Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Am Chem Soc ; 139(30): 10419-10431, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28696673

RESUMO

We investigated the hydration state of the deep, well-accessible hydrophobic S1' specificity pocket of the metalloprotease thermolysin with purposefully designed ligands using high-resolution crystallography and isothermal titration calorimetry. The S1' pocket is known to recognize selectively a very stringent set of aliphatic side chains such as valine, leucine, and isoleucine of putative substrates. We engineered a weak-binding ligand covering the active site of the protease without addressing the S1' pocket, thus transforming it into an enclosed cavity. Its sustained accessibility could be proved by accommodating noble gas atoms into the pocket in the crystalline state. The topology and electron content of the enclosed pocket with a volume of 141 Å3 were analyzed using an experimental MAD-phased electron density map that was calibrated to an absolute electron number scale, enabling access to the total electron content within the cavity. Our analysis indicates that the S1' pocket is virtually vacated, thus free of any water molecules. The thermodynamic signature of the reduction of the void within the pocket by growing aliphatic P1' substituents (H, Me, iPr, iBu) reveals a dramatic, enthalpy-dominated gain in free energy of binding resulting in a factor of 41 000 in Kd for the H-to-iBu transformation. Substituents placing polar decoy groups into the pocket to capture putatively present water molecules could not collect any evidence for a bound solvent molecule.


Assuntos
Termolisina/química , Sítios de Ligação , Calorimetria , Cristalografia por Raios X , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Modelos Moleculares , Termodinâmica , Termolisina/metabolismo
2.
Angew Chem Int Ed Engl ; 56(17): 4887-4890, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28371253

RESUMO

Hydrogen atoms play a key role in protein-ligand recognition. They determine the quality of established H-bonding networks and define the protonation of bound ligands. Structural visualization of H atoms by X-ray crystallography is rarely possible. We used neutron diffraction to determine the positions of the hydrogen atoms in the ligands aniline and 2-aminopyridine bound to the archetypical serine protease trypsin. The resulting structures show the best resolution so far achieved for proteins larger than 100 residues and allow an accurate description of the protonation states and interactions with nearby water molecules. Despite its low pKa of 4.6 and a large distance of 3.6 Što the charged Asp189 at the bottom of the S1 pocket, the amino group of aniline becomes protonated, whereas in 2-aminopyridine, the pyridine nitrogen picks up the proton although its amino group is 1.6 Šcloser to Asp189. Therefore, apart from charge-charge distances, tautomer stability is decisive for the resulting binding poses, an aspect that is pivotal for predicting correct binding.


Assuntos
Aminopiridinas/química , Aminopiridinas/farmacologia , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Prótons , Tripsina/metabolismo , Descoberta de Drogas , Humanos , Ligação de Hidrogênio , Ligantes , Modelos Moleculares , Difração de Nêutrons , Tripsina/química , Inibidores da Tripsina/química , Inibidores da Tripsina/farmacologia
3.
Angew Chem Int Ed Engl ; 56(7): 1908-1913, 2017 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-28097765

RESUMO

With the rising popularity of fragment-based approaches in drug development, more and more attention has to be devoted to the detection of false-positive screening results. In particular, the small size and low affinity of fragments drives screening techniques to their limit. The pursuit of a false-positive hit can cause significant loss of time and resources. Here, we present an instructive and intriguing investigation into the origin of misleading assay results for a fragment that emerged as the most potent binder for the aspartic protease endothiapepsin (EP) across multiple screening assays. This molecule shows its biological effect mainly after conversion into another entity through a reaction cascade that involves major rearrangements of its heterocyclic scaffold. The formed ligand binds EP through an induced-fit mechanism involving remarkable electrostatic interactions. Structural information in the initial screening proved to be crucial for the identification of this false-positive hit.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Desenho de Fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Sordariales/enzimologia , Ácido Aspártico Endopeptidases/química , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacologia , Ligantes , Simulação de Acoplamento Molecular , Ligação Proteica
4.
Biochemistry ; 54(10): 1943-55, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25706582

RESUMO

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


Assuntos
Proteínas de Bactérias/química , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/química , Modelos Moleculares , Staphylococcus aureus/enzimologia , Água/química , Catálise , Domínio Catalítico , Relação Estrutura-Atividade
5.
J Biol Chem ; 289(23): 15987-6005, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24739388

RESUMO

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


Assuntos
Antibacterianos/farmacologia , Desenho de Fármacos , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Piridonas/farmacologia , Animais , Antibacterianos/química , Antibacterianos/farmacocinética , Sequência de Bases , Cristalografia por Raios X , Primers do DNA , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Feminino , Camundongos , Camundongos Endogâmicos ICR , Testes de Sensibilidade Microbiana , Estrutura Molecular , Reação em Cadeia da Polimerase , Piridonas/química , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento
6.
J Biol Chem ; 288(47): 34190-34204, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24108128

RESUMO

The survival of Mycobacterium tuberculosis depends on mycolic acids, very long α-alkyl-ß-hydroxy fatty acids comprising 60-90 carbon atoms. However, despite considerable efforts, little is known about how enzymes involved in mycolic acid biosynthesis recognize and bind their hydrophobic fatty acyl substrates. The condensing enzyme KasA is pivotal for the synthesis of very long (C38-42) fatty acids, the precursors of mycolic acids. To probe the mechanism of substrate and inhibitor recognition by KasA, we determined the structure of this protein in complex with a mycobacterial phospholipid and with several thiolactomycin derivatives that were designed as substrate analogs. Our structures provide consecutive snapshots along the reaction coordinate for the enzyme-catalyzed reaction and support an induced fit mechanism in which a wide cavity is established through the concerted opening of three gatekeeping residues and several α-helices. The stepwise characterization of the binding process provides mechanistic insights into the induced fit recognition in this system and serves as an excellent foundation for the development of high affinity KasA inhibitors.


Assuntos
Antituberculosos/química , Sistemas de Liberação de Medicamentos , Inibidores Enzimáticos/química , Ácido Graxo Sintases/química , Mycobacterium tuberculosis/enzimologia , Ácidos Micólicos/química , Tuberculose/enzimologia , Antituberculosos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/metabolismo , Ácidos Micólicos/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Tuberculose/tratamento farmacológico
7.
Biochemistry ; 52(24): 4217-28, 2013 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-23697754

RESUMO

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


Assuntos
Enoil-(Proteína de Transporte de Acila) Redutase (NADPH, B-Específica)/química , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/química , Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/química , Ácido Graxo Sintase Tipo II/química , Staphylococcus aureus/enzimologia , Catálise , Química Farmacêutica , Cristalografia por Raios X , Desenho de Fármacos , Escherichia coli/metabolismo , Ácidos Graxos/química , Ligação de Hidrogênio , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Termodinâmica , Fatores de Tempo
8.
J Med Chem ; 64(3): 1611-1625, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33471524

RESUMO

In the S1 pocket, the serine proteases thrombin and trypsin commonly feature Asp189 and a Ala190Ser and Glu192Gln exchange. Nevertheless, thrombin cleaves peptide chains solely after Arg, and trypsin after Lys and Arg. Thrombin exhibits a Na+-binding site next to Asp189, which is missing in trypsin. The fragment benzylamine shows direct H-bonding to Asp189 in trypsin, while in thrombin, it forms an H-bond to Glu192. A series of fragments and expanded ligands were studied against both enzymes and mutated variants by crystallography and ITC. The selectivity-determining features of both S1 pockets are difficult to assign to one dominating factor. The Ala190Ser and Glu192Gln replacements may be regarded as highly conserved as no structural and affinity changes are observed between both proteases. With respect to charge distribution, Glu192, together with the thrombin-specific sodium ion, helps in creating an electrostatic gradient across the S1 pocket. This feature is definitely absent in trypsin but important for selectivity along with solvation-pattern differences in the S1 pocket.


Assuntos
Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Trombina/química , Tripsina/química , Sítios de Ligação , Sequência Conservada , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Cinética , Ligantes , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Relação Estrutura-Atividade , Especificidade por Substrato , Trombina/genética , Tripsina/genética
9.
ACS Infect Dis ; 7(4): 746-758, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33710875

RESUMO

The enoyl-acyl carrier protein (ACP) reductase (ENR) is a key enzyme within the bacterial fatty-acid synthesis pathway. It has been demonstrated that small-molecule inhibitors carrying the diphenylether (DPE) scaffold bear a great potential for the development of highly specific and effective drugs against this enzyme class. Interestingly, different substitution patterns of the DPE scaffold have been shown to lead to varying effects on the kinetic and thermodynamic behavior toward ENRs from different organisms. Here, we investigated the effect of a 4'-pyridone substituent in the context of the slow tight-binding inhibitor SKTS1 on the inhibition of the Staphylococcus aureus enoyl-ACP-reductase saFabI and the closely related isoenzyme from Mycobacterium tuberculosis, InhA, and explored a new interaction site of DPE inhibitors within the substrate-binding pocket. Using high-resolution crystal structures of both complexes in combination with molecular dynamics (MD) simulations, kinetic measurements, and quantum mechanical (QM) calculations, we provide evidence that the 4'-pyridone substituent adopts different tautomeric forms when bound to the two ENRs. We furthermore elucidate the structural determinants leading to significant differences in the residence time of SKTS1 on both enzymes.


Assuntos
Inibidores Enzimáticos/farmacologia , Isoenzimas , Oxirredutases/antagonistas & inibidores , Isomerismo , Mycobacterium tuberculosis/enzimologia , Staphylococcus aureus/enzimologia
10.
Nat Commun ; 9(1): 3559, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-30177695

RESUMO

Hydrogen bonds are key interactions determining protein-ligand binding affinity and therefore fundamental to any biological process. Unfortunately, explicit structural information about hydrogen positions and thus H-bonds in protein-ligand complexes is extremely rare and similarly the important role of water during binding remains poorly understood. Here, we report on neutron structures of trypsin determined at very high resolutions ≤1.5 Å in uncomplexed and inhibited state complemented by X-ray and thermodynamic data and computer simulations. Our structures show the precise geometry of H-bonds between protein and the inhibitors N-amidinopiperidine and benzamidine along with the dynamics of the residual solvation pattern. Prior to binding, the ligand-free binding pocket is occupied by water molecules characterized by a paucity of H-bonds and high mobility resulting in an imperfect hydration of the critical residue Asp189. This phenomenon likely constitutes a key factor fueling ligand binding via water displacement and helps improving our current view on water influencing protein-ligand recognition.


Assuntos
Cristalografia , Ligantes , Difração de Nêutrons , Ligação Proteica , Tripsina/química , Água , Benzamidinas/farmacologia , Simulação por Computador , Cristalografia por Raios X , Ligação de Hidrogênio , Inibidores de Serina Proteinase/farmacologia , Termodinâmica , Tripsina/efeitos dos fármacos , Tripsina/metabolismo
12.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 5): 346-55, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27139825

RESUMO

Crystallographic screening of the binding of small organic compounds (termed fragments) to proteins is increasingly important for medicinal chemistry-oriented drug discovery. To enable such experiments in a widespread manner, an affordable 96-compound library has been assembled for fragment screening in both academia and industry. The library is selected from already existing protein-ligand structures and is characterized by a broad ligand diversity, including buffer ingredients, carbohydrates, nucleotides, amino acids, peptide-like fragments and various drug-like organic compounds. When applied to the model protease endothiapepsin in a crystallographic screening experiment, a hit rate of nearly 10% was obtained. In comparison to other fragment libraries and considering that no pre-screening was performed, this hit rate is remarkably high. This demonstrates the general suitability of the selected compounds for an initial fragment-screening campaign. The library composition, experimental considerations and time requirements for a complete crystallographic fragment-screening campaign are discussed as well as the nine fully refined obtained endothiapepsin-fragment structures. While most of the fragments bind close to the catalytic centre of endothiapepsin in poses that have been observed previously, two fragments address new sites on the protein surface. ITC measurements show that the fragments bind to endothiapepsin with millimolar affinity.


Assuntos
Ácido Aspártico Endopeptidases/química , Fragmentos de Peptídeos/química , Calorimetria , Cristalografia por Raios X , Ligação Proteica , Conformação Proteica
13.
Chem Sci ; 7(9): 5945-5954, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27547299

RESUMO

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

14.
ACS Chem Biol ; 11(6): 1693-701, 2016 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-27028906

RESUMO

Fragment-based lead discovery (FBLD) has become a pillar in drug development. Typical applications of this method comprise at least two biophysical screens as prefilter and a follow-up crystallographic experiment on a subset of fragments. Clearly, structural information is pivotal in FBLD, but a key question is whether such a screening cascade strategy will retrieve the majority of fragment-bound structures. We therefore set out to screen 361 fragments for binding to endothiapepsin, a representative of the challenging group of aspartic proteases, employing six screening techniques and crystallography in parallel. Crystallography resulted in the very high number of 71 structures. Yet alarmingly, 44% of these hits were not detected by any biophysical screening approach. Moreover, any screening cascade, building on the results from two or more screening methods, would have failed to predict at least 73% of these hits. We thus conclude that, at least in the present case, the frequently applied biophysical prescreening filters deteriorate the number of possible X-ray hits while only the immediate use of crystallography enables exhaustive retrieval of a maximum of fragment structures, which represent a rich source guiding hit-to-lead-to-drug evolution.


Assuntos
Descoberta de Drogas/métodos , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/química , Biofísica , Calorimetria , Cristalografia por Raios X , Bases de Dados de Compostos Químicos , Modelos Moleculares , Inibidores de Proteases/química , Ligação Proteica , Relação Estrutura-Atividade
15.
J Med Chem ; 59(16): 7561-75, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27463859

RESUMO

Successful optimization of a given lead scaffold requires thorough binding-site mapping of the target protein particular in regions remote from the catalytic center where high conservation across protein families is given. We screened a 361-entry fragment library for binding to the aspartic protease endothiapepsin by crystallography. This enzyme is frequently used as a surrogate for the design of renin and ß-secretase inhibitors. A hit rate of 20% was achieved, providing 71 crystal structures. Here, we discuss 45 binding poses of fragments accommodated in pockets remote from the catalytic dyad. Three major hot spots are discovered in remote binding areas: Asp81, Asp119, and Phe291. Compared to the dyad binders, bulkier fragments occupy these regions. Many of the discovered fragments suggest an optimization concept on how to grow them into larger ligands occupying adjacent binding pockets that will possibly endow them with the desired selectivity for one given member of a protein family.


Assuntos
Ácido Aspártico Endopeptidases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Ácido Aspártico Endopeptidases/metabolismo , Biocatálise , Domínio Catalítico/efeitos dos fármacos , Cristalografia por Raios X , Ligantes , Modelos Moleculares , Inibidores de Proteases/química , Relação Estrutura-Atividade
16.
Structure ; 24(8): 1398-1409, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27452405

RESUMO

Today the identification of lead structures for drug development often starts from small fragment-like molecules raising the chances to find compounds that successfully pass clinical trials. At the heart of the screening for fragments binding to a specific target, crystallography delivers structural information essential for subsequent drug design. While it is common to search for bound ligands in electron densities calculated directly after an initial refinement cycle, we raise the important question whether this strategy is viable for fragments characterized by low affinities. Here, we describe and provide a collection of high-quality diffraction data obtained from 364 protein crystals treated with diverse fragments. Subsequent data analysis showed that ∼25% of all hits would have been missed without further refining the resulting structures. To enable fast and reliable hit identification, we have designed an automated refinement pipeline that will inspire the development of optimized tools facilitating the successful application of fragment-based methods.


Assuntos
Cristalografia por Raios X/estatística & dados numéricos , Ensaios de Triagem em Larga Escala , Bibliotecas de Moléculas Pequenas/química , Água/química , Cristalografia por Raios X/métodos , Conjuntos de Dados como Assunto , Desenho de Fármacos , Humanos , Difração de Raios X
17.
J Med Chem ; 59(21): 9743-9759, 2016 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-27726357

RESUMO

Crystallography is frequently used as follow-up method to validate hits identified by biophysical screening cascades. The capacity of crystallography to directly screen fragment libraries is often underestimated, due to its supposed low-throughput and need for high-quality crystals. We applied crystallographic fragment screening to map the protein-binding site of the aspartic protease endothiapepsin by individual soaking experiments. Here, we report on 41 fragments binding to the catalytic dyad and adjacent specificity pockets. The analysis identifies already known warheads but also reveals hydrazide, pyrazole, or carboxylic acid fragments as novel functional groups binding to the dyad. A remarkable swapping of the S1 and S1' pocket between structurally related fragments is explained by either steric demand, required displacement of a well-bound water molecule, or changes of trigonal-planar to tetrahedral geometry of an oxygen functional group in a side chain. Some warheads simultaneously occupying both S1 and S1' are promising starting points for fragment-growing strategies.


Assuntos
Ácido Aspártico Proteases/química , Ácido Aspártico Proteases/metabolismo , Biocatálise , Ácidos Carboxílicos/metabolismo , Hidrazinas/metabolismo , Pirazóis/metabolismo , Calorimetria , Ácidos Carboxílicos/química , Domínio Catalítico , Cristalografia por Raios X , Hidrazinas/química , Modelos Moleculares , Pirazóis/química
18.
Structure ; 23(1): 21-33, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25482540

RESUMO

With the exception of HIV, tuberculosis (TB) is the leading cause of mortality among infectious diseases. The urgent need to develop new antitubercular drugs is apparent due to the increasing number of drug-resistant Mycobacterium tuberculosis (Mtb) strains. Proteins involved in cholesterol import and metabolism have recently been discovered as potent targets against TB. FadA5, a thiolase from Mtb, is catalyzing the last step of the ß-oxidation reaction of the cholesterol side-chain degradation under release of critical metabolites and was shown to be of importance during the chronic stage of TB infections. To gain structural and mechanistic insight on FadA5, we characterized the enzyme in different stages of the cleavage reaction and with a steroid bound to the binding pocket. Structural comparisons to human thiolases revealed that it should be possible to target FadA5 specifically, and the steroid-bound structure provides a solid basis for the development of inhibitors against FadA5.


Assuntos
Acetil-CoA C-Acetiltransferase/química , Acetil-CoA C-Acetiltransferase/metabolismo , Antituberculosos , Descoberta de Drogas , Mycobacterium tuberculosis/enzimologia , Domínios e Motivos de Interação entre Proteínas , Domínio Catalítico , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica , Esteroides/metabolismo , Água/metabolismo
19.
ChemMedChem ; 10(9): 1511-21, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26259992

RESUMO

Fragment-based lead discovery is gaining momentum in drug development. Typically, a hierarchical cascade of several screening techniques is consulted to identify fragment hits which are then analyzed by crystallography. Because crystal structures with bound fragments are essential for the subsequent hit-to-lead-to-drug optimization, the screening process should distinguish reliably between binders and non-binders. We therefore investigated whether different screening methods would reveal similar collections of putative binders. First we used a biochemical assay to identify fragments that bind to endothiapepsin, a surrogate for disease-relevant aspartic proteases. In a comprehensive screening approach, we then evaluated our 361-entry library by using a reporter-displacement assay, saturation-transfer difference NMR, native mass spectrometry, thermophoresis, and a thermal shift assay. While the combined results of these screening methods retrieve 10 of the 11 crystal structures originally predicted by the biochemical assay, the mutual overlap of individual hit lists is surprisingly low, highlighting that each technique operates on different biophysical principles and conditions.


Assuntos
Bioquímica/métodos , Biofísica/métodos , Ensaios de Triagem em Larga Escala/métodos , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Descoberta de Drogas/métodos , Espectroscopia de Ressonância Magnética , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Espectrometria de Massas por Ionização por Electrospray/métodos
20.
Structure ; 20(5): 802-13, 2012 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-22579249

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) infections constitute a serious health threat worldwide, and novel antibiotics are therefore urgently needed. The enoyl-ACP reductase (saFabI) is essential for the S. aureus fatty acid biosynthesis and, hence, serves as an attractive drug target. We have obtained a series of snapshots of this enzyme that provide a mechanistic picture of ligand and inhibitor binding, including a dimer-tetramer transition combined with extensive conformational changes. Significantly, our results reveal key differences in ligand binding and recognition compared to orthologous proteins. The remarkable observed protein flexibility rationalizes our finding that saFabI is capable of efficiently reducing branched-chain fatty acid precursors. Importantly, branched-chain fatty acids represent a major fraction of the S. aureus cell membrane and are crucial for its in vivo fitness. Our discovery thus addresses a long-standing controversy regarding the essentiality of the fatty acid biosynthesis pathway in S. aureus rationalizing saFabI as a drug target.


Assuntos
Proteínas de Bactérias/química , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/antagonistas & inibidores , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/química , Staphylococcus aureus/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Enoil-(Proteína de Transporte de Acila) Redutase (NADH)/metabolismo , Ácidos Graxos/metabolismo , Cinética , Ligantes , Dados de Sequência Molecular , Conformação Proteica , Staphylococcus aureus/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA