Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 11(21)2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36359850

RESUMO

The turn-on mutations of the KRAS gene, coding a small GTPase coupling growth factor signaling, are contributing to nearly 25% of all human cancers, leading to highly malignant tumors with poor outcomes. Targeting of oncogenic KRAS remains a most challenging task in oncology. Recently, the specific G12C mutant KRAS inhibitors have been developed but with a limited clinical outcome because they acquire drug resistance. Alternatively, exploiting a metabolic breach of KRAS-mutant cancer cells related to a glucose-dependent sensitivity to oxidative stress is becoming a promising indirect cancer targeting approach. Here, we discuss the use of a vitamin C (VC) acting in high dose as an oxidative "Trojan horse" agent for KRAS-mutant cancer cells that can be potentiated with another oxidizing drug arsenic trioxide (ATO) to obtain a potent and selective cytotoxic impact. Moreover, we outline the advantages of VC's non-natural enantiomer, D-VC, because of its distinctive pharmacokinetics and lower toxicity. Thus, the D-VC and ATO combination shows a promising path to treat KRAS-mutant cancers in clinical settings.


Assuntos
Ácido Ascórbico , Neoplasias , Humanos , Trióxido de Arsênio/farmacologia , Trióxido de Arsênio/uso terapêutico , Ácido Ascórbico/farmacologia , Ácido Ascórbico/uso terapêutico , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Estresse Oxidativo , Vitaminas/farmacologia , Oxirredução , Neoplasias/tratamento farmacológico , Neoplasias/genética
2.
RSC Adv ; 11(42): 25921-25932, 2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-35479483

RESUMO

Recent advances in convolutional neural networks have inspired the application of deep learning to other disciplines. Even though image processing and natural language processing have turned out to be the most successful, there are many other domains that have also benefited; among them, life sciences in general and chemistry and drug design in particular. In concordance with this observation, from 2018 the scientific community has seen a surge of methodologies related to the generation of diverse molecular libraries using machine learning. However to date, attention mechanisms have not been employed for the problem of de novo molecular generation. Here we employ a variant of transformers, an architecture recently developed for natural language processing, for this purpose. Our results indicate that the adapted Transmol model is indeed applicable for the task of generating molecular libraries and leads to statistically significant increases in some of the core metrics of the MOSES benchmark. The presented model can be tuned to either input-guided or diversity-driven generation modes by applying a standard one-seed and a novel two-seed approach, respectively. Accordingly, the one-seed approach is best suited for the targeted generation of focused libraries composed of close analogues of the seed structure, while the two-seeds approach allows us to dive deeper into under-explored regions of the chemical space by attempting to generate the molecules that resemble both seeds. To gain more insights about the scope of the one-seed approach, we devised a new validation workflow that involves the recreation of known ligands for an important biological target vitamin D receptor. To further benefit the chemical community, the Transmol algorithm has been incorporated into our cheML.io web database of ML-generated molecules as a second generation on-demand methodology.

3.
Arch Immunol Ther Exp (Warsz) ; 68(4): 25, 2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32815043

RESUMO

Cathepsin G (CatG) is involved in controlling numerous processes of the innate and adaptive immune system. These features include the proteolytic activity of CatG and play a pivotal role in alteration of chemokines as well as cytokines, clearance of exogenous and internalized pathogens, platelet activation, apoptosis, and antigen processing. This is in contrast to the capability of CatG acting in a proteolytic-independent manner due to the net charge of arginine residues in the CatG sequence which interferes with bacteria. CatG is a double-edged sword; CatG is also responsible in pathophysiological conditions, such as autoimmunity, chronic pulmonary diseases, HIV infection, tumor progression and metastasis, photo-aged human skin, Papillon-Lefèvre syndrome, and chronic inflammatory pain. Here, we summarize the latest findings for functional responsibilities of CatG in immunity, including bivalent regulation of major histocompatibility complex class I molecules, which underscore an additional novel role of CatG within the immune system.


Assuntos
Catepsina G/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/imunologia , Neoplasias/metabolismo , Linfócitos T Reguladores/imunologia , Viroses/metabolismo , Animais , Apresentação de Antígeno , Autoimunidade , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Lactoferrina/metabolismo
4.
Mol Pharmacol ; 96(5): 655-663, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31575620

RESUMO

Aldehyde hydrogenases (ALDHs) belong to a large gene family involved in oxidation of both endogenous and exogenous compounds in mammalian tissues. Among ALDHs, the rat ALDH1A7 gene displays a curious strain dependence in phenobarbital (PB)-induced hepatic expression: the responsive RR strains exhibit induction of both ALDH1A7 and CYP2B mRNAs and activities, whereas the nonresponsive rr strains show induction of CYP2B only. Here, we investigated the responsiveness of ALDH1A1, ALDH1A7, CYP2B1, and CYP3A23 genes to prototypical P450 inducers, expression of nuclear receptors CAR and pregnane X receptor, and structure of the ALDH1A7 promoter in both rat strains. ALDH1A7 mRNA, associated protein and activity were strongly induced by PB and modestly induced by pregnenolone 16α-carbonitrile in the RR strain but negligibly in the rr strain, whereas induction of ALDH1A1 and P450 mRNAs was similar between the strains. Reporter gene and chromatin immunoprecipitation assays indicated that the loss of ALDH1A7 inducibility in the rr strain is profoundly linked with a 16-base pair deletion in the proximal promoter and inability of the upstream DNA sequences to recruit constitutive androstane receptor-retinoid X receptor heterodimers. SIGNIFICANCE STATEMENT: Genetic variation in rat ALDH1A7 promoter sequences underlie the large strain-dependent differences in expression and inducibility by phenobarbital of the aldehyde dehydrogenase activity. This finding has implications for the design and interpretation of pharmacological and toxicological studies on the effects and disposition of aldehydes.


Assuntos
Família Aldeído Desidrogenase 1/biossíntese , Família Aldeído Desidrogenase 1/genética , Regulação Enzimológica da Expressão Gênica , Variação Genética/fisiologia , Animais , Masculino , Ratos , Ratos Wistar , Especificidade da Espécie
5.
J Med Chem ; 62(15): 6854-6875, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30916559

RESUMO

For many individuals, in particular during winter, supplementation with the secosteroid vitamin D3 is essential for the prevention of bone disorders, muscle weakness, autoimmune diseases, and possibly also different types of cancer. Vitamin D3 acts via its metabolite 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3] as potent agonist of the transcription factor vitamin D receptor (VDR). Thus, vitamin D directly affects chromatin structure and gene regulation at thousands of genomic loci, i.e., the epigenome and transcriptome of its target tissues. Modifications of 1,25(OH)2D3 at its side-chain, A-ring, triene system, or C-ring, alone and in combination, as well as nonsteroidal mimics provided numerous potent VDR agonists and some antagonists. The nearly 150 crystal structures of VDR's ligand-binding domain with various vitamin D compounds allow a detailed molecular understanding of their action. This review discusses the most important vitamin D analogs presented during the past 10 years and molecular insight derived from new structural information on the VDR protein.


Assuntos
Receptores de Calcitriol/química , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Vitamina D/metabolismo , Animais , Calcifediol/análogos & derivados , Calcifediol/metabolismo , Calcitriol/análogos & derivados , Calcitriol/metabolismo , Humanos , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
6.
Mol Cell Endocrinol ; 453: 22-35, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28257826

RESUMO

1α,25-Dihydroxvitamin D3 (1,25(OH)2D3) is the hormonally active form of vitamin D3. Its synthesis and its metabolites, their transport and elimination as well as action on transcriptional regulation involves the harmonic cooperation of diverse proteins with vitamin D binding capacities such as vitamin D binding protein (DBP), cytochrome P450 enzymes or the nuclear vitamin receptor (VDR). The genomic mechanism of 1,25(OH)2D3 action involves its binding to VDR that functionally acts as a heterodimer with retinoid X receptor. The crystal structures of the most important proteins for vitamin D3, VDR, DBP, CYP2R1 and CYP24A1, have provided identification of mechanisms of actions of these proteins and those mediating VDR-regulated transcription. This review will present the structural information on recognition of the vitamin D3 and metabolites by CYP proteins and DBP as well as the structural basis of VDR activation by 1,25(OH)2D3 and metabolites. Additionally, we will describe, the implications of the VDR mutants associated with hereditary vitamin D-resistant rickets (HVDRR) that display impaired function.


Assuntos
Colecalciferol/química , Colecalciferol/metabolismo , Receptores de Calcitriol/metabolismo , Proteína de Ligação a Vitamina D/química , Regulação Alostérica , Colecalciferol/genética , Colestanotriol 26-Mono-Oxigenase/química , Colestanotriol 26-Mono-Oxigenase/metabolismo , Família 2 do Citocromo P450/química , Família 2 do Citocromo P450/metabolismo , Regulação da Expressão Gênica , Humanos , Modelos Moleculares , Mutação , Receptores de Calcitriol/química , Receptores de Calcitriol/genética , Raquitismo Hipofosfatêmico/genética , Proteína de Ligação a Vitamina D/metabolismo , Vitamina D3 24-Hidroxilase/química , Vitamina D3 24-Hidroxilase/metabolismo
7.
Expert Opin Ther Pat ; 26(11): 1291-1306, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27454349

RESUMO

INTRODUCTION: Vitamin D3 activates via its hormonal form 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3), the transcription factor vitamin D receptor (VDR). VDR is expressed in most human tissues and has more than 1,000 target genes. Thus, 1α,25(OH)2D3 and its synthetic analogs have a broad physiological impact. The crystal structures of the VDR ligand-binding domain (LBD), and its various ligands, allows further the understanding of the receptor's molecular actions. Areas covered: We discuss the most important novel VDR ligands and the further insight derived from new structural information on VDR. Expert opinion: There is an increasing appreciation of the impact of vitamin D and its receptor VDR not only in bone biology, but also for metabolic diseases, immunological disorders, and cancer. Detailed structural analysis of the interaction of additional novel ligands with VDR highlight helices 6 and 7 of the LBD as being most critical for stabilizing the receptor for an efficient interaction with co-activator proteins, i.e. for efficient agonistic action. This permits the design of even more effective VDR agonists. In addition, chemists took more liberty in replacing major parts of the 1α,25(OH)2D3 molecule, such as the A- and CD-rings or the side chain, with significantly different structures, such as carboranes, and still obtained functional VDR agonists.


Assuntos
Calcitriol/análogos & derivados , Desenho de Fármacos , Receptores de Calcitriol/agonistas , Animais , Calcitriol/metabolismo , Calcitriol/farmacologia , Colecalciferol/metabolismo , Humanos , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/patologia , Ligantes , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/patologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Patentes como Assunto , Receptores de Calcitriol/metabolismo
8.
Eur J Pharm Sci ; 95: 72-81, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27221369

RESUMO

Microscale freeze-drying makes rapid process cycles possible for early-stage formulation development. To investigate the effects of equipment scale and cooling rate on the solid state properties and the protein's secondary structure of a sample, three binary formulations of catalase were prepared and freeze-dried with sucrose, mannitol, or (2-hydroxypropyl)-ß-cyclodextrin (HP-ß-CD). The protein's secondary structure was assessed using attenuated total reflection Fourier transform infrared spectroscopy (FTIR-ATR). The solid state properties were assessed using differential scanning calorimetry (DSC) and X-ray diffraction (XRD). The results were interpreted with respect to the biological activity of catalase after its reconstitution. According to the results of both the protein secondary structure and the reconstituted biological activity, scale-up could be achieved with the sucrose-catalase formulation when it was prepared at a high cooling rate and with the mannitol-catalase formulation when prepared at a low cooling rate. However, differences in the polymorph composition of crystalline mannitol were noted. No cooling rate influence was found with the HP-ß-CD formulation. The results clearly indicate that the effects of the cooling rate should be closely examined during microscale formulation development and scale-up of the freeze-drying process.


Assuntos
Temperatura Baixa , Excipientes/química , Liofilização/métodos , Composição de Medicamentos , Excipientes/análise , Liofilização/tendências , Estrutura Secundária de Proteína , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Espectroscopia de Infravermelho com Transformada de Fourier/tendências , Difração de Raios X/métodos , Difração de Raios X/tendências
9.
Xenobiotica ; 46(3): 200-10, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26153444

RESUMO

1. Nuclear receptors CAR (NR1I3) and PXR (NR1I2) are major ligand-activated transcriptional regulators of xenobiotic metabolism and disposition and modulators of endobiotic metabolism. Differences in xenobiotic selectivity between the human and rodent receptors are well recognized but there is lack of such information on properties of CAR and PXR in important domestic animals. 2. The pig and bovine receptors were cloned and their ligand profiles were systematically compared to corresponding human and mouse forms utilizing a panel of xenobiotics and structural analysis. 3. Pig CAR and PXR resemble their human counterparts which can be rationalized by only modest amino acid changes between critical residues of the human ligand-binding pockets (H203Q for CAR, L210V and M243I for PXR). 4. In contrast, bovine CAR shows a blunted response to CAR agonists and inverse agonists. These changes are likely due to disruptive mutations at or near critical hydrogen bond-forming residues (N165I, Y326F). The unresponsiveness of bovine PXR to human- and mouse-selective agonists may be related to substitutions at important ligand-contacting residues R410Q and F305V, respectively. 5. Our findings have implications for regulation of drug-metabolizing enzymes and transporters and pharmacokinetics in cattle and pigs.


Assuntos
Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Clonagem Molecular , Receptor Constitutivo de Androstano , Regulação da Expressão Gênica , Humanos , Inativação Metabólica , Ligantes , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Receptor de Pregnano X , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/genética , Alinhamento de Sequência , Suínos
10.
J Pharm Sci ; 104(11): 3710-3721, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26305147

RESUMO

Microscale (MS) freeze-drying offers rapid process cycles for early-stage formulation development. The effects of the MS approach on the secondary structures of two model proteins, lysozyme and catalase, were compared with pilot-scale (PS) vial freeze-drying. The secondary structures were assessed by attenuated total reflection Fourier transformed infrared spectroscopy. Formulations were made with increasing sucrose-protein ratios. Freeze-drying protocols involved regular cooling without thermal treatment and annealing with MS and PS equipment, and cooling rate variations with the MS. Principal component analysis of smoothed second-derivative amide I spectra revealed sucrose-protein ratio-dependent shifts toward α-helical structures. Transferability of sucrose-protein formulations from MS to PS vial freeze-drying was evidenced at regular cooling rates. Local differences in protein secondary structures between the bottom and top of sucrose-catalase samples could be detected at the sucrose-catalase ratios of 1 and 2, this being related to the initial filling height and ice crystal morphology. Annealing revealed temperature, protein, formulation, and sample location-dependent effects influencing surface morphology at the top, or causing protein secondary structure perturbation at the bottom. With the MS approach, protein secondary structure differences at different cooling rates could be detected for sucrose-lysozyme samples at the sucrose-lysozyme ratio of 1.


Assuntos
Catalase/química , Liofilização/métodos , Muramidase/química , Animais , Bovinos , Galinhas , Excipientes/química , Liofilização/economia , Análise de Componente Principal , Estrutura Secundária de Proteína , Sacarose/química
11.
Can J Physiol Pharmacol ; 93(5): 311-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25741777

RESUMO

Vitamin D3 is one of the few natural compounds that has, via its metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) and the transcription factor vitamin D receptor (VDR), a direct effect on gene regulation. For efficiently applying the therapeutic and disease-preventing potential of 1,25(OH)2D3 and its synthetic analogs, the key steps in vitamin D signaling need to be understood. These are the different types of molecular interactions with the VDR, such as (i) the complex formation of VDR with genomic DNA, (ii) the interaction of VDR with its partner transcription factors, (iii) the binding of 1,25(OH)2D3 or its synthetic analogs within the ligand-binding pocket of the VDR, and (iv) the resulting conformational change on the surface of the VDR leading to a change of the protein-protein interaction profile of the receptor with other proteins. This review will present the latest genome-wide insight into vitamin D signaling, and will discuss its therapeutic implications.


Assuntos
Estudo de Associação Genômica Ampla , Receptores de Calcitriol/química , Receptores de Calcitriol/metabolismo , Transdução de Sinais/fisiologia , Animais , Estudo de Associação Genômica Ampla/tendências , Humanos , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores de Calcitriol/genética
12.
Cell Rep ; 10(4): 516-26, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25620699

RESUMO

The bioactive form of vitamin D [1,25(OH)2D3] regulates mineral and bone homeostasis and exerts potent anti-inflammatory and antiproliferative properties through binding to the vitamin D receptor (VDR). The 3D structures of the VDR ligand-binding domain with 1,25(OH)2D3 or gemini analogs unveiled the molecular mechanism underlying ligand recognition. On the basis of structure-function correlations, we generated a point-mutated VDR (VDR(gem)) that is unresponsive to 1,25(OH)2D3, but the activity of which is efficiently induced by the gemini ligands. Moreover, we show that many VDR target genes are repressed by unliganded VDR(gem) and that mineral ion and bone homeostasis are more impaired in VDR(gem) mice than in VDR null mice, demonstrating that mutations abolishing VDR ligand binding result in more severe skeletal defects than VDR null mutations. As gemini ligands induce VDR(gem) transcriptional activity in mice and normalize their serum calcium levels, VDR(gem) is a powerful tool to further unravel both liganded and unliganded VDR signaling.


Assuntos
Receptores de Calcitriol/química , Receptores de Calcitriol/metabolismo , Animais , Imunoprecipitação da Cromatina , Cristalografia por Raios X , Polarização de Fluorescência , Genótipo , Células HEK293 , Humanos , Células MCF-7 , Camundongos , Camundongos Knockout , Mutação/genética , Ligação Proteica/genética , Receptores de Calcitriol/genética , Espectrometria de Massas por Ionização por Electrospray , Vitamina D/metabolismo
13.
Front Physiol ; 5: 191, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936188

RESUMO

Crystal structures represent the static picture in the life of a molecule giving a sneak preview what it might be in reality. Hence, it is very hard to extrapolate from these photos toward dynamic processes such as transcriptional regulation. Mechanistically VDR may be considered as molecular machine able to perform ligand-, DNA- and protein recognition, and interaction in a multi-task manner. Taking this into account the functional net effect will be the combination of all these processes. The long awaited answer to explain the differences in physiological effects for various ligands was one of the biggest disappointment that crystal structures provided since no substantial distinction could be made for the conformation of the active VDR-ligand complexes. This may have come from the limitation on the complexity of the available ligand-VDR structures. The recent studies with full length VDR-RXRα showed somewhat more comprehensive perspective for the 3D organization and possible function of the VDR-RXRα-cofactor complex. In addition to in vitro approaches, also computational tools had been introduced with the aim to get understanding on the mechanic and dynamic properties of the VDR complexes with some success. Using these methods and based on measurable descriptors such as pocket size and positions of side chains it is possible to note subtle differences between the structures. The meaning of these differences has not been fully understood yet but the possibility of a "butterfly effect" may have more extreme consequences in terms of VDR signaling. In this review, the three functional aspects (ligand-, DNA- and protein recognition, and binding) will be discussed with respect to available data as well as possible implication and questions that may be important to address in the future.

14.
FEBS Lett ; 588(9): 1523-8, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24681097

RESUMO

The modulation of protein deacetylase SIRT1 has a vast therapeutic potential in treatment of several aging-associated diseases. Active regulator of SIRT1 (AROS) is a small endogenous protein which was originally reported to activate SIRT1 through a direct interaction in cancer cells. We show that the interaction between the two proteins is weak and does not alter the activity of SIRT1 in non-cancerous human cells. The results of different in vitro SIRT1 activity assays disclosed AROS as an inhibitor of SIRT1. The functional relationship between AROS and SIRT1 proved to be dependent on the biological context and experimental setting.


Assuntos
Proteínas Nucleares/metabolismo , Sirtuína 1/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Células Epiteliais/enzimologia , Células HEK293 , Histonas/química , Humanos , NAD/química , Proteínas Nucleares/química , Ligação Proteica , Processamento de Proteína Pós-Traducional , Epitélio Pigmentado da Retina/citologia , Sirtuína 1/química , Fatores de Transcrição/química , Proteína Supressora de Tumor p53/metabolismo
15.
Eur J Pharm Biopharm ; 87(2): 347-56, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24607807

RESUMO

Downscaled freeze-drying was demonstrated to be a valuable alternative for formulation development and optimization. Although the pore structure is known to exert a major influence on the freeze-drying cycle, little is known about the ones of microscale preparations. This study describes morphology evaluation methods for lysozyme formulations prepared in one microscale processing option and the assessment of fundamental product quality criteria. Scanning electron microscopy (SEM) revealed cooling rate dependent pore size variations at the nucleation site which diminished as the rate increased. Micro-X-ray computed tomography (µ-CT) showed that porosity generally increased in the sample from bottom to top, the pore size fractions shifted toward larger pores in elevated sample levels, and horizontal homogeneity was found throughout each sample with minor deviations in the bottom region. Furthermore, the event of microcollapse could be identified and quantified. Low residual moisture was achieved repeatedly and the procedure did not influence the post freeze-drying bioactivity. This microscale heating stage is a valuable option to reduce overall cycle times and cost, and to prepare freeze-drying formulations with high reproducibility. The mapping tools permit a quick but detailed insight into the structural features resulting from the process environment and processing conditions.


Assuntos
Liofilização , Muramidase/química , Tecnologia Farmacêutica/métodos , Animais , Química Farmacêutica , Galinhas , Desenho de Equipamento , Liofilização/instrumentação , Manitol/química , Microscopia Eletrônica de Varredura , Miniaturização , Porosidade , Conformação Proteica , Sacarose/química , Propriedades de Superfície , Tecnologia Farmacêutica/instrumentação , Fatores de Tempo , Trealose/química , Água/química , Microtomografia por Raio-X
16.
Drug Metabol Drug Interact ; 28(2): 79-93, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23729557

RESUMO

The constitutive androstane receptor (CAR; NR1I3) has emerged as one of the main drug- and xenobiotic-sensitive transcriptional regulators. It has a major effect on the expression of several oxidative and conjugative enzymes and transporters, and hence, CAR can contribute to drug/drug interactions. Novel functions for CAR are also emerging: it is able to modulate the metabolic fate of glucose, lipids, and bile acids, and it is also involved in cell-cell communication, regulation of the cell cycle, and chemical carcinogenesis. Here, we will review the recent information available on CAR and its target gene expression, its interactions with partner proteins and mechanisms of action, interindividual and species variation, and current advances in CAR ligand selectivity and methods used in interrogation of its ligands.


Assuntos
Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Evolução Biológica , Receptor Constitutivo de Androstano , DNA/metabolismo , Humanos , Ligantes , Modelos Moleculares , Receptores Citoplasmáticos e Nucleares/química , Especificidade da Espécie
17.
Expert Opin Ther Pat ; 22(4): 417-35, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22449247

RESUMO

INTRODUCTION: In the past years, the biologically active form of vitamin D(3), 1α,25-dihydroxyvitamin D(3) (1α,25(OH)(2)D(3)), has received large appreciation due to the broad physiological impact of the hormone and its nuclear receptor, the transcription factor vitamin D receptor (VDR). Recently, the understanding of VDR actions has progressed greatly, due to VDR crystal structures with various ligands. AREAS COVERED: This review will present and discuss new synthetic agonistic and antagonistic 1α,25(OH)(2)D(3) analogs in the context of the recent insights provided by VDR crystal structures. EXPERT OPINION: During the last 5 years, a large number of new 1α,25(OH)(2)D(3) analogs, many of which have an interesting functional profile, have been patented. Moreover, for a surprisingly high number of 1α,25(OH)(2)D(3) analogs, the crystal structure data of their complex with the VDR is available. This structural information provides important insight into the functional potential of the VDR ligands and explains their agonistic and antagonistic action. However, so far, only for a few VDR ligands, a rational design, based on crystal structure information, has been applied. The design of future analogs may also take the specificity of co-factor interaction into account, in order to create selective VDR modulators.


Assuntos
Calcitriol/farmacologia , Receptores de Calcitriol/efeitos dos fármacos , Animais , Calcitriol/análogos & derivados , Calcitriol/síntese química , Cristalização , Desenho de Fármacos , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular , Receptores de Calcitriol/química , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Relação Estrutura-Atividade
18.
Curr Top Med Chem ; 12(6): 528-47, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22242854

RESUMO

Vitamin D and in particular its biologically most active metabolite, 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3), are central endocrine molecules that influence many aspects of human physiology, which are not only the well-known calcium and phosphorus up-take and transport controlling bone formation, but also the control of immune functions and of cellular growth and differentiation. Basically all actions of 1α,25(OH)2D3 are mediated by the transcription factor vitamin D receptor (VDR). The crystal structure of the VDR and detailed knowledge on its molecular interactions with the ligand provide significant insight into the mechanisms of vitamin D signaling. This applies also on the action of the huge number of synthetic 1α,25(OH)2D3 analogues, which have been developed with the goal of a therapeutic application in hyper-proliferative diseases, such as psoriasis, benign prostate hyperplasia and different types of cancer, in immune functions, such as autoimmune diseases and microbial infections, or in bone disorders, such as osteoporosis. Moreover, detailed investigations on many VDR target genes and in particular the recently available genome-wide view on vitamin D signaling allows a more complete view on the potential of the nuclear hormone. In this review we discuss the latest insight into vitamin D signaling in context with the most prominent 1α,25(OH)2D3 analogues.


Assuntos
Transdução de Sinais , Vitamina D/metabolismo , Vitamina D/uso terapêutico , Animais , Doença , Humanos , Ligantes , Receptores de Calcitriol/química , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados
19.
Chemistry ; 18(2): 603-12, 2012 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-22162241

RESUMO

Based on the crystal structures of human vitamin D receptor (hVDR) bound to 1α,25-dihydroxy-vitamin D(3) (1,25 D) and superagonist ligands, we previously designed new superagonist ligands with a tetrahydrofuran ring at the side chain that optimize the aliphatic side-chain conformation through an entropy benefit. Following a similar strategy, four novel vitamin D analogues with aromatic furan side chains (3a, 3b, 4a, 4b) have now been developed. The triene system has been constructed by an efficient stereoselective intramolecular cyclization of an enol triflate (A-ring precursor) followed by a Suzuki-Miyaura coupling of the resulting intermediate with an alkenyl boronic ester (CD-side chain, upper fragment). The furan side chains have been constructed by gold chemistry. These analogues exhibit significant pro-differentiation effects and transactivation potency. The crystal structure of 3a in a complex with the ligand-binding domain of hVDR revealed that the side-chain furanic ring adopts two conformations.


Assuntos
Furanos/química , Furanos/farmacologia , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Vitamina D/farmacologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Modelos Moleculares , Ligação Proteica , Ativação Transcricional/efeitos dos fármacos
20.
Mol Pharm ; 8(6): 2424-33, 2011 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-22044162

RESUMO

The human constitutive androstane receptor (CAR, NR1I3) is one of the key regulators of xenobiotic and endobiotic metabolism. The unique properties of human CAR, such as the high constitutive activity and the complexity of signaling, as well as the lack of functional and predictive cell-based assays to study the properties of the receptor, have hindered the discovery of selective human CAR ligands. Here we report a novel human CAR inverse agonist, 1-[(2-methylbenzofuran-3-yl)methyl]-3-(thiophen-2-ylmethyl) urea (S07662), which suppresses human CAR activity, recruits the corepressor NCoR in cell-based assays, and attenuates the phenytoin- and 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime (CITCO)-induced expression of CYP2B6 mRNA in human primary hepatocytes. The properties of S07662 are also compared with those of known human CAR inverse agonists by using an array of different in vitro and in silico assays. The identified compound S07662 can be used as a chemical tool to study the biological functions of human CAR and also as a starting point for the development of new drugs for various conditions involving the receptor.


Assuntos
Descoberta de Drogas , Compostos de Metilureia/química , Receptores Citoplasmáticos e Nucleares/agonistas , Tiofenos/química , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Receptor Constitutivo de Androstano , Humanos , Isoquinolinas/química , Modelos Moleculares , Simulação de Dinâmica Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...