Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
J Med Chem ; 66(14): 9797-9822, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37440686

RESUMEN

In cystic fibrosis (CF), deletion of phenylalanine 508 (F508del) in the CF transmembrane conductance regulator (CFTR) is associated to misfolding and defective gating of the mutant channel. One of the most promising CF drug targets is the ubiquitin ligase RNF5, which promotes F508del-CFTR degradation. Recently, the first ever reported inhibitor of RNF5 was discovered, i.e., the 1,2,4-thiadiazol-5-ylidene inh-2. Here, we designed and synthesized a series of new analogues to explore the structure-activity relationships (SAR) of this class of compounds. SAR efforts ultimately led to compound 16, which showed a greater F508del-CFTR corrector activity than inh-2, good tolerability, and no toxic side effects. Analogue 16 increased the basal level of autophagy similar to what has been described with RNF5 silencing. Furthermore, co-treatment with 16 significantly improved the F508del-CFTR rescue induced by the triple combination elexacaftor/tezacaftor/ivacaftor in CFBE41o- cells. These findings validate the 1,2,4-thiadiazolylidene scaffold for the discovery of novel RNF5 inhibitors and provide evidence to pursue this unprecedented strategy for the treatment of CF.


Asunto(s)
Fibrosis Quística , Tiadiazoles , Humanos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Tiadiazoles/farmacología , Tiadiazoles/uso terapéutico , Ubiquitina-Proteína Ligasas/metabolismo , Relación Estructura-Actividad , Aminofenoles , Benzodioxoles/farmacología , Mutación , Proteínas de Unión al ADN/metabolismo
2.
WIREs Mech Dis ; 15(4): e1607, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36958762

RESUMEN

From the (patho)physiological point of view, diseases can be considered as emergent properties of living systems stemming from the complexity of these systems. Complex systems display some typical features, including the presence of emergent behavior and the organization in successive hierarchic levels. Drug treatments increase this complexity scenario, and from some years the use of network models has been introduced to describe drug-disease systems and to make predictions about them with regard to several aspects related to drug discovery. Here, we review some recent examples thereof with the aim to illustrate how network science tools can be very effective in addressing both tasks. We will examine the use of bipartite networks that lead to the important concept of "disease module", as well as the introduction of more articulated models, like multi-scale and multiplex networks, able to describe disease systems at increasing levels of organization. Examples of predictive models will then be discussed, considering both those that exploit approaches purely based on graph theory and those that integrate machine learning methods. A short account of both kinds of methodological applications will be provided. Finally, the point will be made on the present situation of modeling complex drug-disease systems highlighting some open issues. This article is categorized under: Neurological Diseases > Computational Models Infectious Diseases > Computational Models Cardiovascular Diseases > Computational Models.


Asunto(s)
Enfermedades Cardiovasculares , Modelos Biológicos , Humanos , Descubrimiento de Drogas/métodos , Aprendizaje Automático
3.
QRB Discov ; 3: e22, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37529286

RESUMEN

RNA molecules play many functional and regulatory roles in cells, and hence, have gained considerable traction in recent times as therapeutic interventions. Within drug discovery, structure-based approaches have successfully identified potent and selective small-molecule modulators of pharmaceutically relevant protein targets. Here, we embrace the perspective of computational chemists who use these traditional approaches, and we discuss the challenges of extending these methods to target RNA molecules. In particular, we focus on recognition between RNA and small-molecule binders, on selectivity, and on the expected properties of RNA ligands.

4.
Sci Rep ; 11(1): 19426, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34593915

RESUMEN

The COVID-19 pandemic poses a huge problem of public health that requires the implementation of all available means to contrast it, and drugs are one of them. In this context, we observed an unmet need of depicting the continuously evolving scenario of the ongoing drug clinical trials through an easy-to-use, freely accessible online tool. Starting from this consideration, we developed COVIDrugNet ( http://compmedchem.unibo.it/covidrugnet ), a web application that allows users to capture a holistic view and keep up to date on how the clinical drug research is responding to the SARS-CoV-2 infection. Here, we describe the web app and show through some examples how one can explore the whole landscape of medicines in clinical trial for the treatment of COVID-19 and try to probe the consistency of the current approaches with the available biological and pharmacological evidence. We conclude that careful analyses of the COVID-19 drug-target system based on COVIDrugNet can help to understand the biological implications of the proposed drug options, and eventually improve the search for more effective therapies.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Biología Computacional/métodos , Ensayos Clínicos como Asunto , Biología Computacional/instrumentación , Bases de Datos Farmacéuticas , Reposicionamiento de Medicamentos , Humanos , Internet , Proteínas Virales/metabolismo
5.
J Inorg Biochem ; 223: 111554, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34325209

RESUMEN

The survival of several pathogenic bacteria, such as Helicobacter pylori (Hp), relies on the activity of the nickel-dependent enzyme urease. Nickel insertion into urease is mediated by a multimeric chaperone complex (HpUreDFG) that is responsible for the transport of Ni(II) from a conserved metal binding motif located in the UreG dimer (CPH motif) to the catalytic site of the enzyme. The X-ray structure of HpUreDFG revealed the presence of water-filled tunnels that were proposed as a route for Ni(II) translocation. Here, we probe the transport of Ni(II) through the internal tunnels of HpUreDFG, from the CPH motif to the external surface of the complex, using microsecond-long enhanced molecular dynamics simulations. The results suggest a "bucket-brigade" mechanism whereby Ni(II) can be transported through a series of stations found along these internal pathways.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Chaperonas Moleculares/metabolismo , Níquel/metabolismo , Simulación de Dinámica Molecular , Níquel/química , Unión Proteica , Conformación Proteica
6.
J Chem Inf Model ; 61(6): 3091-3108, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-33998810

RESUMEN

Janus kinases (JAKs) are a family of proinflammatory enzymes able to mediate the immune responses and the inflammatory cascade by modulating multiple cytokine expressions as well as various growth factors. In the present study, the inhibition of the JAK-signal transducer and activator of transcription (STAT) signaling pathway is explored as a potential strategy for treating autoimmune and inflammatory disorders. A computationally driven approach aimed at identifying novel JAK inhibitors based on molecular topology, docking, and molecular dynamics simulations was carried out. For the best candidates selected, the inhibitory activity against JAK2 was evaluated in vitro. Two hit compounds with a novel chemical scaffold, 4 (IC50 = 0.81 µM) and 7 (IC50 = 0.64 µM), showed promising results when compared with the reference drug Tofacitinib (IC50 = 0.031 µM).


Asunto(s)
Quinasas Janus , Inhibidores de Proteínas Quinasas , Quinasas Janus/metabolismo , Ligandos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Transductores
7.
J Chem Inf Model ; 60(12): 6532-6543, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-33295174

RESUMEN

In recent years, the K2P family of potassium channels has been the subject of intense research activity. Owing to the complex function and regulation of this family of ion channels, it is common practice to complement experimental findings with the atomistic description provided by computational approaches such as molecular dynamics (MD) simulations, especially, in light of the unprecedented timescales accessible at present. However, despite recent substantial improvements, the accuracy of MD simulations is still undermined by the intrinsic limitations of force fields. Here, we systematically assessed the performance of the most popular force fields employed to study ion channels at timescales that are orders of magnitude greater than the ones accessible when these energy functions were first developed. Using 32 µs of trajectories, we investigated the dynamics of a member of the K2P ion channel family, the TRAAK channel, using two established force fields in simulations of biological systems: AMBER and CHARMM. We found that while results are comparable on the nanosecond timescales, significant inconsistencies arise at microsecond timescales.


Asunto(s)
Simulación de Dinámica Molecular , Canales de Potasio , Canales Iónicos
8.
Molecules ; 25(12)2020 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-32599898

RESUMEN

Urease is a nickel-containing enzyme that is essential for the survival of several and often deadly pathogenic bacterial strains, including Helicobacter pylori. Notwithstanding several attempts, the development of direct urease inhibitors without side effects for the human host remains, to date, elusive. The recently solved X-ray structure of the HpUreDFG accessory complex involved in the activation of urease opens new perspectives for structure-based drug discovery. In particular, the quaternary assembly and the presence of internal tunnels for nickel translocation offer an intriguing possibility to target the HpUreDFG complex in the search of indirect urease inhibitors. In this work, we adopted a theoretical framework to investigate such a hypothesis. Specifically, we searched for putative binding sites located at the protein-protein interfaces on the HpUreDFG complex, and we challenged their druggability through structure-based virtual screening. We show that, by virtue of the presence of tunnels, some protein-protein interfaces on the HpUreDFG complex are intrinsically well suited for hosting small molecules, and, as such, they possess good potential for future drug design endeavors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Helicobacter pylori/metabolismo , Complejos Multiproteicos/metabolismo , Ureasa/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Simulación de Dinámica Molecular , Complejos Multiproteicos/química , Proteínas de Unión a Fosfato/química , Proteínas de Unión a Fosfato/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Ureasa/química , Ureasa/metabolismo
9.
J Med Chem ; 63(16): 8653-8666, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32338900

RESUMEN

Network theory provides one of the most potent analysis tools for the study of complex systems. In this paper, we illustrate the network-based perspective in drug research and how it is coherent with the new paradigm of drug discovery. We first present data sources from which networks are built, then show some examples of how the networks can be used to investigate drug-related systems. A section is devoted to network-based inference applications, i.e., prediction methods based on interactomes, that can be used to identify putative drug-target interactions without resorting to 3D modeling. Finally, we present some aspects of Boolean networks dynamics, anticipating that it might become a very potent modeling framework to develop in silico screening protocols able to simulate phenotypic screening experiments. We conclude that network applications integrated with machine learning and 3D modeling methods will become an indispensable tool for computational drug discovery in the next years.


Asunto(s)
Descubrimiento de Drogas/métodos , Modelos Biológicos , Simulación por Computador , Bases de Datos Factuales , Humanos , Aprendizaje Automático
10.
J Chem Inf Model ; 60(4): 2294-2303, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32233432

RESUMEN

γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the central nervous system (CNS). Dysfunctional GABAergic neurotransmission is associated with numerous neurological and neuropsychiatric disorders. The GABAB receptor (GABAB-R) is a heterodimeric class C G protein-coupled receptor (GPCR) comprised of GABAB1a/b and GABAB2 subunits. The orthosteric binding site for GABA is located in the extracellular Venus flytrap (VFT) domain of the GABAB1a/b. Knowledge about molecular mechanisms and druggable receptor conformations associated with activation is highly important to understand the receptor function and for rational drug design. Currently, the conformational changes of the receptor upon activation are not well described. On the basis of other class C members, the VFT is proposed to fluctuate between an open/inactive and closed/active state and one of these conformations is stabilized upon ligand binding. In the present study, we investigated the dynamics of the GABAB1b-R VFT in the apo form by combining unbiased molecular dynamics with path-metadynamics. Our simulations confirmed the open/inactive and closed/active state as the main conformations adopted by the receptor. Sizeable energy barriers were found between stable minima, suggesting a relatively slow interconversion. Previously undisclosed metastable states were also identified, which might hold potential for future drug discovery efforts.


Asunto(s)
Droseraceae , Receptores de GABA-B , Modelos Moleculares , Receptores de GABA , Ácido gamma-Aminobutírico
11.
Eur J Med Chem ; 188: 111975, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31940507

RESUMEN

Local changes in the structure of G-protein coupled receptors (GPCR) binders largely affect their pharmacological profile. While the sought efficacy can be empirically obtained by introducing local modifications, the underlining structural explanation can remain elusive. Here, molecular dynamics (MD) simulations of the eticlopride-bound inactive state of the Dopamine D3 Receptor (D3DR) have been clustered using a machine learning-based approach in the attempt to rationalize the efficacy change in four congeneric modulators. Accumulating extended MD trajectories of receptor-ligand complexes, we observed how the increase in ligand flexibility progressively destabilized the crystal structure of the inactivated receptor. To prospectively validate this model, a partial agonist was rationally designed based on structural insights and computational modeling, and eventually synthesized and tested. Results turned out to be in line with the predictions. This case study suggests that the investigation of ligand flexibility in the framework of extended MD simulations can assist and inform drug design strategies, highlighting its potential role as a powerful in silico counterpart to functional assays.


Asunto(s)
Carbamatos/metabolismo , Agonistas de Dopamina/metabolismo , Antagonistas de Dopamina/metabolismo , Piperazinas/metabolismo , Receptores de Dopamina D3/metabolismo , Animales , Sitios de Unión , Células CHO , Carbamatos/química , Cricetulus , Agonistas de Dopamina/química , Antagonistas de Dopamina/química , Diseño de Fármacos , Humanos , Ligandos , Aprendizaje Automático , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Piperazinas/química , Conformación Proteica , Receptores de Dopamina D3/química , Salicilamidas/metabolismo
12.
J Chem Theory Comput ; 15(10): 5689-5702, 2019 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-31436987

RESUMEN

Unveiling the mechanistic features of drug-target binding is of central interest in biophysics and drug discovery. Herein, we address this challenge by combining two major computational approaches, namely, Molecular Dynamics (MD) simulations and Markov State Models (MSM), with a Path Collective Variables (PCVs) description coupled with metadynamics. We apply our methodology to reconstruct the binding process of the antagonist alprenolol to the ß2-adrenergic receptor, a well-established pharmaceutical target. The devised protocol allowed us to estimate the binding free energy and identify the minimum free energy path leading to the protein-ligand complex. In summary, we show that MSM and PCVs can be efficiently integrated to shed light upon mechanistic and energetic details underlying complex recognition processes in biological systems.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 2/química , Alprenolol/química , Cadenas de Markov , Simulación de Dinámica Molecular , Receptores Adrenérgicos beta 2/química , Termodinámica
13.
J Chem Inf Model ; 59(1): 535-549, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30500211

RESUMEN

Computational approaches currently assist medicinal chemistry through the entire drug discovery pipeline. However, while several computational tools and strategies are available to predict binding affinity, predicting the drug-target binding kinetics is still a matter of ongoing research. Here, we challenge scaled molecular dynamics simulations to assess the off-rates for a series of structurally diverse inhibitors of the heat shock protein 90 (Hsp90) covering 3 orders of magnitude in their experimental residence times. The derived computational predictions are in overall good agreement with experimental data. Aside from the estimation of exit times, unbinding pathways were assessed through dimensionality reduction techniques. The data analysis framework proposed in this work could lead to better understanding of the mechanistic aspects related to the observed kinetic behavior.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Simulación de Dinámica Molecular , Preparaciones Farmacéuticas/metabolismo , Proteínas HSP90 de Choque Térmico/química , Humanos , Cinética , Ligandos , Unión Proteica , Conformación Proteica
15.
J Chem Inf Model ; 58(11): 2255-2265, 2018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30339750

RESUMEN

Traditionally, a drug potency is expressed in terms of thermodynamic quantities, mostly Kd, and empirical IC50 values. Although binding affinity as an estimate of drug activity remains relevant, it is increasingly clear that it is also important to include (un)binding kinetic parameters in the characterization of potential drug-like molecules. Herein, we used standard in silico screening to identify a series of structurally related inhibitors of hDAAO, a flavoprotein involved in schizophrenia and neuropathic pain. We applied a novel methodology, based on scaled molecular dynamics, to rank them according to their residence times. Notably, we challenged the application in a prospective fashion for the first time. The good agreement between experimental residence times and the predicted residence times highlighted the procedure's reliability in both predictive and refinement scenarios. Additionally, through further inspection of the performed simulations, we substantiated a previous hypothesis on the involvement of a protein loop during ligand unbinding.


Asunto(s)
D-Aminoácido Oxidasa/antagonistas & inhibidores , D-Aminoácido Oxidasa/metabolismo , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , D-Aminoácido Oxidasa/química , Humanos , Cinética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Termodinámica
16.
J Chem Inf Model ; 58(2): 490-500, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29378136

RESUMEN

Predicting the geometry of protein-ligand binding complexes is of primary importance for structure-based drug discovery. Molecular dynamics (MD) is emerging as a reliable computational tool for use in conjunction with, or an alternative to, docking methods. However, simulating the protein-ligand binding process often requires very expensive simulations. This drastically limits the practical application of MD-based approaches. Here, we propose a general framework to accelerate the generation of putative protein-ligand binding modes using potential-scaled MD simulations. The proposed dynamical protocol has been applied to two pharmaceutically relevant systems (GSK-3ß and the N-terminal domain of HSP90α). Our approach is fully independent of any predefined reaction coordinate (or collective variable). It identified the correct binding mode of several ligands and can thus save valuable computational time in dynamic docking simulations.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas/metabolismo , Sitios de Unión , Ligandos , Unión Proteica
17.
Molecules ; 22(11)2017 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-29165360

RESUMEN

Molecular docking is the methodology of choice for studying in silico protein-ligand binding and for prioritizing compounds to discover new lead candidates. Traditional docking simulations suffer from major limitations, mostly related to the static or semi-flexible treatment of ligands and targets. They also neglect solvation and entropic effects, which strongly limits their predictive power. During the last decade, methods based on full atomistic molecular dynamics (MD) have emerged as a valid alternative for simulating macromolecular complexes. In principle, compared to traditional docking, MD allows the full exploration of drug-target recognition and binding from both the mechanistic and energetic points of view (dynamic docking). Binding and unbinding kinetic constants can also be determined. While dynamic docking is still too computationally expensive to be routinely used in fast-paced drug discovery programs, the advent of faster computing architectures and advanced simulation methodologies are changing this scenario. It is feasible that dynamic docking will replace static docking approaches in the near future, leading to a major paradigm shift in in silico drug discovery. Against this background, we review the key achievements that have paved the way for this progress.


Asunto(s)
Simulación por Computador , Descubrimiento de Drogas , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Algoritmos , Ligandos , Modelos Moleculares , Unión Proteica , Proteínas/química , Proteínas/metabolismo
18.
J Phys Chem B ; 121(41): 9572-9582, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-28926706

RESUMEN

Intrinsically disordered proteins (IDPs) are emerging as an important class of the proteome. Being able to interact with different molecular targets, they participate in many physiological and pathological activities. However, due to their intrinsically heterogeneous nature, determining the equilibrium properties of IDPs is still a challenge for biophysics. Herein, we applied state-of-the-art enhanced sampling methods to Sev NTAIL, a test case of IDPs, and constructed a bin-based kinetic model to unveil the underlying kinetics. To validate our simulation strategy, we compared the predicted NMR properties against available experimental data. Our simulations reveal a rough free-energy surface comprising multiple local minima, which are separated by low energy barriers. Moreover, we identified interconversion rates between the main kinetic states, which lie in the sub-µs time scales, as suggested in previous works for Sev NTAIL. Therefore, the emerging picture is in agreement with the atomic-level properties possessed by the free IDP in solution. By providing both a thermodynamic and kinetic characterization of this IDP test case, our study demonstrates how computational methods can be effective tools for studying this challenging class of proteins.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Cinética , Simulación de Dinámica Molecular , Conformación Proteica , Termodinámica
19.
Eur J Pharm Sci ; 105: 91-98, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28501492

RESUMEN

In normal cells, heat shock response (HSR) is rapidly induced in response to a variety of harmful conditions and represents one of the most efficient defense mechanism. In cancer tissues, constitutive activation converts HSR into a life-threatening process, which plays a major role in helping cell survival and proliferation. Overexpression of heat shock proteins (HSPs) has been widely reported in human cancers and was found to correlate with tumor progression. Hepatocellular carcinoma is one of the conditions in which HSR activation was shown to have the highest clinical significance. Transcription of HSPs is induced by HSF-1, which also activates glycolytic metabolism and increases the expression of LDH-A, the master regulator of the Warburg effect. In this paper, we tried to explore the relationship between HSR and LDH-A. In cultured hepatocellular carcinoma cells, by using two enzyme inhibitors (oxamate and galloflavin), we found that the reduction of LDH-A activity led to decreased level and function of the major HSPs involved in tumorigenesis. Galloflavin (a polyphenol) also inhibited the ATPase activity of two of the examined HSPs. Finally, hindering HSR markedly lowered the alpha-fetoprotein cellular levels and induced senescence. Specific inhibitors of single HSPs are currently under evaluation in different neoplastic diseases. However, one of the effects usually observed during treatment is a compensatory elevation of other HSPs, which decreases treatment efficacy. Our results highlight a connection between LDH and HSR and suggest LDH inhibition as a way to globally impact on this tumor promoting process.


Asunto(s)
Respuesta al Choque Térmico , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Adenosina Trifosfatasas/metabolismo , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Proteínas de Choque Térmico/genética , Humanos , Isocumarinas/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Neoplasias Hepáticas/metabolismo , Compuestos Orgánicos/farmacología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , alfa-Fetoproteínas/genética
20.
J Comput Chem ; 38(21): 1834-1843, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28558120

RESUMEN

Force-field parameters are developed for a multisite model of Ni(II) ions to be used in molecular dynamics simulations combined to enhanced sampling methods. The performances of two charge-partitioning schemes are validated by taking into account structural, thermodynamic, and kinetic observables. One of the two models, featuring partial charges on the dummy atoms only, matches both Ni(II) free energy of solvation and water exchange rates. Such model is particularly suited to study complexation events at a fully dynamic description. © 2017 Wiley Periodicals, Inc.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...