Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(15): e2316662121, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38557187

RESUMO

Drug resistance in HIV type 1 (HIV-1) is a pervasive problem that affects the lives of millions of people worldwide. Although records of drug-resistant mutations (DRMs) have been extensively tabulated within public repositories, our understanding of the evolutionary kinetics of DRMs and how they evolve together remains limited. Epistasis, the interaction between a DRM and other residues in HIV-1 protein sequences, is key to the temporal evolution of drug resistance. We use a Potts sequence-covariation statistical-energy model of HIV-1 protein fitness under drug selection pressure, which captures epistatic interactions between all positions, combined with kinetic Monte-Carlo simulations of sequence evolutionary trajectories, to explore the acquisition of DRMs as they arise in an ensemble of drug-naive patient protein sequences. We follow the time course of 52 DRMs in the enzymes protease, RT, and integrase, the primary targets of antiretroviral therapy. The rates at which DRMs emerge are highly correlated with their observed acquisition rates reported in the literature when drug pressure is applied. This result highlights the central role of epistasis in determining the kinetics governing DRM emergence. Whereas rapidly acquired DRMs begin to accumulate as soon as drug pressure is applied, slowly acquired DRMs are contingent on accessory mutations that appear only after prolonged drug pressure. We provide a foundation for using computational methods to determine the temporal evolution of drug resistance using Potts statistical potentials, which can be used to gain mechanistic insights into drug resistance pathways in HIV-1 and other infectious agents.


Assuntos
Fármacos Anti-HIV , Infecções por HIV , Soropositividade para HIV , HIV-1 , Humanos , HIV-1/genética , Farmacorresistência Viral/genética , Genótipo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Mutação , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico
2.
bioRxiv ; 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38559238

RESUMO

Protein kinases are molecular machines with rich sequence variation that distinguishes the two main evolutionary branches - tyrosine kinases (TKs) from serine/threonine kinases (STKs). Using a sequence co-variation Potts statistical energy model we previously concluded that TK catalytic domains are more likely than STKs to adopt an inactive conformation with the activation loop in an autoinhibitory "folded" conformation, due to intrinsic sequence effects. Here we investigated the structural basis for this phenomenon by integrating the sequence-based model with structure-based molecular dynamics (MD) to determine the effects of mutations on the free energy difference between active and inactive conformations, using a novel thermodynamic cycle involving many (n=108) protein-mutation free energy perturbation (FEP) simulations in the active and inactive conformations. The sequence and structure-based results are consistent and support the hypothesis that the inactive conformation "DFG-out Activation Loop Folded", is a functional regulatory state that has been stabilized in TKs relative to STKs over the course of their evolution via the accumulation of residue substitutions in the activation loop and catalytic loop that facilitate distinct substrate binding modes in trans and additional modes of regulation in cis for TKs.

3.
J Phys Chem B ; 128(7): 1656-1667, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38350894

RESUMO

Single-point mutations in kinase proteins can affect their stability and fitness, and computational analysis of these effects can provide insights into the relationships among protein sequence, structure, and function for this enzyme family. To assess the impact of mutations on protein stability, we used a sequence-based Potts Hamiltonian model trained on a kinase family multiple-sequence alignment (MSA) to calculate the statistical energy (fitness) effects of mutations and compared these against relative folding free energies (ΔΔGs) calculated from all-atom molecular dynamics free energy perturbation (FEP) simulations in explicit solvent. The fitness effects of mutations in the Potts model (ΔEs) showed good agreement with experimental thermostability data (Pearson r = 0.68), similar to the correlation we observed with ΔΔGs predicted from structure-based relative FEP simulations. Recognizing the possible advantages of using Potts models to rapidly estimate protein stability effects of kinase mutations seen in cancer genomics data, we used the Potts statistical energy model to estimate the stability effects of 65 conservative and nonconservative mutations across three distinct kinases (Wee1, Abl1, and Cdc7) with somatic mutations reported in the Genomic Data Commons (GDC) database. The ΔEs of these mutations calculated from the Potts model are consistent with the corresponding ΔΔGs from FEP simulations (Pearson ratio of 0.72). The agreement between these methods suggests that the Potts model may be used as a sequence-based tool for high-throughput screening of mutational effects as part of a computational pipeline for predicting the stability effects of mutations. We also demonstrate how the scalability of the fitness-based Potts model calculations permits analyses that are not easily accessed using FEP simulations. To this end, we employed site-saturation mutagenesis in the Potts model in order to investigate the relative stability effects of mutations seen in different cancer evolutionary scenarios. We used this approach to analyze the effects of drug pressure in Abl kinase by contrasting the relative fitness penalties of somatic mutations seen in miscellaneous cancer types with those calculated for mutations associated with cancer drug resistance. We observed that, in contrast to somatic mutations of Abl seen in various tumors that appear to have evolved neutrally, cancer mutations that evolved under drug pressure in Abl-targeted therapies tend to preserve enzyme stability.


Assuntos
Simulação de Dinâmica Molecular , Neoplasias , Humanos , Proteínas Quinases/genética , Termodinâmica , Mutação , Estabilidade Proteica
4.
Sci Adv ; 9(29): eadg5953, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37478179

RESUMO

HIV-1 infection depends on the integration of viral DNA into host chromatin. Integration is mediated by the viral enzyme integrase and is blocked by integrase strand transfer inhibitors (INSTIs), first-line antiretroviral therapeutics widely used in the clinic. Resistance to even the best INSTIs is a problem, and the mechanisms of resistance are poorly understood. Here, we analyze combinations of the mutations E138K, G140A/S, and Q148H/K/R, which confer resistance to INSTIs. The investigational drug 4d more effectively inhibited the mutants compared with the approved drug Dolutegravir (DTG). We present 11 new cryo-EM structures of drug-resistant HIV-1 intasomes bound to DTG or 4d, with better than 3-Å resolution. These structures, complemented with free energy simulations, virology, and enzymology, explain the mechanisms of DTG resistance involving E138K + G140A/S + Q148H/K/R and show why 4d maintains potency better than DTG. These data establish a foundation for further development of INSTIs that potently inhibit resistant forms in integrase.


Assuntos
Inibidores de Integrase de HIV , Integrase de HIV , Inibidores de Integrase de HIV/farmacologia , Inibidores de Integrase de HIV/química , Oxazinas/farmacologia , Mutação , Integrase de HIV/genética , Integrase de HIV/química , Integrase de HIV/metabolismo
5.
Elife ; 112022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36562610

RESUMO

Inactive conformations of protein kinase catalytic domains where the DFG motif has a "DFG-out" orientation and the activation loop is folded present a druggable binding pocket that is targeted by FDA-approved 'type-II inhibitors' in the treatment of cancers. Tyrosine kinases (TKs) typically show strong binding affinity with a wide spectrum of type-II inhibitors while serine/threonine kinases (STKs) usually bind more weakly which we suggest here is due to differences in the folded to extended conformational equilibrium of the activation loop between TKs vs. STKs. To investigate this, we use sequence covariation analysis with a Potts Hamiltonian statistical energy model to guide absolute binding free-energy molecular dynamics simulations of 74 protein-ligand complexes. Using the calculated binding free energies together with experimental values, we estimated free-energy costs for the large-scale (~17-20 Å) conformational change of the activation loop by an indirect approach, circumventing the very challenging problem of simulating the conformational change directly. We also used the Potts statistical potential to thread large sequence ensembles over active and inactive kinase states. The structure-based and sequence-based analyses are consistent; together they suggest TKs evolved to have free-energy penalties for the classical 'folded activation loop' DFG-out conformation relative to the active conformation, that is, on average, 4-6 kcal/mol smaller than the corresponding values for STKs. Potts statistical energy analysis suggests a molecular basis for this observation, wherein the activation loops of TKs are more weakly 'anchored' against the catalytic loop motif in the active conformation and form more stable substrate-mimicking interactions in the inactive conformation. These results provide insights into the molecular basis for the divergent functional properties of TKs and STKs, and have pharmacological implications for the target selectivity of type-II inhibitors.


Assuntos
Proteínas Serina-Treonina Quinases , Tirosina , Proteínas Serina-Treonina Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Simulação de Dinâmica Molecular , Conformação Proteica , Treonina , Serina
6.
J Phys Chem B ; 126(50): 10622-10636, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36493468

RESUMO

The ability of HIV-1 to rapidly mutate leads to antiretroviral therapy (ART) failure among infected patients. Drug-resistance mutations (DRMs), which cause a fitness penalty to intrinsic viral fitness, are compensated by accessory mutations with favorable epistatic interactions which cause an evolutionary trapping effect, but the kinetics of this overall process has not been well characterized. Here, using a Potts Hamiltonian model describing epistasis combined with kinetic Monte Carlo simulations of evolutionary trajectories, we explore how epistasis modulates the evolutionary dynamics of HIV DRMs. We show how the occurrence of a drug-resistance mutation is contingent on favorable epistatic interactions with many other residues of the sequence background and that subsequent mutations entrench DRMs. We measure the time-autocorrelation of fluctuations in the likelihood of DRMs due to epistatic coupling with the sequence background, which reveals the presence of two evolutionary processes controlling DRM kinetics with two distinct time scales. Further analysis of waiting times for the evolutionary trapping effect to reverse reveals that the sequences which entrench (trap) a DRM are responsible for the slower time scale. We also quantify the overall strength of epistatic effects on the evolutionary kinetics for different mutations and show these are much larger for DRM positions than polymorphic positions, and we also show that trapping of a DRM is often caused by the collective effect of many accessory mutations, rather than a few strongly coupled ones, suggesting the importance of multiresidue sequence variations in HIV evolution. The analysis presented here provides a framework to explore the kinetic pathways through which viral proteins like HIV evolve under drug-selection pressure.


Assuntos
Infecções por HIV , HIV-1 , Humanos , Farmacorresistência Viral/genética , Mutação , Infecções por HIV/tratamento farmacológico , HIV-1/genética , Cinética
7.
PLoS One ; 17(1): e0262314, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35041711

RESUMO

The rapid evolution of HIV is constrained by interactions between mutations which affect viral fitness. In this work, we explore the role of epistasis in determining the mutational fitness landscape of HIV for multiple drug target proteins, including Protease, Reverse Transcriptase, and Integrase. Epistatic interactions between residues modulate the mutation patterns involved in drug resistance, with unambiguous signatures of epistasis best seen in the comparison of the Potts model predicted and experimental HIV sequence "prevalences" expressed as higher-order marginals (beyond triplets) of the sequence probability distribution. In contrast, experimental measures of fitness such as viral replicative capacities generally probe fitness effects of point mutations in a single background, providing weak evidence for epistasis in viral systems. The detectable effects of epistasis are obscured by higher evolutionary conservation at sites. While double mutant cycles in principle, provide one of the best ways to probe epistatic interactions experimentally without reference to a particular background, we show that the analysis is complicated by the small dynamic range of measurements. Overall, we show that global pairwise interaction Potts models are necessary for predicting the mutational landscape of viral proteins.


Assuntos
Epistasia Genética , Aptidão Genética , Infecções por HIV/virologia , Protease de HIV/genética , HIV-1/genética , Mutação , Proteínas Virais/genética , Evolução Molecular , Infecções por HIV/genética , Humanos , Replicação Viral
8.
Proteins ; 90(2): 601-614, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34599827

RESUMO

G-protein-coupled receptors (GPCRs) are the largest family of human membrane proteins and represent the primary targets of about one third of currently marketed drugs. Despite the critical importance, experimental structures have been determined for only a limited portion of GPCRs and functional mechanisms of GPCRs remain poorly understood. Here, we have constructed novel sequence coevolutionary models of the A and B classes of GPCRs and compared them with residue contact frequency maps generated with available experimental structures. Significant portions of structural residue contacts were successfully detected in the sequence-based covariational models. "Exception" residue contacts predicted from sequence coevolutionary models but not available structures added missing links that were important for GPCR activation and allosteric modulation. Moreover, we identified distinct residue contacts involving different sets of functional motifs for GPCR activation, such as the Na+ pocket, CWxP, DRY, PIF, and NPxxY motifs in the class A and the HETx and PxxG motifs in the class B. Finally, we systematically uncovered critical residue contacts tuned by allosteric modulation in the two classes of GPCRs, including those from the activation motifs and particularly the extracellular and intracellular loops in class A GPCRs. These findings provide a promising framework for rational design of ligands to regulate GPCR activation and allosteric modulation.


Assuntos
Receptores Acoplados a Proteínas G , Humanos , Ligantes , Receptores Acoplados a Proteínas G/química
9.
Nat Commun ; 12(1): 6302, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728624

RESUMO

Potts models and variational autoencoders (VAEs) have recently gained popularity as generative protein sequence models (GPSMs) to explore fitness landscapes and predict mutation effects. Despite encouraging results, current model evaluation metrics leave unclear whether GPSMs faithfully reproduce the complex multi-residue mutational patterns observed in natural sequences due to epistasis. Here, we develop a set of sequence statistics to assess the "generative capacity" of three current GPSMs: the pairwise Potts Hamiltonian, the VAE, and the site-independent model. We show that the Potts model's generative capacity is largest, as the higher-order mutational statistics generated by the model agree with those observed for natural sequences, while the VAE's lies between the Potts and site-independent models. Importantly, our work provides a new framework for evaluating and interpreting GPSM accuracy which emphasizes the role of higher-order covariation and epistasis, with broader implications for probabilistic sequence models in general.


Assuntos
Mutação , Proteínas/química , Alinhamento de Sequência/métodos , Algoritmos , Sequência de Aminoácidos , Simulação por Computador , Bases de Dados de Proteínas , Humanos , Modelos Estatísticos , Elementos Estruturais de Proteínas , Proteínas/genética , Relação Estrutura-Atividade
10.
Viruses ; 13(5)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34063519

RESUMO

While drug resistance mutations can often be attributed to the loss of direct or solvent-mediated protein-ligand interactions in the drug-mutant complex, in this study we show that a resistance mutation for the picomolar HIV-1 capsid (CA)-targeting antiviral (GS-6207) is mainly due to the free energy cost of the drug-induced protein side chain reorganization in the mutant protein. Among several mutations, M66I causes the most suppression of the GS-6207 antiviral activity (up to ~84,000-fold), and only 83- and 68-fold reductions for PF74 and ZW-1261, respectively. To understand the molecular basis of this drug resistance, we conducted molecular dynamics free energy simulations to study the structures, energetics, and conformational free energy landscapes involved in the inhibitors binding at the interface of two CA monomers. To minimize the protein-ligand steric clash, the I66 side chain in the M66I-GS-6207 complex switches to a higher free energy conformation from the one adopted in the apo M66I. In contrast, the binding of GS-6207 to the wild-type CA does not lead to any significant M66 conformational change. Based on an analysis that decomposes the absolute binding free energy into contributions from two receptor conformational states, it appears that it is the free energy cost of side chain reorganization rather than the reduced protein-ligand interaction that is largely responsible for the drug resistance against GS-6207.


Assuntos
Proteínas do Capsídeo/genética , Capsídeo/efeitos dos fármacos , Farmacorresistência Viral/genética , HIV-1/genética , Simulação de Dinâmica Molecular , Mutação , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Sítios de Ligação , Capsídeo/química , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Humanos , Ligantes , Ligação Proteica , Conformação Proteica
11.
J Phys Chem Lett ; 12(18): 4368-4377, 2021 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-33938761

RESUMO

We introduce a method called restrain-free energy perturbation-release 2.0 (R-FEP-R 2.0) to estimate conformational free energy changes of protein loops via an alchemical path. R-FEP-R 2.0 is a generalization of the method called restrain-free energy perturbation-release (R-FEP-R) that can only estimate conformational free energy changes of protein side chains but not loops. The reorganization of protein loops is a central feature of many biological processes. Unlike other advanced sampling algorithms such as umbrella sampling and metadynamics, R-FEP-R and R-FEP-R 2.0 do not require predetermined collective coordinates and transition pathways that connect the two endpoint conformational states. The R-FEP-R 2.0 method was applied to estimate the conformational free energy change of a ß-turn flip in the protein ubiquitin. The result obtained by R-FEP-R 2.0 agrees with the benchmarks very well. We also comment on problems commonly encountered when applying umbrella sampling to calculate protein conformational free energy changes.


Assuntos
Proteases Específicas de Ubiquitina/química , Algoritmos , Cinética , Simulação de Dinâmica Molecular , Conformação Proteica , Solventes/química , Termodinâmica
12.
Comput Phys Commun ; 2602021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33716309

RESUMO

Inverse Ising inference is a method for inferring the coupling parameters of a Potts/Ising model based on observed site-covariation, which has found important applications in protein physics for detecting interactions between residues in protein families. We introduce Mi3-GPU ("mee-three", for MCMC Inverse Ising Inference) software for solving the inverse Ising problem for protein-sequence datasets with few analytic approximations, by parallel Markov-Chain Monte-Carlo sampling on GPUs. We also provide tools for analysis and preparation of protein-family Multiple Sequence Alignments (MSAs) to account for finite-sampling issues, which are a major source of error or bias in inverse Ising inference. Our method is "generative" in the sense that the inferred model can be used to generate synthetic MSAs whose mutational statistics (marginals) can be verified to match the dataset MSA statistics up to the limits imposed by the effects of finite sampling. Our GPU implementation enables the construction of models which reproduce the covariation patterns of the observed MSA with a precision that is not possible with more approximate methods. The main components of our method are a GPU-optimized algorithm to greatly accelerate MCMC sampling, combined with a multi-step Quasi-Newton parameter-update scheme using a "Zwanzig reweighting" technique. We demonstrate the ability of this software to produce generative models on typical protein family datasets for sequence lengths L ~ 300 with 21 residue types with tens of millions of inferred parameters in short running times.

13.
J Phys Chem B ; 124(52): 11771-11782, 2020 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-33306906

RESUMO

Solvation thermodynamics is concerned with the evaluation and physical interpretation of solvation free energies. Endpoints DFT provides a framework for computing solvation free energies by combining molecular simulations with a version of the classical density-functional theory of solutions which focuses on ω, the indirect (solvent-mediated) part of the solute-solvent potential of mean force (indirect PMF). The simulations are performed at the endpoints of a hypothetical charging process which transforms the solvent density from the pure liquid state to that of the solution state. The endpoints DFT expression for solvation free energy can be shown to be equivalent to the standard expression for which the key quantity is the direct correlation function, but it has the advantage that the indirect term ω is more focused on the change in solvent-solvent correlations with respect to the pure liquid as the solute is inserted into the solution. In this Perspective, we review recent developments of endpoints DFT, highlighting a series of papers we have written together beginning in 2017. We emphasize the importance of dimensionality reduction as the key to the evaluation of endpoints DFT expressions and present a recently developed, spatially resolved version of the theory. The role of interfacial water at certain positions which stabilize or destabilize a solute in solution can be analyzed with the spatially resolved version, and it is of considerable interest to investigate how changes in solvation affect protein-ligand binding and conformational landscapes from an endpoints DFT perspective. Endpoints DFT can also be employed in materials science; an example involving the rational design strategy for polymer membrane separation is described. The endpoints DFT method is a scheme to evaluate the solvation free energy by introducing approximations to integrate the classical density functional over a charging parameter. We have further proposed a new functional which captures the correct dependence of the indirect PMF ω at both endpoints of the charging process, and we review how it might be employed in future work.


Assuntos
Água , Ligantes , Soluções , Solventes , Termodinâmica
14.
J Phys Chem B ; 124(25): 5220-5237, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32469519

RESUMO

Endpoints density functional theory (DFT) provides a framework for calculating the excess chemical potential of a solute in solution using solvent distribution functions obtained from both physical endpoints of a hypothetical charging process which transforms the solvent density from that of the pure liquid to the solution state. In this work, the endpoints DFT equations are formulated in terms of the indirect (solvent-mediated) contribution ω(x) to the solute-solvent potential of mean force, and their connections are established with the conventional DFT expressions which are based on the use of direct correlation functions. ω actually corresponds to the free-energy cost to move a cavity particle (a tagged solvent molecule which interacts with the other solvent molecules but not the solute) from the bulk to the configuration x of a solvent molecule relative to the solute and is a suitable variable to describe the solvent effects on the solute-solvent interactions. HNC and PY type approximations are then used to integrate the DFT charging integral involved in the exact expression for the excess chemical potential. With these approximations, molecular simulations are to be performed at the two endpoints of solute insertion: pure solvent without the solute and the solution system with the fully coupled solute-solvent interaction. An endpoints method thus utilizes the ensembles of intermolecular configurations of physical interest, which are often readily accessible with MD simulations given the present computational power. To illustrate properties of the formulation, we perform simulations of model systems consisting of a cavity particle in an aqueous solution containing a spherical hydrophobic solute of three different sizes from which ω(x) and the solute chemical potential can be calculated using endpoints DFT expressions. These are compared with corresponding results obtained using the approximations needed in order to evaluate the endpoints DFT charging integral when cavity particle simulation data is not available. We analyze a new approximation (two-points quadratic HNC) to the DFT charging integral which captures the correct behavior of the cavity distributions at both endpoints of the solute insertion. The behavior of the cavity particle in simple and complex liquids plays an important role in various theoretical treatments of the solute chemical potential. For pure Lennard-Jones fluids, the free energy to bring a cavity particle from the bulk to the center of a fluid particle is negative. However, for solutes of varying size, this is not generally true for Lennard-Jones fluids or the systems studied in this work. We carry out energetic and structural analyses of the cavity particle in aqueous solution with hydrophobic solutes of varying size and discuss the results in the context of the hydrophobic effect.


Assuntos
Interações Hidrofóbicas e Hidrofílicas , Água , Soluções , Solventes , Termodinâmica
16.
J Chem Theory Comput ; 16(1): 67-79, 2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-31743019

RESUMO

Free energy perturbation (FEP) simulations have been widely applied to obtain predictions of the relative binding free energy for a series of congeneric ligands binding to the same receptor, which is an essential component for the lead optimization process in computer-aided drug discovery. In the case of several congeneric ligands forming a perturbation map involving a closed thermodynamic cycle, the summation of the estimated free energy change along each edge in the cycle using Bennett acceptance ratio (BAR) usually will deviate from zero due to systematic and random errors, which is the hysteresis of cycle closure. In this work, the advanced reweighting techniques binless weighted histogram analysis method (UWHAM) and locally weighted histogram analysis method (LWHAM) are applied to provide statistical estimators of the free energy change along each edge in order to eliminate the hysteresis effect. As an example, we analyze a closed thermodynamic cycle involving four congeneric ligands which bind to HIV-1 integrase, a promising target which has emerged for antiviral therapy. We demonstrate that, compared with FEP and BAR, more accurate and hysteresis-free estimates of free energy differences can be achieved by using UWHAM to find a single estimate of the density of states based on all of the data in the cycle. Furthermore, by comparison of LWHAM results obtained from the inclusion of different numbers of neighboring states with UWHAM estimation involving all the states, we show how to determine the optimal neighborhood size in the LWHAM analysis to balance the trade-offs between computational cost and accuracy of the free energy prediction. Even with the smallest neighborhood, LWHAM can improve the BAR free energy estimates using the same input data as BAR. We introduce an overlapping states matrix that is constructed by using the global jump formula of LWHAM and plot its heat map. The heat map provides a quantitative measure of the overlap between pairs of alchemical/thermodynamic states. We explain how to identify and improve the FEP calculations along the edges that most likely cause large systematic errors by using the heat map of the overlapping states matrix and by comparing the BAR and UWHAM estimates of the free energy change.

17.
J Comput Chem ; 41(1): 56-68, 2020 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-31621932

RESUMO

We propose a free energy calculation method for receptor-ligand binding, which have multiple binding poses that avoids exhaustive enumeration of the poses. For systems with multiple binding poses, the standard procedure is to enumerate orientations of the binding poses, restrain the ligand to each orientation, and then, calculate the binding free energies for each binding pose. In this study, we modify a part of the thermodynamic cycle in order to sample a broader conformational space of the ligand in the binding site. This modification leads to more accurate free energy calculation without performing separate free energy simulations for each binding pose. We applied our modification to simple model host-guest systems as a test, which have only two binding poses, by using a single decoupling method (SDM) in implicit solvent. The results showed that the binding free energies obtained from our method without knowing the two binding poses were in good agreement with the benchmark results obtained by explicit enumeration of the binding poses. Our method is applicable to other alchemical binding free energy calculation methods such as the double decoupling method (DDM) in explicit solvent. We performed a calculation for a protein-ligand system with explicit solvent using our modified thermodynamic path. The results of the free energy simulation along our modified path were in good agreement with the results of conventional DDM, which requires a separate binding free energy calculation for each of the binding poses of the example of phenol binding to T4 lysozyme in explicit solvent. © 2019 Wiley Periodicals, Inc.


Assuntos
Simulação de Dinâmica Molecular , Muramidase/química , Fenóis/química , Termodinâmica , Sítios de Ligação , Ligantes , Muramidase/metabolismo
18.
Elife ; 82019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31591964

RESUMO

The development of drug resistance in HIV is the result of primary mutations whose effects on viral fitness depend on the entire genetic background, a phenomenon called 'epistasis'. Based on protein sequences derived from drug-experienced patients in the Stanford HIV database, we use a co-evolutionary (Potts) Hamiltonian model to provide direct confirmation of epistasis involving many simultaneous mutations. Building on earlier work, we show that primary mutations leading to drug resistance can become highly favored (or entrenched) by the complex mutation patterns arising in response to drug therapy despite being disfavored in the wild-type background, and provide the first confirmation of entrenchment for all three drug-target proteins: protease, reverse transcriptase, and integrase; a comparative analysis reveals that NNRTI-induced mutations behave differently from the others. We further show that the likelihood of resistance mutations can vary widely in patient populations, and from the population average compared to specific molecular clones.


Assuntos
Fármacos Anti-HIV/farmacologia , Farmacorresistência Viral , Epistasia Genética , HIV-1/efeitos dos fármacos , HIV-1/genética , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Proteínas Mutantes/genética , Mutação
19.
Elife ; 82019 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-31120420

RESUMO

Allosteric HIV-1 integrase (IN) inhibitors (ALLINIs) are a promising new class of antiretroviral agents that disrupt proper viral maturation by inducing hyper-multimerization of IN. Here we show that lead pyridine-based ALLINI KF116 exhibits striking selectivity for IN tetramers versus lower order protein oligomers. IN structural features that are essential for its functional tetramerization and HIV-1 replication are also critically important for KF116 mediated higher-order IN multimerization. Live cell imaging of single viral particles revealed that KF116 treatment during virion production compromises the tight association of IN with capsid cores during subsequent infection of target cells. We have synthesized the highly active (-)-KF116 enantiomer, which displayed EC50 of ~7 nM against wild type HIV-1 and ~10 fold higher, sub-nM activity against a clinically relevant dolutegravir resistant mutant virus suggesting potential clinical benefits for complementing dolutegravir therapy with pyridine-based ALLINIs.


HIV-1 inserts its genetic code into human genomes, turning healthy cells into virus factories. To do this, the virus uses an enzyme called integrase. Front-line treatments against HIV-1 called "integrase strand-transfer inhibitors" stop this enzyme from working. These inhibitors have helped to revolutionize the treatment of HIV/AIDS by protecting the cells from new infections. But, the emergence of drug resistance remains a serious problem. As the virus evolves, it changes the shape of its integrase protein, substantially reducing the effectiveness of the current therapies. One way to overcome this problem is to develop other therapies that can kill the drug resistant viruses by targeting different parts of the integrase protein. It should be much harder for the virus to evolve the right combination of changes to escape two or more treatments at once. A promising class of new compounds are "allosteric integrase inhibitors". These chemical compounds target a part of the integrase enzyme that the other treatments do not yet reach. Rather than stopping the integrase enzyme from inserting the viral code into the human genome, the new inhibitors make integrase proteins clump together and prevent the formation of infectious viruses. At the moment, these compounds are still experimental. Before they are ready for use in people, researchers need to better understand how they work, and there are several open questions to answer. Integrase proteins work in groups of four and it is not clear how the new compounds make the integrases form large clumps, or what this does to the virus. Understanding this should allow scientists to develop improved versions of the drugs. To answer these questions, Koneru et al. first examined two of the new compounds. A combination of molecular analysis and computer modelling revealed how they work. The compounds link many separate groups of four integrases with each other to form larger and larger clumps, essentially a snowball effect. Live images of infected cells showed that the clumps of integrase get stuck outside of the virus's protective casing. This leaves them exposed, allowing the cell to destroy the integrase enzymes. Koneru et al. also made a new compound, called (-)-KF116. Not only was this compound able to tackle normal HIV-1, it could block viruses resistant to the other type of integrase treatment. In fact, in laboratory tests, it was 10 times more powerful against these resistant viruses. Together, these findings help to explain how allosteric integrase inhibitors work, taking scientists a step closer to bringing them into the clinic. In the future, new versions of the compounds, like (-)-KF116, could help to tackle drug resistance in HIV-1.


Assuntos
Antivirais/farmacologia , Inibidores de Integrase de HIV/farmacologia , Integrase de HIV/metabolismo , Multimerização Proteica , Piridinas/farmacologia , Regulação Alostérica/efeitos dos fármacos , Antivirais/química , Células HEK293 , Integrase de HIV/química , Inibidores de Integrase de HIV/química , Células HeLa , Humanos , Modelos Moleculares , Domínios Proteicos , Piridinas/química , Estereoisomerismo
20.
Phys Rev E ; 99(3-1): 032405, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30999494

RESUMO

Potts statistical models have become a popular and promising way to analyze mutational covariation in protein multiple sequence alignments (MSAs) in order to understand protein structure, function, and fitness. But the statistical limitations of these models, which can have millions of parameters and are fit to MSAs of only thousands or hundreds of effective sequences using a procedure known as inverse Ising inference, are incompletely understood. In this work we predict how model quality degrades as a function of the number of sequences N, sequence length L, amino-acid alphabet size q, and the degree of conservation of the MSA, in different applications of the Potts models: in "fitness" predictions of individual protein sequences, in predictions of the effects of single-point mutations, in "double mutant cycle" predictions of epistasis, and in 3D contact prediction in protein structure. We show how as MSA depth N decreases an "overfitting" effect occurs such that sequences in the training MSA have overestimated fitness, and we predict the magnitude of this effect and discuss how regularization can help correct for it, using a regularization procedure motivated by statistical analysis of the effects of finite sampling. We find that as N decreases the quality of point-mutation effect predictions degrade least, fitness and epistasis predictions degrade more rapidly, and contact predictions are most affected. However, overfitting becomes negligible for MSA depths of more than a few thousand effective sequences, as often used in practice, and regularization becomes less necessary. We discuss the implications of these results for users of Potts covariation analysis.


Assuntos
Modelos Moleculares , Modelos Estatísticos , Proteínas/genética , Proteínas/metabolismo , Algoritmos , Sequência de Aminoácidos , Simulação por Computador , Modelos Genéticos , Mutação , Conformação Proteica , Proteínas/química , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA