevivência Celular , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Linfoma de Células T/genética , Camundongos , Camundongos Knockout , Alinhamento de Sequência , Transdução de Sinais , Células-Tronco/metabolismo
13.
Expert Opin Drug Metab Toxicol ; 2(3): 381-97, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16863441

RESUMO

The pregnane X receptor (PXR; NR1I2) is a nuclear hormone receptor (NR) that transcriptionally regulates genes encoding transporters and drug-metabolising enzymes in the liver and intestine. PXR activation leads to enhanced metabolism and elimination of xenobiotics and endogenous compounds such as hormones and bile salts. Relative to other vertebrate NRs, PXR has the broadest specificity for ligand activators by virtue of a large, flexible ligand-binding cavity. In addition, PXR has the most extensive sequence diversity across vertebrate species in the ligand-binding domain of any NR, with significant pharmacological differences between human and rodent PXRs, and especially marked divergence between mammalian and nonmammalian PXRs. The unusual properties of PXR complicate the use of in silico and animal models to predict in vivo human PXR pharmacology. Research into the evolutionary history of the PXR gene has also provided insight into the function of PXR in humans and other animals.


Assuntos
Ácidos e Sais Biliares/farmacologia , Citocromo P-450 CYP3A/metabolismo , Evolução Molecular , Regulação Enzimológica da Expressão Gênica , Hormônios Esteroides Gonadais/farmacologia , Fígado/efeitos dos fármacos , Receptores de Esteroides/agonistas , Sequência de Aminoácidos , Animais , Simulação por Computador , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Intestinos/efeitos dos fármacos , Intestinos/enzimologia , Ligantes , Fígado/enzimologia , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Filogenia , Receptor de Pregnano X , Conformação Proteica , Receptores de Esteroides/química , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Especificidade da Espécie , Xenopus laevis/metabolismo
14.
Blood ; 107(10): 4115-21, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16449526

RESUMO

Loss of E2A transcription factor activity or activation of the intracellular form of Notch1 (ICN) leads to the development of leukemia or lymphoma in humans or mice, respectively. Current models propose that ICN functions by suppressing E2A through a pre-T cell receptor (TCR)-dependent mechanism. Here we show that lymphomas arising in E2A(-/-) mice require the activation of Notch1 for their survival and have accumulated mutations in, or near, the Notch1 PEST domain, resulting in increased stability and signaling. In contrast, lymphomas arising in p53(-/-) mice show the activation of Notch1, but no mutations were identified in ICN. The requirement for Notch1 signaling in E2A(-/-) lymphomas cannot be overcome by ectopic expression of pTalpha; however, pTalpha is required for optimal survival and expansion of these cells. Our findings indicate that the activation of Notch1 is an important "second hit" for the transformation of E2A(-/-) T cell lymphomas and that Notch1 promotes survival through pre-TCR-dependent and -independent mechanisms.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Linfoma de Células T/imunologia , Receptor Notch1/genética , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Sequência de Bases , Northern Blotting , Sobrevivência Celular , Transformação Celular Neoplásica , Primers do DNA , Humanos , Linfoma de Células T/patologia , Camundongos , Camundongos Knockout , Proteína Supressora de Tumor p53/deficiência
15.
J Biol Chem ; 277(12): 9645-54, 2002 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-11744719

RESUMO

Mice have a monodisperse high density lipoprotein (HDL) profile, whereas humans have two major subfractions designated HDL(2) and HDL(3). Human apoA-I transgenic mice exhibit a human-like HDL profile, indicating that the amino acid sequence of apoA-I is a determinant of the HDL profile. Comparison of the primary sequence of mouse and human apoA-I and the previously designated "hinge" domain of apoA-I led us to hypothesize that alpha-helices 7 and 8 (7/8) are determinants of HDL subclass distribution. The following proteins were expressed in Escherichia coli: human apoA-I, T7-hAI; mouse apoA-I, T7-mAI; chimeric human apoA-I containing murine helices 7/8 in place of human helices 7/8, T7-hAI(m7/8); and the reciprocal chimera, T7-mAI(h7/8). The recombinant proteins were examined for their association with human plasma HDL subclasses. The results demonstrated that T7-hAI bound HDL(2) and HDL(3) equally well, whereas T7-mAI bound to HDL(2) preferentially. T7-hAI(m7/8) behaved like T7-mAI, and T7-mAI(h7/8) behaved like T7-hAI. Thus, alpha-helices 7/8 are strong contributors to the pattern of HDL subclass association. Self-association, alpha-helicity, cholesterol efflux, and lecithin-cholesterol acyltransferase activity of the recombinant proteins were also assessed. Human apoA-I self-associates more and activates human lecithin-cholesterol acyltransferase better than mouse apoA-I. These differential characteristics of human and mouse apoA-I are not dependent on helices 7/8.


Assuntos
Apolipoproteína A-I/química , HDL-Colesterol/química , Lipoproteínas HDL/química , Sequência de Aminoácidos , Animais , Apolipoproteína A-I/metabolismo , Western Blotting , Colesterol/metabolismo , Dicroísmo Circular , Reagentes de Ligações Cruzadas/farmacologia , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Humanos , Cinética , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...