Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Commun ; 15(1): 4307, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811567

RESUMO

G protein-coupled receptors (GPCRs) are sophisticated signaling machines able to simultaneously elicit multiple intracellular signaling pathways upon activation. Complete (in)activation of all pathways can be counterproductive for specific therapeutic applications. This is the case for the serotonin 2 A receptor (5-HT2AR), a prominent target for the treatment of schizophrenia. In this study, we elucidate the complex 5-HT2AR coupling signature in response to different signaling probes, and its physiological consequences by combining computational modeling, in vitro and in vivo experiments with human postmortem brain studies. We show how chemical modification of the endogenous agonist serotonin dramatically impacts the G protein coupling profile of the 5-HT2AR and the associated behavioral responses. Importantly, among these responses, we demonstrate that memory deficits are regulated by Gαq protein activation, whereas psychosis-related behavior is modulated through Gαi1 stimulation. These findings emphasize the complexity of GPCR pharmacology and physiology and open the path to designing improved therapeutics for the treatment of stchizophrenia.


Assuntos
Transtornos da Memória , Transtornos Psicóticos , Receptor 5-HT2A de Serotonina , Serotonina , Animais , Feminino , Humanos , Masculino , Camundongos , Encéfalo/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Células HEK293 , Transtornos da Memória/metabolismo , Transtornos Psicóticos/metabolismo , Transtornos Psicóticos/tratamento farmacológico , Receptor 5-HT2A de Serotonina/metabolismo , Esquizofrenia/metabolismo , Serotonina/metabolismo , Transdução de Sinais
2.
Nat Commun ; 14(1): 3318, 2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-37308482

RESUMO

p38α is a versatile protein kinase that can control numerous processes and plays important roles in the cellular responses to stress. Dysregulation of p38α signaling has been linked to several diseases including inflammation, immune disorders and cancer, suggesting that targeting p38α could be therapeutically beneficial. Over the last two decades, numerous p38α inhibitors have been developed, which showed promising effects in pre-clinical studies but results from clinical trials have been disappointing, fueling the interest in the generation of alternative mechanisms of p38α modulation. Here, we report the in silico identification of compounds that we refer to as non-canonical p38α inhibitors (NC-p38i). By combining biochemical and structural analyses, we show that NC-p38i efficiently inhibit p38α autophosphorylation but weakly affect the activity of the canonical pathway. Our results demonstrate how the structural plasticity of p38α can be leveraged to develop therapeutic opportunities targeting a subset of the functions regulated by this pathway.


Assuntos
Inflamação , Transdução de Sinais , Humanos , Fosforilação
3.
J Chem Inf Model ; 63(1): 321-334, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36576351

RESUMO

Mutations in the kinase domain of the epidermal growth factor receptor (EGFR) can be drivers of cancer and also trigger drug resistance in patients receiving chemotherapy treatment based on kinase inhibitors. A priori knowledge of the impact of EGFR variants on drug sensitivity would help to optimize chemotherapy and design new drugs that are effective against resistant variants before they emerge in clinical trials. To this end, we explored a variety of in silico methods, from sequence-based to "state-of-the-art" atomistic simulations. We did not find any sequence signal that can provide clues on when a drug-related mutation appears or the impact of such mutations on drug activity. Low-level simulation methods provide limited qualitative information on regions where mutations are likely to cause alterations in drug activity, and they can predict around 70% of the impact of mutations on drug efficiency. High-level simulations based on nonequilibrium alchemical free energy calculations show predictive power. The integration of these "state-of-the-art" methods into a workflow implementing an interface for parallel distribution of the calculations allows its automatic and high-throughput use, even for researchers with moderate experience in molecular simulations.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Resistência a Medicamentos/genética , Receptores ErbB/metabolismo , Mutação , Resistencia a Medicamentos Antineoplásicos/genética
4.
ACS Pharmacol Transl Sci ; 4(6): 1818-1834, 2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34927013

RESUMO

Lysine-specific demethylase 1 (LSD1 or KDM1A) is a chromatin modifying enzyme playing a key role in the cell cycle and cell differentiation and proliferation through the demethylation of histones and nonhistone substrates. In addition to its enzymatic activity, LSD1 plays a fundamental scaffolding role as part of transcription silencing complexes such as rest co-repressor (CoREST) and nucleosome remodeling and deacetylase (NuRD). A host of classical amine oxidase inhibitors such as tranylcypromine, pargyline, and phenelzine together with LSD1 tool compounds such as SP-2509 and GSK-LSD1 have been extensively utilized in LSD1 mechanistic cancer studies. Additionally, several optimized new chemical entities have reached clinical trials in oncology such as ORY-1001 (iadademstat), GSK2879552, SP-2577 (seclidemstat), IMG-7289 (bomedemstat), INCB059872, and CC-90011 (pulrodemstat). Despite this, no single study exists that characterizes them all under the same experimental conditions, preventing a clear interpretation of published results. Herein, we characterize the whole LSD1 small molecule compound class as inhibitors of LSD1 catalytic activity, disruptors of SNAIL/GFI1 (SNAG)-scaffolding protein-protein interactions, inducers of cell differentiation, and potential anticancer treatments for hematological and solid tumors to yield an updated, unified perspective of this field. Our results highlight significant differences in potency and selectivity among the clinical compounds with iadademstat being the most potent and reveal that most of the tool compounds have very low activity and selectivity, suggesting some conclusions derived from their use should be taken with caution.

5.
J Chem Theory Comput ; 16(10): 6575-6585, 2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-32786895

RESUMO

By using a combination of classical Hamiltonian replica exchange with high-level quantum mechanical calculations on more than one hundred drug-like molecules, we explored here the energy cost associated with binding of drug-like molecules to target macromolecules. We found that, in general, the drug-like molecules present bound to proteins in the Protein Data Bank (PDB) can access easily the bioactive conformation and in fact for 73% of the studied molecules the "bioactive" conformation is within 3kBT from the most-stable conformation in solution as determined by DFT/SCRF calculations. Cases with large differences between the most-stable and the bioactive conformations appear in ligands recognized by ionic contacts, or very large structures establishing many favorable interactions with the protein. There are also a few cases where we observed a non-negligible uncertainty related to the experimental structure deposited in PDB. Remarkably, the rough automatic force field used here provides reasonable estimates of the conformational ensemble of drugs in solution. The outlined protocol can be used to better estimate the cost of adopting the bioactive conformation.


Assuntos
Bibliotecas de Moléculas Pequenas/química , Bases de Dados de Proteínas , Teoria da Densidade Funcional , Ligantes , Modelos Moleculares , Conformação Molecular , Peso Molecular , Proteínas/química
6.
J Chem Theory Comput ; 16(10): 6586-6597, 2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-32786900

RESUMO

Modern high-throughput structure-based drug discovery algorithms consider ligand flexibility, but typically with low accuracy, which results in a loss of performance in the derived models. Here we present the bioactive conformational ensemble (BCE) server and its associated database. The server creates conformational ensembles of drug-like ligands and stores them in the BCE database, where a variety of analyses are offered to the user. The workflow implemented in the BCE server combines enhanced sampling molecular dynamics with self-consistent reaction field quantum mechanics (SCRF/QM) calculations. The server automatizes all of the steps to transform one-dimensional (1D) or 2D representation of drugs into 3D molecules, which are then titrated, parametrized, hydrated, and optimized before being subjected to Hamiltonian replica-exchange (HREX) molecular dynamics simulations. Ensembles are collected and subjected to a clustering procedure to derive representative conformers, which are then analyzed at the SCRF/QM level of theory. All structural data are organized in a noSQL database accessible through a graphical interface and in a programmatic manner through a REST API. The server allows the user to define a private workspace and offers a deposition protocol as well as input files for "in house" calculations in those cases where confidentiality is a must. The database and the associated server are available at https://mmb.irbbarcelona.org/BCE.


Assuntos
Descoberta de Drogas , Preparações Farmacêuticas/química , Bases de Dados Factuais , Ensaios de Triagem em Larga Escala , Conformação Molecular , Simulação de Dinâmica Molecular , Teoria Quântica
7.
J Chem Theory Comput ; 16(10): 6598-6608, 2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-32856910

RESUMO

We present drug force-field recalibration (DFFR), a new method for refining of automatic force-fields used to represent small drugs in docking and molecular dynamics simulations. The method is based on fine-tuning of torsional terms to obtain ensembles that reproduce observables derived from reference data. DFFR is fast and flexible and can be easily automatized for a high-throughput regime, making it useful in drug-design projects. We tested the performance of the method in a few model systems and also in a variety of druglike molecules using reference data derived from: (i) density functional theory coupled to a self-consistent reaction field (DFT/SCRF) calculations on highly populated conformers and (ii) enhanced sampling quantum mechanical/molecular mechanics (QM/MM) where the drug is reproduced at the QM level, while the solvent is represented by classical force-fields. Extension of the method to include other sources of reference data is discussed.


Assuntos
Automação , Ensaios de Triagem em Larga Escala , Preparações Farmacêuticas/química , Calibragem , Teoria da Densidade Funcional , Simulação de Dinâmica Molecular
8.
Br J Pharmacol ; 177(17): 4034-4054, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32510591

RESUMO

BACKGROUND AND PURPOSE: Δ9 -Tetrahydrocannabinolic acid (Δ9 -THCA-A), the precursor of Δ9 -THC, is a non-psychotropic phytocannabinoid that shows PPARγ agonist activity. Here, we investigated the ability of Δ9 -THCA-A to modulate the classic cannabinoid CB1 and CB2 receptors and evaluated its anti-arthritis activity in vitro and in vivo. EXPERIMENTAL APPROACH: Cannabinoid receptors binding and intrinsic activity, as well as their downstream signalling, were analysed in vitro and in silico. The anti-arthritis properties of Δ9 -THCA-A were studied in human chondrocytes and in the murine model of collagen-induced arthritis (CIA). Plasma disease biomarkers were identified by LC-MS/MS based on proteomic and elisa assays. KEY RESULTS: Functional and docking analyses showed that Δ9 -THCA-A can act as an orthosteric CB1 receptor agonist and also as a positive allosteric modulator in the presence of CP-55,940. Also, Δ9 -THCA-A seemed to be an inverse agonist for CB2 receptors. In vivo, Δ9 -THCA-A reduced arthritis in CIA mice, preventing the infiltration of inflammatory cells, synovium hyperplasia, and cartilage damage. Furthermore, Δ9 -THCA-A inhibited expression of inflammatory and catabolic genes on knee joints. The anti-arthritic effect of Δ9 -THCA-A was blocked by either SR141716 or T0070907. Analysis of plasma biomarkers, and determination of cytokines and anti-collagen antibodies confirmed that Δ9 -THCA-A mediated its activity mainly through PPARγ and CB1 receptor pathways. CONCLUSION AND IMPLICATIONS: Δ9 -THCA-A modulates CB1 receptors through the orthosteric and allosteric binding sites. In addition, Δ9 -THCA-A exerts anti-arthritis activity through CB1 receptors and PPARγ pathways, highlighting its potential for the treatment of chronic inflammatory diseases such as rheumatoid arthritis.


Assuntos
Artrite Experimental , Dronabinol , Animais , Artrite Experimental/tratamento farmacológico , Cromatografia Líquida , Dronabinol/farmacologia , Camundongos , PPAR gama , Proteômica , Receptor CB1 de Canabinoide , Receptor CB2 de Canabinoide , Espectrometria de Massas em Tandem
9.
J Chem Inf Model ; 60(3): 1728-1736, 2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-32027130

RESUMO

The early stages of drug discovery rely on hit-to-lead programs, where initial hits undergo partial optimization to improve binding affinities for their biological target. This is an expensive and time-consuming process, requiring multiple iterations of trial and error designs, an ideal scenario for applying computer simulation. However, most state-of-the-art modeling techniques fail to provide a fast and reliable answer to the Induced-Fit protein-ligand problem. To aid in this matter, we present FragPELE, a new tool for in silico hit-to-lead drug design, capable of growing a fragment from a bound core while exploring the protein-ligand conformational space. We tested the ability of FragPELE to predict crystallographic data, even in cases where cryptic sub-pockets open because of the presence of particular R-groups. Additionally, we evaluated the potential of the software on growing and scoring five congeneric series from the 2015 FEP+ dataset, comparing them to FEP+, SP and Induced-Fit Glide, and MMGBSA simulations. Results show that FragPELE could be useful not only for finding new cavities and novel binding modes in cases where standard docking tools cannot but also to rank ligand activities in a reasonable amount of time and with acceptable precision.


Assuntos
Desenho de Fármacos , Software , Sítios de Ligação , Simulação por Computador , Ligantes , Simulação de Acoplamento Molecular , Ligação Proteica
10.
RSC Adv ; 10(12): 7058-7064, 2020 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-35493910

RESUMO

In silico binding site location and pose prediction for a molecule targeted at a large protein surface is a challenging task. We report a blind test with two peptidomimetic molecules that bind the flu virus hemagglutinin (HA) surface antigen, JNJ7918 and JNJ4796 (recently disclosed in van Dongen et al., Science, 2019, 363). Tests with a series of conventional approaches such as rigid (receptor) docking against available X-ray crystal structures or against an ensemble of structures generated by quick methodologies (NMA, homology modeling) gave mixed results, due to the shallowness and flexibility of the binding site and the sheer size of the target. However, tests with our Monte Carlo platform PELE in two protocols involving either exploration of the whole protein surface (global exploration), or the latter followed by refinement of best solutions (local exploration) yielded remarkably good results by locating the actual binding site and generating binding modes that recovered all native contacts found in the X-ray structures. Thus, the Monte Carlo scheme of PELE seems promising as a quick methodology to overcome the challenge of identifying entirely unknown binding sites and modes for protein-protein disruptors.

11.
Sci Rep ; 9(1): 17249, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31754232

RESUMO

The molecular recognition of the RORγ nuclear hormone receptor (NHR) ligand-binding domain (LBD) has been extensively studied with numerous X-ray crystal structures. However, the picture afforded by these complexes is static and does not fully explain the functional behavior of the LBD. In particular, the apo structure of the LBD seems to be in a fully active state, with no obvious differences to the agonist-bound structure. Further, several atypical in vivo inverse agonists have surprisingly been found to co-crystallize with the LBD in agonist mode (with co-activator), leading to a disconnection between molecular recognition and functional activity. Moreover, the experimental structures give no clues on how RORγ LBD binders access the interior of the LBD. To address all these points, we probe here, with a variety of simulation techniques, the fine structural balance of the RORγ LBD in its apo vs. holo form, the differences in flexibility and stability of the LBD in complex with agonists vs. inverse agonists and how binders diffuse in and out of the LBD in unbiased simulations. Our data conclusively point to the stability afforded by the so-called "agonist lock" between H479 and Y502 and the precise location of Helix 12 (H12) for the competence of the LBD to bind co-activator proteins. We observe the "water trapping" mechanism suggested previously for the atypical inverse agonists and discover a different behavior for the latter when co-activator is present or absent, which might help explain their conflicting data. Additionally, we unveil the same entry/exit path for agonists and inverse agonist into and out of the LBD for RORγ, suggesting it belongs to the type III NHR sub-family.


Assuntos
Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Ligação Proteica/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Sítios de Ligação/fisiologia , Humanos , Ligantes , Conformação Proteica
12.
J Chem Theory Comput ; 15(11): 6243-6253, 2019 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-31589430

RESUMO

In this study, we present a fully automatic platform based on our Monte Carlo algorithm, the Protein Energy Landscape Exploration method (PELE), for the estimation of absolute protein-ligand binding free energies, one of the most significant challenges in computer aided drug design. Based on a ligand pathway approach, an initial short enhanced sampling simulation is performed to identify reasonable starting positions for more extended sampling. This stepwise approach allows for a significant faster convergence of the free energy estimation using the Markov State Model (MSM) technique. PELE-MSM was applied on four diverse protein and ligand systems, successfully ranking compounds for two systems. Based on the results, current limitations and challenges with physics-based methods in computational structural biology are discussed. Overall, PELE-MSM constitutes a promising step toward computing absolute binding free energies and in their application into drug discovery projects.


Assuntos
Algoritmos , Proteínas/química , Desenho de Fármacos , Ligantes , Cadeias de Markov , Método de Monte Carlo , Ligação Proteica , Proteínas/metabolismo , Termodinâmica
13.
J Comput Aided Mol Des ; 33(7): 613-626, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31270654

RESUMO

Peptide-protein interactions are ubiquitous in living cells and essential to a wide range of biological processes, as well as pathologies such as cancer or cardiovascular disease. Yet, obtaining reliable binding mode predictions in peptide-protein docking remains a great challenge for most computational docking programs. The main goal of this study was to assess the performance of the small molecule docking program rDock in comparison to other widely used small molecule docking programs, using 100 peptide-protein systems with peptides ranging from 2 to 12 residues. As we used two large independent benchmark sets previously published for other small-molecule docking programs (AutoDockVina benchmark and LEADSPEP), the performance of rDock could directly be compared to the performances of AutoDockVina, Surflex, GOLD, and Glide, as well as to the peptide docking protocol PIPER-FlexPepDock and the webserver HPepDock. Our benchmark reveals that rDock can dock the 100 peptides with an overall backbone RMSD below 2.5 Å in 58.5% of the cases (76% for the 47 systems of the AutoDockVina benchmark set and 43% for the 53 systems of the LEADSPEP benchmark set). More specifically, rDock docks up to 11-residue peptides with a backbone RMSD below 2.5 Å in 60.75% of the cases. rDock displays higher accuracy than most available small molecule docking programs for 6-10-residue peptides and can sometimes perform similarly to the peptide docking tool, especially at a high level of exhaustiveness (100 or 150 runs). Its performance, as is the case for many other unguided small molecule docking tools, is compromised when the peptides adopt secondary structures upon binding. However, our analyses suggest that rDock could be used for predicting how medium-sized biologically relevant peptides bind to their respective protein targets when the latter bind in an extended mode.


Assuntos
Simulação de Acoplamento Molecular , Peptídeos/metabolismo , Proteínas/metabolismo , Software , Bases de Dados de Proteínas , Peptídeos/química , Ligação Proteica , Conformação Proteica , Proteínas/química
14.
J Chem Inf Model ; 58(8): 1707-1715, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30053380

RESUMO

Cryo-electron microscopy (cryo-EM) is emerging as a real alternative for structural elucidation. In spite of this, very few cryo-EM structures have been described so far as successful platforms for in silico drug design. Gabapentin and pregabalin are some of the most successful drugs in the treatment of epilepsy and neuropathic pain. Although both are in clinical use and are known to exert their effects by binding to the regulatory α2δ subunit of voltage gated calcium channels, their binding modes have never been characterized. We describe here the successful use of an exhaustive protein-ligand sampling algorithm on the α2δ-1 subunit of the recently published cryo-EM structure, with the goal of characterizing the ligand entry path and binding mode for gabapentin, pregabalin, and several other amino acidic α2δ-1 ligands. Our studies indicate that (i) all simulated drugs explore the same path for accessing the occluded binding site on the interior of the α2δ-1 subunit; (ii) they all roughly occupy the same pocket; (iii) the plasticity of the binding site allows the accommodation of a variety of amino acidic modulators, driven by the flexible "capping loop" delineated by residues Tyr426-Val435 and the floppy nature of Arg217; (iv) the predicted binding modes are in line with previously available mutagenesis data, confirming Arg217 as key for binding, with Asp428 and Asp467 highlighted as additional anchoring points for all amino acidic drugs. The study is one of the first proofs that latest-generation cryo-EM structures combined with exhaustive computational methods can be exploited in early drug discovery.


Assuntos
Analgésicos/farmacologia , Canais de Cálcio/metabolismo , Gabapentina/farmacologia , Pregabalina/farmacologia , Algoritmos , Analgésicos/química , Sítios de Ligação , Canais de Cálcio/química , Canais de Cálcio/ultraestrutura , Microscopia Crioeletrônica , Gabapentina/química , Humanos , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Pregabalina/química , Ligação Proteica
15.
Bioorg Med Chem ; 24(20): 4961-4969, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27545443

RESUMO

After decades of work, the correct determination of the binding mode of a small molecule into a target protein is still a challenging problem, whose difficulty depends on: (i) the sizes of the binding site and the ligand; (ii) the flexibility of both interacting partners, and (iii) the differential solvation of bound and unbound partners. We have evaluated the performance of standard rigid(receptor)/flexible(ligand) docking approaches with respect to last-generation fully flexible docking methods to obtain reasonable poses in a very challenging case: soluble Epoxide Hydrolase (sEH), a flexible protein showing different binding sites. We found that full description of the flexibility of both protein and ligand and accurate description of solvation leads to significant improvement in the ability of docking to reproduce well known binding modes, and at the same time capture the intrinsic binding promiscuity of the protein.


Assuntos
Epóxido Hidrolases/química , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas/química , Sítios de Ligação , Epóxido Hidrolases/antagonistas & inibidores , Epóxido Hidrolases/metabolismo , Humanos , Ligantes , Estrutura Molecular , Bibliotecas de Moléculas Pequenas/farmacologia
16.
J Chem Inf Model ; 56(5): 915-23, 2016 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-27082876

RESUMO

The development of mutations in HIV-1 protease (PR) hinders the activity of antiretroviral drugs, forcing changes in drug prescription. Most resistance assessments used to date rely on expert-based rules on predefined sets of stereotypical mutations; such an information-driven approach cannot capture new polymorphisms or be applied for new drugs. Computational modeling could provide a more general assessment of drug resistance and could be made available to clinicians through the Internet. We have created a protocol involving sequence comparison and all-atom protein-ligand induced fit simulations to predict resistance at the molecular level. We first compared our predictions with the experimentally determined IC50 values of darunavir, amprenavir, ritonavir, and indinavir from reference PR mutants displaying different resistance levels. We then performed analyses on a large set of variants harboring more than 10 mutations. Finally, several sequences from real patients were analyzed for amprenavir and darunavir. Our computational approach detected all of the genotype changes triggering high-level resistance, even those involving a large number of mutations.


Assuntos
Biologia Computacional/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Farmacorresistência Viral/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , Protease de HIV/metabolismo , HIV-1/efeitos dos fármacos , HIV-1/enzimologia , Sequência de Aminoácidos , Carbamatos/farmacologia , Darunavir/farmacologia , Furanos , Protease de HIV/química , Protease de HIV/genética , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Mutação , Multimerização Proteica , Estrutura Quaternária de Proteína , Sulfonamidas/farmacologia
17.
Proteins ; 80(1): 269-80, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22072623

RESUMO

Protein kinases play critical roles in cellular activation and differentiation, and are involved in numerous pathophysiological processes. As a critical component of the regulatory circuitry of the cell, the kinase domain has the ability to integrate multiple signals, yielding a predetermined output. In PKC and other protein kinases of the AGC family, several phosphorylation sites control the activity, but these are in turn influenced by the presence of ligands in the binding pocket, which promotes phosphorylation. Here, we take PKC-theta as a prototypical member of the family and use molecular dynamics simulations to investigate the cross-talk that exists between regulatory and functional sites. We first show how the apo-unphosphorylated form of the kinase is populating a conformational space in which access to the ATP binding site and to the activation loop (AL) are simultaneously hindered. This could explain why the inactive state is not only catalytically incompetent but also resistant to activation. AL phosphorylation induces ATP binding site opening, which can then readily accept the cofactor. But the signal transmission mechanism works both ways, and if ligand binding to the unphosphorylated form occurs first, the AL is de-protected and becomes exposed to phosphorylation, thus providing an explanation for the paradoxical activation of PKCs by their inhibitors.


Assuntos
Isoenzimas/química , Simulação de Dinâmica Molecular , Proteína Quinase C/química , Regulação Alostérica , Sítio Alostérico , Sequência de Aminoácidos , Domínio Catalítico , Sequência Conservada , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Dados de Sequência Molecular , Peptídeos/química , Fosforilação , Análise de Componente Principal , Ligação Proteica , Proteína Quinase C-theta , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência , Termodinâmica
18.
J Med Chem ; 50(2): 283-93, 2007 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-17228870

RESUMO

The main recognition characteristics of the ATP binding site of p38 mitogen activated protein kinase alpha (p38alpha MAPK) have been explored by a combination of modeling and bioinformatics techniques, making special emphasis in the characteristics of the site that justifies binding specificity with respect to other MAP kinases. Particularly, we have analyzed the binding mode of a new family of p38 MAPK inhibitors based on the pyridinyl-heterocycle core. This family of compounds has a marked pseudosymmetry and can exist in different tautomeric forms, which makes the determination of the binding mode especially challenging. A combination of homology modeling, quantum mechanics, classical docking, and molecular dynamics calculations allowed us to determine the main characteristics defining the binding mode of this new scaffold in the ATP binding site of p38alpha. A set of free energy calculations allowed us to verify the binding mode proposed, giving an overall excellent agreement with the experimental values. Finally, the binding mode of this new family of compounds was compared to that of other members of the pyridinyl and pyrimidinyl heterocycle class.


Assuntos
Modelos Moleculares , Pirazóis/química , Piridinas/química , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/química , Trifosfato de Adenosina/química , Sítios de Ligação , Isomerismo , Ligação Proteica , Teoria Quântica , Relação Estrutura-Atividade , Termodinâmica
19.
Protein Sci ; 15(11): 2525-33, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17001031

RESUMO

A fast method for the calculation of residue contributions to protein solvation is presented. The approach uses the exposed polar and apolar surface of protein residues and has been parametrized from the fractional contributions to solvation determined from linear response theory coupled to molecular dynamics simulations. Application of the method to a large subset of proteins taken from the Protein Data Bank allowed us to compute the expected fractional solvation of residues. This information is used to discuss when a residue or a group of residues presents an uncommon solvation profile.


Assuntos
Modelos Químicos , Conformação Proteica , Solubilidade , Sequência de Aminoácidos , Simulação por Computador , Estrutura Secundária de Proteína , Solventes/química , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...