Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(5): 4255-4269, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35188371

ABSTRACT

Gallinamide A, a metabolite of the marine cyanobacterium Schizothrix sp., selectively inhibits cathepsin L-like cysteine proteases. We evaluated the potency of gallinamide A and 23 synthetic analogues against intracellular Trypanosoma cruzi amastigotes and the cysteine protease, cruzain. We determined the co-crystal structures of cruzain with gallinamide A and two synthetic analogues at ∼2 Å. SAR data revealed that the N-terminal end of gallinamide A is loosely bound and weakly contributes in drug-target interactions. At the C-terminus, the intramolecular π-π stacking interactions between the aromatic substituents at P1' and P1 restrict the bioactive conformation of the inhibitors, thus minimizing the entropic loss associated with target binding. Molecular dynamics simulations showed that in the absence of an aromatic group at P1, the substituent at P1' interacts with tryptophan-184. The P1-P1' interactions had no effect on anti-cruzain activity, whereas anti-T. cruzi potency increased by ∼fivefold, likely due to an increase in solubility/permeability of the analogues.


Subject(s)
Cysteine Proteases , Trypanosoma cruzi , Antimicrobial Cationic Peptides/chemistry , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Protozoan Proteins
2.
J Comput Aided Mol Des ; 35(10): 1067-1079, 2021 10.
Article in English | MEDLINE | ID: mdl-34617191

ABSTRACT

Falcipain-2 (FP-2) is a Plasmodium falciparum hemoglobinase widely targeted in the search for antimalarials. FP-2 can be allosterically modulated by various noncompetitive inhibitors that have been serendipitously identified. Moreover, the crystal structures of two inhibitors bound to an allosteric site, termed site 6, of the homolog enzyme human cathepsin K (hCatK) suggest that the equivalent region in FP-2 might play a similar role. Here, we conduct the rational identification of FP-2 inhibitors through virtual screenings (VS) of compounds into several pocket-like conformations of site 6, sampled during molecular dynamics (MD) simulations of the free enzyme. Two noncompetitive inhibitors, ZINC03225317 and ZINC72290660, were confirmed using in vitro enzymatic assays and their poses into site 6 led to calculated binding free energies matching the experimental ones. Our results provide strong evidence about the allosteric inhibition of FP-2 through binding of small molecules to site 6, thus opening the way toward the discovery of new inhibitors against this enzyme.


Subject(s)
Antimalarials/pharmacology , Computer Simulation , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Plasmodium falciparum/drug effects , Allosteric Site , Antimalarials/chemistry , Cysteine Proteinase Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Plasmodium falciparum/enzymology , Protein Binding , Structure-Activity Relationship
3.
Comput Struct Biotechnol J ; 19: 2027-2044, 2021.
Article in English | MEDLINE | ID: mdl-33995900

ABSTRACT

During their life cycle, Leishmania parasites display a fine-tuned regulation of the mRNA translation through the differential expression of isoforms of eukaryotic translation initiation factor 4E (LeishIF4Es). The interaction between allosteric modulators such as 4E-interacting proteins (4E-IPs) and LeishIF4E affects the affinity of this initiation factor for the mRNA cap. Here, several computational approaches were employed to elucidate the molecular bases of the previously-reported allosteric modulation in L. major exerted by 4E-IP1 (Lm4E-IP1) on eukaryotic translation initiation factor 4E 1 (LmIF4E-1). Molecular dynamics (MD) simulations and accurate binding free energy calculations (ΔGbind ) were combined with network-based modeling of residue-residue correlations. We also describe the differences in internal motions of LmIF4E-1 apo form, cap-bound, and Lm4E-IP1-bound systems. Through community network calculations, the differences in the allosteric pathways of allosterically-inhibited and active forms of LmIF4E-1 were revealed. The ΔGbind values show significant differences between the active and inhibited systems, which are in agreement with the available experimental data. Our study thoroughly describes the dynamical perturbations of LmIF4E-1 cap-binding site triggered by Lm4E-IP1. These findings are not only essential for the understanding of a critical process of trypanosomatids' gene expression but also for gaining insight into the allostery of eukaryotic IF4Es, which could be useful for structure-based design of drugs against this protein family.

4.
Mol Pharmacol ; 98(6): 770-780, 2020 12.
Article in English | MEDLINE | ID: mdl-33008918

ABSTRACT

Cytochromes P450 (P450, CYP) metabolize a wide variety of endogenous and exogenous lipophilic molecules, including most drugs. Sterol 14α-demethylase (CYP51) is a target for antifungal drugs known as conazoles. Using X-ray crystallography, we have discovered a domain-swap homodimerization mode in CYP51 from a human pathogen, Acanthamoeba castellanii CYP51 (AcCYP51). Recombinant AcCYP51 with a truncated transmembrane helix was purified as a heterogeneous mixture corresponding to the dimer and monomer units. Spectral analyses of these two populations have shown that the CO-bound ferrous form of the dimeric protein absorbed at 448 nm (catalytically competent form), whereas the monomeric form absorbed at 420 nm (catalytically incompetent form). AcCYP51 dimerized head-to-head via N-termini swapping, resulting in formation of a nonplanar protein-protein interface exceeding 2000 Å2 with a total solvation energy gain of -35.4 kcal/mol. In the dimer, the protomers faced each other through the F and G α-helices, thus blocking the substrate access channel. In the presence of the drugs clotrimazole and isavuconazole, the AcCYP51 drug complexes crystallized as monomers. Although clotrimazole-bound AcCYP51 adopted a typical CYP monomer structure, isavuconazole-bound AcCYP51 failed to refold 74 N-terminal residues. The failure of AcCYP51 to fully refold upon inhibitor binding in vivo would cause an irreversible loss of a structurally aberrant enzyme through proteolytic degradation. This assumption explains the superior potency of isavuconazole against A. castellanii The dimerization mode observed in this work is compatible with membrane association and may be relevant to other members of the CYP family of biologic, medical, and pharmacological importance. SIGNIFICANCE STATEMENT: We investigated the mechanism of action of antifungal drugs in the human pathogen Acanthamoeba castellanii. We discovered that the enzyme target [Acanthamoeba castellanii sterol 14α-demethylase (AcCYP51)] formed a dimer via an N-termini swap, whereas drug-bound AcCYP51 was monomeric. In the AcCYP51-isavuconazole complex, the protein target failed to refold 74 N-terminal residues, suggesting a fundamentally different mechanism of AcCYP51 inactivation than only blocking the active site. Proteolytic degradation of a structurally aberrant enzyme would explain the superior potency of isavuconazole against A. castellanii.


Subject(s)
14-alpha Demethylase Inhibitors/pharmacology , Acanthamoeba castellanii/drug effects , Amebiasis/drug therapy , Protozoan Proteins/antagonists & inhibitors , Sterol 14-Demethylase/metabolism , 14-alpha Demethylase Inhibitors/therapeutic use , Acanthamoeba castellanii/metabolism , Amebiasis/parasitology , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Nitriles/pharmacology , Nitriles/therapeutic use , Protein Binding , Protein Domains/physiology , Protein Multimerization/drug effects , Protein Multimerization/physiology , Proteolysis/drug effects , Protozoan Proteins/metabolism , Protozoan Proteins/ultrastructure , Pyridines/pharmacology , Pyridines/therapeutic use , Recombinant Proteins , Sterol 14-Demethylase/ultrastructure , Triazoles/pharmacology , Triazoles/therapeutic use
5.
J Chem Inf Model ; 60(11): 5499-5512, 2020 11 23.
Article in English | MEDLINE | ID: mdl-32634311

ABSTRACT

Falcipain-2 (FP-2) is hemoglobinase considered an attractive drug target of Plasmodium falciparum. Recently, it has been shown that peptidomimetic nitriles containing a 3-pyridyl (3Pyr) moiety at P2 display high affinity and selectivity for FP-2 with respect to human cysteine cathepsins (hCats), outperforming other P2-Pyr isomers and analogs. Further characterization demonstrated that certain P3 variants of these compounds possess micromolar inhibition of parasite growth in vitro and no cytotoxicity against human cell lines. However, the structural determinants underlying the selectivity of the 3Pyr-containing nitriles for FP-2 remain unknown. In this work, we conduct a thorough computational study combining MD simulations and free energy calculations to decipher the bases of the selectivity of the aforementioned nitriles. Our results reveal that water bridges involving the nitrogen and one carboxyl oxygen of I85 and D234 of FP-2, respectively, and the nitrogen of the neutral 3Pyr moiety, which are either less prevalent or nonexistent in the other complexes, explain the experimental activity profiles. The presence of crystallographic waters close to the bridging water positions in the studied proteases strongly supports the occurrence of such interactions. Overall, our findings suggest that selective FP-2 inhibitors can be designed by promoting water bridge formation at the bottom of the S2 subsite and/or by introducing complementary groups that displace the bridging water.


Subject(s)
Antimalarials , Peptide Hydrolases , Cysteine Endopeptidases , Cysteine Proteinase Inhibitors/pharmacology , Humans , Plasmodium falciparum , Water
6.
Edumecentro ; 11(4): 204-221, oct.-dic. 2019. graf
Article in Spanish | LILACS | ID: biblio-1089983

ABSTRACT

RESUMEN Fundamento: el profesional de Trabajo Social en salud requiere una formación que potencie su inteligencia emocional para enfrentar y ofrecer solución a problemas que se les presentarán de manera continua en el ejercicio de su labor. Objetivo: caracterizar el dominio de la inteligencia emocional de los futuros profesionales de Trabajo Social en salud. Métodos: se realizó una investigación de tipo descriptiva longitudinal en la Facultad de Tecnología y Enfermería de la Universidad de Ciencias Médicas de Villa Clara, en el período comprendido enero 2017-diciembre 2018. Se emplearon métodos teóricos: análisis-síntesis, inductivo-deductivo e histórico-lógico; empíricos: la observación científica y la encuesta en forma de cuestionario; y matemático-estadísticos para el análisis de las frecuencias absolutas y relativas. Resultados: los componentes más afectados de la inteligencia emocional resultaron ser la motivación y la autorregulación; y dentro de ellos, los indicadores: confianza en sí mismos, autocontrol, innovación, adaptación, compromiso, iniciativa, optimismo, comunicación y liderazgo; los menos afectados: conciencia emocional, valoración adecuada de sí mismos, confiabilidad, integridad, motivación de logro, influencia, manejo de conflictos, catalización del cambio, establecimiento de vínculos, colaboración y comprensión, y capacidades de equipo. Conclusiones: se caracterizó el dominio de la inteligencia emocional en los futuros profesionales de Trabajo Social en salud, y se constató la necesidad de ofrecer una atención sistemática a los alumnos con carencias en su desarrollo emocional, con el propósito de lograr egresados aptos para la realización de su labor social.


ABSTRACT Background: the Social Work in Health professional requires a training that enhances their emotional intelligence to face and offer solutions to problems that will be addressed in a continuous way in the exercise of their work. Objective: to characterize the command of emotional intelligence of future professionals of Social Work in health. Methods: a descriptive longitudinal research was carried out in the Faculty of Technology and Nursing of Villa Clara University of Medical Sciences, from January 2017 to December 2018. Theoretical methods were used: analysis-synthesis, inductive-deductive and historical-logical; empirical: scientific observation and the survey in the form of a questionnaire; and mathematical-statistics for the analysis of absolute and relative frequencies. Results: the most affected components of emotional intelligence turned out to be motivation and self-regulation; and within them, the indicators: self-confidence, self-control, innovation, adaptation, commitment, initiative, optimism, communication and leadership; the least affected: emotional awareness, adequate self-assessment, reliability, integrity, achievement motivation, influence, conflict management, change catalysis, bonding, collaboration and understanding, and team skills. Conclusions: the command of emotional intelligence was characterized in the future professionals of Social Work in health, and it was confirmed the need to offer a systematic attention to the students with lacks in their emotional development, in order to achieve graduates able to carry out their social work.


Subject(s)
Social Work , Students , Education, Medical , Emotional Intelligence
7.
J Phys Chem B ; 123(34): 7327-7342, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31366200

ABSTRACT

Falcipain-2 (FP-2) is a Plasmodium falciparum cysteine protease that has been extensively targeted to identify novel antimalarials. Remarkably, previous reports have shown that FP-2 can be allosterically modulated and, for a particular noncompetitive chalcone inhibitor, the existing lines of experimental evidence can guide the prediction of its unknown binding mode to the enzyme in a reliable fashion. In this work, we propose a structure of FP-2 in complex with the aforementioned compound that fulfills all of the experimental data, by employing a combination of molecular modeling tools, such as pocket volume measurements, docking, molecular dynamics (MD) simulations, and free energy calculations. Our results show that the studied inhibitor binds a transient pocket occluded in all of the available FP-2 crystal structures and lying in a region previously characterized as a potential allosteric site in related cysteine proteases. In addition, we detected in silico the occurrence of significant community reorganization in FP-2, increased signal transmission between the allosteric pocket and the active site, and change in loop motions and residue pKa values upon the compound binding, thus providing insight into the uncharacterized allosteric mechanism. Overall, this study yields valuable predictions for the design of novel allosteric inhibitors against FP-2 and other cysteine proteases.


Subject(s)
Allosteric Regulation/drug effects , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Plasmodium falciparum/enzymology , Trypanosoma cruzi/enzymology , Binding Sites/drug effects , Cysteine Endopeptidases/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Thermodynamics
8.
PLoS One ; 14(1): e0211227, 2019.
Article in English | MEDLINE | ID: mdl-30682119

ABSTRACT

Trypanosoma cruzi is the causative agent of Chagas disease, a neglected infection affecting millions of people in tropical regions. There are several chemotherapeutic agents for the treatment of this disease, but most of them are highly toxic and generate resistance. Currently, the development of allosteric inhibitors constitutes a promising research field, since it can improve the accessibility to more selective and less toxic medicines. To date, the allosteric drugs prediction is a state-of-the-art topic in rational structure-based computational design. In this work, a simulation strategy was developed for computational discovery of allosteric inhibitors, and it was applied to cruzain, a promising target and the major cysteine protease of T. cruzi. Molecular dynamics simulations, binding free energy calculations and network-based modelling of residue interactions were combined to characterize and compare molecular distinctive features of the apo form and the cruzain-allosteric inhibitor complexes. By using geometry-based criteria on trajectory snapshots, we predicted two main allosteric sites suitable for drug targeting. The results suggest dissimilar mechanisms exerted by the same allosteric site when binding different potential allosteric inhibitors. Finally, we identified the residues involved in suboptimal paths linking the identified site and the orthosteric site. The present study constitutes the first approximation to the design of cruzain allosteric inhibitors and may serve for future pharmacological intervention. Here, no major effects on active site structure were observed due to compound binding (modification of distance and angles between catalytic residues), which indicates that allosteric regulation in cruzain might be mediated via alterations of its dynamical properties similarly to allosteric inhibition of human cathepsin K (HCatK). The current findings are particularly relevant for the design of allosteric modulators of papain-like cysteine proteases.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemistry , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Trypanosoma cruzi/metabolism , Allosteric Regulation/drug effects , Catalytic Domain/drug effects , Cathepsin K/chemistry , Cathepsin K/drug effects , Computer-Aided Design , Cysteine Proteinase Inhibitors/pharmacology , Humans , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Structure-Activity Relationship , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects
9.
Sci Rep ; 7(1): 17887, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263428

ABSTRACT

Classical swine fever (CSF) is one of the most important infectious diseases causing significant economic losses. Its causal agent, CSF virus (CSFV), is a member of the Pestivirus genus included into the Flaviviridae family. Previous molecular epidemiology studies have revealed the CSFV diversity is divided into three main genotypes and different subgenotypes. However, the classification system for CSFV has not yet been harmonized internationally. Similarly, the phylogeny and evolutionary dynamics of CSFV remain unclear. The current study provides novel and significant insights into the origin, diversification and evolutionary process of CSFV. In addition, the best phylogenetic marker for CSFV capable of reproducing the same phylogenetic and evolutionary information as the complete viral genome is characterized. Also, a reliable cut-off to accurately classify CSFV at genotype and subgenotype levels is established. Based on the time for the most recent common ancestor (tMRCA) reconstruction and cophylogenetic analysis, it was determined that CSFV emerged around 225 years ago when the Tunisian Sheep Virus jumped from its natural host to swine. CSFV emergence was followed by a genetic expansion in three main lineages, driven by the action of positive selection pressure and functional divergence, as main natural forces.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever/epidemiology , Genetic Variation/genetics , Genome, Viral/genetics , Animals , Biodiversity , Biological Evolution , Classical Swine Fever/virology , Genotype , Molecular Epidemiology/methods , Phylogeny , Swine
10.
Proteins ; 85(9): 1666-1683, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28543724

ABSTRACT

Falcipain-2 (FP-2) is a major hemoglobinase of Plasmodium falciparum, considered an important drug target for the development of antimalarials. A previous study reported a novel series of 20 reversible peptide-based inhibitors of FP-2. However, the lack of tridimensional structures of the complexes hinders further optimization strategies to enhance the inhibitory activity of the compounds. Here we report the prediction of the binding modes of the aforementioned inhibitors to FP-2. A computational approach combining previous knowledge on the determinants of binding to the enzyme, docking, and postdocking refinement steps, is employed. The latter steps comprise molecular dynamics simulations and free energy calculations. Remarkably, this approach leads to the identification of near-native ligand conformations when applied to a validation set of protein-ligand structures. Overall, we proposed substrate-like binding modes of the studied compounds fulfilling the structural requirements for FP-2 binding and yielding free energy values that correlated well with the experimental data. Proteins 2017; 85:1666-1683. © 2017 Wiley Periodicals, Inc.


Subject(s)
Antimalarials/chemistry , Cysteine Endopeptidases/chemistry , Malaria, Falciparum/drug therapy , Peptides/chemistry , Animals , Antimalarials/therapeutic use , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/metabolism , Humans , Malaria, Falciparum/parasitology , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Plasmodium falciparum/drug effects , Protein Binding , Structure-Activity Relationship
11.
Biomed Res Int ; 2015: 124082, 2015.
Article in English | MEDLINE | ID: mdl-26576414

ABSTRACT

CIGB-552 is a cell-penetrating peptide that exerts in vitro and in vivo antitumor effect on cancer cells. In the present work, the mechanism involved in such anticancer activity was studied using chemical proteomics and expression-based proteomics in culture cancer cell lines. CIGB-552 interacts with at least 55 proteins, as determined by chemical proteomics. A temporal differential proteomics based on iTRAQ quantification method was performed to identify CIGB-552 modulated proteins. The proteomic profile includes 72 differentially expressed proteins in response to CIGB-552 treatment. Proteins related to cell proliferation and apoptosis were identified by both approaches. In line with previous findings, proteomic data revealed that CIGB-552 triggers the inhibition of NF-κB signaling pathway. Furthermore, proteins related to cell invasion were differentially modulated by CIGB-552 treatment suggesting new potentialities of CIGB-552 as anticancer agent. Overall, the current study contributes to a better understanding of the antitumor action mechanism of CIGB-552.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Sequence Data , Neoplasms, Experimental/genetics , Protein Binding , Protein Interaction Mapping/methods , Proteome/chemistry , Proteome/metabolism , Proteomics/methods , Sequence Analysis, Protein/methods , Treatment Outcome
12.
PLoS Negl Trop Dis ; 9(5): e0003759, 2015 May.
Article in English | MEDLINE | ID: mdl-25978322

ABSTRACT

BACKGROUND: Fasciola hepatica is the causative agent of fascioliasis, a disease affecting grazing animals, causing economic losses in global agriculture and currently being an important human zoonosis. Overuse of chemotherapeutics against fascioliasis has increased the populations of drug resistant parasites. F. hepatica cathepsin L3 is a protease that plays important roles during the life cycle of fluke. Due to its particular collagenolytic activity it is considered an attractive target against the infective phase of F. hepatica. METHODOLOGY/PRINCIPAL FINDINGS: Starting with a three dimensional model of FhCL3 we performed a structure-based design of novel inhibitors through a computational study that combined virtual screening, molecular dynamics simulations, and binding free energy (ΔGbind) calculations. Virtual screening was carried out by docking inhibitors obtained from the MYBRIDGE-HitFinder database inside FhCL3 and human cathepsin L substrate-binding sites. On the basis of dock-scores, five compounds were predicted as selective inhibitors of FhCL3. Molecular dynamic simulations were performed and, subsequently, an end-point method was employed to predict ΔGbind values. Two compounds with the best ΔGbind values (-10.68 kcal/mol and -7.16 kcal/mol), comparable to that of the positive control (-10.55 kcal/mol), were identified. A similar approach was followed to structurally and energetically characterize the interface of FhCL3 in complex with a peptidic substrate. Finally, through pair-wise and per-residue free energy decomposition we identified residues that are critical for the substrate/ligand binding and for the enzyme specificity. CONCLUSIONS/SIGNIFICANCE: The present study is the first computer-aided drug design approach against F. hepatica cathepsins. Here we predict the principal determinants of binding of FhCL3 in complex with a natural substrate by detailed energetic characterization of protease interaction surface. We also propose novel compounds as FhCL3 inhibitors. Overall, these results will foster the future rational design of new inhibitors against FhCL3, as well as other F. hepatica cathepsins.


Subject(s)
Cathepsin L/antagonists & inhibitors , Computer-Aided Design , Drug Discovery/methods , Fasciola hepatica/drug effects , Animal Husbandry , Animals , Binding Sites/genetics , Cathepsin L/metabolism , Fasciola hepatica/enzymology , Fascioliasis/diagnosis , Fascioliasis/drug therapy , Fascioliasis/parasitology , Humans , Life Cycle Stages , Molecular Dynamics Simulation , Parasitic Sensitivity Tests , Zoonoses/drug therapy , Zoonoses/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...