Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Mol Model ; 20(6): 2255, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24842325

RESUMO

PqsD mediates the conversion of anthraniloyl-coenzyme A (ACoA) to 2-heptyl-4-hydroxyquinoline (HHQ), a precursor of the Pseudomonas quinolone signal (PQS) molecule. Due to the role of the quinolone signaling pathway of Pseudomonas aeruginosa in the expression of several virulence factors and biofilm formation, PqsD is a potential target for controlling this nosocomial pathogen, which exhibits a low susceptibility to standard antibiotics. PqsD belongs to the ß-ketoacyl-ACP synthase family and is similar in structure to homologous FabH enzymes in E. coli and Mycobacterium tuberculosis. Here, we used molecular dynamics simulations to obtain the structural position of the substrate ACoA in the binding pocket of PqsD, and semiempirical molecular orbital calculations to study the reaction mechanism for the catalytic cleavage of ACoA. Our findings suggest a nucleophilic attack of the deprotonated sulfur of Cys112 at the carbonyl carbon of ACoA and a switch in the protonation pattern of His257 whereby Nδ is protonated and the proton of Nε is shifted to the sulfur of CoA during the reaction. This is in agreement with the experimentally determined decreased catalytic activity of the Cys112Ser mutant, whereas the Cys112Ala, His257Phe, and Asn287Ala mutants are all inactive. ESI mass-spectrometric measurements of the Asn287Ala mutant show that anthraniloyl remains covalently bound to Cys112, thus further supporting the inference from our computed mechanism that Asn287 does not take part in the cleavage of ACoA. Since this mutant is inactive, we suggest instead that Asn287 must play an essential role in the subsequent formation of HHQ in vitro.


Assuntos
3-Oxoacil-(Proteína de Transporte de Acila) Sintase/metabolismo , Proteínas de Bactérias/metabolismo , Coenzima A/metabolismo , Hidroxiquinolinas/metabolismo , Pseudomonas aeruginosa/enzimologia , ortoaminobenzoatos/metabolismo , 3-Oxoacil-(Proteína de Transporte de Acila) Sintase/antagonistas & inibidores , 3-Oxoacil-(Proteína de Transporte de Acila) Sintase/química , 3-Oxoacil-(Proteína de Transporte de Acila) Sintase/genética , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Catálise , Coenzima A/química , Desenho Assistido por Computador , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Hidroxiquinolinas/química , Simulação de Dinâmica Molecular , Estrutura Molecular , Terapia de Alvo Molecular , Mutação , Ligação Proteica , Conformação Proteica , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Percepção de Quorum , Espectrometria de Massas por Ionização por Electrospray , Relação Estrutura-Atividade , Especificidade por Substrato , ortoaminobenzoatos/química
2.
BMC Biophys ; 6(1): 10, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23916145

RESUMO

BACKGROUND: PQS (PseudomonasQuinolone Signal) and its precursor HHQ are signal molecules of the P. aeruginosa quorum sensing system. They explicate their role in mammalian pathogenicity by binding to the receptor PqsR that induces virulence factor production and biofilm formation. The enzyme PqsD catalyses the biosynthesis of HHQ. RESULTS: Enzyme kinetic analysis and surface plasmon resonance (SPR) biosensor experiments were used to determine mechanism and substrate order of the biosynthesis. Comparative analysis led to the identification of domains involved in functionality of PqsD. A kinetic cycle was set up and molecular dynamics (MD) simulations were used to study the molecular bases of the kinetics of PqsD. Trajectory analysis, pocket volume measurements, binding energy estimations and decompositions ensured insights into the binding mode of the substrates anthraniloyl-CoA and ß-ketodecanoic acid. CONCLUSIONS: Enzyme kinetics and SPR experiments hint at a ping-pong mechanism for PqsD with ACoA as first substrate. Trajectory analysis of different PqsD complexes evidenced ligand-dependent induced-fit motions affecting the modified ACoA funnel access to the exposure of a secondary channel. A tunnel-network is formed in which Ser317 plays an important role by binding to both substrates. Mutagenesis experiments resulting in the inactive S317F mutant confirmed the importance of this residue. Two binding modes for ß-ketodecanoic acid were identified with distinct catalytic mechanism preferences.

3.
J Med Chem ; 56(17): 6761-74, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23919758

RESUMO

The human pathogen Pseudomonas aeruginosa employs alkyl quinolones for cell-to-cell communication. The Pseudomonas quinolone signal (PQS) regulates various virulence factors via interaction with the transcriptional regulator PqsR. Therefore, we consider the development of PqsR antagonists a novel strategy to limit the pathogenicity of P. aeruginosa. A fragment identification approach using surface plasmon resonance screening led to the discovery of chemically diverse PqsR ligands. The optimization of the most promising hit (5) resulted in the oxadiazole-2-amine 37 showing pure antagonistic activity in Escherichia coli (EC50 = 7.5 µM) and P. aeruginosa (EC50 = 38.5 µM) reporter gene assays. 37 was able to diminish the production of the PQS precursor HHQ in a PqsH-deficient P. aeruginosa mutant. The level of the major virulence factor pyocyanin was significantly reduced in wild-type P. aeruginosa. In addition, site-directed mutagenesis in combination with isothermal titration calorimetry and NMR INPHARMA experiments revealed that the identified ligands bind to the same site of PqsR by adopting different binding modes. These findings will be utilized in a future fragment-growing approach aiming at novel therapeutic options for the treatment of P. aeruginosa infections.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Descoberta de Drogas , Oxidiazóis/farmacologia , Pseudomonas aeruginosa/patogenicidade , Biofísica , Ressonância de Plasmônio de Superfície , Virulência
4.
ACS Chem Biol ; 7(9): 1496-501, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22765028

RESUMO

The Gram-negative pathogen Pseudomonas aeruginosa produces an intercellular alkyl quinolone signaling molecule, the Pseudomonas quinolone signal. The pqs quorum sensing communication system that is characteristic for P. aeruginosa regulates the production of virulence factors. Therefore, we consider the pqs system a novel target to limit P. aeruginosa pathogenicity. Here, we present small molecules targeting a key player of the pqs system, PqsR. A rational design strategy in combination with surface plasmon resonance biosensor analysis led to the identification of PqsR binders. Determination of thermodynamic binding signatures and functional characterization in E. coli guided the hit optimization, resulting in the potent hydroxamic acid derived PqsR antagonist 11 (IC(50) = 12.5 µM). Remarkably it displayed a comparable potency in P. aeruginosa (IC(50) = 23.6 µM) and reduced the production of the virulence factor pyocyanin. Beyond this, site-directed mutagenesis together with thermodynamic analysis provided insights into the energetic characteristics of protein-ligand interactions. Thus the identified PqsR antagonists are promising scaffolds for further drug design efforts against this important pathogen.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Humanos , Mutagênese Sítio-Dirigida , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Quinolonas/metabolismo , Termodinâmica
5.
Anal Biochem ; 428(1): 28-30, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22677626

RESUMO

Surface plasmon resonance (SPR) as a label-free biosensor technique has become an important tool in drug discovery campaigns during the last couple of years. For good assay performance, it is of high interest to verify the functional activity on the immobilization of the target protein on the chip. This study illustrates the verification of the catalytic activity of the drug target protein PqsD by monitoring substrate conversion as a decrease in SPR signal and product detection by ultra high-performance liquid chromatography coupled to tandem mass spectrometry (UHPLC-MS(2)). This assay would be applicable to control surface activity of immobilized ligands.


Assuntos
Proteínas de Bactérias/metabolismo , Biocatálise , Técnicas Biossensoriais/instrumentação , Espectrometria de Massas/métodos , Pseudomonas aeruginosa/enzimologia , Ressonância de Plasmônio de Superfície/métodos , Cromatografia Líquida de Alta Pressão , Enzimas Imobilizadas/metabolismo
6.
Chem Biol ; 19(3): 381-90, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22444593

RESUMO

The pqs quorum sensing communication system of Pseudomonas aeruginosa controls virulence factor production and is involved in biofilm formation, therefore playing an important role for pathogenicity. In order to attenuate P. aeruginosa pathogenicity, we followed a ligand-based drug design approach and synthesized a series of compounds targeting PqsR, the receptor of the pqs system. In vitro evaluation using a reporter gene assay in Escherichia coli led to the discovery of the first competitive PqsR antagonists, which are highly potent (K(d,app) of compound 20: 7 nM). These antagonists are able to reduce the production of the virulence factor pyocyanin in P. aeruginosa. Our finding offers insights into the ligand-receptor interaction of PqsR and provides a promising starting point for further drug design.


Assuntos
4-Quinolonas/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/metabolismo , Percepção de Quorum/efeitos dos fármacos , 4-Quinolonas/síntese química , 4-Quinolonas/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Desenho de Fármacos , Genes Reporter , Cinética , Piocianina/biossíntese
7.
J Med Chem ; 55(7): 3307-18, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22380653

RESUMO

The reduction of estrone to estradiol, the most potent estrogen in human, is catalyzed by 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1). A promising approach for the treatment of estrogen-dependent diseases is the reduction of intracellular estradiol formation by inhibition of 17ß-HSD1. For the species-specific optimization of the (hydroxyphenyl)naphthols, a combinatorial approach was applied and enhanced by a focused synthesis that resulted in the aromatic-substituted (hydroxyphenyl)naphthol sulfonamides. Rigidification of 12 led to the 4-indolylsulfonamide 30, which is a highly active and selective human 17ß-HSD1 inhibitor, as well as a highly potent and selective inhibitor of 17ß-HSD1 from Callithrix jacchus. It shows no affinity to the estrogen receptors α and ß and good intracellular activity (T47D). Thus, compound 30 shows good properties for further ADMET studies and might be a candidate for the in vivo proof of concept in C. jacchus.


Assuntos
Endometriose/tratamento farmacológico , Estradiol Desidrogenases/antagonistas & inibidores , Naftóis/síntese química , Sulfonamidas/síntese química , Animais , Callithrix , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Naftóis/química , Naftóis/farmacologia , Placenta/enzimologia , Gravidez , Especificidade da Espécie , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia
8.
PLoS One ; 6(8): e22990, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21857977

RESUMO

17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) catalyzes the reduction of estrone to estradiol, which is the most potent estrogen in humans. Inhibition of 17ß-HSD1 and thereby reducing the intracellular estradiol concentration is thus a promising approach for the treatment of estrogen dependent diseases. In the past, several steroidal and non-steroidal inhibitors of 17ß-HSD1 have been described but so far there is no cocrystal structure of the latter in complex with 17ß-HSD1. However, a distinct knowledge of active site topologies and protein-ligand interactions is a prerequisite for structure-based drug design and optimization. An elegant strategy to enhance this knowledge is to compare inhibition values obtained for one compound toward ortholog proteins from various species, which are highly conserved in sequence and differ only in few residues. In this study the inhibitory potencies of selected members of different non-steroidal inhibitor classes toward marmoset 17ß-HSD1 were determined and the data were compared with the values obtained for the human enzyme. A species specific inhibition profile was observed in the class of the (hydroxyphenyl)naphthols. Using a combination of computational methods, including homology modelling, molecular docking, MD simulation, and binding energy calculation, a reasonable model of the three-dimensional structure of marmoset 17ß-HSD1 was developed and inhibition data were rationalized on the structural basis. In marmoset 17ß-HSD1, residues 190 to 196 form a small α-helix, which induces conformational changes compared to the human enzyme. The docking poses suggest these conformational changes as determinants for species specificity and energy decomposition analysis highlighted the outstanding role of Asn152 as interaction partner for inhibitor binding. In summary, this strategy of comparing the biological activities of inhibitors toward highly conserved ortholog proteins might be an alternative to laborious x-ray or site-directed mutagenesis experiments in certain cases. Additionally, it facilitates inhibitor design and optimization by offering new information on protein-ligand interactions.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Relação Estrutura-Atividade , Sequência de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Sítios de Ligação/genética , Callithrix , Simulação por Computador , Cristalografia por Raios X , Estradiol Desidrogenases/química , Estradiol Desidrogenases/metabolismo , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Especificidade por Substrato
9.
ChemMedChem ; 6(3): 476-87, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21337522

RESUMO

An attractive target that has still to be explored for the treatment of estrogen-dependent diseases, such as breast cancer and endometriosis, is the enzyme responsible for the last step in the biosynthesis of estradiol (E2): 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1). It catalyzes the reduction of the weakly active estrone (E1) into E2, which is the most potent estrogen in humans. Inhibition of 17ß-HSD1 lowers intracellular E2 concentrations and thus presents a therapeutic target for estrogen-dependent pathologies. Recently, we reported a new class of highly active and selective 17ß-HSD1 inhibitors: bicyclic substituted hydroxyphenylmethanones. Here, further structural variations on the bicyclic moiety are described, especially focusing on the exchange of its hydroxy function. Twenty-nine novel inhibitors were synthesized and evaluated for 17ß-HSD1 inhibition in a cell-free and cellular assay, for selectivity toward 17ßHSD2 and estrogen receptors (ER) alpha and beta, as well as for metabolic stability. The best compound exhibited IC50 values of 12 nM (cell-free assay) and 78 nM (cellular assay), high selectivity for 17ß-HSD1, and reasonable metabolic stability. A molecular docking study provided insight into the protein-ligand interactions of this compound with 17ß-HSD1.


Assuntos
Compostos Bicíclicos com Pontes/química , Inibidores Enzimáticos/química , Estradiol Desidrogenases/antagonistas & inibidores , Sítios de Ligação , Simulação por Computador , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/metabolismo , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/química , Receptor beta de Estrogênio/metabolismo , Humanos , Microssomos Hepáticos/metabolismo , Relação Estrutura-Atividade
10.
J Steroid Biochem Mol Biol ; 125(1-2): 66-82, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21193039

RESUMO

17ß-Hydroxysteroid dehydrogenases (17ß-HSDs) are oxidoreductases, which play a key role in estrogen and androgen steroid metabolism by catalyzing final steps of the steroid biosynthesis. Up to now, 14 different subtypes have been identified in mammals, which catalyze NAD(P)H or NAD(P)(+) dependent reductions/oxidations at the 17-position of the steroid. Depending on their reductive or oxidative activities, they modulate the intracellular concentration of inactive and active steroids. As the genomic mechanism of steroid action involves binding to a steroid nuclear receptor, 17ß-HSDs act like pre-receptor molecular switches. 17ß-HSDs are thus key enzymes implicated in the different functions of the reproductive tissues in both males and females. The crucial role of estrogens and androgens in the genesis and development of hormone dependent diseases is well recognized. Considering the pivotal role of 17ß-HSDs in steroid hormone modulation and their substrate specificity, these proteins are promising therapeutic targets for diseases like breast cancer, endometriosis, osteoporosis, and prostate cancer. The selective inhibition of the concerned enzymes might provide an effective treatment and a good alternative to the existing endocrine therapies. Herein, we give an overview of functional and structural aspects for the different 17ß-HSDs. We focus on steroidal and non-steroidal inhibitors recently published for each subtype and report on existing animal models for the different 17ß-HSDs and the respective diseases. Article from the Special issue on Targeted Inhibitors.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Inibidores Enzimáticos/metabolismo , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , 17-Hidroxiesteroide Desidrogenases/classificação , Sequência de Aminoácidos , Androgênios/química , Androgênios/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Domínio Catalítico , Inibidores Enzimáticos/química , Estrogênios/química , Estrogênios/metabolismo , Feminino , Humanos , Isoenzimas/classificação , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Filogenia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Conformação Proteica , Alinhamento de Sequência
11.
J Med Chem ; 54(2): 534-47, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21189020

RESUMO

Inhibition of 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) is a novel and attractive approach to reduce the local levels of the active estrogen 17ß-estradiol in patients with estrogen-dependent diseases like breast cancer or endometriosis. With the aim of optimizing the biological profile of 17ß-HSD1 inhibitors from the hydroxyphenylnaphthol class, structural optimizations were performed at the 1-position of the naphthalene by introduction of different heteroaromatic rings as well as substituted phenyl groups. In the latter class of compounds, which were synthesized applying Suzuki-cross coupling, the 3-methanesulfonamide 15 turned out to be a highly potent 17ß-HSD1 inhibitor (IC(50) = 15 nM in a cell-free assay). It was also very active in the cellular assay (T47D cells, IC(50) = 71 nM) and selective toward 17ß-HSD2 and the estrogen receptors α and ß. It showed a good membrane permeation and metabolic stability and was orally available in the rat.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Estrogênios/fisiologia , Naftóis/síntese química , Fenóis/síntese química , Esteroides/química , Administração Oral , Animais , Disponibilidade Biológica , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Sistema Livre de Células , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Fígado/metabolismo , Masculino , Microssomos/efeitos dos fármacos , Microssomos/enzimologia , Mimetismo Molecular , Naftóis/química , Naftóis/farmacologia , Fenóis/química , Fenóis/farmacologia , Placenta/enzimologia , Gravidez , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Proteínas Recombinantes/antagonistas & inibidores , Relação Estrutura-Atividade
12.
Resuscitation ; 58(2): 209-17, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12909384

RESUMO

Intestinal ischaemia is a major complication of shock syndromes causing translocation of bacteria and endotoxins and multiple organ failure in intensive care patients. The present study was designed to use microdialysis as a tool to monitor intestinal ischaemia after cardiac arrest and resuscitation in pigs. For this purpose, microdialysis probes were implanted in pig jejunal wall, peritoneum, skeletal muscle and brain, and interstitial fluid was obtained during circulatory arrest (induced by ventricular fibrillation) and after return of spontaneous circulation (ROSC). Cardiac arrest for 4 min caused a prolonged (60 min) reduction of blood flow in jejunal wall, muscle and brain as determined by the ethanol technique. This was accompanied by cellular damage in heart muscle and brain as indicated by increased levels of troponin-I and protein S-100, respectively. Plasma levels of glucose, lactate and choline were increased at 15-60 min following cardiac arrest. In contrast, cardiac arrest induced a rapid but variable decrease of interstitial glucose levels in all monitored organs; this decrease was followed by an increase over baseline during reperfusion. In the intestine, lactate, glutamate and choline levels were increased during ischaemia and reperfusion for 60-120 min; intestinal and peritoneal samples yielded parallel changes of lactate levels. Brain and muscle samples showed similar changes as in intestinum and peritoneum except for glutamate, which was increased in brain but not in muscle. We conclude that intestinal ischaemia occurs as a consequence of cardiac arrest and resuscitation and can be monitored by in vivo microdialysis. Comparative analysis by multi-site microdialysis reveals that the intestine is equally or even more sensitive to ischaemia than brain or muscle.


Assuntos
Reanimação Cardiopulmonar , Parada Cardíaca/complicações , Intestinos/irrigação sanguínea , Isquemia/metabolismo , Animais , Encéfalo/metabolismo , Colina/metabolismo , Espaço Extracelular/metabolismo , Glucose/metabolismo , Ácido Glutâmico/metabolismo , Mucosa Intestinal/metabolismo , Ácido Láctico/metabolismo , Microdiálise , Músculo Esquelético/metabolismo , Peritônio/metabolismo , Suínos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA