Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Molecules ; 23(4)2018 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-29584709

RESUMO

There is an urgent need for the discovery of new antileishmanial drugs with a new mechanism of action. Type 2 NADH dehydrogenase from Leishmania infantum (LiNDH2) is an enzyme of the parasite's respiratory system, which catalyzes the electron transfer from NADH to ubiquinone without coupled proton pumping. In previous studies of the related NADH: ubiquinone oxidoreductase crystal structure from Saccharomyces cerevisiae, two ubiquinone-binding sites (UQI and UQII) were identified and shown to play an important role in the NDH-2-catalyzed oxidoreduction reaction. Based on the available structural data, we developed a three-dimensional structural model of LiNDH2 using homology detection methods and performed an in silico virtual screening campaign to search for potential inhibitors targeting the LiNDH2 ubiquinone-binding site 1-UQI. Selected compounds displaying favorable properties in the computational screening experiments were assayed for inhibitory activity in the structurally similar recombinant NDH-2 from S. aureus and leishmanicidal activity was determined in the wild-type axenic amastigotes and promastigotes of L. infantum. The identified compound, a substituted 6-methoxy-quinalidine, showed promising nanomolar leishmanicidal activity on wild-type axenic promastigotes and amastigotes of L. infantum and the potential for further development.


Assuntos
Antiprotozoários/química , Leishmania infantum/enzimologia , NADH Desidrogenase/metabolismo , Quinaldinas/química , Antiprotozoários/farmacologia , Domínio Catalítico/efeitos dos fármacos , Simulação por Computador , Avaliação Pré-Clínica de Medicamentos , Leishmania infantum/efeitos dos fármacos , Modelos Moleculares , NADH Desidrogenase/química , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Quinaldinas/farmacologia , Homologia Estrutural de Proteína , Relação Estrutura-Atividade
2.
J Med Chem ; 58(15): 6179-94, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26126187

RESUMO

Bacterial DNA gyrase is a well-known and validated target in the design of antibacterial drugs. However, inhibitors of its ATP binding subunit, DNA gyrase B (GyrB), have so far not reached clinical use. In the present study, three different series of N-phenyl-4,5-dibromopyrrolamides and N-phenylindolamides were designed and prepared as potential DNA gyrase B inhibitors. The IC50 values of compounds on DNA gyrase from Escherichia coli were in the low micromolar range, with the best compound, (4-(4,5-dibromo-1H-pyrrole-2-carboxamido)benzoyl)glycine (18a), displaying an IC50 of 450 nM. For this compound, a high-resolution crystal structure in complex with E. coli DNA gyrase B was obtained, revealing details of its binding mode within the active site. The binding affinities of three compounds with GyrB were additionally evaluated by surface plasmon resonance, and the results were in good agreement with the determined enzymatic activities. For the most promising compounds, the inhibitory activities against DNA gyrase from Staphylococcus aureus and topoisomerases IV from E. coli and S. aureus were determined. Antibacterial activities of the most potent compounds of each series were evaluated against two Gram-positive and two Gram-negative bacterial strains. The results obtained in this study provide valuable information on the binding mode and structure-activity relationship of N-phenyl-4,5-dibromopyrrolamides and N-phenylindolamides as promising classes of ATP competitive GyrB inhibitors.


Assuntos
Trifosfato de Adenosina/química , Indóis/química , Indóis/farmacologia , Pirróis/química , Pirróis/farmacologia , Amidas/química , Cristalografia por Raios X , Desenho de Fármacos , Indóis/síntese química , Modelos Moleculares , Pirróis/síntese química , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/química , Inibidores da Topoisomerase II/farmacologia
3.
Bioorg Med Chem ; 23(15): 4218-4229, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26183545

RESUMO

Human DNA topoisomerase IIα (htIIα) is a validated target for the development of novel anticancer agents. Starting from our discovered 4-amino-1,3,5-triazine inhibitors of htIIα, we investigated a library of 2,4,6-trisubstituted-1,3,5-triazines for novel inhibitors that bind to the htIIα ATP binding site using a combination of structure-based and ligand-based pharmacophore models and molecular docking. 4,6-substituted-1,3,5-triazin-2(1H)-ones 8, 9 and 14 were identified as novel inhibitors with activity comparable to the established drug etoposide (1). Compound 8 inhibits the htIIα decatenation in a superior fashion to etoposide. Cleavage assays demonstrated that selected compounds 8 and 14 do not act as poisons and antagonize the poison effect of etoposide. Microscale thermophoresis (MST) confirmed binding of compound 8 to the htIIα ATPase domain and compound 14 effectively inhibits the htIIα mediated ATP hydrolysis. The molecular dynamics simulation study provides further insight into the molecular recognition. The 4,6-disubstituted-1,3,5-triazin-2(1H)-ones represent the first validated monocyclic class of catalytic inhibitors that bind to the to the htIIα ATPase domain.


Assuntos
Trifosfato de Adenosina/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores da Topoisomerase II/química , Inibidores da Topoisomerase II/farmacologia , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Sítios de Ligação , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Etoposídeo/farmacologia , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Estrutura Terciária de Proteína , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Inibidores da Topoisomerase II/metabolismo , Triazinas
4.
J Med Chem ; 58(14): 5501-21, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26098163

RESUMO

Bacterial DNA gyrase and topoisomerase IV are essential enzymes that control the topological state of DNA during replication and validated antibacterial drug targets. Starting from a library of marine alkaloid oroidin analogues, we identified low micromolar inhibitors of Escherichia coli DNA gyrase based on the 5,6,7,8-tetrahydroquinazoline and 4,5,6,7-tetrahydrobenzo[1,2-d]thiazole scaffolds. Structure-based optimization of the initial hits resulted in low nanomolar E. coli DNA gyrase inhibitors, some of which exhibited micromolar inhibition of E. coli topoisomerase IV and of Staphylococcus aureus homologues. Some of the compounds possessed modest antibacterial activity against Gram positive bacterial strains, while their evaluation against wild-type, impA and ΔtolC E. coli strains suggests that they are efflux pump substrates and/or do not possess the physicochemical properties necessary for cell wall penetration. Our study provides a rationale for optimization of this class of compounds toward balanced dual DNA gyrase and topoisomerase IV inhibitors with antibacterial activity.


Assuntos
Trifosfato de Adenosina/metabolismo , DNA Girase/metabolismo , Desenho de Fármacos , Tiazóis/química , Tiazóis/farmacologia , Inibidores da Topoisomerase II/química , Inibidores da Topoisomerase II/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Sítios de Ligação , DNA Girase/química , DNA Topoisomerase IV/antagonistas & inibidores , Escherichia coli/efeitos dos fármacos , Escherichia coli/enzimologia , Concentração Inibidora 50 , Modelos Moleculares , Conformação Proteica , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/enzimologia , Relação Estrutura-Atividade
5.
J Comput Aided Mol Des ; 29(6): 541-60, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25851408

RESUMO

Bacterial resistance to the available antibiotic agents underlines an urgent need for the discovery of novel antibacterial agents. Members of the bacterial Mur ligase family MurC-MurF involved in the intracellular stages of the bacterial peptidoglycan biosynthesis have recently emerged as a collection of attractive targets for novel antibacterial drug design. In this study, we have first extended the knowledge of the class of furan-based benzene-1,3-dicarboxylic acid derivatives by first showing a multiple MurC-MurF ligase inhibition for representatives of the extended series of this class. Steady-state kinetics studies on the MurD enzyme were performed for compound 1, suggesting a competitive inhibition with respect to ATP. To the best of our knowledge, compound 1 represents the first ATP-competitive MurD inhibitor reported to date with concurrent multiple inhibition of all four Mur ligases (MurC-MurF). Subsequent molecular dynamic (MD) simulations coupled with interaction energy calculations were performed for two alternative in silico models of compound 1 in the UMA/D-Glu- and ATP-binding sites of MurD, identifying binding in the ATP-binding site as energetically more favorable in comparison to the UMA/D-Glu-binding site, which was in agreement with steady-state kinetic data. In the final stage, based on the obtained MD data novel furan-based benzene monocarboxylic acid derivatives 8-11, exhibiting multiple Mur ligase (MurC-MurF) inhibition with predominantly superior ligase inhibition over the original series, were discovered and for compound 10 it was shown to possess promising antibacterial activity against S. aureus. These compounds represent novel leads that could by further optimization pave the way to novel antibacterial agents.


Assuntos
Antibacterianos/química , Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Furanos/química , Ligases/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Sítios de Ligação , Ácidos Carboxílicos/química , Avaliação Pré-Clínica de Medicamentos/métodos , Ligases/química , Ligases/metabolismo , Simulação de Dinâmica Molecular , Relação Estrutura-Atividade
6.
ChemMedChem ; 10(2): 345-59, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25522133

RESUMO

Human DNA topoisomerase IIα (htIIα) is a validated target for the development of anticancer agents. Based on structural data regarding the binding mode of AMP-PNP (5'-adenylyl-ß,γ-imidodiphosphate) to htIIα, we designed a two-stage virtual screening campaign that combines structure-based pharmacophores and molecular docking. In the first stage, we identified several monosubstituted 9H-purine compounds and a novel class of 1H-pyrazolo[3,4]pyrimidines as inhibitors of htIIα. In the second stage, disubstituted analogues with improved cellular activities were discovered. Compounds from both classes were shown to inhibit htIIα-mediated DNA decatenation, and surface plasmon resonance (SPR) experiments confirmed binding of these two compounds on the htIIα ATPase domain. Proposed complexes and interaction patterns between both compounds and htIIα were further analyzed in molecular dynamics simulations. Two compounds identified in the second stage showed promising anticancer activities in hepatocellular carcinoma (HepG2) and breast cancer (MCF-7) cell lines. The discovered compounds are suitable starting points for further hit-to-lead development in anticancer drug discovery.


Assuntos
Antineoplásicos/química , Proteínas de Ligação a DNA/antagonistas & inibidores , Purinas/química , Pirazóis/química , Pirimidinas/química , Inibidores da Topoisomerase II/química , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacologia , Sítios de Ligação , Sobrevivência Celular/efeitos dos fármacos , DNA Girase/química , DNA Girase/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Células MCF-7 , Simulação de Acoplamento Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Purinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície , Inibidores da Topoisomerase II/farmacologia
7.
Bioorg Med Chem Lett ; 24(24): 5762-5768, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25453816

RESUMO

Human DNA topoisomerase IIα (htIIα) is a validated target for the development of anticancer agents. Starting from the available information about the binding of the purine-based htIIα inhibitors in the ATP binding site we designed a virtual screening campaign combining structure-based and ligand-based pharmacophores with a molecular docking calculation searching for compounds that would contain a monocycle mimetic of the purine moiety. We discovered novel 4-amino-6-(phenylamino)-1,3,5-triazines 6, 7 and 11 as monocyclic htIIα inhibitors targeting the ATP binding site. Compound 6 from the 1,3,5-triazine series also displayed cytotoxicity properties in hepatocellular carcinoma (HepG2) cell lines and selectivity against human umbilical vein endothelial (HUVEC) cell lines.


Assuntos
Antineoplásicos/química , Proteínas de Ligação a DNA/antagonistas & inibidores , Purinas/química , Tiocarbamatos/química , Inibidores da Topoisomerase II/química , Triazinas/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Antígenos de Neoplasias/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Sítios de Ligação , Sobrevivência Celular/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Desenho de Fármacos , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Tiocarbamatos/síntese química , Tiocarbamatos/toxicidade , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/farmacologia , Triazinas/síntese química , Triazinas/farmacologia , Triazinas/toxicidade
8.
Bioorg Med Chem ; 22(15): 4124-34, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24953950

RESUMO

Enzymes catalyzing the biosynthesis of bacterial peptidoglycan represent traditionally a collection of highly selective targets for novel antibacterial drug design. Four members of the bacterial Mur ligase family-MurC, MurD, MurE and MurF-are involved in the intracellular steps of peptidoglycan biosynthesis, catalyzing the synthesis of the peptide moiety of the Park's nucleotide. In our previous virtual screening campaign, a chemical class of benzene-1,3-dicarboxylic acid 2,5-dimethylpyrrole derivatives exhibiting dual MurD/MurE inhibition properties was discovered. In the present study we further investigated this class of compounds by performing inhibition assays on all four Mur ligases (MurC-MurF). Furthermore, molecular dynamics (MD) simulation studies of one of the initially discovered compound 1 were performed to explore its geometry as well as its energetic behavior based on the Linear Interaction Energy (LIE) method. Further in silico virtual screening (VS) experiments based on the parent active compound 1 were conducted to optimize the discovered series. Selected hits were assayed against all Escherichia coli MurC-MurF enzymes in biochemical inhibition assays and molecules 10-14 containing benzene-1,3-dicarboxylic acid 2,5-dimethylpyrrole coupled with five member-ring rhodanine moiety were found to be multiple inhibitors of the whole MurC-MurF cascade of bacterial enzymes in the micromolar range. Steady-state kinetics studies suggested this class to act as competitive inhibitors of the MurD enzyme towards d-Glu. These compounds represent novel valuable starting point in the development of novel antibacterial agents.


Assuntos
Inibidores Enzimáticos/química , Proteínas de Escherichia coli/antagonistas & inibidores , Peptídeo Sintases/antagonistas & inibidores , Pirróis/química , Sítios de Ligação , Inibidores Enzimáticos/metabolismo , Escherichia coli/enzimologia , Proteínas de Escherichia coli/metabolismo , Cinética , Simulação de Dinâmica Molecular , Peptídeo Sintases/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Pirróis/metabolismo , Relação Estrutura-Atividade
9.
J Chem Inf Model ; 54(5): 1451-66, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24724969

RESUMO

Increasing bacterial resistance to available antibiotics stimulated the discovery of novel efficacious antibacterial agents. The biosynthesis of the bacterial peptidoglycan, where the MurD enzyme is involved in the intracellular phase of the UDP-MurNAc-pentapeptide formation, represents a collection of highly selective targets for novel antibacterial drug design. In our previous computational studies, the C-terminal domain motion of the MurD ligase was investigated using Targeted Molecular Dynamic (TMD) simulation and the Off-Path Simulation (OPS) technique. In this study, we present a drug design strategy using multiple protein structures for the identification of novel MurD ligase inhibitors. Our main focus was the ATP-binding site of the MurD enzyme. In the first stage, three MurD protein conformations were selected based on the obtained OPS/TMD data as the initial criterion. Subsequently, a two-stage virtual screening approach was utilized combining derived structure-based pharmacophores with molecular docking calculations. Selected compounds were then assayed in the established enzyme binding assays, and compound 3 from the aminothiazole class was discovered to act as a dual MurC/MurD inhibitor in the micomolar range. A steady-state kinetic study was performed on the MurD enzyme to provide further information about the mechanistic aspects of its inhibition. In the final stage, all used conformations of the MurD enzyme with compound 3 were simulated in classical molecular dynamics (MD) simulations providing atomistic insights of the experimental results. Overall, the study depicts several challenges that need to be addressed when trying to hit a flexible moving target such as the presently studied bacterial MurD enzyme and show the possibilities of how computational tools can be proficiently used at all stages of the drug discovery process.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Escherichia coli/enzimologia , Peptídeo Sintases/antagonistas & inibidores , Peptídeo Sintases/química , Trifosfato de Adenosina/metabolismo , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Peptídeo Sintases/metabolismo , Conformação Proteica , Tiazóis/química , Tiazóis/metabolismo , Tiazóis/farmacologia , Interface Usuário-Computador
10.
J Comput Aided Mol Des ; 27(8): 723-38, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23990043

RESUMO

The biosynthetic pathway of the bacterial peptidoglycan, where MurD is an enzyme involved at the intracellular stage of its construction, represents a collection of highly selective macromolecular targets for novel antibacterial drug design. In this study as part of our investigation of the MurD bacterial target two recently discovered classes of the MurD ligase inhibitors were investigated resulting from the lead optimization phases of the N-sulfonamide D-Glu MurD inhibitors. Molecular dynamics simulations, based on novel structural data, in conjunction with the linear interaction energy (LIE) method suggested the transferability of our previously obtained LIE coefficients to further D-Glu based classes of MurD inhibitors. Analysis of the observed dynamical behavior of these compounds in the MurD active site was supported by static drug design techniques. These results complement the current knowledge of the MurD inhibitory mechanism and provide valuable support for the D-Glu paradigm of the inhibitor design.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Desenho de Fármacos , Escherichia coli/enzimologia , Simulação de Dinâmica Molecular , Peptídeo Sintases/antagonistas & inibidores , Escherichia coli/efeitos dos fármacos , Infecções por Escherichia coli/tratamento farmacológico , Ácido Glutâmico/química , Ácido Glutâmico/farmacologia , Humanos , Peptídeo Sintases/metabolismo
11.
Eur J Med Chem ; 67: 208-20, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23867605

RESUMO

The D-aspartate ligase of Enterococcus faecium (Aslfm) is an attractive target for the development of narrow-spectrum antibacterial agents that are active against multidrug-resistant E. faecium. Although there is currently little available information regarding the structural characteristics of Aslfm, we exploited the knowledge that this enzyme belongs to the ATP-grasp superfamily to target its ATP binding site. In the first design stage, we synthesized and screened a small library of known ATP-competitive inhibitors of ATP-grasp enzymes. A series of amino-oxazoles derived from bacterial biotin carboxylase inhibitors showed low micromolar activity. The most potent inhibitor compound 12, inhibits Aslfm with a Ki value of 2.9 µM. In the second design stage, a validated ligand-based pharmacophore modeling approach was used, taking the newly available inhibition data of an initial series of compounds into account. Experimental evaluation of the virtual screening hits identified two novel structural types of Aslfm inhibitors with 7-amino-9H-purine (18) and 7-amino-1H-pyrazolo[3,4-d]pyrimidine (30 and 34) scaffolds, and also with Ki values in the low micromolar range. Investigation the inhibitors modes of action confirmed that these compounds are competitive with respect to the ATP molecule. The binding of inhibitors to the target enzyme was also studied using isothermal titration calorimetry (ITC). Compounds 6, 12, 18, 30 and 34 represent the first inhibitors of Aslfm reported to date, and are an important step forward in combating infections due to E. faecium.


Assuntos
Ácido D-Aspártico/metabolismo , Descoberta de Drogas , Enterococcus faecium/enzimologia , Inibidores Enzimáticos/farmacologia , Ligases/antagonistas & inibidores , Relação Dose-Resposta a Droga , Enterococcus faecium/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Ligases/metabolismo , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
12.
Biochim Biophys Acta ; 1828(11): 2609-19, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23774522

RESUMO

Membrane proteins represent about a third of the gene products in most organisms, as revealed by the genome sequencing projects. They account for up to two thirds of known drugable targets, which emphasizes their critical pharmaceutical importance. Here we present a study on bilitranslocase (BTL) (TCDB 2.A.65), a membrane protein primarily involved in the transport of bilirubin from blood to liver cells. Bilitranslocase has also been identified as a potential membrane transporter for cellular uptake of several drugs and due to its implication in drug uptake, it is extremely important to advance the knowledge about its 3D structure. However, at present, only a limited knowledge is available beyond the primary structure of BTL. It has been recently confirmed experimentally that one of the four computationally predicted transmembrane segments of bilitranslocase, TM3, has a helical structure with hydrophilic amino acid residues oriented towards one side, which is typical for transmembrane domains of membrane proteins. In this study we confirmed by the use of multidimensional NMR spectroscopy that the second transmembrane segment, TM2, also appears in a form of α-helix. The stability of this polypeptide chain was verified by molecular dynamics (MD) simulation in dipalmitoyl phosphatidyl choline (DPPC) and in sodium dodecyl sulfate (SDS) micelles. The two α-helices, TM2 corroborated in this study, and TM3 confirmed in our previous investigation, provide reasonable building blocks of a potential transmembrane channel for transport of bilirubin and small hydrophilic molecules, including pharmaceutically active compounds.


Assuntos
Proteínas de Membrana/química , Ressonância Magnética Nuclear Biomolecular/métodos , Sequência de Aminoácidos , Transporte Biológico Ativo , Ceruloplasmina , Dicroísmo Circular , Micelas , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Conformação Proteica , Dodecilsulfato de Sódio
13.
Curr Pharm Des ; 19(13): 2474-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23363399

RESUMO

DNA topoisomerases are an important family of enzymes that catalyze the induction of topological changes in the DNA molecule. Their ability to modulate the topology of the DNA makes DNA topoisomerases a key player in several vital cell processes such as replication, transcription, chromosome separation and segregation. Consequently, they already represent an important collection of macromolecular targets for some of the established anticancer drugs on the market as well as serve as templates in the development of novel anticancer drugs especially supported by recent structural advances in the field. The aim of this review is to provide an overview of the recent developments in the field of DNA poisons - a major class of human topoisomerase IIα inhibitors - of which several are already in clinical use. Due to frequently experienced occurrence of serious side effects of these molecules during therapy, especially cardiotoxicity issues, further drug design efforts were initiated already yielding novel promising compounds that have overcome this issue and already entered into clinical studies. Some of the presented and discussed chemical classes include intercalators, non-intercalators and redox-dependent poisons of human topoisomerase IIα. In particular, this review focuses on the currently available structure-based standpoint of molecular design and on the medicinal chemist's perspective of this field of anticancer drug design.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Inibidores da Topoisomerase II/farmacologia , Antígenos de Neoplasias/química , Antineoplásicos/química , Antineoplásicos/uso terapêutico , DNA Topoisomerases Tipo II/química , Proteínas de Ligação a DNA/química , Humanos , Conformação Molecular , Neoplasias/tratamento farmacológico , Inibidores da Topoisomerase II/química , Inibidores da Topoisomerase II/uso terapêutico
14.
J Comput Chem ; 34(9): 790-801, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23280926

RESUMO

A classical protein sequence alignment and homology modeling strategy were used for building three Mycobacterium tuberculosis-DNA gyrase protein models using the available topoII-DNA-6FQ crystal structure complexes originating from different organisms. The recently determined M. tuberculosis-DNA gyrase apoprotein structures and topoII-DNA-6FQ complexes were used for defining the 6-fluoroquinolones (6-FQs) binding pockets. The quality of the generated models was initially validated by docking of the cocrystallized ligands into their binding site, and subsequently by quantitative evaluation of their discriminatory performances (identification of active/inactive 6-FQs) for a set of 145 6-FQs with known biological activity values. The M. tuberculosis-DNA gyrase model with the highest estimated discriminatory power was selected and used afterwards in an additional molecular docking experiment on a mixed combinatorial set of 427 drug-like 6-FQ analogs for which the biological activity values were predicted using a prebuilt counter-propagation artificial neural network model. A novel three-level Boolean-based [T/F (true/false)] clustering algorithm was used to assess the generated binding poses: Level 1 (geometry properties assessment), Level 2 (score-based clustering and selection of the (T)-signed highly scored Level 1 poses), and Level 3 (activity-based clustering and selection of the most "active" (T)-signed Level 2 hits). The frequency analysis of occurrence of the fragments attached at R(1) and R(7) position of the (T)-signed 6-FQs selected in Level 3 revealed several novel attractive fragments and confirmed some previous findings. We believe that this methodology could be successfully used in establishing novel possible structure-activity relationship recommendations in the 6-FQs optimization, which could be of great importance in the current antimycobacterial hit-to-lead processes.


Assuntos
Antituberculosos/química , Proteínas de Bactérias/química , DNA Girase/química , Inibidores Enzimáticos/química , Fluoroquinolonas/química , Simulação de Acoplamento Molecular , Mycobacterium tuberculosis/química , Algoritmos , Proteínas de Bactérias/antagonistas & inibidores , Sítios de Ligação , Análise por Conglomerados , Desenho de Fármacos , Ligantes , Conformação Molecular , Mycobacterium tuberculosis/enzimologia , Redes Neurais de Computação , Ligação Proteica , Relação Estrutura-Atividade , Inibidores da Topoisomerase II
15.
Bioorg Med Chem ; 20(17): 5220-8, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22841617

RESUMO

A set of 16 previously synthesized aryl-aminopyridine and aryl-aminoquinoline derivatives have been evaluated for cytotoxic activity against three cancer cell lines (human cervical cancer-HeLa; human chronic myeloid leukemia-K562; human melanoma-Fem-x) and two types of normal peripheral blood mononuclear cells, with and without phytohemaglutinin (PBMC-PHA; PBMC+PHA). Twelve of the studied compounds showed moderate cytotoxicity, with selectivity against K562 but not the remaining two cancer cell lines. Four compounds were not active in cytotoxicity assays, presumably due to high predicted lipophilicity and low solubility. To rationalize the observed cytotoxic effects, structure-based virtual screening was carried out against a pool of potential targets constructed using the inverse docking program Tarfisdock and bibliographical references. The putative targets were identified on the basis of the best correlation between docking scores and in vitro cytotoxicity. It is proposed that the mechanism of action of the studied aminopyridines involves the disruption of signaling pathways and cancer cell cycle through the inhibition of cyclin-dependent kinases and several tyrosine kinases, namely Bcr-Abl kinase and KIT receptor kinase. The obtained results can guide further structural modifications of the studied compounds aimed at developing selective agents targeting proteins involved in cancer cell survival and proliferation.


Assuntos
Aminopiridinas/farmacologia , Antineoplásicos/farmacologia , Aminopiridinas/síntese química , Aminopiridinas/química , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células HeLa , Humanos , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade , Células Tumorais Cultivadas
16.
PLoS One ; 7(6): e38967, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22745694

RESUMO

Using a combination of genomic and post-genomic approaches is rapidly altering the number of identified human influx carriers. A transmembrane protein bilitranslocase (TCDB 2.A.65) has long attracted attention because of its function as an organic anion carrier. It has also been identified as a potential membrane transporter for cellular uptake of several drugs and due to its implication in drug uptake, it is extremely important to advance the knowledge about its structure. However, at present, only the primary structure of bilitranslocase is known. In our work, transmembrane subunits of bilitranslocase were predicted by a previously developed chemometrics model and the stability of these polypeptide chains were studied by molecular dynamics (MD) simulation. Furthermore, sodium dodecyl sulfate (SDS) micelles were used as a model of cell membrane and herein we present a high-resolution 3D structure of an 18 amino acid residues long peptide corresponding to the third transmembrane part of bilitranslocase obtained by use of multidimensional NMR spectroscopy. It has been experimentally confirmed that one of the transmembrane segments of bilitranslocase has alpha helical structure with hydrophilic amino acid residues oriented towards one side, thus capable of forming a channel in the membrane.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana Transportadoras/química , Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Ceruloplasmina , Espectroscopia de Ressonância Magnética , Micelas , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Dodecilsulfato de Sódio/química
17.
J Med Chem ; 55(14): 6413-26, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22731783

RESUMO

Bacterial DNA gyrase is a well-established and validated target for the development of novel antibacterials. Starting from the available structural information about the binding of the natural product inhibitor, clorobiocin, we identified a novel series of 4'-methyl-N(2)-phenyl-[4,5'-bithiazole]-2,2'-diamine inhibitors of gyrase B with a low micromolar inhibitory activity by implementing a two-step structure-based design procedure. This novel class of DNA gyrase inhibitors was extensively investigated by various techniques (differential scanning fluorimetry, surface plasmon resonance, and microscale thermophoresis). The binding mode of the potent inhibitor 18 was revealed by X-ray crystallography, confirming our initial in silico binding model. Furthermore, the high resolution of the complex structure allowed for the placement of the Gly97-Ser108 flexible loop, thus revealing its role in binding of this class of compounds. The crystal structure of the complex protein G24 and inhibitor 18 provides valuable information for further optimization of this novel class of DNA gyrase B inhibitors.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Tiazóis/química , Tiazóis/farmacologia , Inibidores da Topoisomerase II , Trifosfato de Adenosina/metabolismo , Antibacterianos/química , Antibacterianos/farmacologia , DNA Girase/química , DNA Girase/metabolismo , Avaliação Pré-Clínica de Medicamentos , Modelos Moleculares , Novobiocina/análogos & derivados , Novobiocina/metabolismo , Conformação Proteica , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
18.
Bioorg Med Chem ; 20(8): 2572-80, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22444877

RESUMO

Bacterial DNA gyrase is an established and validated target for the development of novel antibacterials. In our previous work, we identified a novel series of bacterial gyrase inhibitors from the class of 4-(2,4-dihydroxyphenyl) thiazoles. Our ongoing effort was designated to search for synthetically more available compounds with possibility of hit to lead development. By using the virtual screening approach, new potential inhibitors were carefully selected from the focused chemical library and tested for biological activity. Herein we report on a novel class of 5-(2-hydroxybenzylidene) rhodanines as gyrase B inhibitors with activity in low micromolar range and moderate antibacterial activity. The binding of the two most active compounds to the enzyme target was further characterised using surface plasmon resonance (SPR) and differential scanning fluorimetry methods (DSF).


Assuntos
Antibacterianos/farmacologia , Compostos de Benzilideno/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Rodanina/análogos & derivados , Inibidores da Topoisomerase II , Antibacterianos/síntese química , Antibacterianos/química , Compostos de Benzilideno/síntese química , Compostos de Benzilideno/química , DNA Girase/metabolismo , Relação Dose-Resposta a Droga , Enterococcus faecalis/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Haemophilus influenzae/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , Rodanina/síntese química , Rodanina/química , Rodanina/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Relação Estrutura-Atividade
19.
J Mol Model ; 18(5): 1735-53, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21833830

RESUMO

The virtual combinatorial chemistry approach as a methodology for generating chemical libraries of structurally-similar analogs in a virtual environment was employed for building a general mixed virtual combinatorial library with a total of 53.871 6-FQ structural analogs, introducing the real synthetic pathways of three well known 6-FQ inhibitors. The druggability properties of the generated combinatorial 6-FQs were assessed using an in-house developed drug-likeness filter integrating the Lipinski/Veber rule-sets. The compounds recognized as drug-like were used as an external set for prediction of the biological activity values using a neural-networks (NN) model based on an experimentally-determined set of active 6-FQs. Furthermore, a subset of compounds was extracted from the pool of drug-like 6-FQs, with predicted biological activity, and subsequently used in virtual screening (VS) campaign combining pharmacophore modeling and molecular docking studies. This complex scheme, a powerful combination of chemometric and molecular modeling approaches provided novel QSAR guidelines that could aid in the further lead development of 6-FQs agents.


Assuntos
Antituberculosos/química , Técnicas de Química Combinatória/métodos , Fluoroquinolonas/química , Simulação de Dinâmica Molecular , Bibliotecas de Moléculas Pequenas , Software , Desenho de Fármacos , Bibliotecas Digitais , Estrutura Molecular , Redes Neurais de Computação , Relação Quantitativa Estrutura-Atividade
20.
Mol Divers ; 15(2): 417-26, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20229318

RESUMO

Quantitative structure-activity relationship study on three diverse sets of structurally similar fluoroquinolones was performed using a comprehensive set of molecular descriptors. Multiple linear regression technique was applied as a preprocessing tool to find the set of relevant descriptors (10) which are subsequently used in the artificial neural networks approach (non-linear procedure). The biological activity in the series (minimal inhibitory concentration (µg/mL) was treated as negative decade logarithm, pMIC). Using the non-linear technique counter propagation artificial neural networks, we obtained good predictive models. All models were validated using cross validation leave-one-out procedure. The results (the best models: Assay1, R = 0.8108; Assay2, R = 0.8454, and Assay3, R = 0.9212) obtained on external, previously excluded test datasets show the ability of these models in providing structure-activity relationship of fluoroquinolones. Thus, we demonstrated the advantage of non-linear approach in prediction of biological activity in these series. Furthermore, these validated models could be proficiently used for the design of novel structurally similar fluoroquinolone analogues with potentially higher activity.


Assuntos
Antituberculosos/química , Antituberculosos/metabolismo , Fluoroquinolonas/química , Fluoroquinolonas/metabolismo , Relação Quantitativa Estrutura-Atividade , Algoritmos , Desenho de Fármacos , Humanos , Modelos Estatísticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...