Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
J Chem Inf Model ; 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717640

ABSTRACT

Accurate force field parameters, potential energy functions, and receptor-ligand models are essential for modeling the solvation and binding of drug-like molecules to a receptor. A large and ever-growing chemical space of medicinally relevant scaffolds has also required these factors, especially force field parameters, to be highly transferable. Generalized force fields such as the CHARMM General Force Field (CGenFF) and the generalized AMBER force field (GAFF) have accomplished this feat along with other contemporaneous ones like OPLS. Here, we analyze the limits in the parametrization of drug-like small molecules by CGenFF and GAFF in terms of the various functional groups represented within them. Specifically, we link the presence of specific functional groups to the error in the absolute hydration free energy of over 600 small molecules, predicted by alchemical free energy methods implemented in the CHARMM program. Our investigation reveals that molecules with (i) a nitro group in CGenFF and GAFF are, respectively, over- or undersolubilized in aqueous medium, (ii) amine groups are undersolubilized more so in CGenFF than in GAFF, and (iii) carboxyl groups are more oversolubilized in GAFF than in CGenFF. We present our analyses of the potential factors underlying these trends. We also showcase the use of a machine-learning-based approach combined with the SHapley Additive exPlanations framework to attribute these trends to specific functional groups, which can be easily adopted to explore the limits of other general force fields.

2.
J Am Chem Soc ; 146(4): 2728-2735, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38237569

ABSTRACT

3-Hydroxyindolenines can be used to access several structural motifs that are featured in natural products and pharmaceutical compounds, yet the chemical synthesis of 3-hydroxyindolenines is complicated by overoxidation, rearrangements, and complex product mixtures. The selectivity possible in enzymatic reactions can overcome these challenges and deliver enantioenriched products. Herein, we present the development of an asymmetric biocatalytic oxidation of 2-arylindole substrates aided by a curated library of flavin-dependent monooxygenases (FDMOs) sampled from an ancestral sequence space, a sequence similarity network, and a deep-learning-based latent space model. From this library of FDMOs, a previously uncharacterized enzyme, Champase, from the Valley fever fungus, Coccidioides immitis strain RS, was found to stereoselectively catalyze the oxidation of a variety of substituted indole substrates. The promiscuity of this enzyme is showcased by the oxidation of a wide variety of substituted 2-arylindoles to afford the respective 3-hydroxyindolenine products in moderate to excellent yields and up to 95:5 er.


Subject(s)
Biological Products , Mixed Function Oxygenases , Oxidation-Reduction , Mixed Function Oxygenases/chemistry , Biocatalysis , Catalysis
3.
Proc Natl Acad Sci U S A ; 121(3): e2312029121, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38194446

ABSTRACT

Understanding natural protein evolution and designing novel proteins are motivating interest in development of high-throughput methods to explore large sequence spaces. In this work, we demonstrate the application of multisite λ dynamics (MSλD), a rigorous free energy simulation method, and chemical denaturation experiments to quantify evolutionary selection pressure from sequence-stability relationships and to address questions of design. This study examines a mesophilic phylogenetic clade of ribonuclease H (RNase H), furthering its extensive characterization in earlier studies, focusing on E. coli RNase H (ecRNH) and a more stable consensus sequence (AncCcons) differing at 15 positions. The stabilities of 32,768 chimeras between these two sequences were computed using the MSλD framework. The most stable and least stable chimeras were predicted and tested along with several other sequences, revealing a designed chimera with approximately the same stability increase as AncCcons, but requiring only half the mutations. Comparing the computed stabilities with experiment for 12 sequences reveals a Pearson correlation of 0.86 and root mean squared error of 1.18 kcal/mol, an unprecedented level of accuracy well beyond less rigorous computational design methods. We then quantified selection pressure using a simple evolutionary model in which sequences are selected according to the Boltzmann factor of their stability. Selection temperatures from 110 to 168 K are estimated in three ways by comparing experimental and computational results to evolutionary models. These estimates indicate selection pressure is high, which has implications for evolutionary dynamics and for the accuracy required for design, and suggests accurate high-throughput computational methods like MSλD may enable more effective protein design.


Subject(s)
Escherichia coli , Ribonuclease H , Escherichia coli/genetics , Phylogeny , Computer Simulation , Consensus Sequence , Ribonuclease H/genetics
4.
J Chem Inf Model ; 64(3): 621-626, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38276895

ABSTRACT

Using a combination of multisite λ-dynamics (MSλD) together with in vitro IC50 assays, we evaluated the polypharmacological potential of a scaffold currently in clinical trials for inhibition of human neutrophil elastase (HNE), targeting cardiopulmonary disease, for efficacious inhibition of Proteinase 3 (PR3), a related neutrophil serine proteinase. The affinities we observe suggest that the dihydropyrimidinone scaffold can serve as a suitable starting point for the establishment of polypharmacologically targeting both enzymes and enhancing the potential for treatments addressing diseases like chronic obstructive pulmonary disease.


Subject(s)
Polypharmacology , Humans , Myeloblastin , Proteinase Inhibitory Proteins, Secretory
5.
Biochim Biophys Acta Gen Subj ; 1868(2): 130534, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38065235

ABSTRACT

The relevance of motions in biological macromolecules has been clear since the early structural analyses of proteins by X-ray crystallography. Computer simulations have been applied to provide a deeper understanding of the dynamics of biological macromolecules since 1976, and are now a standard tool in many labs working on the structure and function of biomolecules. In this mini-review we highlight some areas of current interest and active development for simulations, in particular all-atom molecular dynamics simulations.


Subject(s)
Molecular Dynamics Simulation , Proteins , Proteins/chemistry , Crystallography, X-Ray
6.
J Chem Inf Model ; 63(22): 7219-7227, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37939386

ABSTRACT

Allostery is involved in innumerable biological processes and plays a fundamental role in human disease. Thus, the exploration of allosteric modulation is crucial for research on biological mechanisms and in the development of novel therapeutics. The development of small-molecule allosteric effectors can be used as tools to probe biological mechanisms of interest. One of the main limitations in targeting allosteric sites is the difficulty in uncovering them for specific receptors. Furthermore, upon discovery of novel allosteric modulation, early lead generation is made more difficult as compared to that at orthosteric sites because there is likely no information about the types of molecules that can bind at the site. In the work described here, we present a novel drug discovery pipeline, FASTDock, which allows one to uncover ligandable sites as well as small molecules that target the given site without requiring pre-existing knowledge of ligands that can bind in the targeted site. By using a hierarchical screening strategy, this method has the potential to enable high-throughput screens of an exceptionally large database of targeted ligand space.


Subject(s)
Drug Discovery , Humans , Allosteric Regulation , Allosteric Site , Drug Discovery/methods , Ligands
7.
ACS Med Chem Lett ; 14(6): 860-866, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37284689

ABSTRACT

The COVID-19 pandemic has highlighted the need for new antiviral approaches because many of the currently approved drugs have proven ineffective against mitigating SARS-CoV-2 infections. The host transmembrane serine protease TMPRSS2 is a promising antiviral target because it plays a role in priming the spike protein before viral entry occurs for the most virulent variants. Further, TMPRSS2 has no established physiological role, thereby increasing its attractiveness as a target for antiviral agents. Here, we utilize virtual screening to curate large libraries into a focused collection of potential inhibitors. Optimization of a recombinant expression and purification protocol for the TMPRSS2 peptidase domain facilitates subsequent biochemical screening and characterization of selected compounds from the curated collection in a kinetic assay. In doing so, we identify new noncovalent TMPRSS2 inhibitors that block SARS-CoV-2 infectivity in a cellular model. One such inhibitor, debrisoquine, has high ligand efficiency, and an initial structure-activity relationship study demonstrates that debrisoquine is a tractable hit compound for TMPRSS2.

8.
J Chem Theory Comput ; 19(12): 3752-3762, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37267404

ABSTRACT

CHARMM is rich in methodology and functionality as one of the first programs addressing problems of molecular dynamics and modeling of biological macromolecules and their partners, e.g., small molecule ligands. When combined with the highly developed CHARMM parameters for proteins, nucleic acids, small molecules, lipids, sugars, and other biologically relevant building blocks, and the versatile CHARMM scripting language, CHARMM has been a trendsetting platform for modeling studies of biological macromolecules. To further enhance the utility of accessing and using CHARMM functionality in increasingly complex workflows associated with modeling biological systems, we introduce pyCHARMM, Python bindings, functions, and modules to complement and extend the extensive set of modeling tools and methods already available in CHARMM. These include access to CHARMM function-generated variables associated with the system (psf), coordinates, velocities and forces, atom selection variables, and force field related parameters. The ability to augment CHARMM forces and energies with energy terms or methods derived from machine learning or other sources, written in Python, CUDA, or OpenCL and expressed as Python callable routines is introduced together with analogous functions callable during dynamics calculations. Integration of Python-based graphical engines for visualization of simulation models and results is also accessible. Loosely coupled parallelism is available for workflows such as free energy calculations, using MBAR/TI approaches or high-throughput multisite λ-dynamics (MSλD) free energy methods, string path optimization calculations, replica exchange, and molecular docking with a new Python-based CDOCKER module. CHARMM accelerated platform kernels through the CHARMM/OpenMM API, CHARMM/DOMDEC, and CHARMM/BLaDE API are also readily integrated into this Python framework. We anticipate that pyCHARMM will be a robust platform for the development of comprehensive and complex workflows utilizing Python and its extensive functionality as well as an optimal platform for users to learn molecular modeling methods and practices within a Python-friendly environment such as Jupyter Notebooks.


Subject(s)
Molecular Dynamics Simulation , Nucleic Acids , Molecular Docking Simulation , Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 120(15): e2218248120, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37014851

ABSTRACT

Controlling the selectivity of a reaction is critical for target-oriented synthesis. Accessing complementary selectivity profiles enables divergent synthetic strategies, but is challenging to achieve in biocatalytic reactions given enzymes' innate preferences of a single selectivity. Thus, it is critical to understand the structural features that control selectivity in biocatalytic reactions to achieve tunable selectivity. Here, we investigate the structural features that control the stereoselectivity in an oxidative dearomatization reaction that is key to making azaphilone natural products. Crystal structures of enantiocomplementary biocatalysts guided the development of multiple hypotheses centered on the structural features that control the stereochemical outcome of the reaction; however, in many cases, direct substitutions of active site residues in natural proteins led to inactive enzymes. Ancestral sequence reconstruction (ASR) and resurrection were employed as an alternative strategy to probe the impact of each residue on the stereochemical outcome of the dearomatization reaction. These studies suggest that two mechanisms are active in controlling the stereochemical outcome of the oxidative dearomatization reaction: one involving multiple active site residues in AzaH and the other dominated by a single Phe to Tyr switch in TropB and AfoD. Moreover, this study suggests that the flavin-dependent monooxygenases (FDMOs) adopt simple and flexible strategies to control stereoselectivity, which has led to stereocomplementary azaphilone natural products produced by fungi. This paradigm of combining ASR and resurrection with mutational and computational studies showcases sets of tools for understanding enzyme mechanisms and provides a solid foundation for future protein engineering efforts.


Subject(s)
Biological Products , Mixed Function Oxygenases , Mixed Function Oxygenases/metabolism , Oxidation-Reduction , Flavins/metabolism , Proteins/metabolism , Biocatalysis , Organic Chemicals , Biological Products/chemistry
10.
Protein Sci ; 32(4): e4623, 2023 04.
Article in English | MEDLINE | ID: mdl-36906820

ABSTRACT

Multisite λ-dynamics (MSλD) is a novel method for the calculation of relative free energies of binding for ligands to their targeted receptors. It can be readily used to examine a large number of molecules with multiple functional groups at multiple sites around a common core. This makes MSλD a powerful tool in structure-based drug design. In the present study, MSλD is applied to calculate the relative binding free energies of 1296 inhibitors to the testis specific serine kinase 1B (TSSK1B), a validated target for male contraception. For this system, MSλD requires significantly fewer computational resources compared to traditional free energy methods like free energy perturbation or thermodynamic integration. From MSλD simulations, we examined whether modifications of a ligand at two different sites are coupled or not. Based on our calculations, we established a quantitative structure-activity relationship (QSAR) for this set of molecules and identified a site in the ligand where further modification, such as adding more polar groups, may lead to increased binding affinity.


Subject(s)
Molecular Dynamics Simulation , Quantitative Structure-Activity Relationship , Humans , Male , Entropy , Ligands , Protein Binding , Thermodynamics , Protein Serine-Threonine Kinases/metabolism
11.
J Phys Chem Lett ; 13(40): 9303-9308, 2022 Oct 13.
Article in English | MEDLINE | ID: mdl-36174129

ABSTRACT

A recently proposed lipid-chaperone hypothesis suggests that free lipid molecules, not bound to membranes, affect the aggregation of amyloidogenic peptides such as amyloid-ß (Aß) peptides, whose aggregates are the hallmarks of Alzheimer's disease. Here, we combine experiments with all-atom molecular dynamics simulations in explicit solvent to explore the effects of neuronal ganglioside GM1, abundant in mammalian brains, on the aggregation of two principal isoforms of Aß, Aß40 and Aß42. Our simulations show that free GM1 forms stable, highly water-soluble complexes with both isoforms, and nuclear magnetic resonance experiments support the formation of well-ordered, structurally compact GM1+Aß complexes. By simulation, we also show that Aß40 monomers display a preference for binding to GM1-containing hetero-oligomers over GM1-lacking homo-oligomers, while Aß42 monomers have the opposite preference. These observations explain why GM1 dose-dependently inhibits Aß40 aggregation but has no effect on Aß42 aggregation, as assessed by thioflavin T fluorescence.


Subject(s)
Alzheimer Disease , G(M1) Ganglioside , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Animals , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Gangliosides/metabolism , Mammals/metabolism , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Solvents , Water
12.
Proc Natl Acad Sci U S A ; 119(26): e2119686119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35737838

ABSTRACT

Allostery is the phenomenon of coupling between distal binding sites in a protein. Such coupling is at the crux of protein function and regulation in a myriad of scenarios, yet determining the molecular mechanisms of coupling networks in proteins remains a major challenge. Here, we report mechanisms governing pH-dependent myristoyl switching in monomeric hisactophilin, whereby the myristoyl moves between a sequestered state, i.e., buried within the core of the protein, to an accessible state, in which the myristoyl has increased accessibility for membrane binding. Measurements of the pH and temperature dependence of amide chemical shifts reveal protein local structural stability and conformational heterogeneity that accompany switching. An analysis of these measurements using a thermodynamic cycle framework shows that myristoyl-proton coupling at the single-residue level exists in a fine balance and extends throughout the protein. Strikingly, small changes in the stereochemistry or size of core and surface hydrophobic residues by point mutations readily break, restore, or tune myristoyl switch energetics. Synthesizing the experimental results with those of molecular dynamics simulations illuminates atomistic details of coupling throughout the protein, featuring a large network of hydrophobic interactions that work in concert with key electrostatic interactions. The simulations were critical for discerning which of the many ionizable residues in hisactophilin are important for switching and identifying the contributions of nonnative interactions in switching. The strategy of using temperature-dependent NMR presented here offers a powerful, widely applicable way to elucidate the molecular mechanisms of allostery in proteins at high resolution.


Subject(s)
Microfilament Proteins , Protozoan Proteins , Genes, Switch , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Signal Transduction , Static Electricity
13.
PLoS Comput Biol ; 18(4): e1009977, 2022 04.
Article in English | MEDLINE | ID: mdl-35452454

ABSTRACT

The coactivator KIX of CBP uses two binding surfaces to recognize multiple activators and exhibits allostery in ternary complex formation. Activator•coactivator interactions are central to transcriptional regulation, yet the microscopic origins of allostery in dynamic proteins like KIX are largely unknown. Here, we investigate the molecular recognition and allosteric manifestations involved in two KIX ternary systems c-Myb•KIX•MLL and pKID•KIX•MLL. Exploring the hypothesis that binary complex formation prepays an entropic cost for positive cooperativity, we utilize molecular dynamics simulations, side chain methyl order parameters, and differential scanning fluorimetry (DSF) to explore conformational entropy changes in KIX. The protein's configurational micro-states from structural clustering highlight the utility of protein plasticity in molecular recognition and allostery. We find that apo KIX occupies a wide distribution of lowly-populated configurational states. Each binding partner has its own suite of KIX states that it selects, building a model of molecular recognition fingerprints. Allostery is maximized with MLL pre-binding, which corresponds to the observation of a significant reduction in KIX micro-states observed when MLL binds. With all binding partners, the changes in KIX conformational entropy arise predominantly from changes in the most flexible loop. Likewise, we find that a small molecule and mutations allosterically inhibit/enhance activator binding by tuning loop dynamics, suggesting that loop-targeting chemical probes could be developed to alter KIX•activator interactions. Experimentally capturing KIX stabilization is challenging, particularly because of the disordered nature of particular activators. However, DSF melting curves allow for inference of relative entropic changes that occur across complexes, which we compare to our computed entropy changes using simulation methyl order parameters.


Subject(s)
CREB-Binding Protein , Molecular Dynamics Simulation , Binding Sites , CREB-Binding Protein/chemistry , CREB-Binding Protein/metabolism , Molecular Conformation , Protein Binding
14.
J Chem Inf Model ; 62(6): 1479-1488, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35286093

ABSTRACT

With the ability to sample combinations of alchemical perturbations at multiple sites off a small molecule core, multisite λ-dynamics (MSλD) has become an attractive alternative to conventional alchemical free energy methods for exploring large combinatorial chemical spaces. However, current software implementations dictate that combinatorial sampling with MSλD must be performed with a multiple topology model (MTM), which is nontrivial to create by hand, especially for a series of ligand analogues which may have diverse functional groups attached. This work introduces an automated workflow, referred to as msld_py_prep, to assist in the creation of a MTM for use with MSλD. One approach for partitioning partial atomic charges between ligands to create a MTM, called charge renormalization, is also presented and rigorously evaluated. We find that msld_py_prep greatly accelerates the preparation of MSλD ready-to-use files and that charge renormalization can provide a successful approach for MTM generation, as long as bookending calculations are applied to correct small differences introduced by charge renormalization. Charge renormalization also facilitates the use of many different force field parameters with MSλD, broadening the applicability of MSλD for computer-aided drug design.


Subject(s)
Drug Design , Molecular Dynamics Simulation , Entropy , Ligands , Thermodynamics
15.
J Chem Inf Model ; 62(6): 1458-1470, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35258972

ABSTRACT

Accurate and rapid predictions of the binding affinity of a compound to a target are one of the ultimate goals of computer aided drug design. Alchemical approaches to free energy estimations follow the path from an initial state of the system to the final state through alchemical changes of the energy function during a molecular dynamics simulation. Herein, we explore the accuracy and efficiency of two such techniques: relative free energy perturbation (FEP) and multisite lambda dynamics (MSλD). These are applied to a series of inhibitors for the bromodomain-containing protein 4 (BRD4). We demonstrate a procedure for obtaining accurate relative binding free energies using MSλD when dealing with a change in the net charge of the ligand. This resulted in an impressive comparison with experiment, with an average difference of 0.4 ± 0.4 kcal mol-1. In a benchmarking study for the relative FEP calculations, we found that using 20 lambda windows with 0.5 ns of equilibration and 1 ns of data collection for each window gave the optimal compromise between accuracy and speed. Overall, relative FEP and MSλD predicted binding free energies with comparable accuracy, an average of 0.6 kcal mol-1 for each method. However, MSλD makes predictions for a larger molecular space over a much shorter time scale than relative FEP, with MSλD requiring a factor of 18 times less simulation time for the entire molecule space.


Subject(s)
Nuclear Proteins , Transcription Factors , Entropy , Ligands , Molecular Dynamics Simulation , Protein Binding , Thermodynamics
16.
J Chem Theory Comput ; 18(4): 2114-2123, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35255214

ABSTRACT

Alchemical free energy methods are playing a growing role in molecular design, both for computer-aided drug design of small molecules and for computational protein design. Multisite λ dynamics (MSλD) is a uniquely scalable alchemical free energy method that enables more efficient exploration of combinatorial alchemical spaces encountered in molecular design, but simulations have typically been limited to a few hundred ligands or sequences. Here, we focus on coupling between sites to enable scaling to larger alchemical spaces. We first discuss updates to the biasing potentials that facilitate MSλD sampling to include coupling terms and show that this can provide more thorough sampling of alchemical states. We then harness coupling between sites by developing a new free energy estimator based on the Potts models underlying direct coupling analysis, a method for predicting contacts from sequence coevolution, and find it yields more accurate free energies than previous estimators. The sampling requirements of the Potts model estimator scale with the square of the number of sites, a substantial improvement over the exponential scaling of the standard estimator. This opens up exploration of much larger alchemical spaces with MSλD for molecular design.


Subject(s)
Molecular Dynamics Simulation , Proteins , Entropy , Ligands , Proteins/chemistry , Thermodynamics
17.
J Chem Theory Comput ; 17(11): 6799-6807, 2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34709046

ABSTRACT

There is an accelerating interest in practical applications of alchemical free energy methods to problems in protein design, constant pH simulations, and especially computer-aided drug design. In the present paper, we describe a basic lambda dynamics engine (BLaDE) that enables alchemical free energy simulations, including multisite λ dynamics (MSλD) simulations, on graphical processor units (GPUs). We find that BLaDE is 5 to 8 times faster than the current GPU implementation of MSλD-based free energy calculations in CHARMM. We also demonstrate that BLaDE running standard molecular dynamics attains a performance competitive with and sometimes exceeding that of the highly optimized OpenMM GPU code. BLaDE is available as a standalone program and through an API in CHARMM.

18.
J Chem Inf Model ; 61(11): 5535-5549, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34704754

ABSTRACT

The binding of small-molecule ligands to protein or nucleic acid targets is important to numerous biological processes. Accurate prediction of the binding modes between a ligand and a macromolecule is of fundamental importance in structure-based structure-function exploration. When multiple ligands with different sizes are docked to a target receptor, it is reasonable to assume that the residues in the binding pocket may adopt alternative conformations upon interacting with the different ligands. In addition, it has been suggested that the entropic contribution to binding can be important. However, only a few attempts to include the side chain conformational entropy upon binding within the application of flexible receptor docking methodology exist. Here, we propose a new physics-based scoring function that includes both enthalpic and entropic contributions upon binding by considering the conformational variability of the flexible side chains within the ensemble of docked poses. We also describe a novel hybrid searching algorithm that combines both molecular dynamics (MD)-based simulated annealing and genetic algorithm crossovers to address the enhanced sampling of the increased search space. We demonstrate improved accuracy in flexible cross-docking experiments compared with rigid cross-docking. We test our developments by considering five protein targets, thrombin, dihydrofolate reductase(DHFR), T4 L99A, T4 L99A/M102Q, and PDE10A, which belong to different enzyme classes with different binding pocket environments, as a representative set of diverse ligands and receptors. Each target contains dozens of different ligands bound to the same binding pocket. We also demonstrate that this flexible docking algorithm may be applicable to RNA docking with a representative riboswitch example. Our findings show significant improvements in top ranking accuracy across this set, with the largest improvement relative to rigid, 23.64%, occurring for ligands binding to DHFR. We then evaluate the ability to identify lead compounds among a large chemical space for the proposed flexible receptor docking algorithm using a subset of the DUD-E containing receptor targets MCR, GCR, and ANDR. We demonstrate that our new algorithms show improved performance in modeling flexible binding site residues compared to DOCK. Finally, we select the T4 L99A and T4 L99A/M102Q decoy sets, containing dozens of binders and experimentally validated nonbinders, to test our approach in distinguishing binders from nonbinders. We illustrate that our new algorithms for searching and scoring have superior performance to rigid receptor CDOCKER as well as AutoDock Vina. Finally, we suggest that flexible CDOCKER is sufficiently fast to be utilized in high-throughput docking screens in the context of hierarchical approaches.


Subject(s)
Algorithms , Binding Sites , Entropy , Ligands , Molecular Conformation , Molecular Docking Simulation , Protein Binding , Protein Conformation
19.
J Chem Theory Comput ; 17(7): 3895-3907, 2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34101448

ABSTRACT

In this work, the discrete λ variant of the Gibbs sampler-based λ-dynamics (d-GSλD) method is developed to enable multiple functional group perturbations to be investigated at one or more sites of substitution off a common ligand core. The theoretical framework and special considerations for constructing discrete λ states for multisite d-GSλD are presented. The precision and accuracy of the d-GSλD method is evaluated with three test cases of increasing complexity. Specifically, methyl → methyl symmetric perturbations in water, 1,4-benzene hydration free energies and protein-ligand binding affinities for an example HIV-1 reverse transcriptase inhibitor series are computed with d-GSλD. Complementary MSλD calculations were also performed to compare with d-GSλD's performance. Excellent agreement between d-GSλD and MSλD is observed, with mean unsigned errors of 0.12 and 0.22 kcal/mol for computed hydration and binding free energy test cases, respectively. Good agreement with experiment is also observed, with errors of 0.5-0.7 kcal/mol. These findings support the applicability of the d-GSλD free energy method for a variety of molecular design problems, including structure-based drug design. Finally, a discussion of d-GSλD versus MSλD approaches is presented to compare and contrast features of both methods.

20.
J Am Chem Soc ; 143(25): 9297-9302, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34137598

ABSTRACT

Inhibitors of transcriptional protein-protein interactions (PPIs) have high value both as tools and for therapeutic applications. The PPI network mediated by the transcriptional coactivator Med25, for example, regulates stress-response and motility pathways, and dysregulation of the PPI networks contributes to oncogenesis and metastasis. The canonical transcription factor binding sites within Med25 are large (∼900 Å2) and have little topology, and thus, they do not present an array of attractive small-molecule binding sites for inhibitor discovery. Here we demonstrate that the depsidone natural product norstictic acid functions through an alternative binding site to block Med25-transcriptional activator PPIs in vitro and in cell culture. Norstictic acid targets a binding site comprising a highly dynamic loop flanking one canonical binding surface, and in doing so, it both orthosterically and allosterically alters Med25-driven transcription in a patient-derived model of triple-negative breast cancer. These results highlight the potential of Med25 as a therapeutic target as well as the inhibitor discovery opportunities presented by structurally dynamic loops within otherwise challenging proteins.


Subject(s)
Lactones/pharmacology , Mediator Complex/metabolism , Protein Binding/drug effects , Salicylates/pharmacology , Transcription, Genetic/drug effects , Allosteric Regulation , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Mediator Complex/chemistry , Molecular Dynamics Simulation , Protein Domains , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...