Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 74
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cell Calcium ; 123: 102947, 2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39226841

RÉSUMÉ

S100A1, a calcium-binding protein, plays a crucial role in regulating Ca2+ signaling pathways in skeletal and cardiac myocytes via interactions with the ryanodine receptor (RyR) to affect Ca2+ release and contractile performance. Biophysical studies strongly suggest that S100A1 interacts with RyRs but have been inconclusive about both the nature of this interaction and its competition with another important calcium-binding protein, calmodulin (CaM). Thus, high-resolution cryo-EM studies of RyRs in the presence of S100A1, with or without additional CaM, were needed. The elegant work by Weninger et al. demonstrates the interaction between S100A1 and RyR1 through various experiments and confirms that S100A1 activates RyR1 at sub-micromolar Ca2+ concentrations, increasing the open probability of RyR1 channels.

2.
Elife ; 132024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39240197

RÉSUMÉ

Small-molecule drug design hinges on obtaining co-crystallized ligand-protein structures. Despite AlphaFold2's strides in protein native structure prediction, its focus on apo structures overlooks ligands and associated holo structures. Moreover, designing selective drugs often benefits from the targeting of diverse metastable conformations. Therefore, direct application of AlphaFold2 models in virtual screening and drug discovery remains tentative. Here, we demonstrate an AlphaFold2-based framework combined with all-atom enhanced sampling molecular dynamics and Induced Fit docking, named AF2RAVE-Glide, to conduct computational model-based small-molecule binding of metastable protein kinase conformations, initiated from protein sequences. We demonstrate the AF2RAVE-Glide workflow on three different mammalian protein kinases and their type I and II inhibitors, with special emphasis on binding of known type II kinase inhibitors which target the metastable classical DFG-out state. These states are not easy to sample from AlphaFold2. Here, we demonstrate how with AF2RAVE these metastable conformations can be sampled for different kinases with high enough accuracy to enable subsequent docking of known type II kinase inhibitors with more than 50% success rates across docking calculations. We believe the protocol should be deployable for other kinases and more proteins generally.


Sujet(s)
Découverte de médicament , Conformation des protéines , Découverte de médicament/méthodes , Simulation de docking moléculaire , Liaison aux protéines , Simulation de dynamique moléculaire , Humains , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Ligands , Protein kinases/composition chimique , Protein kinases/métabolisme
3.
Chem Sci ; 2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39234215

RÉSUMÉ

In this work we examine the nucleation from NaCl aqueous solutions within nano-confined environments, employing enhanced sampling molecular dynamics simulations integrated with machine learning-derived reaction coordinates. Through our simulations, we successfully induce phase transitions between solid, liquid, and a hydrated phase, typically observed at lower temperatures in bulk environments. Interestingly, while generally speaking nano-confinement serves to stabilize the solid phase and elevate melting points, there are subtle variations in the thermodynamics of competing phases with the precise extent of confinement. Our simulations explain these findings by underscoring the significant role of water, alongside ion aggregation and subtle, anisotropic dielectric behavior, in driving nucleation within nano-confined environments. This report thus provides a framework for sampling, analyzing and understanding nucleation processes under nano-confinement.

4.
J Phys Chem B ; 128(34): 8207-8214, 2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39163635

RÉSUMÉ

We investigate crystal nucleation in supersaturated colloid suspensions using enhanced molecular dynamics simulations augmented with machine learning techniques. The simulations reveal that crystallization in the model colloidal system studied here, with particles interacting through a repulsive screened Coulomb Yukawa potential, proceeds from vapor to dense liquid droplet to crystalline phases across multiple high barriers. Employing a one-dimensional reaction coordinate derived from the State Predictive Information Bottleneck framework, our simulations capture back-and-forth phase transitions across multiple barriers effectively in biased metadynamics simulations. We obtain relative free energy differences between different phases and also quantify the roles of different molecular level features in driving the phase changes.

6.
J Chem Theory Comput ; 20(14): 6341-6349, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38991145

RÉSUMÉ

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.


Sujet(s)
Simulation de dynamique moléculaire , Protéines , Ligands , Protéines/composition chimique , Protéines/métabolisme , Mésilate d'imatinib/composition chimique , Algorithmes , Liaison aux protéines
7.
ArXiv ; 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38947932

RÉSUMÉ

Markov state models (MSMs) have proven valuable in studying dynamics of protein conformational changes via statistical analysis of molecular dynamics (MD) simulations. In MSMs, the complex configuration space is coarse-grained into conformational states, with dynamics modeled by a series of Markovian transitions among these states at discrete lag times. Constructing the Markovian model at a specific lag time necessitates defining states that circumvent significant internal energy barriers, enabling internal dynamics relaxation within the lag time. This process effectively coarse-grains time and space, integrating out rapid motions within metastable states. Thus, MSMs possess a multi-resolution nature, where the granularity of states can be adjusted according to the time-resolution, offering flexibility in capturing system dynamics. This work introduces a continuous embedding approach for molecular conformations using the state predictive information bottleneck (SPIB), a framework that unifies dimensionality reduction and state space partitioning via a continuous, machine learned basis set. Without explicit optimization of the VAMP-based scores, SPIB demonstrates state-of-the-art performance in identifying slow dynamical processes and constructing predictive multi-resolution Markovian models. Through applications to well-validated mini-proteins, SPIB showcases unique advantages compared to competing methods. It autonomously and self-consistently adjusts the number of metastable states based on specified minimal time resolution, eliminating the need for manual tuning. While maintaining efficacy in dynamical properties, SPIB excels in accurately distinguishing metastable states and capturing numerous well-populated macrostates. This contrasts with existing VAMP-based methods, which often emphasize slow dynamics at the expense of incorporating numerous sparsely populated states. Furthermore, SPIB's ability to learn a low-dimensional continuous embedding of the underlying MSMs enhances the interpretation of dynamic pathways. With these benefits, we propose SPIB as an easy-to-implement methodology for end-to-end MSMs construction.

8.
ArXiv ; 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38947925

RÉSUMÉ

The weighted ensemble (WE) method stands out as a widely used segment-based sampling technique renowned for its rigorous treatment of kinetics. The WE framework typically involves initially mapping the configuration space onto a low-dimensional collective variable (CV) space and then partitioning it into bins. The efficacy of WE simulations heavily depends on the selection of CVs and binning schemes. The recently proposed State Predictive Information Bottleneck (SPIB) method has emerged as a promising tool for automatically constructing CVs from data and guiding enhanced sampling through an iterative manner. In this work, we advance this data-driven pipeline by incorporating prior expert knowledge. Our hybrid approach combines SPIB-learned CVs to enhance sampling in explored regions with expert-based CVs to guide exploration in regions of interest, synergizing the strengths of both methods. Through benchmarking on alanine dipeptide and chignoin systems, we demonstrate that our hybrid approach effectively guides WE simulations to sample states of interest, and reduces run-to-run variances. Moreover, our integration of the SPIB model also enhances the analysis and interpretation of WE simulation data by effectively identifying metastable states and pathways, and offering direct visualization of dynamics.

9.
J Chem Theory Comput ; 20(12): 5352-5367, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38859575

RÉSUMÉ

Markov state models (MSMs) have proven valuable in studying the dynamics of protein conformational changes via statistical analysis of molecular dynamics simulations. In MSMs, the complex configuration space is coarse-grained into conformational states, with dynamics modeled by a series of Markovian transitions among these states at discrete lag times. Constructing the Markovian model at a specific lag time necessitates defining states that circumvent significant internal energy barriers, enabling internal dynamics relaxation within the lag time. This process effectively coarse-grains time and space, integrating out rapid motions within metastable states. Thus, MSMs possess a multiresolution nature, where the granularity of states can be adjusted according to the time-resolution, offering flexibility in capturing system dynamics. This work introduces a continuous embedding approach for molecular conformations using the state predictive information bottleneck (SPIB), a framework that unifies dimensionality reduction and state space partitioning via a continuous, machine learned basis set. Without explicit optimization of the VAMP-based scores, SPIB demonstrates state-of-the-art performance in identifying slow dynamical processes and constructing predictive multiresolution Markovian models. Through applications to well-validated mini-proteins, SPIB showcases unique advantages compared to competing methods. It autonomously and self-consistently adjusts the number of metastable states based on a specified minimal time resolution, eliminating the need for manual tuning. While maintaining efficacy in dynamical properties, SPIB excels in accurately distinguishing metastable states and capturing numerous well-populated macrostates. This contrasts with existing VAMP-based methods, which often emphasize slow dynamics at the expense of incorporating numerous sparsely populated states. Furthermore, SPIB's ability to learn a low-dimensional continuous embedding of the underlying MSMs enhances the interpretation of dynamic pathways. With these benefits, we propose SPIB as an easy-to-implement methodology for end-to-end MSM construction.


Sujet(s)
Chaines de Markov , Simulation de dynamique moléculaire , Protéines/composition chimique , Conformation des protéines
10.
Proc Natl Acad Sci U S A ; 121(23): e2408742121, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38809708
11.
ArXiv ; 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38659642

RÉSUMÉ

Small molecule drug design hinges on obtaining co-crystallized ligand-protein structures. Despite AlphaFold2's strides in protein native structure prediction, its focus on apo structures overlooks ligands and associated holo structures. Moreover, designing selective drugs often benefits from the targeting of diverse metastable conformations. Therefore, direct application of AlphaFold2 models in virtual screening and drug discovery remains tentative. Here, we demonstrate an AlphaFold2 based framework combined with all-atom enhanced sampling molecular dynamics and induced fit docking, named AF2RAVE-Glide, to conduct computational model based small molecule binding of metastable protein kinase conformations, initiated from protein sequences. We demonstrate the AF2RAVE-Glide workflow on three different protein kinases and their type I and II inhibitors, with special emphasis on binding of known type II kinase inhibitors which target the metastable classical DFG-out state. These states are not easy to sample from AlphaFold2. Here we demonstrate how with AF2RAVE these metastable conformations can be sampled for different kinases with high enough accuracy to enable subsequent docking of known type II kinase inhibitors with more than 50% success rates across docking calculations. We believe the protocol should be deployable for other kinases and more proteins generally.

12.
bioRxiv ; 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38659748

RÉSUMÉ

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.

13.
J Chem Theory Comput ; 20(9): 3503-3513, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38649368

RÉSUMÉ

While representation learning has been central to the rise of machine learning and artificial intelligence, a key problem remains in making the learned representations meaningful. For this, the typical approach is to regularize the learned representation through prior probability distributions. However, such priors are usually unavailable or are ad hoc. To deal with this, recent efforts have shifted toward leveraging the insights from physical principles to guide the learning process. In this spirit, we propose a purely dynamics-constrained representation learning framework. Instead of relying on predefined probabilities, we restrict the latent representation to follow overdamped Langevin dynamics with a learnable transition density─a prior driven by statistical mechanics. We show that this is a more natural constraint for representation learning in stochastic dynamical systems, with the crucial ability to uniquely identify the ground truth representation. We validate our framework for different systems including a real-world fluorescent DNA movie data set. We show that our algorithm can uniquely identify orthogonal, isometric, and meaningful latent representations.

14.
J Chem Inf Model ; 64(7): 2637-2644, 2024 04 08.
Article de Anglais | MEDLINE | ID: mdl-38453912

RÉSUMÉ

Identifying and discovering druggable protein binding sites is an important early step in computer-aided drug discovery, but it remains a difficult task where most campaigns rely on a priori knowledge of binding sites from experiments. Here, we present a binding site prediction method called Graph Attention Site Prediction (GrASP) and re-evaluate assumptions in nearly every step in the site prediction workflow from data set preparation to model evaluation. GrASP is able to achieve state-of-the-art performance at recovering binding sites in PDB structures while maintaining a high degree of precision which will minimize wasted computation in downstream tasks such as docking and free energy perturbation.


Sujet(s)
Agents antiVIH , Sites de fixation , Découverte de médicament , , Force de la main
15.
J Phys Chem B ; 128(12): 3037-3045, 2024 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-38502931

RÉSUMÉ

In this study, we present a graph neural network (GNN)-based learning approach using an autoencoder setup to derive low-dimensional variables from features observed in experimental crystal structures. These variables are then biased in enhanced sampling to observe state-to-state transitions and reliable thermodynamic weights. In our approach, we used simple convolution and pooling methods. To verify the effectiveness of our protocol, we examined the nucleation of various allotropes and polymorphs of iron and glycine in their molten states. Our graph latent variables, when biased in well-tempered metadynamics, consistently show transitions between states and achieve accurate thermodynamic rankings in agreement with experiments, both of which are indicators of dependable sampling. This underscores the strength and promise of our GNN variables for improved sampling. The protocol shown here should be applicable for other systems and other sampling methods.

16.
Annu Rev Phys Chem ; 75(1): 347-370, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38382572

RÉSUMÉ

Molecular dynamics (MD) enables the study of physical systems with excellent spatiotemporal resolution but suffers from severe timescale limitations. To address this, enhanced sampling methods have been developed to improve the exploration of configurational space. However, implementing these methods is challenging and requires domain expertise. In recent years, integration of machine learning (ML) techniques into different domains has shown promise, prompting their adoption in enhanced sampling as well. Although ML is often employed in various fields primarily due to its data-driven nature, its integration with enhanced sampling is more natural with many common underlying synergies. This review explores the merging of ML and enhanced MD by presenting different shared viewpoints. It offers a comprehensive overview of this rapidly evolving field, which can be difficult to stay updated on. We highlight successful strategies such as dimensionality reduction, reinforcement learning, and flow-based methods. Finally, we discuss open problems at the exciting ML-enhanced MD interface.

17.
J Phys Chem B ; 128(3): 755-767, 2024 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-38205806

RÉSUMÉ

Ligand unbinding is mediated by its free energy change, which has intertwined contributions from both energy and entropy. It is important, but not easy, to quantify their individual contributions to the free energy profile. We model hydrophobic ligand unbinding for two systems, a methane particle and a C60 fullerene, both unbinding from hydrophobic pockets in all-atom water. Using a modified deep learning framework, we learn a thermodynamically optimized reaction coordinate to describe the hydrophobic ligand dissociation for both systems. Interpretation of these reaction coordinates reveals the roles of entropic and enthalpic forces as the ligand and pocket sizes change. In both cases, we observe that the free-energy barrier to unbinding is dominated by entropy considerations. Furthermore, the process of methane unbinding is driven by methane solvation, while fullerene unbinding is driven first by pocket wetting and then fullerene wetting. For both solutes, the direct importance of the distance from the binding pocket to the learned reaction coordinate is present, but low. Our framework and subsequent feature important analysis thus give useful thermodynamic insight into hydrophobic ligand dissociation problems that are otherwise difficult to glean.

18.
J Phys Chem B ; 128(4): 1012-1021, 2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38262436

RÉSUMÉ

Even though nucleation is ubiquitous in different science and engineering problems, investigating nucleation is extremely difficult due to the complicated ranges of time and length scales involved. In this work, we simulate NaCl nucleation in both molten and aqueous environments using enhanced sampling of all-atom molecular dynamics with deep-learning-based estimation of reaction coordinates. By incorporating various structural order parameters and learning the reaction coordinate as a function thereof, we achieve significantly improved sampling relative to traditional ad hoc descriptions of what drives nucleation, particularly in an aqueous medium. Our results reveal a one-step nucleation mechanism in both environments, with reaction coordinate analysis highlighting the importance of local ion density in distinguishing solid and liquid states. However, although fluctuations in the local ion density are necessary to drive nucleation, they are not sufficient. Our analysis shows that near the transition states, descriptors such as enthalpy and local structure become crucial. Our protocol proposed here enables robust nucleation analysis and phase sampling and could offer insights into nucleation mechanisms for generic small molecules in different environments.

19.
J Chem Inf Model ; 64(7): 2789-2797, 2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-37981824

RÉSUMÉ

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).


Sujet(s)
Oligopeptides , Inhibiteurs de protéines kinases , Humains , Modèles moléculaires , Inhibiteurs de protéines kinases/composition chimique , Conformation des protéines , Oligopeptides/composition chimique
20.
J Chem Theory Comput ; 19(24): 9093-9101, 2023 Dec 26.
Article de Anglais | MEDLINE | ID: mdl-38084039

RÉSUMÉ

Understanding nucleation from aqueous solutions is of fundamental importance in a multitude of fields, ranging from materials science to biophysics. The complex solvent-mediated interactions in aqueous solutions hamper the development of a simple physical picture, elucidating the roles of different interactions in nucleation processes. In this work, we make use of three complementary techniques to disentangle the role played by short- and long-range interactions in solvent-mediated nucleation. Specifically, the first approach we utilize is the local molecular field (LMF) theory to renormalize long-range Coulomb electrostatics. Second, we use well-tempered metadynamics to speed up rare events governed by short-range interactions. Third, the deep learning-based State Predictive Information Bottleneck approach is employed in analyzing the reaction coordinate of the nucleation processes obtained from the LMF treatment coupled with well-tempered metadynamics. We find that the two-step nucleation mechanism can largely be captured by the short-range interactions, while the long-range interactions further contribute to the stability of the primary crystal state under ambient conditions. Furthermore, by analyzing the reaction coordinate obtained from the combined LMF-metadynamics treatment, we discern the fluctuations on different time scales, highlighting the need for long-range interactions when accounting for metastability.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE