Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chembiochem ; 23(21): e202200351, 2022 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-35951472

RESUMO

The zika virus (ZIKV), transmitted to humans from the bites of Aedes Aegypti and Aedes Albopictus mosquitoes produces Zika fever and neurodegenerative disorders that despite affecting millions of people, most recently in Africa and the Americas, has been declared a neglected tropical disease by the World Health Organization. In this work, atomistic molecular dynamics simulations followed by rigorous analysis of the intermolecular interactions reveal crucial aspects of the initial virus⋯cell molecular recognition and attachment, events that trigger the infectious cycle. Previous experimental studies have shown that Dermatan Sulfate (DS) and Chondroitin Sulfate A (CSA), two glycosaminoglycans which are actually epimers to each other and that are structural constituents of receptors expressed in cell membranes, are the preferred anchorage sites, with a marked preference for DS. Our calculations rationalize this preference from a molecular perspective as follows: when free of the virus, DS has one sulfate group that does not participate in intramolecular strong hydrogen bonds, thus, it is readily available to interact with the envelope protein of the virus (Zika-E), then, after formation of the complexes, Zika-E⋯DS exhibits ten strong salt brides connecting the two fragments against only six salt bridges and two hydrogen bonds in Zika-E⋯CSA. Our results complement the current view of the interaction between the virus and the receptor glycosoaminoglycans revealing that the negatively charged carboxylate groups in CSA and DS are just as important as the sulfates because of the formation of equally strong salt bridges with the positively charged Arginine and Lysine aminoacids in the envelope protein of the virus.


Assuntos
Aedes , Infecção por Zika virus , Zika virus , Animais , Humanos , Zika virus/metabolismo , Simulação de Dinâmica Molecular , Aedes/metabolismo , Glicosaminoglicanos
2.
Comput Struct Biotechnol J ; 19: 4360-4372, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34429853

RESUMO

Tubulin is a well-validated target for herbicides, fungicides, anti-parasitic, and anti-tumor drugs. Many of the non-cancer tubulin drugs bind to its colchicine site but no colchicine-site anticancer drug is available. The colchicine site is composed of three interconnected sub-pockets that fit their ligands and modify others' preference, making the design of molecular hybrids (that bind to more than one sub-pocket) a difficult task. Taking advantage of the more than eighty published X-ray structures of tubulin in complex with ligands bound to the colchicine site, we generated an ensemble of pharmacophore representations that flexibly sample the interactional space between the ligands and target. We searched the ZINC database for scaffolds able to fit several of the subpockets, such as tetrazoles, sulfonamides and diarylmethanes, selected roughly ~8000 compounds with favorable predicted properties. A Flexi-pharma virtual screening, based on ensemble pharmacophore, was performed by two different methodologies. Combining the scaffolds that best fit the ensemble pharmacophore-representation, we designed a new family of ligands, resulting in a novel tubulin modulator. We synthesized tetrazole 5 and tested it as a tubulin inhibitor in vitro. In good agreement with the design principles, it demonstrated micromolar activity against in vitro tubulin polymerization and nanomolar anti-proliferative effect against human epithelioid carcinoma HeLa cells through microtubule disruption, as shown by immunofluorescence confocal microscopy. The integrative methodology succedes in the design of new scaffolds for flexible proteins with structural coupling between pockets, thus expanding the way in which computational methods can be used as significant tools in the drug design process.

3.
J Alzheimers Dis ; 82(s1): S321-S333, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33337368

RESUMO

BACKGROUND: The most important hallmark in the neuropathology of Alzheimer's disease (AD) is the formation of amyloid-ß (Aß) fibrils due to the misfolding/aggregation of the Aß peptide. Preventing or reverting the aggregation process has been an active area of research. Naturally occurring products are a potential source of molecules that may be able to inhibit Aß42 peptide aggregation. Recently, we and others reported the anti-aggregating properties of curcumin and some of its derivatives in vitro, presenting an important therapeutic avenue by enhancing these properties. OBJECTIVE: To computationally assess the interaction between Aß peptide and a set of curcumin derivatives previously explored in experimental assays. METHODS: The interactions of ten ligands with Aß monomers were studied by combining molecular dynamics and molecular docking simulations. We present the in silico evaluation of the interaction between these derivatives and the Aß42 peptide, both in the monomeric and fibril forms. RESULTS: The results show that a single substitution in curcumin could significantly enhance the interaction between the derivatives and the Aß42 monomers when compared to a double substitution. In addition, the molecular docking simulations showed that the interaction between the curcumin derivatives and the Aß42 monomers occur in a region critical for peptide aggregation. CONCLUSION: Results showed that a single substitution in curcumin improved the interaction of the ligands with the Aß monomer more so than a double substitution. Our molecular docking studies thus provide important insights for further developing/validating novel curcumin-derived molecules with high therapeutic potential for AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Simulação por Computador , Curcumina/metabolismo , Simulação de Acoplamento Molecular/métodos , Amiloide/química , Peptídeos beta-Amiloides/química , Curcumina/química , Humanos , Simulação de Dinâmica Molecular , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína
4.
PLoS Comput Biol ; 16(8): e1007898, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32797038

RESUMO

New treatments for diseases caused by antimicrobial-resistant microorganisms can be developed by identifying unexplored therapeutic targets and by designing efficient drug screening protocols. In this study, we have screened a library of compounds to find ligands for the flavin-adenine dinucleotide synthase (FADS) -a potential target for drug design against tuberculosis and pneumonia- by implementing a new and efficient virtual screening protocol. The protocol has been developed for the in silico search of ligands of unexplored therapeutic targets, for which limited information about ligands or ligand-receptor structures is available. It implements an integrative funnel-like strategy with filtering layers that increase in computational accuracy. The protocol starts with a pharmacophore-based virtual screening strategy that uses ligand-free receptor conformations from molecular dynamics (MD) simulations. Then, it performs a molecular docking stage using several docking programs and an exponential consensus ranking strategy. The last filter, samples the conformations of compounds bound to the target using MD simulations. The MD conformations are scored using several traditional scoring functions in combination with a newly-proposed score that takes into account the fluctuations of the molecule with a Morse-based potential. The protocol was optimized and validated using a compound library with known ligands of the Corynebacterium ammoniagenes FADS. Then, it was used to find new FADS ligands from a compound library of 14,000 molecules. A small set of 17 in silico filtered molecules were tested experimentally. We identified five inhibitors of the activity of the flavin adenylyl transferase module of the FADS, and some of them were able to inhibit growth of three bacterial species: C. ammoniagenes, Mycobacterium tuberculosis, and Streptococcus pneumoniae, where the last two are human pathogens. Overall, the results show that the integrative VS protocol is a cost-effective solution for the discovery of ligands of unexplored therapeutic targets.


Assuntos
Antibacterianos , Proteínas de Bactérias , Nucleotidiltransferases , Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Corynebacterium/efeitos dos fármacos , Corynebacterium/enzimologia , Desenho de Fármacos , Farmacorresistência Bacteriana/efeitos dos fármacos , Ligantes , Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/enzimologia , Nucleotidiltransferases/antagonistas & inibidores , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo
5.
J Comput Aided Mol Des ; 34(10): 1063-1077, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32656619

RESUMO

Computer-aided strategies are useful for reducing the costs and increasing the success-rate in drug discovery. Among these strategies, methods based on pharmacophores (an ensemble of electronic and steric features representing the target active site) are efficient to implement over large compound libraries. However, traditional pharmacophore-based methods require knowledge of active compounds or ligand-receptor structures, and only few ones account for target flexibility. Here, we developed a pharmacophore-based virtual screening protocol, Flexi-pharma, that overcomes these limitations. The protocol uses molecular dynamics (MD) simulations to explore receptor flexibility, and performs a pharmacophore-based virtual screening over a set of MD conformations without requiring prior knowledge about known ligands or ligand-receptor structures for building the pharmacophores. The results from the different receptor conformations are combined using a "voting" approach, where a vote is given to each molecule that matches at least one pharmacophore from each MD conformation. Contrarily to other approaches that reduce the pharmacophore ensemble to some representative models and score according to the matching models or molecule conformers, the Flexi-pharma approach takes directly into account the receptor flexibility by scoring in regards to the receptor conformations. We tested the method over twenty systems, finding an enrichment of the dataset for 19 of them. Flexi-pharma is computationally efficient allowing for the screening of thousands of compounds in minutes on a single CPU core. Moreover, the ranking of molecules by vote is a general strategy that can be applied with any pharmacophore-filtering program.


Assuntos
Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Preparações Farmacêuticas/análise , Preparações Farmacêuticas/química , Humanos , Ligantes , Modelos Moleculares , Preparações Farmacêuticas/metabolismo , Ligação Proteica
6.
Front Mol Biosci ; 7: 38, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32211419

RESUMO

As a class C GPCR and regulator of synaptic activity, mGlu5 is an attractive drug target, potentially offering treatment for several neurologic and psychiatric disorders. As little is known about the activation mechanism of mGlu5 at a structural level, potential of mean force calculations linked to molecular dynamics simulations were performed on the mGlu5 transmembrane domain crystal structure to explore various internal mechanisms responsible for its activation. Our results suggest that the hydrophilic interactions between intracellular loop 1 and the intracellular side of TM6 have to be disrupted to reach a theoretically active-like conformation. In addition, interactions between residues that are key for mGlu5 activation (Tyr6593.44 and Ile7515.51) and mGlu5 inactivation (Tyr6593.44 and Ser8097.39) have been identified. Inasmuch as mGlu5 receptor signaling is poorly understood, potentially showing a complex network of micro-switches and subtle structure-activity relationships, the present study represents a step forward in the understanding of mGlu5 transmembrane domain activation.

7.
Sci Rep ; 9(1): 5142, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30914702

RESUMO

Consensus-scoring methods are commonly used with molecular docking in virtual screening campaigns to filter potential ligands for a protein target. Traditional consensus methods combine results from different docking programs by averaging the score or rank of each molecule obtained from individual programs. Unfortunately, these methods fail if one of the docking programs has poor performance, which is likely to occur due to training-set dependencies and scoring-function parameterization. In this work, we introduce a novel consensus method that overcomes these limitations. We combine the results from individual docking programs using a sum of exponential distributions as a function of the molecule rank for each program. We test the method over several benchmark systems using individual and ensembles of target structures from diverse protein families with challenging decoy/ligand datasets. The results demonstrate that the novel method outperforms the best traditional consensus strategies over a wide range of systems. Moreover, because the novel method is based on the rank rather than the score, it is independent of the score units, scales and offsets, which can hinder the combination of results from different structures or programs. Our method is simple and robust, providing a theoretical basis not only for molecular docking but also for any consensus strategy in general.

8.
Phys Chem Chem Phys ; 20(38): 24869-24876, 2018 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-30232484

RESUMO

Understanding the migration of exogenous molecules to the interior of cell membranes is of pivotal importance to the design of new drugs and to the improvement of the capabilities of existing ones. This research dissects, from a molecular perspective, using classical molecular dynamics, the thermodynamic factors driving the insertion of ibuprofen into a model phosphatidylcholine membrane in an aqueous environment. We suggest an analysis of the insertion path that focuses on the net resulting force acting on the tertiary drug/water/membrane system; this allows us to understand the opposition that ibuprofen has to overcome as it inserts into the membrane. We provide conclusive evidence that entropy changes, arising from an increase of the number of possible microstates due to structural reorganization of the tertiary system, are the main factor driving this process. Our results allow us to unambiguously rationalize long standing conflicting experimental reports not understood up to now.


Assuntos
Anti-Inflamatórios não Esteroides/química , Entropia , Ibuprofeno/química , Membranas Artificiais , Modelos Teóricos , Dimiristoilfosfatidilcolina/química , Termodinâmica , Água/química
9.
Biophys J ; 115(6): 988-995, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30177440

RESUMO

Flavin mononucleotide (FMN) and flavin-adenine dinucleotide (FAD) are essential flavoprotein cofactors. A riboflavin kinase (RFK) activity catalyzes riboflavin phosphorylation to FMN, which can then be transformed into FAD by an FMN:adenylyltransferase (FMNAT) activity. Two enzymes are responsible for each one of these activities in eukaryotes, whereas prokaryotes have a single bifunctional enzyme, FAD synthase (FADS). FADS folds in two independent modules: the C-terminal with RFK activity and the N-terminal with FMNAT activity. Differences in structure and chemistry for the FMNAT catalysis among prokaryotic and eukaryotic enzymes pointed to the FMNAT activity of prokaryotic FADS as a potential antimicrobial target, making the structural model of the bacterial FMNAT module in complex with substrates relevant to understand the FADS catalytic mechanism and to the discovery of antimicrobial drugs. However, such a crystallographic complex remains elusive. Here, we have used molecular docking and molecular dynamics simulations to generate energetically stable interactions of the FMNAT module of FADS from Corynebacterium ammoniagenes with ATP/Mg2+ and FMN in both the monomeric and dimer-of-trimers assemblies reported for this protein. For the monomer, we have identified the residues that accommodate the reactive phosphates in a conformation compatible with catalysis. Interestingly, for the dimer-of-trimers conformation, we have found that the RFK module negatively influences FMN binding at the interacting FMNAT module. These results agree with calorimetric data of purified samples containing nearly 100% monomer or nearly 100% dimer-of-trimers, indicating that FMN binds to the monomer but not to the dimer-of-trimers. Such observations support regulation of flavin homeostasis by quaternary C. ammoniagenes FADS assemblies.


Assuntos
Biocatálise , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Multimerização Proteica , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Coenzimas/metabolismo , Corynebacterium/enzimologia , Mononucleotídeo de Flavina/metabolismo , Simulação de Acoplamento Molecular , Estrutura Quaternária de Proteína
10.
J Enzyme Inhib Med Chem ; 33(1): 241-254, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29258359

RESUMO

The increase of bacterial strains resistant to most of the available antibiotics shows a need to explore novel antibacterial targets to discover antimicrobial drugs. Bifunctional bacterial FAD synthetases (FADSs) synthesise the flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). These cofactors act in vital processes as part of flavoproteins, making FADS an essential enzyme. Bacterial FADSs are potential antibacterial targets because of differences to mammalian enzymes, particularly at the FAD producing site. We have optimised an activity-based high throughput screening assay targeting Corynebacterium ammoniagenes FADS (CaFADS) that identifies inhibitors of its different activities. We selected the three best high-performing inhibitors of the FMN:adenylyltransferase activity (FMNAT) and studied their inhibition mechanisms and binding properties. The specificity of the CaFADS hits was evaluated by studying also their effect on the Streptococcus pneumoniae FADS activities, envisaging differences that can be used to discover species-specific antibacterial drugs. The antimicrobial effect of these compounds was also evaluated on C. ammoniagenes, S. pneumoniae, and Mycobacterium tuberculosis cultures, finding hits with favourable antimicrobial properties.


Assuntos
Antibacterianos/farmacologia , Corynebacterium/enzimologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Nucleotidiltransferases/antagonistas & inibidores , Antibacterianos/síntese química , Antibacterianos/química , Corynebacterium/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Estrutura Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Nucleotidiltransferases/metabolismo , Streptococcus pneumoniae/efeitos dos fármacos , Relação Estrutura-Atividade
11.
Biophys J ; 110(3): 561-571, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26840722

RESUMO

Characterization by electron paramagnetic resonance techniques of several variants of Anabaena flavodoxin, where the naturally occurring FMN cofactor is substituted by different analogs, makes it possible to improve the details of the spin distribution map in the isoallosazine ring in its semiquinone state. The analyzed variants were selected to monitor the effects of intrinsic changes in the flavin ring electronic structure, as well as perturbations in the apoflavodoxin-flavin interaction, on the spin populations. When these effects were analyzed together with the functional properties of the different flavodoxin variants, a relationship between spin population and biochemical parameters, as the reduction potential, could be envisaged.


Assuntos
Proteínas de Bactérias/química , Dinitrocresóis/química , Flavoproteínas/química , Desacopladores/química , Sequência de Aminoácidos , Anabaena/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Ligação Proteica
12.
Curr Neuropharmacol ; 14(5): 441-54, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26391742

RESUMO

Metabotropic glutamate receptors (mGluRs) are important drug targets because of their involvement in several neurological diseases. Among mGluRs, mGlu5 is a particularly high-profile target because its positive or negative allosteric modulation can potentially treat schizophrenia or anxiety and chronic pain, respectively. Here, we computationally and experimentally probe the functional binding of a novel photoswitchable mGlu5 NAM, termed alloswitch-1, which loses its NAM functionality under violet light. We show alloswitch-1 binds deep in the allosteric pocket in a similar fashion to mavoglurant, the co-crystallized NAM in the mGlu5 transmembrane domain crystal structure. Alloswitch-1, like NAM 2-Methyl-6-(phenylethynyl)pyridine (MPEP), is significantly affected by P655M mutation deep in the allosteric pocket, eradicating its functionality. In MD simulations, we show alloswitch-1 and MPEP stabilize the co-crystallized water molecule located at the bottom of the allosteric site that is seemingly characteristic of the inactive receptor state. Furthermore, both NAMs form H-bonds with S809 on helix 7, which may constitute an important stabilizing interaction for NAM-induced mGlu5 inactivation. Alloswitch-1, through isomerization of its amide group from trans to cis is able to form an additional interaction with N747 on helix 5. This may be an important interaction for amide-containing mGlu5 NAMs, helping to stabilize their binding in a potentially unusual cis-amide state. Simulated conformational switching of alloswitch-1 in silico suggests photoisomerization of its azo group from trans to cis may be possible within the allosteric pocket. However, photoexcited alloswitch-1 binds in an unstable fashion, breaking H-bonds with the protein and destabilizing the co-crystallized water molecule. This suggests photoswitching may have destabilizing effects on mGlu5 binding and functionality.


Assuntos
Regulação Alostérica , Luz , Processos Fotoquímicos , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptor de Glutamato Metabotrópico 5/efeitos da radiação , Sítio Alostérico , Antagonistas de Aminoácidos Excitatórios/farmacologia , Células HEK293 , Humanos , Ligação de Hidrogênio , Isomerismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutação , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Piridinas/farmacologia , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Receptor de Glutamato Metabotrópico 5/genética , Água/química
13.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 12): 2526-42, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26627660

RESUMO

Riboflavin kinases (RFKs) catalyse the phosphorylation of riboflavin to produce FMN. In most bacteria this activity is catalysed by the C-terminal module of a bifunctional enzyme, FAD synthetase (FADS), which also catalyses the transformation of FMN into FAD through its N-terminal FMN adenylyltransferase (FMNAT) module. The RFK module of FADS is a homologue of eukaryotic monofunctional RFKs, while the FMNAT module lacks homologyto eukaryotic enzymes involved in FAD production. Previously, the crystal structure of Corynebacterium ammoniagenes FADS (CaFADS) was determined in its apo form. This structure predicted a dimer-of-trimers organization with the catalytic sites of two modules of neighbouring protomers approaching each other, leading to a hypothesis about the possibility of FMN channelling in the oligomeric protein. Here, two crystal structures of the individually expressed RFK module of CaFADS in complex with the products of the reaction, FMN and ADP, are presented. Structures are complemented with computational simulations, binding studies and kinetic characterization. Binding of ligands triggers dramatic structural changes in the RFK module, which affect large portions of the protein. Substrate inhibition and molecular-dynamics simulations allowed the conformational changes that take place along the RFK catalytic cycle to be established. The influence of these conformational changes in the FMNAT module is also discussed in the context of the full-length CaFADS protomer and the quaternary organization.


Assuntos
Difosfato de Adenosina/química , Proteínas de Bactérias/química , Corynebacterium/química , Mononucleotídeo de Flavina/química , Nucleotidiltransferases/química , Fosfotransferases (Aceptor do Grupo Álcool)/química , Difosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Corynebacterium/enzimologia , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Mononucleotídeo de Flavina/metabolismo , Expressão Gênica , Cinética , Ligantes , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
14.
J Struct Biol ; 192(3): 545-553, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26522273

RESUMO

A collection of crystal structures of rhodopsin, ß2-adrenergic and adenosine A2A receptors in active, intermediate and inactive states were selected for structural and energetic analyses to identify the changes involved in the activation/deactivation of Class A GPCRs. A set of helix interactions exclusive to either inactive or active/intermediate states were identified. The analysis of these interactions distinguished some local conformational changes involved in receptor activation, in particular, a packing between the intracellular domains of transmembrane helices H3 and H7 and a separation between those of H2 and H6. Also, differential movements of the extracellular and intracellular domains of these helices are apparent. Moreover, a segment of residues in helix H3, including residues L/I3.40 to L3.43, is identified as a key component of the activation mechanism, acting as a conformational hinge between extracellular and intracellular regions. Remarkably, the influence on the activation process of some glutamic and aspartic acidic residues and, as a consequence, the influence of variations on local pH is highlighted. Structural hypotheses that arose from the analysis of rhodopsin, ß2-adrenergic and adenosine A2A receptors were tested on the active and inactive M2 muscarinic acetylcholine receptor structures and further discussed in the context of the new mechanistic insights provided by the recently determined active and inactive crystal structures of the µ-opioid receptor. Overall, the structural and energetic analyses of the interhelical interactions present in this collection of Class A GPCRs suggests the existence of a common general activation mechanism featuring a chemical space useful for drug discovery exploration.


Assuntos
Receptor A2A de Adenosina/ultraestrutura , Receptor Muscarínico M2/ultraestrutura , Receptores Adrenérgicos beta 2/ultraestrutura , Receptores Opioides mu/ultraestrutura , Rodopsina/ultraestrutura , Sítios de Ligação , Cristalografia por Raios X , Ativação Enzimática/fisiologia , Modelos Moleculares , Conformação Proteica , Estrutura Terciária de Proteína , Receptor A2A de Adenosina/metabolismo , Receptor Muscarínico M2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores Opioides mu/metabolismo , Rodopsina/metabolismo , Transdução de Sinais/fisiologia
15.
J Phys Chem B ; 119(30): 9510-9, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26140747

RESUMO

Rhodopsin, the visual pigment in the retina, is a Class A G protein-coupled receptor (GPCR) covalently bound to retinal chromophore. In dark conditions, retinal is in the cis-isomeric state, stabilizing the rhodopsin inactive state as an inverse agonist. After light absorption, retinal undergoes an isomerization photoreaction to trans-retinal, which includes a conformational change of the receptor to its active state. In the absence of retinal, the apoprotein opsin presents a low level of constitutive activity, which depends on pH, with higher propensity of activation at acidic pH. To examine the effect and the underlying mechanism that protonation may have on opsin activation, a number of MD simulations were run varying the number and identity of acidic residues selected for protonation. Results show that the combined protonation of D83, E113, and E247 is of special relevance for the induction of receptor activation. Subsequent conformational analysis of the MD trajectories provides a structural mechanistic insight into the opsin activation process. Furthermore, because protonation seems to be a determining step in the activation of other GPCRs, the methodology and rationale used herein can be extended to mechanistic studies of GPCRs in general.


Assuntos
Simulação de Dinâmica Molecular , Opsinas/química , Opsinas/metabolismo , Prótons , Concentração de Íons de Hidrogênio , Estrutura Secundária de Proteína
16.
BMC Bioinformatics ; 16: 124, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25902715

RESUMO

BACKGROUND: G-protein-coupled receptors (GPCRs) are important drug targets and a better understanding of their molecular mechanisms would be desirable. The crystallization rate of GPCRs has accelerated in recent years as techniques have become more sophisticated, particularly with respect to Class A GPCRs interacting with G-proteins. These developments have made it possible for a quantitative analysis of GPCR geometrical features and binding-site conformations, including a statistical comparison between Class A GPCRs in active (agonist-bound) and inactive (antagonist-bound) states. RESULTS: Here we implement algorithms for the analysis of interhelical angles, distances, interactions and binding-site volumes in the transmembrane domains of 25 Class A GPCRs (7 active and 18 inactive). Two interhelical angles change in a statistically significant way between average inactive and active states: TM3-TM6 (by -9°) and TM6-TM7 (by +12°). A third interhelical angle: TM5-TM6 shows a trend, changing by -9°. In the transition from inactive to active states, average van der Waals interactions between TM3 and TM7 significantly increase as the average distance between them decreases by >2 Å. Average H-bonding between TM3 and TM6 decreases but is seemingly compensated by an increase in H-bonding between TM5 and TM6. In five Class A GPCRs, crystallized in both active and inactive states, increased H-bonding of agonists to TM6 and TM7, relative to antagonists, is observed. These protein-agonist interactions likely favour a change in the TM6-TM7 angle, which creates a narrowing in the binding pocket of activated receptors and an average ~200 Å(3) reduction in volume. CONCLUSIONS: In terms of similar conformational changes and agonist binding pattern, Class A GPCRs appear to share a common mechanism of activation, which can be exploited in future drug development.


Assuntos
Conformação Proteica , Receptores Adrenérgicos beta 2/química , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Sítios de Ligação , Domínio Catalítico , Simulação por Computador , Humanos , Ligação de Hidrogênio , Ligantes , Modelos Moleculares , Ligação Proteica , Receptores Adrenérgicos beta 2/metabolismo
17.
J Am Chem Soc ; 134(50): 20544-53, 2012 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-23181670

RESUMO

During photosynthesis, ferredoxin-NADP(+) reductase (FNR) catalyzes the electron transfer from ferredoxin to NADP(+) via its FAD cofactor. The final hydride transfer event between FNR and the nucleotide is a reversible process. Two different transient charge-transfer complexes form prior to and upon hydride transfer, FNR(rd)-NADP(+) and FNR(ox)-NADPH, regardless of the hydride transfer direction. Experimental structures of the FNR(ox):NADP(+) interaction have suggested a series of conformational rearrangements that might contribute to attaining the catalytically competent complex, but to date, no direct experimental information about the structure of this complex is available. Recently, a molecular dynamics (MD) theoretical approach was used to provide a putative organization of the active site that might represent a structure close to the transient catalytically competent interaction of Anabaena FNR with its coenzyme, NADP(+). Using this structure, we performed fully microscopic simulations of the hydride transfer processes between Anabaena FNR(rd)/FNR(ox) and NADP(+)/H, accounting also for the solvation. A dual-level QM/MM hybrid approach was used to describe the potential energy surface of the whole system. MD calculations using the finite-temperature string method combined with the WHAM method provided the potential of mean force for the hydride transfer processes. The results confirmed that the structural model of the reactants evolves to a catalytically competent transition state through very similar free energy barriers for both the forward and reverse reactions, in good agreement with the experimental hydride transfer rate constants reported for this system. This theoretical approach additionally provides subtle structural details of the mechanism in wild-type FNR and provides an explanation why Tyr303 makes possible the photosynthetic reaction, a process that cannot occur when this Tyr is replaced by a Ser.


Assuntos
Ferredoxina-NADP Redutase/metabolismo , NADP/metabolismo , Tirosina/fisiologia , Modelos Moleculares , Simulação de Dinâmica Molecular
18.
Biochim Biophys Acta ; 1817(12): 2118-27, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22982476

RESUMO

The chemical versatility of flavin cofactors within the flavoprotein environment allows them to play main roles in the bioenergetics of all type of organisms, particularly in energy transformation processes such as photosynthesis or oxidative phosphorylation. Despite the large diversity of properties shown by flavoproteins and of the biological processes in which they are involved, only two flavin cofactors, FMN and FAD (both derived from the 7,8-dimethyl-10-(1'-D-ribityl)-isoalloxazine), are usually found in these proteins. Using theoretical and experimental approaches we have carried out an evaluation of the effects introduced upon substituting the 7- and/or 8-methyls of the isoalloxazine ring in the chemical and oxido-reduction properties of the different atoms of the ring on free flavins and on the photosynthetic Anabaena Flavodoxin (a flavoprotein that replaces Ferredoxin as electron carrier from Photosystem I to Ferredoxin-NADP(+) reductase). In Anabaena Flavodoxin both the protein environment and the redox state contribute to modulate the chemical reactivity of the isoalloxazine ring. Anabaena apoflavodoxin is shown to be designed to stabilise/destabilise each one of the FMN redox states (but not of the analogues produced upon substitution of the 7- and/or 8-methyls groups) in the adequate proportions to provide Flavodoxin with the particular properties required for the functions in which it is involved in vivo. The 7- and/or 8-methyl groups of the ixoalloxazine can be discarded as the gate for electrons exchange in Anabaena Fld, but a key role in this process is envisaged for the C6 atom of the flavin and the backbone atoms of Asn58.


Assuntos
Anabaena/metabolismo , Apoproteínas/química , Mononucleotídeo de Flavina/química , Flavina-Adenina Dinucleotídeo/química , Flavodoxina/química , Apoproteínas/metabolismo , Elétrons , Mononucleotídeo de Flavina/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Flavinas/química , Flavinas/metabolismo , Flavodoxina/metabolismo , Cinética , Modelos Moleculares , Oxirredução , Fotossíntese , Ligação Proteica , Conformação Proteica
19.
Eur Biophys J ; 41(1): 117-28, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21538059

RESUMO

Ferredoxin-NADP(+) reductase (FNR) catalyses the electron transfer from ferredoxin to NADP(+) via its flavin FAD cofactor. A molecular dynamics theoretical approach is applied here to visualise the transient catalytically competent interaction of Anabaena FNR with its coenzyme, NADP(+). The particular role of some of the residues identified as key in binding and accommodating the 2'P-AMP moiety of the coenzyme is confirmed in molecular terms. Simulations also indicate that the architecture of the active site precisely contributes to the orientation of the N5 of the FAD isoalloxazine ring and the C4 of the coenzyme nicotinamide ring in the conformation of the catalytically competent hydride transfer complex and, therefore, contributes to the efficiency of the process. In particular, the side chain of the C-terminal Y303 in Anabaena FNR appears key to providing the optimum geometry by reducing the stacking probability between the isoalloxazine and nicotinamide rings, thus providing the required co-linearity and distance among the N5 of the flavin cofactor, the C4 of the coenzyme nicotinamide and the hydride that has to be transferred between them. All these factors are highly related to the reaction efficiency, mechanism and reversibility of the process.


Assuntos
Anabaena/enzimologia , Biocatálise , Domínio Catalítico , Coenzimas/metabolismo , Ferredoxina-NADP Redutase/química , Ferredoxina-NADP Redutase/metabolismo , Simulação de Dinâmica Molecular , Monofosfato de Adenosina/metabolismo , Substituição de Aminoácidos , Ferredoxina-NADP Redutase/genética , Flavinas/metabolismo , Niacinamida/metabolismo , Fatores de Tempo
20.
J Phys Chem B ; 114(9): 3368-79, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20163096

RESUMO

The flavoenzyme ferredoxin-NADP(+) reductase (FNR) catalyzes the production of NADPH during photosynthesis. The hydride-transfer reactions between the Anabaena mutant Tyr303Ser FNR(rd)/FNR(ox) and NADP(+)/H have been studied both experimentally and theoretically. Stopped-flow pre-steady-state kinetic measurements have shown that, in contrast to that observed for WT FNR, the physiological hydride transfer from Tyr303Ser FNR(rd) to NADP(+) does not occur. Conversely, the reverse reaction does take place with a rate constant just slightly slower than for WT FNR. This latter process shows temperature-dependent rates, but essentially temperature independent kinetic isotope effects, suggesting the reaction takes place following the vibration-driven tunneling model. In turn, ensemble-averaged variational transition-state theory with multidimensional tunneling calculations provide reaction rate constant values and kinetic isotope effects that agree with the experimental results, the experimental and the theoretical values for the reverse process being noticeably similar. The reaction mechanism behind these hydride transfers has been analyzed. The formation of a close contact ionic pair FADH(-):NADP(+) surrounded by the polar environment of the enzyme in the reactant complex of the mutant might be the cause of the huge difference between the direct and the reverse reaction.


Assuntos
Anabaena/enzimologia , Ferredoxinas/química , NADP/química , Substituição de Aminoácidos , Ferredoxinas/metabolismo , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , NADP/metabolismo , Oxirredução , Temperatura , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...