Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Chem Theory Comput ; 20(14): 6341-6349, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38991145

RESUMO

Understanding drug residence times in target proteins is key to improving drug efficacy and understanding target recognition in biochemistry. While drug residence time is just as important as binding affinity, atomic-level understanding of drug residence times through molecular dynamics (MD) simulations has been difficult primarily due to the extremely long time scales. Recent advances in rare event sampling have allowed us to reach these time scales, yet predicting protein-ligand residence times remains a significant challenge. Here we present a semi-automated protocol to calculate the ligand residence times across 12 orders of magnitude of time scales. In our proposed framework, we integrate a deep learning-based method, the state predictive information bottleneck (SPIB), to learn an approximate reaction coordinate (RC) and use it to guide the enhanced sampling method metadynamics. We demonstrate the performance of our algorithm by applying it to six different protein-ligand complexes with available benchmark residence times, including the dissociation of the widely studied anticancer drug Imatinib (Gleevec) from both wild-type Abl kinase and drug-resistant mutants. We show how our protocol can recover quantitatively accurate residence times, potentially opening avenues for deeper insights into drug development possibilities and ligand recognition mechanisms.


Assuntos
Simulação de Dinâmica Molecular , Proteínas , Ligantes , Proteínas/química , Proteínas/metabolismo , Mesilato de Imatinib/química , Algoritmos , Ligação Proteica
2.
bioRxiv ; 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38659748

RESUMO

Understanding drug residence times in target proteins is key to improving drug efficacy and understanding target recognition in biochemistry. While drug residence time is just as important as binding affinity, atomic-level understanding of drug residence times through molecular dynamics (MD) simulations has been difficult primarily due to the extremely long timescales. Recent advances in rare event sampling have allowed us to reach these timescales, yet predicting protein-ligand residence times remains a significant challenge. Here we present a semi-automated protocol to calculate the ligand residence times across 12 orders of magnitudes of timescales. In our proposed framework, we integrate a deep learning-based method, the state predictive information bottleneck (SPIB), to learn an approximate reaction coordinate (RC) and use it to guide the enhanced sampling method metadynamics. We demonstrate the performance of our algorithm by applying it to six different protein-ligand complexes with available benchmark residence times, including the dissociation of the widely studied anti-cancer drug Imatinib (Gleevec) from both wild-type Abl kinase and drug-resistant mutants. We show how our protocol can recover quantitatively accurate residence times, potentially opening avenues for deeper insights into drug development possibilities and ligand recognition mechanisms.

3.
J Chem Inf Model ; 64(7): 2789-2797, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-37981824

RESUMO

Kinases compose one of the largest fractions of the human proteome, and their misfunction is implicated in many diseases, in particular, cancers. The ubiquitousness and structural similarities of kinases make specific and effective drug design difficult. In particular, conformational variability due to the evolutionarily conserved Asp-Phe-Gly (DFG) motif adopting in and out conformations and the relative stabilities thereof are key in structure-based drug design for ATP competitive drugs. These relative conformational stabilities are extremely sensitive to small changes in sequence and provide an important problem for sampling method development. Since the invention of AlphaFold2, the world of structure-based drug design has noticeably changed. In spite of it being limited to crystal-like structure prediction, several methods have also leveraged its underlying architecture to improve dynamics and enhanced sampling of conformational ensembles, including AlphaFold2-RAVE. Here, we extend AlphaFold2-RAVE and apply it to a set of kinases: the wild type DDR1 sequence and three mutants with single point mutations that are known to behave drastically differently. We show that AlphaFold2-RAVE is able to efficiently recover the changes in relative stability using transferable learned order parameters and potentials, thereby supplementing AlphaFold2 as a tool for exploration of Boltzmann-weighted protein conformations (Meller, A.; Bhakat, S.; Solieva, S.; Bowman, G. R. Accelerating Cryptic Pocket Discovery Using AlphaFold. J. Chem. Theory Comput. 2023, 19, 4355-4363).


Assuntos
Oligopeptídeos , Inibidores de Proteínas Quinases , Humanos , Modelos Moleculares , Inibidores de Proteínas Quinases/química , Conformação Proteica , Oligopeptídeos/química
4.
ArXiv ; 2023 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-37731662

RESUMO

Kinases compose one of the largest fractions of the human proteome, and their misfunction is implicated in many diseases, in particular cancers. The ubiquitousness and structural similarities of kinases makes specific and effective drug design difficult. In particular, conformational variability due to the evolutionarily conserved DFG motif adopting in and out conformations and the relative stabilities thereof are key in structure-based drug design for ATP competitive drugs. These relative conformational stabilities are extremely sensitive to small changes in sequence, and provide an important problem for sampling method development. Since the invention of AlphaFold2, the world of structure-based drug design has noticably changed. In spite of it being limited to crystal-like structure prediction, several methods have also leveraged its underlying architecture to improve dynamics and enhanced sampling of conformational ensembles, including AlphaFold2-RAVE. Here, we extend AlphaFold2-RAVE and apply it to a set of kinases: the wild type DDR1 sequence and three mutants with single point mutations that are known to behave drastically differently. We show that AlphaFold2-RAVE is able to efficiently recover the changes in relative stability using transferable learnt order parameters and potentials, thereby supplementing AlphaFold2 as a tool for exploration of Boltzmann-weighted protein conformations.

5.
Org Biomol Chem ; 20(37): 7429-7438, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36097881

RESUMO

We report the molecular recognition properties of Pillar[n]MaxQ (P[n]MQ) toward a series of (methylated) amino acids, amino acid amides, and post-translationally modified peptides by a combination of 1H NMR, isothermal titration calorimetry, indicator displacement assays, and molecular dynamics simulations. We find that P6MQ is a potent receptor for N-methylated amino acid side chains. P6MQ recognized the H3K4Me3 peptide with Kd = 16 nM in phosphate buffered saline.


Assuntos
Aminoácidos , Peptídeos , Amidas , Aminoácidos/química , Calorimetria , Peptídeos/química , Fosfatos
6.
Proc Natl Acad Sci U S A ; 119(32): e2203656119, 2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35925885

RESUMO

Using simulations or experiments performed at some set of temperatures to learn about the physics or chemistry at some other arbitrary temperature is a problem of immense practical and theoretical relevance. Here we develop a framework based on statistical mechanics and generative artificial intelligence that allows solving this problem. Specifically, we work with denoising diffusion probabilistic models and show how these models in combination with replica exchange molecular dynamics achieve superior sampling of the biomolecular energy landscape at temperatures that were never simulated without assuming any particular slow degrees of freedom. The key idea is to treat the temperature as a fluctuating random variable and not a control parameter as is usually done. This allows us to directly sample from the joint probability distribution in configuration and temperature space. The results here are demonstrated for a chirally symmetric peptide and single-strand RNA undergoing conformational transitions in all-atom water. We demonstrate how we can discover transition states and metastable states that were previously unseen at the temperature of interest and even bypass the need to perform further simulations for a wide range of temperatures. At the same time, any unphysical states are easily identifiable through very low Boltzmann weights. The procedure while shown here for a class of molecular simulations should be more generally applicable to mixing information across simulations and experiments with varying control parameters.


Assuntos
Inteligência Artificial , Simulação de Dinâmica Molecular , Peptídeos , RNA , Temperatura , Peptídeos/química , Física , RNA/química
7.
Angew Chem Int Ed Engl ; 61(28): e202200983, 2022 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-35486370

RESUMO

Understanding how mutations render a drug ineffective is a problem of immense relevance. Often the mechanism through which mutations cause drug resistance can be explained purely through thermodynamics. However, the more perplexing situation is when two proteins have the same drug binding affinities but different residence times. In this work, we demonstrate how all-atom molecular dynamics simulations using recent developments grounded in statistical mechanics can provide a detailed mechanistic rationale for such variances. We discover dissociation mechanisms for the anti-cancer drug Imatinib (Gleevec) against wild-type and the N368S mutant of Abl kinase. We show how this point mutation triggers far-reaching changes in the protein's flexibility and leads to a different, much faster, drug dissociation pathway. We believe that this work marks an efficient and scalable approach to obtain mechanistic insight into resistance mutations in biomolecular receptors that are hard to explain using a structural perspective.


Assuntos
Benzamidas , Piperazinas , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Fusão bcr-abl/metabolismo , Mesilato de Imatinib/farmacologia , Mutação , Piperazinas/química , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/química
8.
J Chem Theory Comput ; 18(5): 3231-3238, 2022 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-35384668

RESUMO

An effective implementation of enhanced sampling algorithms for molecular dynamics simulations requires a priori knowledge of the approximate reaction coordinate describing the relevant mechanisms in the system. In this work, we focus on the recently developed artificial intelligence-based State Predictive Information Bottleneck (SPIB) approach and demonstrate how SPIB can learn such a reaction coordinate as a deep neural network even from undersampled trajectories. We exemplify its usefulness by achieving more than 40 times acceleration in simulating two model biophysical systems through well-tempered metadynamics performed by biasing along the SPIB-learned reaction coordinate. These include left- to right-handed chirality transitions in a synthetic helical peptide (Aib)9 and permeation of a small benzoic acid molecule through a synthetic, symmetric phospholipid bilayer. In addition to significantly accelerating the dynamics and achieving back and forth movement between different metastable states, the SPIB-based reaction coordinate gives mechanistic insights into the processes driving these two important problems.


Assuntos
Inteligência Artificial , Simulação de Dinâmica Molecular , Peptídeos , Fosfolipídeos , Termodinâmica
9.
J Phys Chem B ; 125(40): 11150-11158, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34586819

RESUMO

Molecular dynamics (MD) simulations provide a wealth of high-dimensional data at all-atom and femtosecond resolution but deciphering mechanistic information from this data is an ongoing challenge in physical chemistry and biophysics. Theoretically speaking, joint probabilities of the equilibrium distribution contain all thermodynamic information, but they prove increasingly difficult to compute and interpret as the dimensionality increases. Here, inspired by tools in probabilistic graphical modeling, we develop a factor graph trained through belief propagation that helps factorize the joint probability into an approximate tractable form that can be easily visualized and used. We validate the study through the analysis of the conformational dynamics of two small peptides with five and nine residues. Our validations include testing the conditional dependency predictions through an intervention scheme inspired by Judea Pearl. Second, we directly use the belief propagation-based approximate probability distribution as a high-dimensional static bias for enhanced sampling, where we achieve spontaneous back-and-forth motion between metastable states that is up to 350 times faster than unbiased MD. We believe this work opens up useful ways to thinking about and dealing with high-dimensional molecular simulations.


Assuntos
Simulação de Dinâmica Molecular , Peptídeos , Movimento (Física) , Probabilidade , Termodinâmica
10.
J Chem Phys ; 153(23): 234118, 2020 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-33353347

RESUMO

Artificial intelligence (AI)-based approaches have had indubitable impact across the sciences through the ability to extract relevant information from raw data. Recently, AI has also found use in enhancing the efficiency of molecular simulations, wherein AI derived slow modes are used to accelerate the simulation in targeted ways. However, while typical fields where AI is used are characterized by a plethora of data, molecular simulations, per construction, suffer from limited sampling and thus limited data. As such, the use of AI in molecular simulations can suffer from a dangerous situation where the AI-optimization could get stuck in spurious regimes, leading to incorrect characterization of the reaction coordinate (RC) for the problem at hand. When such an incorrect RC is then used to perform additional simulations, one could start to deviate progressively from the ground truth. To deal with this problem of spurious AI-solutions, here, we report a novel and automated algorithm using ideas from statistical mechanics. It is based on the notion that a more reliable AI-solution will be one that maximizes the timescale separation between slow and fast processes. To learn this timescale separation even from limited data, we use a maximum caliber-based framework. We show the applicability of this automatic protocol for three classic benchmark problems, namely, the conformational dynamics of a model peptide, ligand-unbinding from a protein, and folding/unfolding energy landscape of the C-terminal domain of protein G. We believe that our work will lead to increased and robust use of trustworthy AI in molecular simulations of complex systems.


Assuntos
Inteligência Artificial , Modelos Estatísticos , Simulação de Dinâmica Molecular , Peptídeos/química , Proteínas/química , Entropia , Ligantes , Conformação Proteica
11.
Sci Adv ; 3(5): e1700014, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28580424

RESUMO

Obtaining atomistic resolution of drug unbinding from a protein is a much sought-after experimental and computational challenge. We report the unbinding dynamics of the anticancer drug dasatinib from c-Src kinase in full atomistic resolution using enhanced sampling molecular dynamics simulations. We obtain multiple unbinding trajectories and determine a residence time in agreement with experiments. We observe coupled protein-water movement through multiple metastable intermediates. The water molecules form a hydrogen bond bridge, elongating a specific, evolutionarily preserved salt bridge and enabling conformation changes essential to ligand unbinding. This water insertion in the salt bridge acts as a molecular switch that controls unbinding. Our findings provide a mechanistic rationale for why it might be difficult to engineer drugs targeting certain specific c-Src kinase conformations to have longer residence times.


Assuntos
Antineoplásicos/química , Dasatinibe/química , Proteínas de Neoplasias , Inibidores de Proteínas Quinases/química , Quinases da Família src , Sítios de Ligação , Proteína Tirosina Quinase CSK , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/química
12.
Proc Natl Acad Sci U S A ; 113(11): 2839-44, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929365

RESUMO

In modern-day simulations of many-body systems, much of the computational complexity is shifted to the identification of slowly changing molecular order parameters called collective variables (CVs) or reaction coordinates. A vast array of enhanced-sampling methods are based on the identification and biasing of these low-dimensional order parameters, whose fluctuations are important in driving rare events of interest. Here, we describe a new algorithm for finding optimal low-dimensional CVs for use in enhanced-sampling biasing methods like umbrella sampling, metadynamics, and related methods, when limited prior static and dynamic information is known about the system, and a much larger set of candidate CVs is specified. The algorithm involves estimating the best combination of these candidate CVs, as quantified by a maximum path entropy estimate of the spectral gap for dynamics viewed as a function of that CV. The algorithm is called spectral gap optimization of order parameters (SGOOP). Through multiple practical examples, we show how this postprocessing procedure can lead to optimization of CV and several orders of magnitude improvement in the convergence of the free energy calculated through metadynamics, essentially giving the ability to extract useful information even from unsuccessful metadynamics runs.


Assuntos
Algoritmos , Modelos Teóricos , Entropia , Modelos Moleculares , Simulação de Dinâmica Molecular , Peptídeos/química
13.
J Am Chem Soc ; 138(13): 4608-15, 2016 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-26954686

RESUMO

Mutations in the gatekeeper residue of kinases have emerged as a key way through which cancer cells develop resistance to treatment. As such, the design of gatekeeper mutation resistant kinase inhibitors is a crucial way forward in increasing the efficacy of a broad range of anticancer drugs. In this work we use atomistic simulations to provide detailed thermodynamic and structural insight into how two inhibitors of cSrc kinase, namely, a commercial drug and type I kinase inhibitor Dasatinib and the type II inhibitor RL45, respectively fail and succeed in being effective against the T338M gatekeeper residue mutation in the kinase binding site. Given the well-known limitations of atomistic simulations in sampling biomolecular systems, we use an enhanced sampling technique called free energy perturbation with replica exchange solute tempering (FEP/REST). Our calculations find that the type I inhibitor Dasatinib binds favorably to the wild type but unfavorably to T338M mutated kinase, while RL45 binds favorably to both. The predicted relative binding free energies are well within 1 kcal/mol accuracy compared to experiments. We find that Dasatinib's impotency against gatekeeper residue mutations arises from a loss of ligand-kinase hydrogen bonding due to T338M mutation and from steric hindrance due to the presence of an inflexible phenyl ring close to the ligand. On the other hand, in the type II binding RL45, the central phenyl ring has very pronounced flexibility. This leads to the inhibitor overcoming effects of steric clashes on mutation and maintaining an electrostatically favorable "edge-to-face" orientation with a neighboring phenylalanine residue. Our work provides useful insight into the mechanisms of mutation resistant kinase inhibitors and demonstrates the usefulness of enhanced sampling techniques in computational drug design.


Assuntos
Antineoplásicos/farmacologia , Modelos Químicos , Inibidores de Proteínas Quinases/farmacologia , Quinases da Família src/genética , Sítios de Ligação , Proteína Tirosina Quinase CSK , Simulação por Computador , Desenho de Fármacos , Humanos , Ligantes , Mutação , Ligação Proteica , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA