Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Mol Pharm ; 17(12): 4704-4708, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33118829

RESUMEN

Controlling physicochemical properties of light-unresponsive drugs, by light, prima facie, a paradox approach. We expanded light control by ion pairing light-unresponsive salicylate or ibuprofen to photoswitchable azobenzene counterions, thereby reversibly controlling supramolecular structures, hence the drugs' physicochemical and kinetic properties. The resulting ion pairs photoliquefied into room-temperature ionic liquids under ultraviolet light. Aqueous solutions showed trans-cis-dependent supramolecular structures under a light with wormlike aggregates decomposing into small micelles and vice versa. Light control allowed for permeation through membranes of cis-ibuprofen ion pairs within 12 h in contrast to the trans ion pairs requiring 72 h. In conclusion, azobenzene ion-pairing expands light control of physicochemical and kinetic properties to otherwise light-unresponsive drugs.


Asunto(s)
Líquidos Iónicos/efectos de la radiación , Rayos Ultravioleta , Compuestos Azo/química , Compuestos Azo/farmacocinética , Compuestos Azo/efectos de la radiación , Química Farmacéutica , Ibuprofeno/química , Ibuprofeno/farmacocinética , Ibuprofeno/efectos de la radiación , Líquidos Iónicos/química , Líquidos Iónicos/farmacocinética , Estructura Molecular , Permeabilidad , Salicilatos/química , Salicilatos/farmacocinética , Salicilatos/efectos de la radiación , Agua/química
2.
Mol Inform ; 35(11-12): 541-548, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27870243

RESUMEN

The ability to rapidly assess the quality of a protein-ligand complex in terms of its affinity is of fundamental importance for various methods of computer-aided drug design. While simple filtering or matching critieria may be sufficient in fast docking methods or at early stages of virtual screening, estimates of the actual free energy of binding are needed whenever refined docking solutions, ligand rankings or support for the optimization of hit compounds are required. If rigorous free energy calculations based on molecular simulations are impractical, such affinity estimates are provided by scoring functions. The class of empirical scoring functions aims to provide them via a regression-based approach. Using experimental structures and affinity data of protein-ligand complexes and descriptors suitable to capture the essential features of the interaction, these functions are trained with classical linear regression techniques or machine-learning methods. The latter have led to considerable improvements in terms of prediction accuracy for large generic data sets. Nevertheless, many limitations are not yet resolved and pose significant challenges for future developments.


Asunto(s)
Proteínas/química , Sitios de Unión/fisiología , Biología Computacional/métodos , Ligandos , Modelos Lineales , Aprendizaje Automático , Simulación del Acoplamiento Molecular/métodos , Unión Proteica/fisiología , Análisis de Regresión
3.
Bioorg Med Chem ; 24(21): 5134-5147, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27591009

RESUMEN

The bacteria Burkholderia pseudomallei and Legionella pneumophila cause severe diseases like melioidosis and Legionnaire's disease with high mortality rates despite antibiotic treatment. Due to increasing antibiotic resistances against these and other Gram-negative bacteria, alternative therapeutical strategies are in urgent demand. As a virulence factor, the macrophage infectivity potentiator (Mip) protein constitutes an attractive target. The Mip proteins of B. pseudomallei and L. pneumophila exhibit peptidyl-prolyl cis/trans isomerase (PPIase) activity and belong to the PPIase superfamily. In previous studies, the pipecolic acid moiety proved to be a valuable scaffold for inhibiting this PPIase activity. Thus, a library of pipecolic acid derivatives was established guided by structural information and computational analyses of the binding site and possible binding modes. Stability and toxicity considerations were taken into account in iterative extensions of the library. Synthesis and evaluation of the compounds in PPIase assays resulted in highly active inhibitors. The activities can be interpreted in terms of a common binding mode obtained by docking calculations.


Asunto(s)
Burkholderia pseudomallei/enzimología , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Legionella pneumophila/enzimología , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Simulación del Acoplamiento Molecular , Estructura Molecular , Isomerasa de Peptidilprolil/metabolismo , Relación Estructura-Actividad
4.
Chem Sci ; 7(9): 5945-5954, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27547299

RESUMEN

Drug-target kinetics enable time-dependent changes in target engagement to be quantified as a function of drug concentration. When coupled to drug pharmacokinetics (PK), drug-target kinetics can thus be used to predict in vivo pharmacodynamics (PD). Previously we described a mechanistic PK/PD model that successfully predicted the antibacterial activity of an LpxC inhibitor in a model of Pseudomonas aeruginosa infection. In the present work we demonstrate that the same approach can be used to predict the in vivo activity of an enoyl-ACP reductase (FabI) inhibitor in a model of methicillin-resistant Staphylococcus aureus (MRSA) infection. This is significant because the LpxC inhibitors are cidal, whereas the FabI inhibitors are static. In addition P. aeruginosa is a Gram-negative organism whereas MRSA is Gram-positive. Thus this study supports the general applicability of our modeling approach across antibacterial space.

5.
J Comput Aided Mol Des ; 30(3): 251-70, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26923377

RESUMEN

The dengue virus (DENV) has four well-known serotypes, namely DENV1 to DENV4, which together cause 50-100 million infections worldwide each year. DENV NS2B/NS3pro is a protease recognized as a valid target for DENV antiviral drug discovery. However, NS2B/NS3pro conformational flexibility, involving in particular the NS2B region, is not yet completely understood and, hence, a big challenge for any virtual screening (VS) campaign. Molecular dynamics (MD) simulations were performed in this study to explore the DENV3 NS2B/NS3pro binding-site flexibility and obtain guidelines for further VS studies. MD simulations were done with and without the Bz-nKRR-H inhibitor, showing that the NS2B region stays close to the NS3pro core even in the ligand-free structure. Binding-site conformational states obtained from the simulations were clustered and further analysed using GRID/PCA, identifying four conformations of potential importance for VS studies. A virtual screening applied to a set of 31 peptide-based DENV NS2B/NS3pro inhibitors, taken from literature, illustrated that selective alternative pharmacophore models can be constructed based on conformations derived from MD simulations. For the first time, the NS2B/NS3pro binding-site flexibility was evaluated for all DENV serotypes using homology models followed by MD simulations. Interestingly, the number of NS2B/NS3pro conformational states differed depending on the serotype. Binding-site differences could be identified that may be crucial to subsequent VS studies.


Asunto(s)
Virus del Dengue/efectos de los fármacos , Virus del Dengue/enzimología , Inhibidores Enzimáticos/farmacología , Péptidos/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Sitios de Unión , Dengue/tratamiento farmacológico , Dengue/virología , Virus del Dengue/genética , Inhibidores Enzimáticos/química , Humanos , Simulación de Dinámica Molecular , Péptidos/química , Conformación Proteica , Serogrupo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo
6.
J Med Chem ; 59(5): 2067-82, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26886849

RESUMEN

Butyrylcholinesterase (BChE) is a promising target for the treatment of later stage cognitive decline in Alzheimer's disease. A set of pseudo-irreversible BChE inhibitors with high selectivity over hAChE was synthesized based on carbamates attached to tetrahydroquinazoline scaffolds with the 2-thiophenyl compound 2p as the most potent inhibitor of eqBChE (KC = 14.3 nM) and also of hBChE (KC = 19.7 nM). The inhibitors transfer the carbamate moiety onto the active site under release of the phenolic tetrahydroquinazoline scaffolds that themselves act as neuroprotectants. By combination of kinetic data with molecular docking studies, a plausible binding model was probed describing how the tetrahydroquinazoline scaffold guides the carbamate into a close position to the active site. The model explains the influence of the carrier scaffold onto the affinity of an inhibitor just before carbamate transfer. This strategy can be used to utilize the binding mode of other carbamate-based inhibitors.


Asunto(s)
Butirilcolinesterasa/metabolismo , Carbamatos/farmacología , Inhibidores de la Colinesterasa/farmacología , Descubrimiento de Drogas , Animales , Carbamatos/síntesis química , Carbamatos/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/química , Relación Dosis-Respuesta a Droga , Ratones , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
7.
PLoS One ; 10(5): e0127009, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25996598

RESUMEN

An important kinetic parameter for drug efficacy is the residence time of a compound at a drug target, which is related to the dissociation rate constant koff. For the essential antimycobacterial target InhA, this parameter is most likely governed by the ordering of the flexible substrate binding loop (SBL). Whereas the diphenyl ether inhibitors 6PP and triclosan (TCL) do not show loop ordering and thus, no slow-binding inhibition and high koff values, the slightly modified PT70 leads to an ordered loop and a residence time of 24 minutes. To assess the structural differences of the complexes from a dynamic point of view, molecular dynamics (MD) simulations with a total sampling time of 3.0 µs were performed for three ligand-bound and two ligand-free (perturbed) InhA systems. The individual simulations show comparable conformational features with respect to both the binding pocket and the SBL, allowing to define five recurring conformational families. Based on their different occurrence frequencies in the simulated systems, the conformational preferences could be linked to structural differences of the respective ligands to reveal important determinants of residence time. The most abundant conformation besides the stable EI* state is characterized by a shift of Ile202 and Val203 toward the hydrophobic pocket of InhA. The analyses revealed potential directions for avoiding this conformational change and, thus, hindering rapid dissociation: (1) an anchor group in 2'-position of the B-ring for scaffold stabilization, (2) proper occupation of the hydrophobic pocket, and (3) the introduction of a barricade substituent in 5'-position of the diphenyl ether B-ring.


Asunto(s)
Proteínas Bacterianas/química , Simulación de Dinámica Molecular , Mycobacterium tuberculosis , Oxidorreductasas/química , Antituberculosos/química , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Sitios de Unión , Diseño de Fármacos , Humanos , Enlace de Hidrógeno , Ligandos , Simulación del Acoplamiento Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Oxidorreductasas/antagonistas & inhibidores , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
8.
Biochemistry ; 54(10): 1943-55, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25706582

RESUMEN

One third of all drugs in clinical use owe their pharmacological activity to the functional inhibition of enzymes, highlighting the importance of enzymatic targets for drug development. Because of the close relationship between inhibition and catalysis, understanding the recognition and turnover of enzymatic substrates is essential for rational drug design. Although the Staphylococcus aureus enoyl-acyl carrier protein reductase (saFabI) involved in bacterial fatty acid biosynthesis constitutes a very promising target for the development of novel, urgently needed anti-staphylococcal agents, the substrate binding mode and catalytic mechanism remained unclear for this enzyme. Using a combined crystallographic, kinetic, and computational approach, we have explored the chemical properties of the saFabI binding cavity, obtaining a consistent mechanistic model for substrate binding and turnover. We identified a water-molecule network linking the active site with a water basin inside the homo-tetrameric protein, which seems to be crucial for the closure of the flexible substrate binding loop as well as for an effective hydride and proton transfer during catalysis. On the basis of our results, we also derive a new model for the FabI-ACP complex that reveals how the ACP-bound acyl-substrate is injected into the FabI binding crevice. These findings support the future development of novel FabI inhibitors that target the FabI-ACP interface leading to the disruption of the interaction between these two proteins.


Asunto(s)
Proteínas Bacterianas/química , Enoil-ACP Reductasa (NADH)/química , Modelos Moleculares , Staphylococcus aureus/enzimología , Agua/química , Catálisis , Dominio Catalítico , Relación Estructura-Actividad
9.
Parasitol Res ; 114(2): 501-12, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25416330

RESUMEN

Potent compounds do not necessarily make the best drugs in the market. Consequently, with the aim to describe tools that may be fundamental for refining the screening of candidates for animal and preclinical studies and further development, molecules of different structural classes synthesized within the frame of a broad screening platform were evaluated for their trypanocidal activities, cytotoxicities against murine macrophages J774.1 and selectivity indices, as well as for their ligand efficiencies and structural chemical properties. To advance into their modes of action, we also describe the morphological and ultrastructural changes exerted by selected members of each compound class on the parasite Trypanosoma brucei. Our data suggest that the potential organelles targeted are either the flagellar pocket (compound 77, N-Arylpyridinium salt; 15, amino acid derivative with piperazine moieties), the endoplasmic reticulum membrane systems (37, bisquaternary bisnaphthalimide; 77, N-Arylpyridinium salt; 68, piperidine derivative), or mitochondria and kinetoplasts (88, N-Arylpyridinium salt; 68, piperidine derivative). Amino acid derivatives with fumaric acid and piperazine moieties (4, 15) weakly inhibiting cysteine proteases seem to preferentially target acidic compartments. Our results suggest that ligand efficiency indices may be helpful to learn about the relationship between potency and chemical characteristics of the compounds. Interestingly, the correlations found between the physico-chemical parameters of the selected compounds and those of commercial molecules that target specific organelles indicate that our rationale might be helpful to drive compound design toward high activities and acceptable pharmacokinetic properties for all compound families.


Asunto(s)
Fumaratos/farmacología , Piperazinas/farmacología , Piperidinas/farmacología , Tripanocidas/farmacología , Trypanosoma brucei brucei/efectos de los fármacos , Animales , Línea Celular , Proteasas de Cisteína/efectos de los fármacos , Fumaratos/química , Concentración de Iones de Hidrógeno , Macrófagos/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Orgánulos/efectos de los fármacos , Piperazina , Piperazinas/química , Piperidinas/química , Tripanocidas/química , Trypanosoma brucei brucei/ultraestructura
10.
Chemphyschem ; 15(15): 3226-35, 2014 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-25251382

RESUMEN

The inhibition potencies of covalent inhibitors mainly result from the formation of a covalent bond to the enzyme during the inhibition mechanism. This class of inhibitors has essentially been ignored in previous target-directed drug discovery projects because of concerns about possible side effects. However, their advantages, such as higher binding energies and longer drug-target residence times moved them into the focus of recent investigations. While the rational design of non-covalent inhibitors became standard the corresponding design of covalent inhibitors is still in its early stages. Potent covalent inhibitors can be retrieved from large compound libraries by covalent docking approaches but protocols are missing that can reliably predict the influence of variations in the substitution pattern on the affinity and/or reactivity of a given covalent inhibitor. Hence, the wanted property profile can only be obtained from trial-and-error proceedings. This paper presents an appropriate protocol which is able to predict improved covalent inhibitors. It uses hybrid approaches, which mix quantum mechanical (QM) and molecular mechanical (MM) methods to predict variations in the reactivity of the inhibitor. They are also used to compute the required information about the non-covalent enzyme-inhibitor complex. Docking tools are employed to improve the inhibitor with respect to the non-covalent interactions formed in the binding site.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/química , Enzimas/química , Dominio Catalítico , Inhibidores Enzimáticos/metabolismo , Enzimas/metabolismo , Compuestos Epoxi/química , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Nitrofenoles/química , Teoría Cuántica
11.
Bioorg Med Chem ; 22(17): 4867-81, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25047936

RESUMEN

Selective and nanomolar acetylcholinesterase inhibitors were obtained by connecting tri- and tetracyclic quinazolinones-previously described as moderately active and unselective cholinesterase (ChE) inhibitors-via a hydroxyl group in para position to an anilinic nitrogen with different amines linked via a three carbon atom spacer. These tri- and tetracyclic quinazolinones containing different alicyclic ring sizes and connected to tertiary amines were docked to a high-resolution hAChE crystal structure to investigate the preferred binding mode in relation to results obtained by experimental structure-activity relationships. While the 'classical orientation' locating the heterocycle in the active site was rarely found, an alternative binding mode with the basic aliphatic amine in the active center ('inverted' orientation) was obtained for most compounds. Analyses of extended SARs based on this inverted binding mode are able to explain the compounds' binding affinities at AChE.


Asunto(s)
Acetilcolinesterasa/metabolismo , Aminas/química , Butirilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacología , Quinazolinonas/farmacología , Sitios de Unión/efectos de los fármacos , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Quinazolinonas/síntesis química , Quinazolinonas/química , Relación Estructura-Actividad
12.
J Biol Chem ; 289(23): 15987-6005, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24739388

RESUMEN

Determining the molecular basis for target selectivity is of particular importance in drug discovery. The ideal antibiotic should be active against a broad spectrum of pathogenic organisms with a minimal effect on human targets. CG400549, a Staphylococcus-specific 2-pyridone compound that inhibits the enoyl-acyl carrier protein reductase (FabI), has recently been shown to possess human efficacy for the treatment of methicillin-resistant Staphylococcus aureus infections, which constitute a serious threat to human health. In this study, we solved the structures of three different FabI homologues in complex with several pyridone inhibitors, including CG400549. Based on these structures, we rationalize the 65-fold reduced affinity of CG400549 toward Escherichia coli versus S. aureus FabI and implement concepts to improve the spectrum of antibacterial activity. The identification of different conformational states along the reaction coordinate of the enzymatic hydride transfer provides an elegant visual depiction of the relationship between catalysis and inhibition, which facilitates rational inhibitor design. Ultimately, we developed the novel 4-pyridone-based FabI inhibitor PT166 that retained favorable pharmacokinetics and efficacy in a mouse model of S. aureus infection with extended activity against Gram-negative and mycobacterial organisms.


Asunto(s)
Antibacterianos/farmacología , Diseño de Fármacos , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Piridonas/farmacología , Animales , Antibacterianos/química , Antibacterianos/farmacocinética , Secuencia de Bases , Cristalografía por Rayos X , Cartilla de ADN , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Femenino , Ratones , Ratones Endogámicos ICR , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Reacción en Cadena de la Polimerasa , Piridonas/química , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo
13.
ACS Chem Neurosci ; 5(5): 377-89, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24628027

RESUMEN

Photochromic cholinesterase inhibitors were obtained from cis-1,2-α-dithienylethene-based compounds by incorporating one or two aminopolymethylene tacrine groups. All target compounds are potent acetyl- (AChE) and butyrylcholinesterase (BChE) inhibitors in the nanomolar concentration range. Compound 11b bearing an octylene linker exhibited interactions with both the catalytic active site (CAS) and the peripheral anionic site (PAS) of AChE. Yet upon irradiation with light, the mechanism of interaction varied from one photochromic form to another, which was investigated by kinetic studies and proved "photoswitchable". The AChE-induced ß-amyloid (Aß) aggregation assay gave further experimental support to this finding: Aß1-40 aggregation catalyzed by the PAS of AChE might be inhibited by compound 11b in a concentration-dependent manner and seems to occur only with one photochromic form. Computational docking studies provided potential binding modes of the compound. Docking studies and molecular dynamics (MD) simulations for the ring-open and -closed form indicate a difference in binding. Although both forms can interact with the PAS, more stable interactions are observed for the ring-open form based upon stabilization of a water molecule network within the enzyme, whereas the ring-closed form lacks the required conformational flexibility for an analogous binding mode. The photoswitchable inhibitor identified might serve as valuable molecular tool to investigate the different biological properties of AChE as well as its role in pathogenesis of AD in in vitro assays.


Asunto(s)
Inhibidores de la Colinesterasa/farmacología , Modelos Moleculares , Fármacos Fotosensibilizantes/farmacología , Agregado de Proteínas/efectos de los fármacos , Animales , Inhibidores de la Colinesterasa/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Relación Estructura-Actividad
14.
ACS Chem Neurosci ; 5(3): 225-42, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24422467

RESUMEN

Combination of AChE inhibiting and histamine H3 receptor antagonizing properties in a single molecule might show synergistic effects to improve cognitive deficits in Alzheimer's disease, since both pharmacological actions are able to enhance cholinergic neurotransmission in the cortex. However, whereas AChE inhibitors prevent hydrolysis of acetylcholine also peripherally, histamine H3 antagonists will raise acetylcholine levels mostly in the brain due to predominant occurrence of the receptor in the central nervous system. In this work, we designed and synthesized two novel classes of tri- and tetracyclic nitrogen-bridgehead compounds acting as dual AChE inhibitors and histamine H3 antagonists by combining the nitrogen-bridgehead moiety of novel AChE inhibitors with a second N-basic fragment based on the piperidinylpropoxy pharmacophore with different spacer lengths. Intensive structure-activity relationships (SARs) with regard to both biological targets led to compound 41 which showed balanced affinities as hAChE inhibitor with IC50 = 33.9 nM, and hH3R antagonism with Ki = 76.2 nM with greater than 200-fold selectivity over the other histamine receptor subtypes. Molecular docking studies were performed to explain the potent AChE inhibition of the target compounds and molecular dynamics studies to explain high affinity at the hH3R.


Asunto(s)
Inhibidores de la Colinesterasa/química , Antagonistas de los Receptores Histamínicos H3/química , Compuestos de Nitrógeno/química , Acetilcolinesterasa/metabolismo , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacología , GTP Fosfohidrolasas/metabolismo , Antagonistas de los Receptores Histamínicos H3/síntesis química , Antagonistas de los Receptores Histamínicos H3/metabolismo , Antagonistas de los Receptores Histamínicos H3/farmacología , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Compuestos de Nitrógeno/síntesis química , Compuestos de Nitrógeno/farmacocinética , Ensayo de Unión Radioligante , Receptores Histamínicos/genética , Receptores Histamínicos/metabolismo , Receptores Histamínicos H1/genética , Receptores Histamínicos H1/metabolismo , Receptores Histamínicos H2/genética , Receptores Histamínicos H2/metabolismo , Receptores Histamínicos H3/genética , Receptores Histamínicos H3/metabolismo
15.
Biochemistry ; 52(24): 4217-28, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23697754

RESUMEN

Drug-target kinetics has recently emerged as an especially important facet of the drug discovery process. In particular, prolonged drug-target residence times may confer enhanced efficacy and selectivity in the open in vivo system. However, the lack of accurate kinetic and structural data for a series of congeneric compounds hinders the rational design of inhibitors with decreased off-rates. Therefore, we chose the Staphylococcus aureus enoyl-ACP reductase (saFabI)--an important target for the development of new anti-staphylococcal drugs--as a model system to rationalize and optimize the drug-target residence time on a structural basis. Using our new, efficient, and widely applicable mechanistically informed kinetic approach, we obtained a full characterization of saFabI inhibition by a series of 20 diphenyl ethers complemented by a collection of 9 saFabI-inhibitor crystal structures. We identified a strong correlation between the affinities of the investigated saFabI diphenyl ether inhibitors and their corresponding residence times, which can be rationalized on a structural basis. Because of its favorable interactions with the enzyme, the residence time of our most potent compound exceeds 10 h. In addition, we found that affinity and residence time in this system can be significantly enhanced by modifications predictable by a careful consideration of catalysis. Our study provides a blueprint for investigating and prolonging drug-target kinetics and may aid in the rational design of long-residence-time inhibitors targeting the essential saFabI enzyme.


Asunto(s)
Enoil-ACP Reductasa (NADPH Específica B)/química , Enoil-ACP Reductasa (NADH)/química , Inhibidores Enzimáticos/farmacología , Proteínas de Escherichia coli/química , Acido Graso Sintasa Tipo II/química , Staphylococcus aureus/enzimología , Catálisis , Química Farmacéutica , Cristalografía por Rayos X , Diseño de Fármacos , Escherichia coli/metabolismo , Ácidos Grasos/química , Enlace de Hidrógeno , Modelos Moleculares , Unión Proteica , Conformación Proteica , Termodinámica , Factores de Tiempo
16.
J Chem Inf Model ; 53(8): 1923-33, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23705795

RESUMEN

A major shortcoming of empirical scoring functions for protein-ligand complexes is the low degree of correlation between predicted and experimental binding affinities, as frequently observed not only for large and diverse data sets but also for SAR series of individual targets. Improvements can be envisaged by developing new descriptors, employing larger training sets of higher quality, and resorting to more sophisticated regression methods. Herein, we describe the use of SFCscore descriptors to develop an improved scoring function by means of a PDBbind training set of 1005 complexes in combination with random forest for regression. This provided SFCscore(RF) as a new scoring function with significantly improved performance on the PDBbind and CSAR-NRC HiQ benchmarks in comparison to previously developed SFCscore functions. A leave-cluster-out cross-validation and performance in the CSAR 2012 scoring exercise point out remaining limitations but also directions for further improvements of SFCscore(RF) and empirical scoring functions in general.


Asunto(s)
Algoritmos , Proteínas/metabolismo , Análisis por Conglomerados , Bases de Datos Farmacéuticas , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Proteínas/química , Reproducibilidad de los Resultados
17.
Eur J Pharm Sci ; 49(4): 603-13, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23643737

RESUMEN

Given the fundamentally multifactorial character of Alzheimer's disease (AD), addressing more than one target for disease modification or therapy is expected to be highly advantageous. Here, following the cholinergic hypothesis, we aimed to inhibit both acetyl- and butyrylcholinesterase (AChE and BuChE) in order to increase the concentration of acetylcholine in the synaptic cleft. In addition, the formation of the amyloid ß fibrils should be inhibited and already preformed fibrils should be destroyed. Based on a recently identified AChE inhibitor with a 1,4-substituted 4-(1H)-pyridylene-hydrazone skeleton, a substance library has been generated and tested for inhibition of AChE, BuChE, and fibril formation. Blood-brain barrier mobility was ensured by a transwell assay. Whereas the p-nitrosubstituted compound 18C shows an anti-AChE activity in the nanomolar range of concentration (IC50=90 nM), the bisnaphthyl substituted compound 20L was found to be the best overall inhibitor of AChE/BuChE and enhances the fibril destruction.


Asunto(s)
Acetilcolinesterasa/metabolismo , Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Butirilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacología , Hidrazonas/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Células Endoteliales , Células HEK293 , Humanos , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
18.
Int J Mol Sci ; 14(3): 5608-32, 2013 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-23478436

RESUMEN

Cholinesterases are important biological targets responsible for regulation of cholinergic transmission, and their inhibitors are used for the treatment of Alzheimer's disease. To design new cholinesterase inhibitors, of different structure-based design strategies was followed, including the modification of compounds from a previously developed library and a fragment-based design approach. This led to the selection of heterodimeric structures as potential inhibitors. Synthesis and biological evaluation of selected candidates confirmed that the designed compounds were acetylcholinesterase inhibitors with IC50 values in the mid-nanomolar to low micromolar range, and some of them were also butyrylcholinesterase inhibitors.

19.
PLoS One ; 8(2): e56788, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23451087

RESUMEN

The trypanothione synthetase (TryS) catalyses the two-step biosynthesis of trypanothione from spermidine and glutathione and is an attractive new drug target for the development of trypanocidal and antileishmanial drugs, especially since the structural information of TryS from Leishmania major has become available. Unfortunately, the TryS structure was solved without any of the substrates and lacks loop regions that are mechanistically important. This contribution describes docking and molecular dynamics simulations that led to further insights into trypanothione biosynthesis and, in particular, explains the binding modes of substrates for the second catalytic step. The structural model essentially confirm previously proposed binding sites for glutathione, ATP and two Mg(2+) ions, which appear identical for both catalytic steps. The analysis of an unsolved loop region near the proposed spermidine binding site revealed a new pocket that was demonstrated to bind glutathionylspermidine in an inverted orientation. For the second step of trypanothione synthesis glutathionylspermidine is bound in a way that preferentially allows N(1)-glutathionylation of N(8)-glutathionylspermidine, classifying N(8)-glutathionylspermidine as the favoured substrate. By inhibitor docking, the binding site for N(8)-glutathionylspermidine was characterised as druggable.


Asunto(s)
Amida Sintasas/metabolismo , Glutatión/análogos & derivados , Simulación de Dinámica Molecular , Espermidina/análogos & derivados , Biología Computacional , Glutatión/biosíntesis , Glutatión/química , Glutatión/metabolismo , Unión Proteica , Espermidina/biosíntesis , Espermidina/química , Espermidina/metabolismo
20.
Bioinformatics ; 29(1): 62-8, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23104888

RESUMEN

MOTIVATION: With >8 million new cases in 2010, particularly documented in developing countries, tuberculosis (TB) is still a highly present pandemic and often terminal. This is also due to the emergence of antibiotic-resistant strains (MDR-TB and XDR-TB) of the primary causative TB agent Mycobacterium tuberculosis (MTB). Efforts to develop new effective drugs against MTB are restrained by the unique and largely impermeable composition of the mycobacterial cell wall. RESULTS: Based on a database of antimycobacterial substances (CDD TB), 3815 compounds were classified as active and thus permeable. A data mining approach was conducted to gather the physico-chemical similarities of these substances and delimit them from a generic dataset of drug-like molecules. On the basis of the differences in these datasets, a regression model was generated and implemented into the online tool MycPermCheck to predict the permeability probability of small organic compounds. DISCUSSION: Given the current lack of precise molecular criteria determining mycobacterial permeability, MycPermCheck represents an unprecedented prediction tool intended to support antimycobacterial drug discovery. It follows a novel knowledge-driven approach to estimate the permeability probability of small organic compounds. As such, MycPermCheck can be used intuitively as an additional selection criterion for potential new inhibitors against MTB. Based on the validation results, its performance is expected to be of high practical value for virtual screening purposes. AVAILABILITY: The online tool is freely accessible under the URL http://www.mycpermcheck.aksotriffer.pharmazie.uni-wuerzburg.de


Asunto(s)
Antituberculosos/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Programas Informáticos , Antituberculosos/química , Antituberculosos/farmacología , Permeabilidad de la Membrana Celular , Minería de Datos , Descubrimiento de Drogas , Mycobacterium tuberculosis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...