Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Biol Chem ; 292(35): 14362-14370, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28659344

RESUMO

Nitric oxide (NO) modulates many physiological events through production of cGMP from its receptor, the NO-sensitive guanylyl cyclase (GC1). NO also appears to function in a cGMP-independent manner, via S-nitrosation (SNO), a redox-based modification of cysteine thiols. Previously, we have shown that S-nitrosated GC1 (SNO-GC1) is desensitized to NO stimulation following prolonged NO exposure or under oxidative/nitrosative stress. In animal models of nitrate tolerance and angiotensin II-induced hypertension, decreased vasodilation in response to NO correlates with GC1 thiol oxidation, but the physiological mechanism that resensitizes GC1 to NO and restores basal activity is unknown. Because GC1 interacts with the oxidoreductase protein-disulfide isomerase, we hypothesized that thioredoxin-1 (Trx1), a cytosolic oxidoreductase, could be involved in restoring GC1 basal activity and NO sensitivity because the Trx/thioredoxin reductase (TrxR) system maintains thiol redox homeostasis. Here, by manipulating activity and levels of the Trx1/TrxR system and by using a Trx1-Trap assay, we demonstrate that Trx1 modulates cGMP synthesis through an association between Trx1 and GC1 via a mixed disulfide. A proximity ligation assay confirmed the endogenous Trx1-GC1 complex in cells. Mutational analysis suggested that Cys609 in GC1 is involved in the Trx1-GC1 association and modulation of GC1 activity. Functionally, we established that Trx1 protects GC1 from S-nitrosocysteine-induced desensitization. A computational model of Trx1-GC1 interaction illustrates a possible mechanism for Trx1 to maintain basal GC1 activity and prevent/rescue GC1 desensitization to NO. The etiology of some oxidative vascular diseases may very well be explained by the dysfunction of the Trx1-GC1 association.


Assuntos
Gasotransmissores/metabolismo , Modelos Moleculares , Miócitos Cardíacos/enzimologia , Óxido Nítrico/metabolismo , Guanilil Ciclase Solúvel/metabolismo , Tiorredoxinas/metabolismo , Substituição de Aminoácidos , Animais , Animais Recém-Nascidos , Células COS , Domínio Catalítico , Células Cultivadas , Chlorocebus aethiops , Cisteína/química , Cisteína/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Oxirredução , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos Wistar , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Guanilil Ciclase Solúvel/química , Guanilil Ciclase Solúvel/genética , Tiorredoxinas/química , Tiorredoxinas/genética
2.
Bioorg Med Chem Lett ; 28(8): 1303-1308, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29571572

RESUMO

To overcome the chemical and metabolic stability issues of l-cystine dimethyl ester (CDME) and l-cystine methyl ester (CME), a series of l-cystine diamides with or without Nα-methylation was designed, synthesized, and evaluated for their inhibitory activity of l-cystine crystallization. l-Cystine diamides 2a-i without Nα-methylation were found to be potent inhibitors of l-cystine crystallization while Nα-methylation of l-cystine diamides resulted in derivatives 3b-i devoid of any inhibitory activity of l-cystine crystallization. Computational modeling indicates that Nα-methylation leads to significant decrease in binding of the l-cystine diamides to l-cystine crystal surface. Among the l-cystine diamides 2a-i, l-cystine bismorpholide (CDMOR, LH707, 2g) and l-cystine bis(N'-methylpiperazide) (CDNMP, LH708, 2h) are the most potent inhibitors of l-cystine crystallization.


Assuntos
Amidas/química , Cistina/análogos & derivados , Cistina/química , Amidas/síntese química , Cristalização , Cistina/síntese química , Cistinúria/complicações , Cálculos Renais/etiologia , Cálculos Renais/prevenção & controle , Modelos Moleculares
3.
Proc Natl Acad Sci U S A ; 112(9): 2693-8, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25691739

RESUMO

Atherosclerosis, the build-up of occlusive, lipid-rich plaques in arterial walls, is a focal trigger of chronic coronary, intracranial, and peripheral arterial diseases, which together account for the leading causes of death worldwide. Although the directed treatment of atherosclerotic plaques remains elusive, macrophages are a natural target for new interventions because they are recruited to lipid-rich lesions, actively internalize modified lipids, and convert to foam cells with diseased phenotypes. In this work, we present a nanomedicine platform to counteract plaque development based on two building blocks: first, at the single macrophage level, sugar-based amphiphilic macromolecules (AMs) were designed to competitively block oxidized lipid uptake via scavenger receptors on macrophages; second, for sustained lesion-level intervention, AMs were fabricated into serum-stable core/shell nanoparticles (NPs) to rapidly associate with plaques and inhibit disease progression in vivo. An AM library was designed and fabricated into NP compositions that showed high binding and down-regulation of both MSR1 and CD36 scavenger receptors, yielding minimal accumulation of oxidized lipids. When intravenously administered to a mouse model of cardiovascular disease, these AM NPs showed a pronounced increase in lesion association compared with the control nanoparticles, causing a significant reduction in neointimal hyperplasia, lipid burden, cholesterol clefts, and overall plaque occlusion. Thus, synthetic macromolecules configured as NPs are not only effectively mobilized to lipid-rich lesions but can also be deployed to counteract atheroinflammatory vascular diseases, highlighting the promise of nanomedicines for hyperlipidemic and metabolic syndromes.


Assuntos
Aterosclerose/tratamento farmacológico , Carboidratos , Macrófagos/metabolismo , Nanopartículas , Placa Aterosclerótica/tratamento farmacológico , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Antígenos CD36/genética , Antígenos CD36/metabolismo , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/patologia , Lipídeos , Macrófagos/patologia , Camundongos , Camundongos Knockout , Neointima/genética , Neointima/metabolismo , Neointima/patologia , Oxirredução , Placa Aterosclerótica/sangue , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Receptores Depuradores Classe A/genética , Receptores Depuradores Classe A/metabolismo
4.
Nanomedicine ; 13(2): 343-352, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27520722

RESUMO

Inspired by high promise using naturally occurring antimicrobial peptides (AMPs) to treat infections caused by antimicrobial-resistant bacteria, cationic amphiphiles (CAms) were strategically designed as synthetic mimics to overcome associated limitations, including high manufacture cost and low metabolic stability. CAms with facially amphiphilic conformation were expected to demonstrate membrane-lytic properties and thus reduce tendency of resistance development. By systematically tuning the hydrophobicity, CAms with optimized compositions exhibited potent broad-spectrum antimicrobial activity (with minimum inhibitory concentrations in low µg/mL range) as well as negligible hemolytic activity. Electron microscope images revealed the morphological and ultrastructure changes of bacterial membranes induced by CAm treatment and validated their membrane-disrupting mechanism. Additionally, an all-atom molecular dynamics simulation was employed to understand the CAm-membrane interaction on molecular level. This study shows that these CAms can serve as viable scaffolds for designing next generation of AMP mimics as antimicrobial alternatives to combat drug-resistant pathogens.


Assuntos
Anti-Infecciosos/química , Peptídeos Catiônicos Antimicrobianos , Hemólise , Interações Hidrofóbicas e Hidrofílicas , Testes de Sensibilidade Microbiana , Estrutura Molecular
5.
J Biol Chem ; 289(20): 14360-9, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24692561

RESUMO

The glycoprotein hormones are all structurally related heterodimers consisting of an α-subunit and a ligand-specific ß-subunit that confers their unique biological activity. Crystal structures showed how the ß-subunit surrounds a part of the α-subunit, and we showed the existence of the two mechanisms responsible for that assembly. In human choriogonadotropin, the ß-subunit is folded before the subunits dock, and the α-subunit becomes incorporated into the dimer by a mechanism we termed "threading," passing between parts of the preassembled ß-subunit. Here, we show that the human lutropin ß-subunit is not folded completely prior to its interaction with the α-subunit and show that docking of the subunits enables the α-subunit to serve as a chaperone to the ß-subunit. Based on data described here, we propose that the α-subunit facilitates formation of the human lutropin ß-subunit by two mechanisms. First, the cystine knot of the α-subunit potentiates formation of the ß-subunit cystine knot, and second, contacts between α-subunit loop 2 and a hydrophobic tail in the ß-subunit facilitate formation of the seatbelt latch disulfide, which stabilizes the heterodimer. The primary influence of the α-subunit was seen when the hydrophobic tail was present or absent, but the secondary mechanism was required only when the hydrophobic tail of the ß-subunit was present. During the evolution of human choriogonadotropin, neither of these α-subunit roles was necessary for folding of the ß-subunit. The complex mechanism for lutropin assembly may be required to provide an additional control on its positive feedback function in vertebrate reproduction.


Assuntos
Gonadotropina Coriônica/química , Hormônio Luteinizante/química , Multimerização Proteica , Gonadotropina Coriônica/metabolismo , Humanos , Hormônio Luteinizante/metabolismo , Modelos Moleculares , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
6.
Adv Nanobiomed Res ; 2(6)2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36051821

RESUMO

Neuroinflammation is one of the hallmarks contributing to Parkinson's Disease (PD) pathology, where microglial activation occurs as one of the earliest events, triggered by extracellular alpha synuclein (aSYN) binding to the CD36 receptor. Here, CD36-binding nanoparticles (NPs) containing synthetic tartaric acid-based amphiphilic polymers (AMs) were rationally designed to inhibit this aSYN-CD36 binding. In silico docking revealed that four AMs with varying alkyl side chain lengths presented differential levels of CD36 binding affinity and that an optimal alkyl chain length would promote the strongest inhibitory activity towards aSYN-CD36 interactions. In vitro competitive binding assays indicated that the inhibitory activity of AM-based NPs plateaued at intermediate side chain lengths of 12- and 18-carbons, supporting the in silico docking predictions. These 12- and 18-carbon length AM NPs also had significantly stronger effects on reducing aSYN internalization and inhibiting the production of the proinflammatory molecules TNF-α and nitric oxide from aSYN-challenged microglia. All four NPs modulated the gene expression of aSYN-challenged microglia, downregulating the expression of the proinflammatory genes TNF, IL-6, and IL-1ß, and upregulating the expression of the anti-inflammatory genes TGF-ß and Arg1. Overall, this work represents a novel polymeric nanotechnology platform that can be used to modulate aSYN-induced microglial activation in PD.

7.
J Comput Aided Mol Des ; 25(6): 533-54, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21660515

RESUMO

The Online Chemical Modeling Environment is a web-based platform that aims to automate and simplify the typical steps required for QSAR modeling. The platform consists of two major subsystems: the database of experimental measurements and the modeling framework. A user-contributed database contains a set of tools for easy input, search and modification of thousands of records. The OCHEM database is based on the wiki principle and focuses primarily on the quality and verifiability of the data. The database is tightly integrated with the modeling framework, which supports all the steps required to create a predictive model: data search, calculation and selection of a vast variety of molecular descriptors, application of machine learning methods, validation, analysis of the model and assessment of the applicability domain. As compared to other similar systems, OCHEM is not intended to re-implement the existing tools or models but rather to invite the original authors to contribute their results, make them publicly available, share them with other users and to become members of the growing research community. Our intention is to make OCHEM a widely used platform to perform the QSPR/QSAR studies online and share it with other users on the Web. The ultimate goal of OCHEM is collecting all possible chemoinformatics tools within one simple, reliable and user-friendly resource. The OCHEM is free for web users and it is available online at http://www.ochem.eu.


Assuntos
Bases de Dados Factuais , Internet , Modelos Químicos , Disseminação de Informação , Gestão da Informação , Relação Quantitativa Estrutura-Atividade , Interface Usuário-Computador
8.
Antimicrob Agents Chemother ; 54(5): 1965-72, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20231400

RESUMO

We previously demonstrated that the biguanide-based compound NB325 inhibits human immunodeficiency virus type 1 (HIV-1) infection by interacting with the CXCR4 viral coreceptor. This interaction also appeared to be persistent, since HIV-1 infection was inhibited even when the virus was introduced subsequent to the removal of NB325 from the cell culture medium. The present studies were conducted to determine the extent and mechanism of this prolonged antiviral activity. Persistent inhibition of HIV-1 infection by NB325 was concentration dependent and was apparent up to 8 h after removal of the compound. Flow cytometric analyses of stimulated CD4(+) T lymphocytes exposed to NB325 demonstrated concentration-dependent reductions in CXCR4 extracellular loop 2 epitope recognition that were maintained up to 24 h after removal of the compound. CXCL12-induced chemotaxis was also persistently inhibited following pre-exposure to NB325. These results demonstrate that persistent inhibition of X4 HIV-1 infection by NB325 involves extended perturbation of the viral coreceptor CXCR4.


Assuntos
Antivirais/farmacologia , Biguanidas/farmacologia , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Receptores CXCR4/metabolismo , Antivirais/metabolismo , Antivirais/toxicidade , Biguanidas/metabolismo , Biguanidas/toxicidade , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/fisiologia , Epitopos/efeitos dos fármacos , Citometria de Fluxo , Infecções por HIV/metabolismo , Humanos , Concentração Inibidora 50
9.
Antimicrob Agents Chemother ; 53(2): 631-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19047650

RESUMO

The present studies were conducted to better define the mechanism of action of polyethylene hexamethylene biguanide (PEHMB) (designated herein as NB325), which was shown in previous studies to inhibit infection by the human immunodeficiency virus type 1 (HIV-1). Fluorescence-activated flow cytometric analyses of activated human CD4(+) T lymphocytes exposed to NB325 demonstrated concentration-dependent reductions in CXCR4 epitope recognition in the absence of altered recognition of selected CD4 or CD3 epitopes. NB325 also inhibited chemotaxis of CD4(+) T lymphocytes induced by the CXCR4 ligand CXCL12. However, NB325 did not cause CXCR4 internalization (unlike CXCL12) and did not interfere with CXCL12 binding. Additional flow cytometric analyses using antibodies with distinct specificities for extracellular domains of CXCR4 demonstrated that NB325 specifically interfered with antibody binding to extracellular loop 2 (ECL2). This interaction was confirmed using competitive binding analyses, in which a peptide derived from CXCR4 ECL2 competitively inhibited NB325-mediated reductions in CXCR4 epitope recognition. Collectively, these results demonstrate that the biguanide-based compound NB325 inhibits HIV-1 infection by specifically interacting with the HIV-1 coreceptor CXCR4.


Assuntos
Fármacos Anti-HIV/farmacologia , Biguanidas/farmacologia , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Receptores CCR4/efeitos dos fármacos , Ligação Competitiva/efeitos dos fármacos , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Quimiotaxia/efeitos dos fármacos , Citometria de Fluxo , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , Humanos , Peptídeos/química
10.
Drug Metab Dispos ; 37(6): 1295-304, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19251824

RESUMO

The pregnane X receptor (PXR; PXR.1) can be activated by structurally diverse lipophilic ligands. PXR.2, an alternatively spliced form of PXR, lacks 111 nucleotides encoding 37 amino acids in the ligand binding domain. PXR.2 bound a classic CYP3A4 PXR response element (PXRE) in electrophoretic mobility shift assays, but transfected PXR.2 failed to transactivate a CYP3A4-promoter-luciferase reporter plasmid in HepG2 cells treated with various PXR ligands. Cotransfection experiments showed that PXR.2 behaved as a dominant negative, interfering with PXR.1/rifampin activation of CYP3A4-PXRE-LUC. In HepG2 and LS180 cells stably transduced with PXR.1, PXR target genes (CYP3A4, MDR1, CYP2B6, and UGT1A1) were higher than mock-transduced cells in the absence of ligand and were further induced in the presence of rifampin. In contrast, PXR.2 stably introduced into the same host cells failed to induce target genes over levels in mock-transfected cells after drug treatment. Our homology modeling suggests that ligands bind PXR.1 more favorably, probably because of the presence of a key disordered loop region, which is missing in PXR.2. Yeast two-hybrid assays revealed that, even in the presence of ligand, the corepressors remain tightly bound to PXR.2, and coactivators are unable to bind at helix 12. In summary, PXR.2 can bind to PXREs but fails to transactivate target genes because ligands do not bind the ligand binding domain of PXR.2 productively, corepressors remain tightly bound, and coactivators are not recruited to PXR.2.


Assuntos
Processamento Alternativo , Citocromo P-450 CYP3A/química , Isoformas de Proteínas/farmacologia , Receptores de Esteroides/química , Ativação Transcricional/efeitos dos fármacos , Domínio Catalítico , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Humanos , Microssomos Hepáticos , Receptor de Pregnano X , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Conformação Proteica , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Rifampina/farmacologia , Transfecção , Células Tumorais Cultivadas
11.
Pharm Res ; 26(9): 2216-24, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19603258

RESUMO

PURPOSE: Acetylcholinesterase (AChE) is both a therapeutic target for Alzheimer's disease and a target for organophosphorus, carbamates and chemical warfare agents. Prediction of the likelihood of compounds interacting with this enzyme is therefore important from both therapeutic and toxicological perspectives. MATERIALS AND METHODS: Support vector machine classification and regression models with molecular descriptors derived from Shape Signatures and the Molecular Operating Environment (MOE) application software were built and tested using a set of piperidine AChE inhibitors (N = 110). RESULTS: The combination of the alignment free Shape Signatures and 2D MOE descriptors with the Support Vector Regression method outperforms the models based solely on 2D and internal 3D (i3D) MOE descriptors, and is comparable with the best previously reported PLS model based on CoMFA molecular descriptors (r(2)(test,SVR) = 0.48 vs. r(2)(test,PLS) = 0.47 from Sutherland et al. J Med Chem 47:5541-5554, 2004). Support Vector Classification algorithms proved superior to a classifier based on scores from the molecular docking program GOLD, with the overall prediction accuracies being Q(SVC(10CV)) = 74% and Q(SVC(LNO)) = 67% vs. Q(GOLD) = 56%. CONCLUSIONS: These new machine learning models with combined descriptor schemes may find utility for predicting novel AChE inhibitors.


Assuntos
Inibidores da Colinesterase/farmacologia , Humanos , Análise de Regressão
12.
J Mol Graph Model ; 90: 9-17, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30959269

RESUMO

An application of contiguous filling of space with convex polyhedra, also known as Frank-Kasper (FK) atomic domains is demonstrated here for modeling of atomic molecular structures. Both regular, when all polyhedron edges have equal length, and strained, depending on the topology of the polyhedron the length of its edges may slightly fluctuate from the common length, polyhedra are used. Polyhedra are connected to each other in agreement with Plateau's laws to form a contiguous uninterrupted space. An application of a new approach is demonstrated for a modeling of structures of graphite, graphene, graphane, diamond and two types of ice. The proposed approach allows to demonstrate a mutual arrangement of atoms in graphite layers, transitions between allotropic states of carbon atoms, to calculate the distances between layers in graphene and positions of water molecules in a square ice.


Assuntos
Grafite/química , Água/química , Carbono/química , Gelo , Estrutura Molecular
13.
J Med Chem ; 62(15): 7250-7263, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31298541

RESUMO

A series of C2-symmetric inhibitors was designed and evaluated for inhibitory activity against the programmed cell death-1/programmed death-ligand 1(PD-1/PD-L1) protein-protein interaction (PPI) in a homogenous time-resolved fluorescence (HTRF) assay and PD-1 signaling in cell-based coculture assays. C2-symmetric inhibitors 2a (LH1306) and 2b (LH1307) exhibited IC50 values of 25 and 3.0 nM, respectively, in the HTRF assay. While 2a was ∼3.8-fold more potent than previously reported inhibitor 1a, 2b could not be differentiated from 1b due to their high potency and the limit of our HTRF assay conditions. In one cell-based coculture PD-1 signaling assay, 2a and 2b were 8.2- and 2.8-fold more potent in inhibiting PD-1 signaling than 1a and 1b, respectively. NMR and X-ray cocrystal structural studies provided more structural insights into the interaction between 2b and PD-L1; 2b binds to PD-L1 at the PD-1 binding site and induces the formation of a more symmetrically arranged PD-L1 homodimer than that previously reported for other inhibitors.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Desenho de Fármacos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Antígeno B7-H1/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Células Jurkat , Receptor de Morte Celular Programada 1/metabolismo , Domínios e Motivos de Interação entre Proteínas/fisiologia , Estrutura Secundária de Proteína , Difração de Raios X/métodos
14.
Mol Pharmacol ; 74(3): 662-72, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18579710

RESUMO

Very few antagonists have been identified for the human pregnane X receptor (PXR). These molecules may be of use for modulating the effects of therapeutic drugs, which are potent agonists for this receptor (e.g., some anticancer compounds and macrolide antibiotics), with subsequent effects on transcriptional regulation of xenobiotic metabolism and transporter genes. A recent novel pharmacophore for PXR antagonists was developed using three azoles and consisted of two hydrogen bond acceptor regions and two hydrophobic features. This pharmacophore also suggested an overall small binding site that was identified on the outer surface of the receptor at the AF-2 site and validated by docking studies. Using computational approaches to search libraries of known drugs or commercially available molecules is preferred over random screening. We have now described several new smaller antagonists of PXR discovered with the antagonist pharmacophore with in vitro activity in the low micromolar range [S-p-tolyl 3',5-dimethyl-3,5'-biisoxazole-4'-carbothioate (SPB03255) (IC(50), 6.3 microM) and 4-(3-chlorophenyl)-5-(2,4-dichlorobenzylthio)-4H-1,2,4-triazol-3-ol (SPB00574) (IC(50), 24.8 microM)]. We have also used our computational pharmacophore and docking tools to suggest that most of the known PXR antagonists, such as coumestrol and sulforaphane, could also interact on the outer surface of PXR at the AF-2 domain. The involvement of this domain was also suggested by further site-directed mutagenesis work. We have additionally described an FDA approved prodrug, leflunomide (IC(50), 6.8 microM), that seems to be a PXR antagonist in vitro. These observations are important for predicting whether further molecules may interact with PXR as antagonists in vivo with potential therapeutic applications.


Assuntos
Biologia Computacional/métodos , Desenho de Fármacos , Receptores de Esteroides/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Imidazóis/química , Imidazóis/farmacologia , Ligantes , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/metabolismo , Receptor de Pregnano X
15.
Chem Res Toxicol ; 21(6): 1304-14, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18461975

RESUMO

Shape Signatures is a new computational tool that is being evaluated for applications in computational toxicology and drug discovery. The method employs a customized ray-tracing algorithm to explore the volume enclosed by the surface of a molecule and then uses the output to construct compact histograms (i.e., signatures) that encode for molecular shape and polarity. In the present study, we extend the application of the Shape Signatures methodology to the domain of computational models for cardiotoxicity. The Shape Signatures method is used to generate molecular descriptors that are then utilized with widely used classification techniques such as k nearest neighbors ( k-NN), support vector machines (SVM), and Kohonen self-organizing maps (SOM). The performances of these approaches were assessed by applying them to a data set of compounds with varying affinity toward the 5-HT(2B) receptor as well as a set of human ether-a-go-go-related gene (hERG) potassium channel inhibitors. Our classification models for 5-HT(2B) represented the first attempt at global computational models for this receptor and exhibited average accuracies in the range of 73-83%. This level of performance is comparable to using commercially available molecular descriptors. The overall accuracy of the hERG Shape Signatures-SVM models was 69-73%, in line with other computational models published to date. Our data indicate that Shape Signatures descriptors can be used with SVM and Kohonen SOM and perform better in classification problems related to the analysis of highly clustered and heterogeneous property spaces. Such models may have utility for predicting the potential for cardiotoxicity in drug discovery mediated by the 5-HT(2B) receptor and hERG.


Assuntos
Cardiotônicos/química , Cardiotônicos/toxicidade , Simulação por Computador , Coração/efeitos dos fármacos , Biologia Computacional , Humanos , Modelos Biológicos , Estrutura Molecular , Miocárdio/metabolismo , Relação Quantitativa Estrutura-Atividade , Receptor 5-HT2B de Serotonina/metabolismo , Transativadores/metabolismo , Regulador Transcricional ERG
16.
J Mol Graph Model ; 26(2): 591-4, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17433745

RESUMO

Volume learning algorithm (VLA) artificial neural network and partial least squares (PLS) methods were compared using the leave-one-out cross-validation procedure for prediction of relative potency of xenoestrogenic compounds to the estrogen receptor. Using Wilcoxon signed rank test we showed that VLA outperformed PLS by producing models with statistically superior results for a structurally diverse set of compounds comprising eight chemical families. Thus, CoMFA/VLA models are successful in prediction of the endocrine disrupting potential of environmental pollutants and can be effectively applied for testing of prospective chemicals prior their exposure to the environment.


Assuntos
Algoritmos , Estrogênios/química , Análise dos Mínimos Quadrados , Disruptores Endócrinos/química , Disruptores Endócrinos/metabolismo , Poluentes Ambientais/química , Poluentes Ambientais/metabolismo , Estrogênios/metabolismo , Estrogênios não Esteroides/química , Estrogênios não Esteroides/metabolismo , Modelos Moleculares , Estrutura Molecular , Redes Neurais de Computação , Ligação Proteica , Relação Quantitativa Estrutura-Atividade , Receptores de Estrogênio/química , Receptores de Estrogênio/metabolismo , Xenobióticos/química , Xenobióticos/metabolismo
17.
Polymer (Guildf) ; 48(19): 5788-5801, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-19568328

RESUMO

This work is a part of a series of publications devoted to the development of surrogate (semi-empirical) models for the prediction of fibrinogen adsorption onto polymer surfaces. Since fibrinogen is one of the key proteins involved in platelet activation and the formation of thrombosis, the modeling of fibrinogen adsorption on the surface of blood contacting medical devices is of high theoretical and practical significance. We report here, for the first time, on the incorporation three-dimensional structures of polymers obtained from atomistic simulations into conventional mesoscopic-scale calculations. Low energy conformations derived from Molecular Dynamics simulations for 45 representatives of a combinatorial library of polyarylates were used in an improved modeling procedure (referred to as "3D surrogate model") instead of simplistic two-dimensional representations of polymer structures, which were used in several previous models (collectively referred to as "2D surrogate models"). In the framework of this 3D model we created 12 model sets of polymers to account for their chirality, conformational diversity and the structural influence of a solvent. For each polymer set, three-dimensional molecular descriptors were generated and then ranked with respect to the experimental fibrinogen adsorption data by means of a Monte Carlo Decision Tree. The most significant descriptors identified by Decision Tree and the experimental dataset were utilized to predict fibrinogen adsorption using an Artificial Neural Network (ANN). The best prediction achieved by the 3D surrogate model demonstrated a noticeable improvement in the predictive quality as compared to the previously used 2D model (as evidenced by the increase in the average Pearson correlation coefficient from 0.67+/-0.13 to 0.54+/-0.12). The predictive quality of the 3D surrogate model compares favorably with the best results previously reported for extended 2D model that combines an ANN with Partial Least Squares (PLS) regression and principal component (PC) analysis. The significance of the newly developed 3D model is that it allows high accuracy prediction of fibrinogen adsorption without the need for experimentally derived descriptors and it has better predictive quality than the original 2D surrogate model due to utilization of realistic polymer representations.

18.
Front Immunol ; 8: 1521, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29176978

RESUMO

The Tyro3, Axl, and Mertk (TAM) receptors are homologous type I receptor tyrosine kinases that have critical functions in the clearance of apoptotic cells in multicellular organisms. TAMs are activated by their endogenous ligands, growth arrest-specific 6 (Gas6), and protein S (Pros1), that function as bridging molecules between externalized phosphatidylserine (PS) on apoptotic cells and the TAM ectodomains. However, the molecular mechanisms by which Gas6/Pros1 promote TAM activation remains elusive. Using TAM/IFNγR1 reporter cell lines to monitor functional TAM activity, we found that Gas6 activity was exquisitely dependent on vitamin K-mediated γ-carboxylation, whereby replacing vitamin K with anticoagulant warfarin, or by substituting glutamic acid residues involved in PS binding, completely abrogated Gas6 activity as a TAM ligand. Furthermore, using domain and point mutagenesis, Gas6 activity also required both an intact Gla domain and intact EGF-like domains, suggesting these domains function cooperatively in order to achieve TAM activation. Despite the requirement of γ-carboxylation and the functional Gla domain, non-γ-carboxylated Gas6 and Gla deletion/EGF-like domain deletion mutants still retained their ability to bind TAMs and acted as blocking decoy ligands. Finally, we found that distinct sources of PS-positive cells/vesicles (including apoptotic cells, calcium-induced stressed cells, and exosomes) bound Gas6 and acted as cell-derived or exosome-derived ligands to activate TAMs. Taken together, our findings indicate that PS is indispensable for TAM activation by Gas6, and by inference, provides new perspectives on how PS, regulates TAM receptors and efferocytosis.

19.
Sci Rep ; 7: 43908, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28272423

RESUMO

TAM receptors (Tyro-3, Axl, and Mertk) are a family of three homologous type I receptor tyrosine kinases that are implicated in several human malignancies. Overexpression of TAMs and their major ligand Growth arrest-specific factor 6 (Gas6) is associated with more aggressive staging of cancers, poorer predicted patient survival, acquired drug resistance and metastasis. Here we describe small molecule inhibitors (RU-301 and RU-302) that target the extracellular domain of Axl at the interface of the Ig-1 ectodomain of Axl and the Lg-1 of Gas6. These inhibitors effectively block Gas6-inducible Axl receptor activation with low micromolar IC50s in cell-based reporter assays, inhibit Gas6-inducible motility in Axl-expressing cell lines, and suppress H1299 lung cancer tumor growth in a mouse xenograft NOD-SCIDγ model. Furthermore, using homology models and biochemical verifications, we show that RU301 and 302 also inhibit Gas6 inducible activation of Mertk and Tyro3 suggesting they can act as pan-TAM inhibitors that block the interface between the TAM Ig1 ectodomain and the Gas6 Lg domain. Together, these observations establish that small molecules that bind to the interface between TAM Ig1 domain and Gas6 Lg1 domain can inhibit TAM activation, and support the further development of small molecule Gas6-TAM interaction inhibitors as a novel class of cancer therapeutics.


Assuntos
Carcinogênese/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Isoxazóis/farmacologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , c-Mer Tirosina Quinase/metabolismo , Animais , Sítios de Ligação , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas/química , Receptores Proteína Tirosina Quinases/química , Transplante Heterólogo , c-Mer Tirosina Quinase/química , Receptor Tirosina Quinase Axl
20.
J Proteomics ; 138: 40-7, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-26917471

RESUMO

Soluble Guanylyl Cyclase (sGC) is the main receptor for nitric oxide (NO). NO activates sGC to synthesize cGMP, triggering a plethora of signals. Recently, we discovered that NO covalently modifies select sGC cysteines via a post-translational modification termed S-nitrosation or S-nitrosylation. Earlier characterization was conducted on a purified sGC treated with S-nitrosoglutathione, and identified three S-nitrosated cysteines (SNO-Cys). Here we describe a more biologically relevant mapping of sGC SNO-Cys in cells to better understand the multi-faceted interactions between SNO and sGC. Since SNO-Cys are labile during LC/MS/MS, MS analysis of nitrosation typically occurs after a biotin switch reaction, in which a SNO-Cys is converted to a biotin-Cys. Here we report the identification of ten sGC SNO-Cys in rat neonatal cardiomyocytes using an Orbitrap MS. A majority of the SNO-Cys identified is located at the solvent-exposed surface of the sGC, and half of them in the conserved catalytic domain, suggesting biological significance. These findings provide a solid basis for future studies of the regulations and functions of diverse sGC S-nitrosation events in cells.


Assuntos
Miócitos Cardíacos/enzimologia , Óxido Nítrico/metabolismo , Guanilil Ciclase Solúvel/metabolismo , Animais , Cisteína/análogos & derivados , Cisteína/química , Cisteína/metabolismo , Miócitos Cardíacos/citologia , Óxido Nítrico/química , Nitrosação , Ratos , Ratos Wistar , S-Nitrosoglutationa/química , S-Nitrosoglutationa/metabolismo , S-Nitrosotióis/química , S-Nitrosotióis/metabolismo , Guanilil Ciclase Solúvel/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA