Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Chemistry ; 29(3): e202202106, 2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36251739

RESUMEN

The mechanosensitive ion channel of large conductance (MscL) is a promising template for the development of new antibiotics due to its high conservation and uniqueness to microbes. Brilliant green (BG), a triarylmethane dye, has been identified as a new antibiotic targeted MscL. However, the detailed binding sites to MscL and the dynamic pathway of BG through the MscL channel remain unknown. Here, the dynamic interactions between BG and MscL were investigated using solid-state NMR spectroscopy and molecule dynamics (MD) simulations. Residue site-specific binding sites of BG to the MscL channel were identified by solid-state NMR. In addition, MD simulations revealed that BG conducts through the MscL channel via residues along the inner surface of the pore sequentially, in which the strong hydrophobic interactions between BG and hydrophobic residues F23 and I27 in the hydrophobic gate region of the MscL channel are major restrictions. Particularly, it was demonstrated that BG activates the MscL channel by reducing the hydrophobicity of the F23 in the gate region by water molecules that are bound to BG. Taken together, these simulations and experimental data provide novel insights into the dynamic interactions between BG and MscL, based on which new hydrophobic antibiotics and adjuvants targeting MscL can be developed.


Asunto(s)
Proteínas de Escherichia coli , Simulación de Dinámica Molecular , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Canales Iónicos/química , Espectroscopía de Resonancia Magnética , Antibacterianos/química
2.
Proc Natl Acad Sci U S A ; 117(44): 27381-27387, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33051297

RESUMEN

The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global crisis. There is no therapeutic treatment specific for COVID-19. It is highly desirable to identify potential antiviral agents against SARS-CoV-2 from existing drugs available for other diseases and thus repurpose them for treatment of COVID-19. In general, a drug repurposing effort for treatment of a new disease, such as COVID-19, usually starts from a virtual screening of existing drugs, followed by experimental validation, but the actual hit rate is generally rather low with traditional computational methods. Here we report a virtual screening approach with accelerated free energy perturbation-based absolute binding free energy (FEP-ABFE) predictions and its use in identifying drugs targeting SARS-CoV-2 main protease (Mpro). The accurate FEP-ABFE predictions were based on the use of a restraint energy distribution (RED) function, making the practical FEP-ABFE-based virtual screening of the existing drug library possible. As a result, out of 25 drugs predicted, 15 were confirmed as potent inhibitors of SARS-CoV-2 Mpro The most potent one is dipyridamole (inhibitory constant Ki = 0.04 µM) which has shown promising therapeutic effects in subsequently conducted clinical studies for treatment of patients with COVID-19. Additionally, hydroxychloroquine (Ki = 0.36 µM) and chloroquine (Ki = 0.56 µM) were also found to potently inhibit SARS-CoV-2 Mpro We anticipate that the FEP-ABFE prediction-based virtual screening approach will be useful in many other drug repurposing or discovery efforts.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Reposicionamiento de Medicamentos , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , COVID-19 , Cloroquina/farmacología , Proteasas 3C de Coronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Cisteína Endopeptidasas , Dipiridamol/farmacología , Humanos , Hidroxicloroquina/farmacología , Simulación del Acoplamiento Molecular , Estructura Molecular , Pandemias , Neumonía Viral/tratamiento farmacológico , SARS-CoV-2
3.
J Am Chem Soc ; 144(48): 22229-22241, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36413513

RESUMEN

Aquaporins are transmembrane channels that allow for the passive permeation of water and other small molecules across biological membranes. Their channel activities are sensitive to mercury ions. Intriguingly, while most aquaporins are inhibited by mercury ions, several aquaporins are activated by mercury ions. The molecular basis of the opposing aquaporin regulation by mercury remains poorly understood. Herein, we investigated AqpZ inhibition and AQP6 activation upon binding of mercury ions using solid-state NMR (ssNMR) and molecular dynamics (MD) simulations. Based on the structure of the Hg-AqpZ complex constructed by MD simulations and ssNMR, we identified that the pore closure was caused by mercury-induced conformational changes of the key residue R189 in the selectivity filter region, while pore opening was caused by conformational changes of residues H181 and R196 in the selectivity filter region in AQP6. Both conformational changes were caused by the disruption of the H-bond network of R189/R196 by mercury. The molecular details provided a structural basis for mercury-mediated functional changes in aquaporins.


Asunto(s)
Mercurio
4.
Biochem Biophys Res Commun ; 618: 1-7, 2022 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-35714565

RESUMEN

Aquaporins (AQPs) transport water molecules across cell membranes. Although most aquaporins are inhibited by mercury ions, AQP6 was reported to be activated by binding mercury ions to residues C155 and C190. Different from C190 and the other pore-line cysteine residues, C155 is located outside the pore, thus not directly affecting the internal pathway by mercury binding to it. The molecular mechanism of unusual water channel activation by mercury ion binding to the C155 site remains unknown. Here, we investigate the activation of AQP6 by mercury ions binding to C155 by molecular dynamics (MD) simulations. The MD simulation results show that the mercury-induced water permeation activation is derived from the conformational change of a pore-line residue M160, from a point-to-pore conformation before mercury binding to an away-pore conformation after mercury binding. The conformation change of M160 is derived from the reduction of the hydrogen bonding between C155 and S159 in the α-helix with the coordination of C155 to mercury ion altering their conformation significantly. This study reveals the complex mechanism of water channel activation by mercury ion binding to pore-external residues in water channels.


Asunto(s)
Acuaporinas , Mercurio , Acuaporinas/metabolismo , Iones/metabolismo , Mercurio/metabolismo , Simulación de Dinámica Molecular , Agua/química
5.
Acta Pharmacol Sin ; 41(9): 1167-1177, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32737471

RESUMEN

Human infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19) and there is no cure currently. The 3CL protease (3CLpro) is a highly conserved protease which is indispensable for CoVs replication, and is a promising target for development of broad-spectrum antiviral drugs. In this study we investigated the anti-SARS-CoV-2 potential of Shuanghuanglian preparation, a Chinese traditional patent medicine with a long history for treating respiratory tract infection in China. We showed that either the oral liquid of Shuanghuanglian, the lyophilized powder of Shuanghuanglian for injection or their bioactive components dose-dependently inhibited SARS-CoV-2 3CLpro as well as the replication of SARS-CoV-2 in Vero E6 cells. Baicalin and baicalein, two ingredients of Shuanghuanglian, were characterized as the first noncovalent, nonpeptidomimetic inhibitors of SARS-CoV-2 3CLpro and exhibited potent antiviral activities in a cell-based system. Remarkably, the binding mode of baicalein with SARS-CoV-2 3CLpro determined by X-ray protein crystallography was distinctly different from those of known 3CLpro inhibitors. Baicalein was productively ensconced in the core of the substrate-binding pocket by interacting with two catalytic residues, the crucial S1/S2 subsites and the oxyanion loop, acting as a "shield" in front of the catalytic dyad to effectively prevent substrate access to the catalytic dyad within the active site. Overall, this study provides an example for exploring the in vitro potency of Chinese traditional patent medicines and effectively identifying bioactive ingredients toward a specific target, and gains evidence supporting the in vivo studies of Shuanghuanglian oral liquid as well as two natural products for COVID-19 treatment.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus , Medicamentos Herbarios Chinos , Flavanonas , Flavonoides , Pandemias , Neumonía Viral , Replicación Viral/efectos de los fármacos , Administración Oral , Animales , Antivirales/química , Antivirales/farmacología , Betacoronavirus/fisiología , COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Pruebas de Enzimas , Flavanonas/química , Flavanonas/farmacocinética , Flavonoides/química , Flavonoides/farmacocinética , Humanos , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , SARS-CoV-2 , Células Vero , Replicación Viral/fisiología
6.
Chembiochem ; 20(21): 2757-2766, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31161647

RESUMEN

A puzzling feature of prion diseases is the cross-species barriers. The detailed molecular mechanisms underlying these interspecies barriers remain poorly understood because of a lack of high-resolution structural information on the scrapie isoform of the prion protein (PrPSc ). In this study we identified the critical role of the residues 165/167 in the barrier to seeding mouse PrP (mPrP) fibril seeds to human cellular prion protein (PrPC ). Solid-state NMR revealed a C-terminal ß-sheet core spanning residues 165-230 and the packing arrangement of mPrP fibrils. Residues 165/167 are located on one end of the fibril core. Molecular dynamics simulations demonstrated that the stabilities of the seeding-induced ß-strand structures are significantly impacted by hydrogen bonds involving the side chain of residue 167 and steric resistance involving residue 165. These findings suggest that the α2-ß2 loop containing residues 165/167 could be the initial site of seed-template conformational conversion.


Asunto(s)
Amiloide/química , Proteínas Amiloidogénicas/química , Simulación de Dinámica Molecular , Proteínas Priónicas/química , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Amiloide/genética , Amiloide/metabolismo , Proteínas Amiloidogénicas/genética , Proteínas Amiloidogénicas/metabolismo , Animales , Humanos , Ratones , Resonancia Magnética Nuclear Biomolecular/métodos , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Priones/genética , Priones/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido , Especificidad de la Especie
7.
Acta Pharmacol Sin ; 40(6): 850-858, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30796354

RESUMEN

Serine/threonine phosphatase (Stp1) is a member of the bacterial Mg2+- or Mn2+- dependent protein phosphatase/protein phosphatase 2C family, which is involved in the regulation of Staphylococcus aureus virulence. Aurintricarboxylic acid (ATA) is a known Stp1 inhibitor with an IC50 of 1.03 µM, but its inhibitory mechanism has not been elucidated in detail because the Stp1-ATA cocrystal structure has not been determined thus far. In this study, we performed 400 ns molecular dynamics (MD) simulations of the apo-Stp1 and Stp1-ATA complex models. During MD simulations, the flap subdomain of the Stp1-ATA complex experienced a clear conformational transition from an open state to a closed state, whereas the flap domain of apo-Stp1 changed from an open state to a semi-open state. In the Stp1-ATA complex model, the hydrogen bond (H-bond) between D137 and N142 disappeared, whereas critical H-bond interactions were formed between Q160 and H13, Q160/R161 and ATA, as well as N162 and D198. Finally, four residues (D137, N142, Q160, and R161) in Stp1 were mutated to alanine and the mutant enzymes were assessed using phosphate enzyme activity assays, which confirmed their important roles in maintaining Stp1 activity. This study indicated the inhibitory mechanism of ATA targeting Stp1 using MD simulations and sheds light on the future design of allosteric Stp1 inhibitors.


Asunto(s)
Ácido Aurintricarboxílico/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Staphylococcus aureus/enzimología , Secuencia de Aminoácidos , Ácido Aurintricarboxílico/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Inhibidores Enzimáticos/química , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Mutación , Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Unión Proteica , Conformación Proteica , Alineación de Secuencia
8.
Acta Pharmacol Sin ; 38(12): 1673-1682, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28748916

RESUMEN

Phosphoglycerate mutase 1 (PGAM1), an important enzyme in glycolysis, is overexpressed in a number of human cancers, thus has been proposed as a promising metabolic target for cancer treatments. The C-terminal portion of the available crystal structures of PGAM1 and its homologous proteins is partially disordered, as evidenced by weak electron density. In this study, we identified the conformational behavior of the C-terminal region of PGAM1 as well as its role during the catalytic cycle. Using the PONDR-FIT server, we demonstrated that the C-terminal region was intrinsically disordered. We applied the Monte Carlo (MC) method to explore the conformational space of the C-terminus and conducted a series of explicit-solvent molecular dynamics (MD) simulations, and revealed that the C-terminal region is inherently dynamic; large-scale conformational changes in the C-terminal segment led to the structural transition of PGAM1 from the closed state to the open state. Furthermore, the C-terminal segment influenced 2,3-bisphosphoglycerate (2,3-BPG) binding. The proposed swing model illustrated a critical role of the C-terminus in the catalytic cycle through the conformational changes. In conclusion, the C-terminal region induces large movements of PGAM1 from the closed state to the open state and influences cofactor binding during the catalytic cycle. This report describes the dynamic features of the C-terminal region in detail and should aid in design of novel and efficient inhibitors of PGAM1. A swing mechanism of the C-terminal region is proposed, to facilitate further studies of the catalytic mechanism and the physiological functions of its homologues.


Asunto(s)
Simulación de Dinámica Molecular , Fosfoglicerato Mutasa/química , Fosfoglicerato Mutasa/metabolismo , Biocatálisis , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Método de Montecarlo , Fosfoglicerato Mutasa/antagonistas & inhibidores , Análisis de Componente Principal , Conformación Proteica , Electricidad Estática
9.
Nucleic Acids Res ; 43(21): 10157-67, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26464436

RESUMEN

Eukaryotic translation initiation factor eIF4AI, the founding member of DEAD-box helicases, undergoes ATP hydrolysis-coupled conformational changes to unwind mRNA secondary structures during translation initiation. However, the mechanism of its coupled enzymatic activities remains unclear. Here we report that a gating mechanism for Pi release controlled by the inter-domain linker of eIF4AI regulates the coupling between ATP hydrolysis and RNA unwinding. Molecular dynamic simulations and experimental results revealed that, through forming a hydrophobic core with the conserved SAT motif of the N-terminal domain and I357 from the C-terminal domain, the linker gated the release of Pi from the hydrolysis site, which avoided futile hydrolysis cycles of eIF4AI. Further mutagenesis studies suggested this linker also plays an auto-inhibitory role in the enzymatic activity of eIF4AI, which may be essential for its function during translation initiation. Overall, our results reveal a novel regulatory mechanism that controls eIF4AI-mediated mRNA unwinding and can guide further mechanistic studies on other DEAD-box helicases.


Asunto(s)
Factor 4A Eucariótico de Iniciación/química , Iniciación de la Cadena Peptídica Traduccional , ARN Mensajero/química , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Factor 4A Eucariótico de Iniciación/genética , Factor 4A Eucariótico de Iniciación/metabolismo , Simulación de Dinámica Molecular , Mutación , Fosfatos/química , Fosfatos/metabolismo , Unión Proteica , Conformación Proteica , ARN Mensajero/metabolismo
10.
Phys Chem Chem Phys ; 18(6): 4728-38, 2016 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-26799843

RESUMEN

TET proteins iteratively convert 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) in a Fe(ii)/α-ketoglutarate-dependent manner. Our previous biochemical studies revealed that TET proteins are more active on 5mC than on 5hmC and 5fC. However, the source of the substrate preference of TET proteins still remains largely elusive. Here, we investigated the substrate binding and catalytic mechanisms of oxidation reactions mediated by TET2 on different substrates through computational approaches. In accordance with previous experimental reports, our computational results suggest that TET2 can bind to different substrates with comparable binding affinities and the hydrogen abstraction step in the catalytic cycle acts as the rate-limiting step. Further structural characterization of the intermediate structures revealed that the 5-substitution groups on 5hmC and 5fC adopt an unfavorable orientation for hydrogen abstraction, which leads to a higher energy barrier for 5hmC and 5fC (compared to 5mC) and thus a lower catalytic efficiency. In summary, our mechanical insights demonstrate that substrate preference is the intrinsic property of TET proteins and our theoretical calculation results can guide further dry-lab or wet-lab studies on the catalytic mechanism of TET proteins as well as other Fe(ii)/α-ketoglutarate (KG)-dependent dioxygenases.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Catálisis , Dioxigenasas , Oxidación-Reducción , Especificidad por Sustrato
11.
Bioorg Med Chem Lett ; 25(18): 3970-4, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26227779

RESUMEN

A series of compounds with quinazoline scaffold were designed, synthesized and evaluated as novel potent 5-HT2A receptor ligands. N-(4-Chlorophenyl)-2-(piperazin-1-yl)quinazolin-4-amine (5o) has a Ki value of 14.04 ± 0.21 nM, with a selectivity more than 10,000 fold over 5-HT1A receptors (D1 and D2-like receptors). The functional assay showed that this compound is an antagonist to 5-HT2A receptor with an IC50 value of 1.66 µM.


Asunto(s)
Descubrimiento de Drogas , Quinazolinas/química , Receptor de Serotonina 5-HT2A/metabolismo , Antagonistas del Receptor de Serotonina 5-HT2/química , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Estructura Molecular , Quinazolinas/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/síntesis química , Relación Estructura-Actividad
12.
J Chem Inf Model ; 55(12): 2623-32, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26562720

RESUMEN

Protein arginine methyltransferases (PRMTs) catalyze the posttranslational methylation of arginine, which is important in a range of biological processes, including epigenetic regulation, signal transduction, and cancer progression. Although previous studies of PRMT1 mutants suggest that the dimerization arm and the N-terminal region of PRMT1 are important for activity, the contributions of these regions to the structural architecture of the protein and its catalytic methylation activity remain elusive. Molecular dynamics (MD) simulations performed in this study showed that both the dimerization arm and the N-terminal region undergo conformational changes upon dimerization. Because a correlation was found between the two regions despite their physical distance, an allosteric pathway mechanism was proposed based on a network topological analysis. The mutation of residues along the allosteric pathways markedly reduced the methylation activity of PRMT1, which may be attributable to the destruction of dimer formation and accordingly reduced S-adenosyl-L-methionine (SAM) binding. This study provides the first demonstration of the use of a combination of MD simulations, network topological analysis, and biochemical assays for the exploration of allosteric regulation upon PRMT1 dimerization. These findings illuminate the results of mechanistic studies of PRMT1, which have revealed that dimer formation facilitates SAM binding and catalytic methylation, and provided direction for further allosteric studies of the PRMT family.


Asunto(s)
Modelos Moleculares , Simulación de Dinámica Molecular , Proteína-Arginina N-Metiltransferasas/química , S-Adenosilmetionina/metabolismo , Regulación Alostérica , Bioensayo , Secuencia Conservada , Dimerización , Electroforesis en Gel Bidimensional , Fluorescencia , Metilación , Estructura Secundaria de Proteína , S-Adenosilmetionina/química
13.
J Chem Inf Model ; 55(9): 1994-2004, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26288111

RESUMEN

Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine, is an attractive therapeutic target for the treatment of inflammatory diseases. In our previous study, 3-[(biphenyl-4-ylcarbonyl)carbamothioyl]amino benzoic acid (compound 1) was discovered as a potent inhibitor of MIF by docking-based virtual screening and bioassays. Here, a series of analogues of compound 1 derived from similarity search and chemical synthesis were evaluated for their MIF tautomerase activities, and their structure-activity relationships were then analyzed. The most potent inhibitor (compound 5) with an IC50 of 370 nM strongly suppressed lipopolysaccharide (LPS)-induced production of TNF-α and IL-6 in a dose-dependent manner and significantly enhanced the survival rate of mice with LPS-induced endotoxic shock from 0 to 35% at 0.5 mg/kg and to 45% at 1 mg/kg, highlighting the therapeutic potential of the MIF tautomerase inhibition in inflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Formamidas/química , Formamidas/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Animales , Compuestos de Bencidrilo/química , Sitios de Unión , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Concentración 50 Inhibidora , Factores Inhibidores de la Migración de Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Óxido Nítrico/química , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacología
14.
Proteins ; 82(5): 760-70, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24265091

RESUMEN

Two 8-µs all-atom molecular dynamics simulations have been performed on the two highly homologous G protein-coupled receptor (GPCR) subtypes, ß1 - and ß2 -adrenergic receptors, which were embedded in a lipid bilayer with randomly dispersed cholesterol molecules. During the simulations, cholesterol molecules accumulate to different surface regions of the two receptors, suggesting the subtype specificity of cholesterol-ß-adrenergic receptor interaction and providing some clues to the physiological difference of the two subtypes. Meanwhile, comparison between the two receptors in interacting with cholesterols shed some new light on general determinants of cholesterol binding to GPCRs. Our results indicate that although the concave surface, charged residues and aromatic residues are important, neither of these stabilizing factors is indispensable for a cholesterol interaction site. Different combinations of these factors lead to the diversified binding modes of cholesterol binding to the receptors. Our long-time simulations, for the first time, revealed the pathway of a cholesterol molecule entering the consensus cholesterol motif (CCM) site, and the binding process of cholesterol to CCM is accompanied by a side chain flipping of the conserved Trp4.50. Moreover, the simulation results suggest that the I-/V-/L-rich region on the extracellular parts of helix 6 might be an alternatively conserved cholesterol-binding site for the class-A GPCRs.


Asunto(s)
Colesterol/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Colesterol/química , Secuencia de Consenso , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Estructura Secundaria de Proteína , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 2/química , Turquía
15.
Proteins ; 82(2): 312-22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23934928

RESUMEN

Annexins play critical roles in membrane organization, membrane trafficking and vesicle transport. The family members share the ability to bind to membranes with high affinities, but the interactions between annexins and membranes remain unclear. Here, using long-time molecular dynamics simulations, we provide detailed information for the binding of an annexin V trimer to a POPC/POPS lipid bilayer. Calcium ions function as bridges between several negatively charged residues of annexin V and the oxygen atoms of lipids. The preferred calcium-bridges are those formed via the carboxyl oxygen atoms of POPS lipids. H-bonds and hydrophobic interactions formed by several critical residues have also been observed in the annexin-membrane interface. The annexin-membrane binding causes small changes of annexin trimer structures, while has significant effects on lipid bilayer structures. The lipid bilayer shows a bent shape and forms a concave region in the annexin-membrane interaction interface, which provides an atomic-level evidence to support the view that annexins could disturb the stability of lipids and bend membranes. This study provides insights into the commonly occurring PS-dependent and calcium-dependent binding of proteins to membranes.


Asunto(s)
Anexina A5/química , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Calcio , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Fosfatidilcolinas/química , Fosfatidilserinas/química , Unión Proteica , Estabilidad Proteica
16.
Neurosignals ; 22(2): 65-78, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25300231

RESUMEN

The small- and intermediate-conductance Ca(2+)-activated potassium (SK/IK) channels play important roles in the regulation of excitable cells in both the central nervous and cardiovascular systems. Evidence from animal models has implicated SK/IK channels in neurological conditions such as ataxia and alcohol use disorders. Further, genome-wide association studies have suggested that cardiovascular abnormalities such as arrhythmias and hypertension are associated with single nucleotide polymorphisms that occur within the genes encoding the SK/IK channels. The Ca(2+) sensitivity of the SK/IK channels stems from a constitutively bound Ca(2+)-binding protein: calmodulin. Small-molecule positive modulators of SK/IK channels have been developed over the past decade, and recent structural studies have revealed that the binding pocket of these positive modulators is located at the interface between the channel and calmodulin. SK/IK channel positive modulators can potentiate channel activity by enhancing the coupling between Ca(2+) sensing via calmodulin and mechanical opening of the channel. Here, we review binding pocket studies that have provided structural insight into the mechanism of action for SK/IK channel positive modulators. These studies lay the foundation for structure-based drug discovery efforts that can identify novel SK/IK channel positive modulators.


Asunto(s)
Calmodulina/metabolismo , Sistema Cardiovascular/metabolismo , Sistema Nervioso Central/metabolismo , Descubrimiento de Drogas , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Animales , Humanos
17.
Adv Sci (Weinh) ; : e2405596, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39021325

RESUMEN

Excited-state intramolecular proton transfer (ESIPT) has attracted great attention in fluorescent sensors and luminescent materials due to its unique photobiological and photochemical features. However, the current structures are far from meeting the specific demands for ESIPT molecules in different scenarios; the try-and-error development method is labor-intensive and costly. Therefore, it is imperative to devise novel approaches for the exploration of promising ESIPT fluorophores. This research proposes an artificial intelligence approach aiming at exploring ESIPT molecules efficiently. The first high-quality ESIPT dataset and a multi-level prediction system are constructed that realized accurate identification of ESIPT molecules from a large number of compounds under a stepwise distinguishing from conventional molecules to fluorescent molecules and then to ESIPT molecules. Furthermore, key structural features that contributed to ESIPT are revealed by using the SHapley Additive exPlanations (SHAP) method. Then three strategies are proposed to ensure the ESIPT process while keeping good safety, pharmacokinetic properties, and novel structures. With these strategies, >700 previously unreported ESIPT molecules are screened from a large pool of 570 000 compounds. The ESIPT process and biosafety of optimal molecules are successfully validated by quantitative calculation and experiment. This novel approach is expected to bring a new paradigm for exploring ideal ESIPT molecules.

18.
Biochem Biophys Res Commun ; 431(1): 2-7, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23313491

RESUMEN

New Delhi metallo-ß-lactmase-1 (NDM-1) is an enzyme that confers antibiotic resistance to bacteria and is thus a serious threat to human health. Almost all clinically available ß-lactam antibiotics can be hydrolyzed by NDM-1. To determine the mechanism behind the wide substrate diversity and strong catalytic ability of NDM-1, we explored the molecular interactions between NDM-1 and different ß-lactam antibiotics using computational methods. Molecular dynamics simulations and binding free energy calculations were performed on enzyme-substrate (ES) complex models of NDM-1-Meropenem, NDM-1-Nitrocefin, and NDM-1-Ampicillin constructed by molecular docking. Our computational results suggest that mutant residues Ile35 and Lys216, and active site loop L1 residues 65-73 in NDM-1 play crucial roles in substrate recognition and binding. The results of our study provide new insights into the mechanism behind the enhanced substrate binding and wider substrate spectrum of NDM-1 compared with its homologous enzymes CcrA and IMP-1. These insights may be useful in the discovery and design of specific and potent inhibitors against NDM-1.


Asunto(s)
beta-Lactamasas/química , Ampicilina/química , Catálisis , Cefalosporinas/química , Enlace de Hidrógeno , Hidrólisis , Meropenem , Simulación de Dinámica Molecular , Unión Proteica , Estructura Secundaria de Proteína , Especificidad por Sustrato , Tienamicinas/química
19.
J Chem Inf Model ; 53(12): 3202-11, 2013 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-24245825

RESUMEN

The serotonin receptor subtype 1A (5-HT(1A)R) has been implicated in several neurological conditions, and potent 5-HT(1A)R agonists have therapeutic potential for the treatment of depression, anxiety, schizophrenia, and Parkinson's disease. In the present study, a homology model of 5-HT(1A)R was built based on the latest released high-resolution crystal structure of the ß2AR in its active state (PDB: 3SN6). A dynamic pharmacophore model, which takes the receptor flexibility into account, was constructed, validated, and applied to our dynamic pharmacophore-based virtual screening approach with the aim to identify potential 5-5-HT(1A)R agonists. The obtained hits were subjected to 55-HT(1A)R binding and functional assays, and 10 compounds with medium or high K(i) and EC50 values were identified. Among them, FW01 (K(i) = 51.9 nM, EC50 = 7 nM) was evaluated as the strongest agonist for 5-HT(1A)R. The active 5-HT(1A)R model and dynamic pharmacophore model obtained from this study can be used for future discovery and design of novel 5-HT(1A)R agonists. Also, by integrating all computational and available experimental data, a stepwise 5-HT(1A)R signal transduction model induced by agonist FW01 was proposed.


Asunto(s)
Indoles/química , Simulación de Dinámica Molecular , Piperazinas/química , Receptor de Serotonina 5-HT1A/química , Agonistas del Receptor de Serotonina 5-HT1/química , Bibliotecas de Moléculas Pequeñas/química , Interfaz Usuario-Computador , Dominio Catalítico , Bases de Datos de Proteínas , Descubrimiento de Drogas , Guanosina 5'-O-(3-Tiotrifosfato)/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Receptores Adrenérgicos beta 2/química , Homología Estructural de Proteína , Relación Estructura-Actividad
20.
Research (Wash D C) ; 6: 0075, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36930810

RESUMEN

Monitoring the physiological changes of organelles is essential for understanding the local biological information of cells and for improving the diagnosis and therapy of diseases. Currently, fluorescent probes are considered as the most powerful tools for imaging and have been widely applied in biomedical fields. However, the expected targeting effects of these probes are often inconsistent with the real experiments. The design of fluorescent probes mainly depends on the empirical knowledge of researchers, which was inhibited by limited chemical space and low efficiency. Herein, we proposed a novel multilevel framework for the prediction of organelle-targeted fluorescent probes by employing advanced artificial intelligence algorithms. In this way, not only the targeting mechanism could be interpreted beyond intuitions but also a quick evaluation method could be established for the rational design. Furthermore, the targeting and imaging powers of the optimized and synthesized probes based on this methodology were verified by quantitative calculation and experiments.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA