1
|
Nadeem H, Shukla D. Ensemble Adaptive Sampling Scheme: Identifying an Optimal Sampling Strategy via Policy Ranking. J Chem Theory Comput 2025; 21:4626-4639. [PMID: 40261689 DOI: 10.1021/acs.jctc.4c01488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Efficient sampling in biomolecular simulations is critical for accurately capturing the complex dynamic behaviors of biological systems. Adaptive sampling techniques aim to improve efficiency by focusing computational resources on the most relevant regions of the phase space. In this work, we present a framework for identifying the optimal sampling policy through metric-driven ranking. Our approach systematically evaluates the policy ensemble and ranks the policies based on their ability to explore the conformational space effectively. Through a series of biomolecular simulation case studies, we demonstrate that the choice of a different adaptive sampling policy at each round significantly outperforms single policy sampling, leading to faster convergence and improved sampling performance. This approach takes an ensemble of adaptive sampling policies and identifies the optimal policy for the next round based on current data. Beyond presenting this ensemble view of adaptive sampling, we also propose two sampling algorithms that approximate this ranking framework on the fly. The modularity of this framework allows incorporation of any adaptive sampling policy, making it versatile and suitable as a comprehensive adaptive sampling scheme.
Collapse
Affiliation(s)
- Hassan Nadeem
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Diwakar Shukla
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
2
|
Yu W, Kumar S, Zhao M, Weber DJ, MacKerell AD. High-Throughput Ligand Dissociation Kinetics Predictions Using Site Identification by Ligand Competitive Saturation. J Chem Theory Comput 2025; 21:4964-4978. [PMID: 40285712 PMCID: PMC12077591 DOI: 10.1021/acs.jctc.5c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
The dissociation or off rate, koff, of a drug molecule has been shown to be more relevant to efficacy than affinity for selected systems, motivating the development of predictive computational methodologies. These are largely based on enhanced-sampling molecular dynamics (MD) simulations that come at a high computational cost limiting their utility for drug design where a large number of ligands need to be evaluated. To overcome this, presented is a combined physics- and machine learning (ML)-based approach that uses the physics-based site identification by ligand competitive saturation (SILCS) method to enumerate potential ligand dissociation pathways and calculate ligand dissociation free-energy profiles along those pathways. The calculated free-energy profiles along with molecular properties are used as features to train ML models, including tree and neural network approaches, to predict koff values. The protocol is developed and validated using 329 ligands for 13 proteins showing robustness of the ML workflow built upon the SILCS physics-based free-energy profiles. The resulting SILCS-Kinetics workflow offers a highly efficient method to study ligand dissociation kinetics, providing a powerful tool to facilitate drug design including the ability to generate quantitative estimates of atomic and functional groups contributions to ligand dissociation.
Collapse
Affiliation(s)
- Wenbo Yu
- Computer-Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, Maryland 20850, United States
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
| | - Shashi Kumar
- Computer-Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
| | - Mingtian Zhao
- Computer-Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
| | - David J. Weber
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, Maryland 20850, United States
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
| | - Alexander D. MacKerell
- Computer-Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, Maryland 20850, United States
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland Baltimore, Baltimore, Maryland 21201, United States
| |
Collapse
|
3
|
Deganutti G, Pipito L, Rujan RM, Weizmann T, Griffin P, Ciancetta A, Moro S, Reynolds CA. Hidden GPCR structural transitions addressed by multiple walker supervised molecular dynamics (mwSuMD). eLife 2025; 13:RP96513. [PMID: 40305095 PMCID: PMC12043319 DOI: 10.7554/elife.96513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
The structural basis for the pharmacology of human G protein-coupled receptors (GPCRs), the most abundant membrane proteins and the target of about 35% of approved drugs, is still a matter of intense study. What makes GPCRs challenging to study is the inherent flexibility and the metastable nature of interaction with extra- and intracellular partners that drive their effects. Here, we present a molecular dynamics (MD) adaptive sampling algorithm, namely multiple walker supervised molecular dynamics (mwSuMD), to address complex structural transitions involving GPCRs without energy input. We first report the binding and unbinding of the vasopressin peptide from its receptor V2. Successively, we present the complete transition of the glucagon-like peptide-1 receptor (GLP-1R) from inactive to active, agonist and Gs-bound state, and the guanosine diphosphate (GDP) release from Gs. To our knowledge, this is the first time the whole sequence of events leading from an inactive GPCR to the GDP release is simulated without any energy bias. We demonstrate that mwSuMD can address complex binding processes intrinsically linked to protein dynamics out of reach of classic MD.
Collapse
Affiliation(s)
- Giuseppe Deganutti
- Centre for Health and Life Sciences, Coventry UniversityCoventryUnited Kingdom
| | - Ludovico Pipito
- Centre for Health and Life Sciences, Coventry UniversityCoventryUnited Kingdom
| | - Roxana Maria Rujan
- Centre for Health and Life Sciences, Coventry UniversityCoventryUnited Kingdom
| | - Tal Weizmann
- Centre for Health and Life Sciences, Coventry UniversityCoventryUnited Kingdom
| | - Peter Griffin
- Centre for Health and Life Sciences, Coventry UniversityCoventryUnited Kingdom
| | - Antonella Ciancetta
- Dipartimento di Scienze Chimiche, Farmaceutiche ed Agrarie, University of FerraraFerraraItaly
| | - Stefano Moro
- Molecular Modeling Section (MMS), Dipartimento di Scienze del Farmaco, University of Padua via MarzoloPadovaItaly
| | - Christopher Arthur Reynolds
- Centre for Health and Life Sciences, Coventry UniversityCoventryUnited Kingdom
- School of Life Sciences, University of Essex, Wivenhoe ParkColchesterUnited Kingdom
| |
Collapse
|
4
|
Pant S, Dehghani-Ghahnaviyeh S, Trebesch N, Rasouli A, Chen T, Kapoor K, Wen PC, Tajkhorshid E. Dissecting Large-Scale Structural Transitions in Membrane Transporters Using Advanced Simulation Technologies. J Phys Chem B 2025; 129:3703-3719. [PMID: 40100959 DOI: 10.1021/acs.jpcb.5c00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Membrane transporters are integral membrane proteins that act as gatekeepers of the cell, controlling fundamental processes such as recruitment of nutrients and expulsion of waste material. At a basic level, transporters operate using the "alternating access model," in which transported substances are accessible from only one side of the membrane at a time. This model usually involves large-scale structural changes in the transporter, which often cannot be captured using unbiased, conventional molecular simulation techniques. In this article, we provide an overview of some of the major simulation techniques that have been applied to characterize the structural dynamics and energetics involved in the transition of membrane transporters between their functional states. After briefly introducing each technique, we discuss some of their advantages and limitations and provide some recent examples of their application to membrane transporters.
Collapse
Affiliation(s)
- Shashank Pant
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Sepehr Dehghani-Ghahnaviyeh
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Noah Trebesch
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Ali Rasouli
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Tianle Chen
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Karan Kapoor
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Po-Chao Wen
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| |
Collapse
|
5
|
Braza MK, Demir Ö, Ahn SH, Morris CK, Calvó-Tusell C, McGuire KL, de la Peña Avalos B, Carpenter MA, Chen Y, Casalino L, Aihara H, Herzik MA, Harris RS, Amaro RE. Regulatory Interactions between APOBEC3B N- and C-Terminal Domains. J Chem Inf Model 2025; 65:3593-3604. [PMID: 40105360 PMCID: PMC12004528 DOI: 10.1021/acs.jcim.4c02272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
APOBEC3B (A3B) is implicated in DNA mutations that facilitate tumor evolution. Although structures of its individual N- and C-terminal domains (NTD and CTD) have been resolved through X-ray crystallography, the full-length A3B (fl-A3B) structure remains elusive, limiting our understanding of its dynamics and mechanisms. In particular, the APOBEC3B C-terminal domain (A3Bctd) is frequently closed in models and structures. In this study, we built several new models of fl-A3B using integrative structural biology methods and selected a top model for further dynamical investigation. We compared the dynamics of the truncated (A3Bctd) to that of the fl-A3B via conventional and Gaussian accelerated molecular dynamics (MD) simulations. Subsequently, we employed weighted ensemble methods to explore the fl-A3B active site opening mechanism, finding that interactions at the NTD-CTD interface enhance the opening frequency of the fl-A3B active site. Our findings shed light on the structural dynamics and potential druggability of fl-A3B, including observations regarding both the active and allosteric sites, which may offer new avenues for therapeutic intervention in cancer.
Collapse
Affiliation(s)
- Mac Kevin
E. Braza
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Özlem Demir
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Surl-Hee Ahn
- Department
of Chemical Engineering, University of California,
Davis, Davis, California 95616, United States
| | - Clare K. Morris
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Carla Calvó-Tusell
- Department
of Molecular Biology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Kelly L. McGuire
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Bárbara de la Peña Avalos
- Department
of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| | - Michael A. Carpenter
- Department
of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229, United States
- Howard
Hughes Medical Institute, University of
Texas Health San Antonio, San Antonio, Texas 78229, United States
| | - Yanjun Chen
- Department
of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| | - Lorenzo Casalino
- Department
of Molecular Biology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Hideki Aihara
- Department
of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mark A. Herzik
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Reuben S. Harris
- Department
of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229, United States
- Howard
Hughes Medical Institute, University of
Texas Health San Antonio, San Antonio, Texas 78229, United States
| | - Rommie E. Amaro
- Department
of Molecular Biology, University of California,
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
6
|
Leung JG, Frazee NC, Brace A, Bogetti AT, Ramanathan A, Chong LT. Unsupervised Learning of Progress Coordinates during Weighted Ensemble Simulations: Application to NTL9 Protein Folding. J Chem Theory Comput 2025; 21:3691-3699. [PMID: 40105797 PMCID: PMC11983707 DOI: 10.1021/acs.jctc.4c01136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
A major challenge for many rare-event sampling strategies is the identification of progress coordinates that capture the slowest relevant motions. Machine-learning methods that can identify progress coordinates in an unsupervised manner have therefore been of great interest to the simulation community. Here, we developed a general method for identifying progress coordinates "on-the-fly" during weighted ensemble (WE) rare-event sampling via deep learning (DL) of outliers among sampled conformations. Our method identifies outliers in a latent space model of the system's sampled conformations that is periodically trained using a convolutional variational autoencoder. As a proof of principle, we applied our DL-enhanced WE method to simulate the NTL9 protein folding process. To enable rapid tests, our simulations propagated discrete-state synthetic molecular dynamics trajectories using a generative, fine-grained Markov state model. Results revealed that our on-the-fly DL of outliers enhanced the efficiency of WE by >3-fold in estimating the folding rate constant. Our efforts are a significant step forward in the unsupervised learning of slow coordinates during rare event sampling.
Collapse
Affiliation(s)
- Jeremy
M. G. Leung
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Nicolas C. Frazee
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Brace
- Data
Science and Learning Division, Argonne National
Laboratory, Lemont, Illinois 60439, United States
- Department
of Computer Science, University of Chicago, Chicago, Illinois 60637, United States
| | - Anthony T. Bogetti
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Arvind Ramanathan
- Data
Science and Learning Division, Argonne National
Laboratory, Lemont, Illinois 60439, United States
- Department
of Computer Science, University of Chicago, Chicago, Illinois 60637, United States
| | - Lillian T. Chong
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
7
|
Casalino L, Ramos-Guzmán CA, Amaro RE, Simmerling C, Lodola A, Mulholland AJ, Świderek K, Moliner V. A Reflection on the Use of Molecular Simulation to Respond to SARS-CoV-2 Pandemic Threats. J Phys Chem Lett 2025; 16:3249-3263. [PMID: 40118074 PMCID: PMC11973918 DOI: 10.1021/acs.jpclett.4c03654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/23/2025]
Abstract
Molecular simulations play important roles in understanding the lifecycle of the SARS-CoV-2 virus and contribute to the design and development of antiviral agents and diagnostic tests for COVID. Here, we discuss the insights that such simulations have provided and the challenges involved, focusing on the SARS-CoV-2 main protease (Mpro) and the spike glycoprotein. Mpro is the leading target for antivirals, while the spike glycoprotein is the target for vaccine design. Finally, we reflect on lessons from this pandemic for the simulation community. Data sharing initiatives and collaborations across the international research community contributed to advancing knowledge and should be built on to help in future pandemics and other global challenges such as antimicrobial resistance.
Collapse
Affiliation(s)
- Lorenzo Casalino
- Department
of Molecular Biology, University of California
San Diego, La Jolla, California 92093, United States
| | - Carlos A. Ramos-Guzmán
- Centre
for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United
Kingdom
| | - Rommie E. Amaro
- Department
of Molecular Biology, University of California
San Diego, La Jolla, California 92093, United States
| | - Carlos Simmerling
- Department
of Chemistry and Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Alessio Lodola
- Dipartimento
di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, I 43121 Parma, Italy
| | - Adrian J. Mulholland
- Centre
for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United
Kingdom
| | - Katarzyna Świderek
- Biocomp
group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castelló, Spain
| | - Vicent Moliner
- Biocomp
group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castelló, Spain
| |
Collapse
|
8
|
Ma A, Li H. Reaction Coordinates Are Optimal Channels of Energy Flow. Annu Rev Phys Chem 2025; 76:153-179. [PMID: 39903861 DOI: 10.1146/annurev-physchem-082423-010652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Reaction coordinates (RCs) are the few essential coordinates of a protein that control its functional processes, such as allostery, enzymatic reaction, and conformational change. They are critical for understanding protein function and provide optimal enhanced sampling of protein conformational changes and states. Since the pioneering work in the late 1990s, identifying the correct and objectively provable RCs has been a central topic in molecular biophysics and chemical physics. This review summarizes the major advances in identifying RCs over the past 25 years, focusing on methods aimed at finding RCs that meet the rigorous committor criterion, widely accepted as the true RCs. Notably, the newly developed physics-based energy flow theory and generalized work functional method provide a general and rigorous approach for identifying true RCs, revealing their physical nature as the optimal channels of energy flow in biomolecules.
Collapse
Affiliation(s)
- Ao Ma
- Center for Bioinformatics and Quantitative Biology, Richard and Loan Hill Department of Biomedical Engineering, The University of Illinois Chicago, Chicago, Illinois, USA;
| | - Huiyu Li
- Center for Bioinformatics and Quantitative Biology, Richard and Loan Hill Department of Biomedical Engineering, The University of Illinois Chicago, Chicago, Illinois, USA;
| |
Collapse
|
9
|
Singh AN, Das A, Limmer DT. Variational Path Sampling of Rare Dynamical Events. Annu Rev Phys Chem 2025; 76:639-662. [PMID: 39971385 DOI: 10.1146/annurev-physchem-083122-115001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
This article reviews the concepts and methods of variational path sampling. These methods allow computational studies of rare events in systems driven arbitrarily far from equilibrium. Based upon a statistical mechanics of trajectory space and leveraging the theory of large deviations, they provide a perspective from which dynamical phenomena can be studied with the same types of ensemble reweighting ideas that have been used for static equilibrium properties. Applications to chemical, material, and biophysical systems are highlighted.
Collapse
Affiliation(s)
- Aditya N Singh
- Department of Chemistry, University of California, Berkeley, California, USA; , ,
| | - Avishek Das
- Department of Chemistry, University of California, Berkeley, California, USA; , ,
- Current affiliation: Fundamental Research on Matter Institute for Atomic and Molecular Physics (AMOLF), Amsterdam, The Netherlands
| | - David T Limmer
- Department of Chemistry, University of California, Berkeley, California, USA; , ,
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
- Kavli Energy Nanoscience Institute, University of California, Berkeley, California, USA
- Material Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| |
Collapse
|
10
|
Velásquez-Bedoya PA, Zapata-Cardona MI, Monsalve-Escudero LM, Pereañez JA, Guerra-Arias D, Pastrana-Restrepo M, Galeano E, Zapata-Builes W. Antiviral Activity of Halogenated Compounds Derived from L-Tyrosine Against SARS-CoV-2. Molecules 2025; 30:1419. [PMID: 40286029 PMCID: PMC11990460 DOI: 10.3390/molecules30071419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Currently, there are no effective medications for treating all the clinical conditions of patients with COVID-19. We aimed to evaluate the antiviral activity of compounds derived from L-tyrosine against the B.1 lineage of SARS-CoV-2 in vitro and in silico. METHODOLOGY The cytotoxicities of 15 halogenated compounds derived from L-tyrosine were evaluated in Vero-E6 cells by the MTT assay. The antiviral activity of the compounds was evaluated using four strategies, and viral quantification was performed by a plaque assay and qRT-PCR. The toxicity of the compounds was evaluated by ADMET predictor software. The affinity of these compounds for viral or cellular proteins and the stability of their conformations were determined by docking and molecular dynamics, respectively. RESULTS TODC-3M, TODI-2M, and YODC-3M reduced the viral titer >40% and inhibited the replication of viral RNA without significant cytotoxicity. In silico analyses revealed that these compounds presented low toxicity and binding energies between -4.3 and -5.2 Kcal/mol for three viral proteins (spike, Mpro, and RdRp). TODC-3M and YODC-3M presented the most stable conformations with the evaluated proteins. CONCLUSIONS The most promising compounds were TODC-3M, TODI-2M, and YODC-3M, which presented low in vitro and in silico toxicity, antiviral potential through different strategies, and favorable affinities for viral targets. Therefore, they are candidates for in vivo studies.
Collapse
Affiliation(s)
- Paula A. Velásquez-Bedoya
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Calle 50 # 40-74, Medellín 050001, Colombia;
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín 050001, Colombia; (M.I.Z.-C.); (L.M.M.-E.)
| | - María I. Zapata-Cardona
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín 050001, Colombia; (M.I.Z.-C.); (L.M.M.-E.)
| | - Laura M. Monsalve-Escudero
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín 050001, Colombia; (M.I.Z.-C.); (L.M.M.-E.)
| | - Jaime A. Pereañez
- Grupo de Investigación Promoción y Prevención Farmacéutica, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia UdeA, Medellín 050001, Colombia;
| | - Diego Guerra-Arias
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, PTS Granada, 18016 Granada, Spain;
- Programa de Estudio y Control de Enfermedades Tropicales PECET, Facultad de Medicina, Universidad de Antioquia, Medellín 050010, Colombia
| | - Manuel Pastrana-Restrepo
- Grupo Productos Naturales Marinos, Departamento de Farmacia, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia UdeA, Medellín 050001, Colombia; (M.P.-R.); (E.G.)
| | - Elkin Galeano
- Grupo Productos Naturales Marinos, Departamento de Farmacia, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia UdeA, Medellín 050001, Colombia; (M.P.-R.); (E.G.)
| | - Wildeman Zapata-Builes
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Calle 50 # 40-74, Medellín 050001, Colombia;
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín 050001, Colombia; (M.I.Z.-C.); (L.M.M.-E.)
| |
Collapse
|
11
|
Lynch DL, Fan Z, Pavlova A, Gumbart JC. Weighted Ensemble Simulations Reveal Novel Conformations and Modulator Effects in Hepatitis B Virus Capsid Assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.15.643452. [PMID: 40166272 PMCID: PMC11957032 DOI: 10.1101/2025.03.15.643452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Molecular dynamics (MD) simulations provide a detailed description of biophysical processes allowing mechanistic questions to be addressed at the atomic level. The promise of such approaches is partly hampered by well known sampling issues of typical simulations, where time scales available are significantly shorter than the process of interest. For the system of interest here, the binding of modulators of Hepatitis B virus capsid self-assembly, the binding site is at a flexible protein-protein interface. Characterization of the conformational landscape and how it is altered upon ligand binding is thus a prerequisite for a complete mechanistic description of capsid assembly modulation. However, such a description can be difficult due to the aforementioned sampling issues of standard MD, and enhanced sampling strategies are required. Here we employ the Weighted Ensemble methodology to characterize the free-energy landscape of our earlier determined functionally relevant progress coordinates. It is shown that this approach provides conformations outside those sampled by standard MD, as well as an increased number of structures with correspondingly enlarged binding pockets conducive to ligand binding, illustrating the utility of Weighted Ensemble for computational drug development.
Collapse
Affiliation(s)
- Diane L Lynch
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia, 30332, USA
| | - Zixing Fan
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, Georgia, 30332, USA
| | - Anna Pavlova
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia, 30332, USA
| | - James C Gumbart
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia, 30332, USA
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, 30332, USA
| |
Collapse
|
12
|
Ojha AA, Blackwell R, Cruz-Chú ER, Dsouza R, Astore MA, Schwander P, Hanson SM. The ManifoldEM method for cryo-EM: a step-by-step breakdown accompanied by a modern Python implementation. Acta Crystallogr D Struct Biol 2025; 81:89-104. [PMID: 40019002 DOI: 10.1107/s2059798325001469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
Resolving continuous conformational heterogeneity in single-particle cryo-electron microscopy (cryo-EM) is a field in which new methods are now emerging regularly. Methods range from traditional statistical techniques to state-of-the-art neural network approaches. Such ongoing efforts continue to enhance the ability to explore and understand the continuous conformational variations in cryo-EM data. One of the first methods was the manifold embedding approach or ManifoldEM. However, comparing it with more recent methods has been challenging due to software availability and usability issues. In this work, we introduce a modern Python implementation that is user-friendly, orders of magnitude faster than its previous versions and designed with a developer-ready environment. This implementation allows a more thorough evaluation of the strengths and limitations of methods addressing continuous conformational heterogeneity in cryo-EM, paving the way for further community-driven improvements.
Collapse
Affiliation(s)
- Anupam Anand Ojha
- Center for Computational Biology and Center for Computational Mathematics, Flatiron Institute, New York, NY 10010, USA
| | - Robert Blackwell
- Scientific Computing Core, Flatiron Institute, New York, NY 10010, USA
| | - Eduardo R Cruz-Chú
- Department of Physics, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Raison Dsouza
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - Miro A Astore
- Center for Computational Biology and Center for Computational Mathematics, Flatiron Institute, New York, NY 10010, USA
| | - Peter Schwander
- Department of Physics, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Sonya M Hanson
- Center for Computational Biology and Center for Computational Mathematics, Flatiron Institute, New York, NY 10010, USA
| |
Collapse
|
13
|
Bose S, Kilinc C, Dickson A. Markov State Models with Weighted Ensemble Simulation: How to Eliminate the Trajectory Merging Bias. J Chem Theory Comput 2025; 21:1805-1816. [PMID: 39933004 PMCID: PMC11866749 DOI: 10.1021/acs.jctc.4c01141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025]
Abstract
The weighted ensemble (WE) algorithm is gaining popularity as a rare event method for studying long timescale processes with molecular dynamics. WE is particularly useful for determining kinetic properties, such as rates of protein (un)folding and ligand (un)binding, where transition rates can be calculated from the flux of trajectories into a target basin of interest. However, this flux depends exponentially on the number of splitting events that a given trajectory experiences before reaching the target state and can vary by orders of magnitude between WE replicates. Markov state models (MSMs) are helpful tools to aggregate information across multiple WE simulations and have previously been shown to provide more accurate transition rates than WE alone. Discrete-time MSMs are models that coarsely describe the evolution of the system from one discrete state to the next using a discrete lag time, τ. When an MSM is built using conventional MD data, longer values of τ typically provide more accurate results. Combining WE simulations with Markov state modeling presents some additional challenges, especially when using a value of τ that exceeds the lag time between resampling steps in the WE algorithm, τWE. Here, we identify a source of bias that occurs when τ > τWE, which we refer to as "merging bias". We also propose an algorithm to eliminate the merging bias, which results in merging bias-corrected MSMs, or "MBC-MSMs". Using a simple model system, as well as a complex biomolecular example, we show that MBC-MSMs significantly outperform both τ = τWE MSMs and uncorrected MSMs at longer lag times.
Collapse
Affiliation(s)
- Samik Bose
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
- Department
of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Ceren Kilinc
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| | - Alex Dickson
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
- Department
of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
14
|
Han Y, Dawson JR, DeMarco KR, Rouen KC, Ngo K, Bekker S, Yarov-Yarovoy V, Clancy CE, Xiang YK, Ahn SH, Vorobyov I. Molecular simulations reveal intricate coupling between agonist-bound β-adrenergic receptors and G protein. iScience 2025; 28:111741. [PMID: 39898043 PMCID: PMC11787599 DOI: 10.1016/j.isci.2024.111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/24/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
G protein-coupled receptors (GPCRs) and G proteins transmit signals from hormones and neurotransmitters across cell membranes, initiating downstream signaling and modulating cellular behavior. Using advanced computer modeling and simulation, we identified atomistic-level structural, dynamic, and energetic mechanisms of norepinephrine (NE) and stimulatory G protein (Gs) interactions with β-adrenergic receptors (βARs), crucial GPCRs for heart function regulation and major drug targets. Our analysis revealed distinct binding behaviors of NE within β1AR and β2AR despite identical orthosteric binding pockets. β2AR had an additional binding site, explaining variations in NE binding affinities. Simulations showed significant differences in NE dissociation pathways and receptor interactions with the Gs. β1AR binds Gs more strongly, while β2AR induces greater conformational changes in the α subunit of Gs. Furthermore, GTP and GDP binding to Gs may disrupt coupling between NE and βAR, as well as between βAR and Gs. These findings may aid in designing precise βAR-targeted drugs.
Collapse
Affiliation(s)
- Yanxiao Han
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
| | - John R.D. Dawson
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Kevin R. DeMarco
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
| | - Kyle C. Rouen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Khoa Ngo
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Slava Bekker
- American River College, Sacramento, CA 95841, USA
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616, USA
- Center for Precision Medicine and Data Science, University of California, Davis, Davis, CA 95616, USA
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Center for Precision Medicine and Data Science, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
- VA Northern California Health Care System, Mather, CA 95655, USA
| | - Surl-Hee Ahn
- Department of Chemical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
15
|
Mitra S, Biswas R, Chakrabarty S. WeTICA: A directed search weighted ensemble based enhanced sampling method to estimate rare event kinetics in a reduced dimensional space. J Chem Phys 2025; 162:034106. [PMID: 39812249 DOI: 10.1063/5.0239713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Estimating rare event kinetics from molecular dynamics simulations is a non-trivial task despite the great advances in enhanced sampling methods. Weighted Ensemble (WE) simulation, a special class of enhanced sampling techniques, offers a way to directly calculate kinetic rate constants from biased trajectories without the need to modify the underlying energy landscape using bias potentials. Conventional WE algorithms use different binning schemes to partition the collective variable (CV) space separating the two metastable states of interest. In this work, we have developed a new "binless" WE simulation algorithm to bypass the hurdles of optimizing binning procedures. Our proposed protocol (WeTICA) uses a low-dimensional CV space to drive the WE simulation toward the specified target state. We have applied this new algorithm to recover the unfolding kinetics of three proteins: (A) TC5b Trp-cage mutant, (B) TC10b Trp-cage mutant, and (C) Protein G, with unfolding times spanning the range between 3 and 40 μs using projections along predefined fixed Time-lagged Independent Component Analysis (TICA) eigenvectors as CVs. Calculated unfolding times converge to the reported values with good accuracy with more than one order of magnitude less cumulative WE simulation time than the unfolding time scales with or without a priori knowledge of the CVs that can capture unfolding. Our algorithm can be used with other linear CVs, not limited to TICA. Moreover, the new walker selection criteria for resampling employed in this algorithm can be used on more sophisticated nonlinear CV space for further improvements of binless WE methods.
Collapse
Affiliation(s)
- Sudipta Mitra
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block-JD, Sector-III, Salt Lake, Kolkata 700106, India
| | - Ranjit Biswas
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block-JD, Sector-III, Salt Lake, Kolkata 700106, India
| | - Suman Chakrabarty
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Block-JD, Sector-III, Salt Lake, Kolkata 700106, India
| |
Collapse
|
16
|
Ma J, Ayres CM, Brambley CA, Chandran SS, Rosales TJ, Perera WWJG, Eldaly B, Murray WT, Corcelli SA, Kovrigin EL, Klebanoff CA, Baker BM. Dynamic allostery in the peptide/MHC complex enables TCR neoantigen selectivity. Nat Commun 2025; 16:849. [PMID: 39833157 PMCID: PMC11756396 DOI: 10.1038/s41467-025-56004-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
The inherent antigen cross-reactivity of the T cell receptor (TCR) is balanced by high specificity. Surprisingly, TCR specificity often manifests in ways not easily interpreted from static structures. Here we show that TCR discrimination between an HLA-A*03:01 (HLA-A3)-restricted public neoantigen and its wild-type (WT) counterpart emerges from distinct motions within the HLA-A3 peptide binding groove that vary with the identity of the peptide's first primary anchor. These motions create a dynamic gate that, in the presence of the WT peptide, impedes a large conformational change required for TCR binding. The neoantigen is insusceptible to this limiting dynamic, and, with the gate open, upon TCR binding the central tryptophan can transit underneath the peptide backbone to the opposing side of the HLA-A3 peptide binding groove. Our findings thus reveal a novel mechanism driving TCR specificity for a cancer neoantigen that is rooted in the dynamic and allosteric nature of peptide/MHC-I binding grooves, with implications for resolving long-standing and often confounding questions about T cell specificity.
Collapse
Affiliation(s)
- Jiaqi Ma
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - Cory M Ayres
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - Chad A Brambley
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - Smita S Chandran
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Center for Cell Engineering, MSKCC, New York, NY, USA
| | - Tatiana J Rosales
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - W W J Gihan Perera
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - Bassant Eldaly
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA
| | - William T Murray
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Center for Cell Engineering, MSKCC, New York, NY, USA
| | - Steven A Corcelli
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Evgenii L Kovrigin
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Christopher A Klebanoff
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Center for Cell Engineering, MSKCC, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, New York, NY, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA.
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
17
|
Walters BT, Patapoff AW, Kiefer JA, Wu P, Wang W. Integrating Hydrogen Exchange with Molecular Dynamics for Improved Ligand Binding Predictions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632795. [PMID: 39868224 PMCID: PMC11761012 DOI: 10.1101/2025.01.13.632795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
We introduce Hydrogen-Exchange Experimental Structure Prediction (HX-ESP), a method that integrates hydrogen exchange (HX) data with molecular dynamics (MD) simulations to accurately predict ligand binding modes, even for targets requiring significant conformational changes. Benchmarking HX-ESP by fitting two ligands to PAK1 and four ligands to MAP4K1 (HPK1), and comparing the results to X-ray crystallography structures, demonstrated that HX-ESP successfully identified binding modes across a range of affinities significantly outperforming flexible docking for ligands necessitating large conformational adjustments. By objectively guiding simulations with experimental HX data, HX-ESP overcomes the long timescales required for binding predictions using traditional MD. This advancement promises to enhance the accuracy of computational modeling in drug discovery, potentially accelerating the development of effective therapeutics.
Collapse
|
18
|
Ghysbrecht S, Donati L, Keller BG. Accuracy of Reaction Coordinate Based Rate Theories for Modelling Chemical Reactions: Insights From the Thermal Isomerization in Retinal. J Comput Chem 2025; 46:e27529. [PMID: 39659054 PMCID: PMC11632214 DOI: 10.1002/jcc.27529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 12/12/2024]
Abstract
Modern potential energy surfaces have shifted attention to molecular simulations of chemical reactions. While various methods can estimate rate constants for conformational transitions in molecular dynamics simulations, their applicability to studying chemical reactions remains uncertain due to the high and sharp energy barriers and complex reaction coordinates involved. This study focuses on the thermal cis-trans isomerization in retinal, employing molecular simulations and comparing rate constant estimates based on one-dimensional rate theories with those based on sampling transitions and grid-based models for low-dimensional collective variable spaces. Even though each individual method to estimate the rate passes its quality tests, the rate constant estimates exhibit considerable disparities. Rate constant estimates based on one-dimensional reaction coordinates prove challenging to converge, even if the reaction coordinate is optimized. However, consistent estimates of the rate constant are achieved by sampling transitions and by multi-dimensional grid-based models.
Collapse
Affiliation(s)
- Simon Ghysbrecht
- Department of Biology, Chemistry and PharmacyFreie Universität BerlinBerlinGermany
| | - Luca Donati
- Department of Mathematics and Computer ScienceFreie Universität BerlinBerlinGermany
- Modeling and Simulation of Complex ProcessesZuse Institute BerlinBerlinGermany
| | - Bettina G. Keller
- Department of Biology, Chemistry and PharmacyFreie Universität BerlinBerlinGermany
| |
Collapse
|
19
|
Braza MKE, Demir Ö, Ahn SH, Morris CK, Calvó-Tusell C, McGuire KL, de la Peña Avalos B, Carpenter MA, Chen Y, Casalino L, Aihara H, Herzik MA, Harris RS, Amaro RE. Regulatory interactions between APOBEC3B N- and C-terminal domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.628032. [PMID: 39713448 PMCID: PMC11661193 DOI: 10.1101/2024.12.11.628032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
APOBEC3B (A3B) is implicated in DNA mutations that facilitate tumor evolution. Although structures of its individual N- and C-terminal domains (NTD and CTD) have been resolved through X-ray crystallography, the full-length A3B (fl-A3B) structure remains elusive, limiting understanding of its dynamics and mechanisms. In particular, the APOBEC3B C-terminal domain (A3Bctd) active site is frequently closed in models and structures. In this study, we built several new models of fl-A3B using integrative structural biology methods and selected a top model for further dynamical investigation. We compared dynamics of the truncated (A3Bctd) to the fl-A3B via conventional and Gaussian accelerated molecular dynamics (MD) simulations. Subsequently, we employed weighted ensemble methods to explore the fl-A3B active site opening mechanism, finding that interactions at the NTD-CTD interface enhance the opening frequency of the fl-A3B active site. Our findings shed light on the structural dynamics of fl-A3B, which may offer new avenues for therapeutic intervention in cancer.
Collapse
Affiliation(s)
- Mac Kevin E Braza
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA
| | - Özlem Demir
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA
| | - Surl-Hee Ahn
- Department of Chemical Engineering, University of California, Davis, Davis, CA
| | - Clare K Morris
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA
| | - Carla Calvó-Tusell
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA
| | - Kelly L McGuire
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA
| | - Bárbara de la Peña Avalos
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX
| | - Michael A Carpenter
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX
| | - Yanjun Chen
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX
| | - Lorenzo Casalino
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Mark A Herzik
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX
| | - Rommie E Amaro
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA
| |
Collapse
|
20
|
Wang D, Tiwary P. Augmenting Human Expertise in Weighted Ensemble Simulations through Deep Learning-Based Information Bottleneck. J Chem Theory Comput 2024; 20:10371-10383. [PMID: 39589127 DOI: 10.1021/acs.jctc.4c00919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
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 chignolin 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.
Collapse
Affiliation(s)
- Dedi Wang
- Biophysics Program and Institute for Physical Science and Technology, University of Maryland, College Park, Maryland 20742, United States
| | - Pratyush Tiwary
- Department of Chemistry and Biochemistry and Institute for Physical Science and Technology, University of Maryland, College Park, Maryland 20742, United States
- University of Maryland Institute for Health Computing, Bethesda, Maryland 20852, United States
| |
Collapse
|
21
|
Ojha AA, Votapka LW, Amaro RE. Advances and Challenges in Milestoning Simulations for Drug-Target Kinetics. J Chem Theory Comput 2024; 20:9759-9769. [PMID: 39508322 PMCID: PMC11603602 DOI: 10.1021/acs.jctc.4c01108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Molecular dynamics simulations have become indispensable for exploring complex biological processes, yet their limitations in capturing rare events hinder our understanding of drug-target kinetics. In this Perspective, we investigate the domain of milestoning simulations to understand this challenge. The milestoning approach divides the phase space of the drug-target complex into discrete cells, offering extended time scale insights. This Perspective traces the history, applications, and future potential of milestoning simulations in the context of drug-target kinetics. It explores the fundamental principles of milestoning, highlighting the importance of probabilistic transitions and transition time independence. Markovian milestoning with Voronoi tessellations is revisited to address the traditional milestoning challenges. While observing the advancements in this field, this Perspective also addresses impending challenges in estimating drug-target unbinding rate constants through milestoning simulations, paving the way for more effective drug design strategies.
Collapse
Affiliation(s)
- Anupam Anand Ojha
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
- Center
for Computational Biology and Center for Computational Mathematics, Flatiron Institute, New York, New York 10010, United States
| | - Lane W. Votapka
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| | - Rommie E. Amaro
- Department
of Molecular Biology, University of California
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
22
|
Cummins D, Longstreth C, McCarty J. Analysis of transition rates from variational flooding using analytical theory. J Chem Phys 2024; 161:194104. [PMID: 39555758 DOI: 10.1063/5.0238289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024] Open
Abstract
Variational flooding is an enhanced sampling method for obtaining kinetic rates from molecular dynamics simulations. This method is inspired by the idea of conformational flooding that employs a boost potential acting along a chosen reaction coordinate to accelerate rare events. In this work, we show how the empirical distribution of crossing times from variational flooding simulations can be modeled with analytical Kramers' time-dependent rate (KTR) theory. An optimized bias potential that fills metastable free energy basins is constructed from the variationally enhanced sampling (VES) method. This VES-derived flooding potential is then augmented by a switching function that determines the fill level of the boost. Having a prescribed time-dependent fill rate of the flooding potential gives an analytical expression for the distribution of crossing times from KTR theory that is used to extract unbiased rates. In the case of a static boost potential, the distribution of barrier crossing times follows an expected exponential distribution, and unbiased rates are extracted from a series of boosted simulations at discrete fill levels. Introducing a time-dependent boost that increases the fill level gradually over the simulation time leads to a simplified procedure for fitting the biased distribution of crossing times to analytical theory. We demonstrate the approach for the paradigmatic cases of alanine dipeptide in vacuum, the asymmetric SN2 reaction, and the folding of chignolin in explicit solvent.
Collapse
Affiliation(s)
- David Cummins
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225, USA
| | - Carter Longstreth
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225, USA
| | - James McCarty
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225, USA
| |
Collapse
|
23
|
Wang D, Tiwary P. Augmenting Human Expertise in Weighted Ensemble Simulations through Deep Learning based Information Bottleneck. ARXIV 2024:arXiv:2406.14839v2. [PMID: 38947925 PMCID: PMC11213147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
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.
Collapse
Affiliation(s)
- Dedi Wang
- Biophysics Program and Institute for Physical Science and Technology, University of Maryland, College Park 20742, USA
| | - Pratyush Tiwary
- Department of Chemistry and Biochemistry and Institute for Physical Science and Technology, University of Maryland, College Park 20742, USA
- University of Maryland Institute for Health Computing, Bethesda 20852, USA
| |
Collapse
|
24
|
Fiorin G, Marinelli F, Forrest LR, Chen H, Chipot C, Kohlmeyer A, Santuz H, Hénin J. Expanded Functionality and Portability for the Colvars Library. J Phys Chem B 2024; 128:11108-11123. [PMID: 39501453 PMCID: PMC11572706 DOI: 10.1021/acs.jpcb.4c05604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024]
Abstract
Colvars is an open-source C++ library that provides a modular toolkit for collective-variable-based molecular simulations. It allows practitioners to easily create and implement descriptors that best fit a process of interest and to apply a wide range of biasing algorithms in collective variable space. This paper reviews several features and improvements to Colvars that were added since its original introduction. Special attention is given to contributions that significantly expanded the capabilities of this software or its distribution with major MD simulation packages. Collective variables can now be optimized either manually or by machine-learning methods, and the space of descriptors can be explored interactively using the graphical interface included in VMD. Beyond the spatial coordinates of individual molecules, Colvars can now apply biasing forces to mesoscale structures and alchemical degrees of freedom and perform simulations guided by experimental data within ensemble averages or probability distributions. It also features advanced computational schemes to boost the accuracy, robustness, and general applicability of simulation methods, including extended-system and multiple-walker adaptive biasing force, boundary conditions for metadynamics, replica exchange with biasing potentials, and adiabatic bias molecular dynamics. The library is made available directly within the main distributions of the academic software GROMACS, LAMMPS, NAMD, Tinker-HP, and VMD. The robustness of the software and the reliability of the results are ensured through the use of continuous integration with a test suite within the source repository.
Collapse
Affiliation(s)
- Giacomo Fiorin
- National
Institute of Neurological Disorders and Stroke, Bethesda, Maryland 20814, United States
- National
Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States
| | - Fabrizio Marinelli
- National
Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States
- Department
of Biophysics and Data Science Institute, Medical College of Wisconsin, Milwaukee, Wisconsin 53226-3548, United States
| | - Lucy R. Forrest
- National
Institute of Neurological Disorders and Stroke, Bethesda, Maryland 20814, United States
| | - Haochuan Chen
- Theoretical
and Computational Biophysics Group, Beckman Institute, and Department
of Physics, University of Illinois at Urbana−Champaign, Urbana, Illinois 61820, United States
| | - Christophe Chipot
- Theoretical
and Computational Biophysics Group, Beckman Institute, and Department
of Physics, University of Illinois at Urbana−Champaign, Urbana, Illinois 61820, United States
- Laboratoire
International Associé CNRS et University of Illinois at Urbana−Champaign,
UMR 7019, Université de Lorraine, 54506 Vandœuvre-lès-Nancy, France
- Department
of Biochemistry and Molecular Biology, The
University of Chicago, 929 E. 57th Street W225, Chicago, Illinois 60637, United States
- Department
of Chemistry, The University of Hawai’i
at Manoa, 2545 McCarthy
Mall, Honolulu, Hawaii 96822, United States
| | - Axel Kohlmeyer
- Institute
for Computational Molecular Science, Temple
University, Philadelphia, Pennsylvania 19122, United States
| | - Hubert Santuz
- Laboratoire
de Biochimie Théorique UPR 9080, Université Paris Cité, CNRS, 75005 Paris, France
| | - Jérôme Hénin
- Laboratoire
de Biochimie Théorique UPR 9080, Université Paris Cité, CNRS, 75005 Paris, France
| |
Collapse
|
25
|
Mitchell AR, Rotskoff GM. Committor Guided Estimates of Molecular Transition Rates. J Chem Theory Comput 2024; 20:9378-9393. [PMID: 39420582 DOI: 10.1021/acs.jctc.4c00997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The probability that a configuration of a physical system reacts, or transitions from one metastable state to another, is quantified by the committor function. This function contains richly detailed mechanistic information about transition pathways, but a full parametrization of the committor requires the construction of a high-dimensional function, a generically challenging task. Recent efforts to leverage neural networks as a means to solve high-dimensional partial differential equations, often called "physics-informed" machine learning, have brought the committor into computational reach. Here, we build on the semigroup approach to learning the committor and assess its utility for predicting dynamical quantities such as transition rates. We show that a careful reframing of the objective function and improved adaptive sampling strategies provide highly accurate representations of the committor. Furthermore, by directly applying the Hill relation, we show that these committors provide accurate transition rates for molecular systems.
Collapse
Affiliation(s)
- Andrew R Mitchell
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Grant M Rotskoff
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
26
|
Vithani N, Zhang S, Thompson JP, Patel LA, Demidov A, Xia J, Balaeff A, Mentes A, Arnautova YA, Kohlmann A, Lawson JD, Nicholls A, Skillman AG, LeBard DN. Exploration of Cryptic Pockets Using Enhanced Sampling Along Normal Modes: A Case Study of KRAS G12D. J Chem Inf Model 2024; 64:8258-8273. [PMID: 39419500 PMCID: PMC11558672 DOI: 10.1021/acs.jcim.4c01435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
Identification of cryptic pockets has the potential to open new therapeutic opportunities by discovering ligand binding sites that remain hidden in static apo structures of a target protein. Moreover, allosteric cryptic pockets can become valuable for designing target-selective ligands when the natural ligand binding sites are conserved in variants of a protein. For example, before an allosteric cryptic pocket was discovered, KRAS was considered undruggable due to its smooth surface and conservation of the GDP/GTP binding pocket across the wild type and oncogenic isoforms. Recent identification of the Switch-II cryptic pocket in the KRASG12C mutant and FDA approval of anticancer drugs targeting this site underscores the importance of cryptic pockets in solving pharmaceutical challenges. Here, we present a newly developed approach for the exploration of cryptic pockets using weighted ensemble molecular dynamics simulations with inherent normal modes as progress coordinates applied to the wild type KRAS and the G12D mutant. We performed extensive all-atomic simulations (>400 μs) with and without several cosolvents (xenon, ethanol, benzene), and analyzed trajectories using three distinct methods to search for potential binding pockets. These methods have been applied as a proof-of-concept to KRAS and have shown they can predict known cryptic binding sites. Furthermore, we performed ligand-binding simulations of a known inhibitor (MRTX1133) to shed light on the nature of cryptic pockets in KRASG12D and the role of conformational selection vs induced-fit mechanism in the formation of these cryptic pockets.
Collapse
Affiliation(s)
- Neha Vithani
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - She Zhang
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Jeffrey P. Thompson
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Lara A. Patel
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Alex Demidov
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Junchao Xia
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Alexander Balaeff
- Black
Diamond Therapeutics, Cambridge, Massachusetts 02142, United States
| | - Ahmet Mentes
- Black
Diamond Therapeutics, Cambridge, Massachusetts 02142, United States
| | | | - Anna Kohlmann
- Black
Diamond Therapeutics, Cambridge, Massachusetts 02142, United States
| | - J. David Lawson
- Mirati
Therapeutics, Inc., San Diego, California 92121, United States
| | - Anthony Nicholls
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | | | - David N. LeBard
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| |
Collapse
|
27
|
Choe S. Insights into Translocation of Arginine-Rich Cell-Penetrating Peptides across a Model Membrane. J Phys Chem B 2024; 128:10894-10903. [PMID: 39445646 DOI: 10.1021/acs.jpcb.4c04266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
It is well-known that membrane deformation and water pores contribute to the spontaneous translocation of arginine-rich cell-penetrating peptides (CPPs). We confirm this through the observation of the spontaneous translocation of single R9 (nona-arginine) and Tat (48-60) peptides across a model membrane using the weighted ensemble (WE) method within all-atom molecular dynamics (MD) simulations. Furthermore, we demonstrate that membrane deformation and the presence of a water pore reduce the effective charge of the CPP and the bending rigidity of the model membrane during translocation. We find that R9 disturbs the model membrane more than Tat (48-60), leading to more efficient translocation of R9 across the model membrane.
Collapse
Affiliation(s)
- Seungho Choe
- Department of Energy Science & Engineering, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea
- Energy Science & Engineering Research Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
28
|
Silvestrini ML, Solazzo R, Boral S, Cocco MJ, Closson JD, Masetti M, Gardner KH, Chong LT. Gating residues govern ligand unbinding kinetics from the buried cavity in HIF-2α PAS-B. Protein Sci 2024; 33:e5198. [PMID: 39467204 PMCID: PMC11516114 DOI: 10.1002/pro.5198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024]
Abstract
While transcription factors have been generally perceived as "undruggable," an exception is the HIF-2 hypoxia-inducible transcription factor, which contains an internal cavity that is sufficiently large to accommodate a range of small-molecules, including the therapeutically used inhibitor belzutifan. Given the relatively long ligand residence times of these small molecules and the lack of any experimentally observed pathway connecting the cavity to solvent, there has been great interest in understanding how these drug ligands exit the buried receptor cavity. Here, we focus on the relevant PAS-B domain of hypoxia-inducible factor 2α (HIF-2α) and examine how one such small molecule (THS-017) exits from the buried cavity within this domain on the seconds-timescale using atomistic simulations and ZZ-exchange NMR. To enable the simulations, we applied the weighted ensemble path sampling strategy, which generates continuous pathways for a rare-event process [e.g., ligand (un)binding] with rigorous kinetics in orders of magnitude less computing time compared to conventional simulations. Results reveal the formation of an encounter complex intermediate and two distinct classes of pathways for ligand exit. Based on these pathways, we identified two pairs of conformational gating residues in the receptor: one for the major class (N288 and S304) and another for the minor class (L272 and M309). ZZ-exchange NMR validated the kinetic importance of N288 for ligand unbinding. Our results provide an ideal simulation dataset for rational manipulation of ligand unbinding kinetics.
Collapse
Affiliation(s)
| | - Riccardo Solazzo
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐Università di BolognaBolognaItaly
| | - Soumendu Boral
- Structural Biology InitiativeCUNY Advanced Science Research CenterNew YorkNew YorkUSA
| | - Melanie J. Cocco
- Department of Pharmaceutical SciencesUniversity of California, IrvineIrvineCaliforniaUSA
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCaliforniaUSA
| | - Joseph D. Closson
- Structural Biology InitiativeCUNY Advanced Science Research CenterNew YorkNew YorkUSA
- PhD Program in BiochemistryCUNY Graduate CenterNew YorkNew YorkUSA
| | - Matteo Masetti
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐Università di BolognaBolognaItaly
| | - Kevin H. Gardner
- Structural Biology InitiativeCUNY Advanced Science Research CenterNew YorkNew YorkUSA
- Department of Chemistry and BiochemistryCity College of New YorkNew YorkNew YorkUSA
- PhD Programs in Biochemistry, Biology, and ChemistryCUNY Graduate CenterNew YorkNew YorkUSA
| | - Lillian T. Chong
- Department of ChemistryUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
29
|
Gupta A, Ma H, Ramanathan A, Zerze GH. A Deep Learning-Driven Sampling Technique to Explore the Phase Space of an RNA Stem-Loop. J Chem Theory Comput 2024; 20:9178-9189. [PMID: 39374435 DOI: 10.1021/acs.jctc.4c00669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
The folding and unfolding of RNA stem-loops are critical biological processes; however, their computational studies are often hampered by the ruggedness of their folding landscape, necessitating long simulation times at the atomistic scale. Here, we adapted DeepDriveMD (DDMD), an advanced deep learning-driven sampling technique originally developed for protein folding, to address the challenges of RNA stem-loop folding. Although tempering- and order parameter-based techniques are commonly used for similar rare-event problems, the computational costs or the need for a priori knowledge about the system often present a challenge in their effective use. DDMD overcomes these challenges by adaptively learning from an ensemble of running MD simulations using generic contact maps as the raw input. DeepDriveMD enables on-the-fly learning of a low-dimensional latent representation and guides the simulation toward the undersampled regions while optimizing the resources to explore the relevant parts of the phase space. We showed that DDMD estimates the free energy landscape of the RNA stem-loop reasonably well at room temperature. Our simulation framework runs at a constant temperature without external biasing potential, hence preserving the information on transition rates, with a computational cost much lower than that of the simulations performed with external biasing potentials. We also introduced a reweighting strategy for obtaining unbiased free energy surfaces and presented a qualitative analysis of the latent space. This analysis showed that the latent space captures the relevant slow degrees of freedom for the RNA folding problem of interest. Finally, throughout the manuscript, we outlined how different parameters are selected and optimized to adapt DDMD for this system. We believe this compendium of decision-making processes will help new users adapt this technique for the rare-event sampling problems of their interest.
Collapse
Affiliation(s)
- Ayush Gupta
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| | - Heng Ma
- Data Science and Learning Division, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Arvind Ramanathan
- Data Science and Learning Division, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Gül H Zerze
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
30
|
Poruthoor AJ, Stallone JJ, Miaro M, Sharma A, Grossfield A. System size effects on the free energy landscapes from molecular dynamics of phase-separating bilayers. J Chem Phys 2024; 161:145101. [PMID: 39382132 PMCID: PMC11829248 DOI: 10.1063/5.0225753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
The "lipid raft" hypothesis proposes that cell membranes contain distinct domains of varying lipid compositions, where "rafts" of ordered lipids and cholesterol coexist with disordered lipid regions. Experimental and theoretical phase diagrams of model membranes have revealed multiple coexisting phases. Molecular dynamics (MD) simulations can also capture spontaneous phase separation of bilayers. However, these methods merely determine the sign of the free energy change upon phase separation-whether or not it is favorable-but not the amplitude. Recently, we developed a workflow to compute the free energy of phase separation from MD simulations using the weighted ensemble method. However, while theoretical treatments generally focus on infinite systems and experimental measurements on mesoscopic to macroscopic systems, MD simulations are comparatively small. Therefore, if we are to put the results of these calculations into the appropriate context, we need to understand the effects the finite size of the simulation has on the computed free energy landscapes. In this study, we investigate this phenomenon by computing free energy profiles for a model phase-separating system as a function of system size, ranging from 324 to 10 110 lipids. The results suggest that, within the limits of statistical uncertainty, bulk-like behavior emerges once the systems contain roughly 4000 lipids.
Collapse
Affiliation(s)
- Ashlin J. Poruthoor
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Jack J. Stallone
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Megan Miaro
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Akshara Sharma
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Alan Grossfield
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
31
|
Qian R, Xue J, Xu Y, Huang J. Alchemical Transformations and Beyond: Recent Advances and Real-World Applications of Free Energy Calculations in Drug Discovery. J Chem Inf Model 2024; 64:7214-7237. [PMID: 39360948 DOI: 10.1021/acs.jcim.4c01024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Computational methods constitute efficient strategies for screening and optimizing potential drug molecules. A critical factor in this process is the binding affinity between candidate molecules and targets, quantified as binding free energy. Among various estimation methods, alchemical transformation methods stand out for their theoretical rigor. Despite challenges in force field accuracy and sampling efficiency, advancements in algorithms, software, and hardware have increased the application of free energy perturbation (FEP) calculations in the pharmaceutical industry. Here, we review the practical applications of FEP in drug discovery projects since 2018, covering both ligand-centric and residue-centric transformations. We show that relative binding free energy calculations have steadily achieved chemical accuracy in real-world applications. In addition, we discuss alternative physics-based simulation methods and the incorporation of deep learning into free energy calculations.
Collapse
Affiliation(s)
- Runtong Qian
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| | - Jing Xue
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| | - You Xu
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| | - Jing Huang
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
32
|
Yang DT, Goldberg AM, Chong LT. Rare-Event Sampling using a Reinforcement Learning-Based Weighted Ensemble Method. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617475. [PMID: 39416089 PMCID: PMC11482931 DOI: 10.1101/2024.10.09.617475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Despite the power of path sampling strategies in enabling simulations of rare events, such strategies have not reached their full potential. A common challenge that remains is the identification of a progress coordinate that captures the slow relevant motions of a rare event. Here we have developed a weighted ensemble (WE) path sampling strategy that exploits reinforcement learning to automatically identify an effective progress coordinate among a set of potential coordinates during a simulation. We apply our WE strategy with reinforcement learning to three benchmark systems: (i) an egg carton-shaped toy potential, (ii) an S-shaped toy potential, and (iii) a dimer of the HIV-1 capsid protein (C-terminal domain). To enable rapid testing of the latter system at the atomic level, we employed discrete-state synthetic molecular dynamics trajectories using a generative, fine-grained Markov state model that was based on extensive conventional simulations. Our results demonstrate that using concepts from reinforcement learning with a weighted ensemble of trajectories automatically identifies relevant progress co-ordinates among multiple candidates at a given time during a simulation. Due to the rigorous weighting of trajectories, the simulations maintain rigorous kinetics.
Collapse
Affiliation(s)
- Darian T. Yang
- Molecular Biophysics and Structural Biology Graduate Program, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, Pennsylvania 15260
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Alex M. Goldberg
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Lillian T. Chong
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
33
|
Zhang M, Wu H, Wang Y. Enhanced Sampling of Biomolecular Slow Conformational Transitions Using Adaptive Sampling and Machine Learning. J Chem Theory Comput 2024; 20:8569-8582. [PMID: 39301626 DOI: 10.1021/acs.jctc.4c00764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Biomolecular simulations often suffer from the "time scale problem", hindering the study of rare events occurring over extended time scales. Enhanced sampling techniques aim to alleviate this issue by accelerating conformational transitions, yet they typically necessitate well-defined collective variables (CVs), posing a significant challenge. Machine learning offers promising solutions but typically requires rich training data encompassing the entire free energy surface (FES). In this work, we introduce an automated iterative pipeline designed to mitigate these limitations. Our protocol first utilizes a CV-free count-based adaptive sampling method to generate a data set rich in rare events. From this data set, slow modes are identified using Koopman-reweighted time-lagged independent component analysis (KTICA), which are subsequently leveraged by on-the-fly probability enhanced sampling (OPES) to efficiently explore the FES. The effectiveness of our pipeline is demonstrated and further compared with the common Markov State Model (MSM) approach on two model systems with increasing complexity: alanine dipeptide (Ala2) and deca-alanine (Ala10), underscoring its applicability across diverse biomolecular simulations.
Collapse
Affiliation(s)
- Mingyuan Zhang
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Hao Wu
- School of Mathematical Sciences, Institute of Natural Sciences, and MOE-LSC, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
34
|
Capponi S, Wang S. AI in cellular engineering and reprogramming. Biophys J 2024; 123:2658-2670. [PMID: 38576162 PMCID: PMC11393708 DOI: 10.1016/j.bpj.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024] Open
Abstract
During the last decade, artificial intelligence (AI) has increasingly been applied in biophysics and related fields, including cellular engineering and reprogramming, offering novel approaches to understand, manipulate, and control cellular function. The potential of AI lies in its ability to analyze complex datasets and generate predictive models. AI algorithms can process large amounts of data from single-cell genomics and multiomic technologies, allowing researchers to gain mechanistic insights into the control of cell identity and function. By integrating and interpreting these complex datasets, AI can help identify key molecular events and regulatory pathways involved in cellular reprogramming. This knowledge can inform the design of precision engineering strategies, such as the development of new transcription factor and signaling molecule cocktails, to manipulate cell identity and drive authentic cell fate across lineage boundaries. Furthermore, when used in combination with computational methods, AI can accelerate and improve the analysis and understanding of the intricate relationships between genes, proteins, and cellular processes. In this review article, we explore the current state of AI applications in biophysics with a specific focus on cellular engineering and reprogramming. Then, we showcase a couple of recent applications where we combined machine learning with experimental and computational techniques. Finally, we briefly discuss the challenges and prospects of AI in cellular engineering and reprogramming, emphasizing the potential of these technologies to revolutionize our ability to engineer cells for a variety of applications, from disease modeling and drug discovery to regenerative medicine and biomanufacturing.
Collapse
Affiliation(s)
- Sara Capponi
- IBM Almaden Research Center, San Jose, California; Center for Cellular Construction, San Francisco, California.
| | - Shangying Wang
- Bay Area Institute of Science, Altos Labs, Redwood City, California.
| |
Collapse
|
35
|
Xu X, Closson JD, Marcelino LP, Favaro DC, Silvestrini ML, Solazzo R, Chong LT, Gardner KH. Identification of small-molecule ligand-binding sites on and in the ARNT PAS-B domain. J Biol Chem 2024; 300:107606. [PMID: 39059491 PMCID: PMC11381877 DOI: 10.1016/j.jbc.2024.107606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Transcription factors are challenging to target with small-molecule inhibitors due to their structural plasticity and lack of catalytic sites. Notable exceptions include naturally ligand-regulated transcription factors, including our prior work with the hypoxia-inducible factor (HIF)-2 transcription factor, showing that small-molecule binding within an internal pocket of the HIF-2α Per-Aryl hydrocarbon Receptor Nuclear Translocator (ARNT)-Sim (PAS)-B domain can disrupt its interactions with its dimerization partner, ARNT. Here, we explore the feasibility of targeting small molecules to the analogous ARNT PAS-B domain itself, potentially opening a promising route to modulate several ARNT-mediated signaling pathways. Using solution NMR fragment screening, we previously identified several compounds that bind ARNT PAS-B and, in certain cases, antagonize ARNT association with the transforming acidic coiled-coil containing protein 3 transcriptional coactivator. However, these ligands have only modest binding affinities, complicating characterization of their binding sites. We address this challenge by combining NMR, molecular dynamics simulations, and ensemble docking to identify ligand-binding "hotspots" on and within the ARNT PAS-B domain. Our data indicate that the two ARNT/transforming acidic coiled-coil containing protein 3 inhibitors, KG-548 and KG-655, bind to a β-sheet surface implicated in both HIF-2 dimerization and coactivator recruitment. Furthermore, while KG-548 binds exclusively to the β-sheet surface, KG-655 can additionally bind within a water-accessible internal cavity in ARNT PAS-B. Finally, KG-279, while not a coactivator inhibitor, exemplifies ligands that preferentially bind only to the internal cavity. All three ligands promoted ARNT PAS-B homodimerization, albeit to varying degrees. Taken together, our findings provide a comprehensive overview of ARNT PAS-B ligand-binding sites and may guide the development of more potent coactivator inhibitors for cellular and functional studies.
Collapse
Affiliation(s)
- Xingjian Xu
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA; PhD Program in Biochemistry, The Graduate Center, CUNY, New York, New York, USA
| | - Joseph D Closson
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA; PhD Program in Biochemistry, The Graduate Center, CUNY, New York, New York, USA
| | | | - Denize C Favaro
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA
| | - Marion L Silvestrini
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Riccardo Solazzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Bologna, Italy
| | - Lillian T Chong
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin H Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA; Department of Chemistry and Biochemistry, City College of New York, New York, New York, USA; PhD. Programs in Biochemistry, Chemistry and Biology, The Graduate Center, CUNY, New York, New York, USA.
| |
Collapse
|
36
|
Strahan J, Lorpaiboon C, Weare J, Dinner AR. BAD-NEUS: Rapidly converging trajectory stratification. J Chem Phys 2024; 161:084109. [PMID: 39185846 PMCID: PMC11349377 DOI: 10.1063/5.0215975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
An issue for molecular dynamics simulations is that events of interest often involve timescales that are much longer than the simulation time step, which is set by the fastest timescales of the model. Because of this timescale separation, direct simulation of many events is prohibitively computationally costly. This issue can be overcome by aggregating information from many relatively short simulations that sample segments of trajectories involving events of interest. This is the strategy of Markov state models (MSMs) and related approaches, but such methods suffer from approximation error because the variables defining the states generally do not capture the dynamics fully. By contrast, once converged, the weighted ensemble (WE) method aggregates information from trajectory segments so as to yield unbiased estimates of both thermodynamic and kinetic statistics. Unfortunately, errors decay no faster than unbiased simulation in WE as originally formulated and commonly deployed. Here, we introduce a theoretical framework for describing WE that shows that the introduction of an approximate stationary distribution on top of the stratification, as in nonequilibrium umbrella sampling (NEUS), accelerates convergence. Building on ideas from MSMs and related methods, we generalize the NEUS approach in such a way that the approximation error can be reduced systematically. We show that the improved algorithm can decrease the simulation time required to achieve the desired precision by orders of magnitude.
Collapse
Affiliation(s)
- John Strahan
- Department of Chemistry and James Franck Institute, University of Chicago, Chicago, Illinois 60637, USA
| | - Chatipat Lorpaiboon
- Department of Chemistry and James Franck Institute, University of Chicago, Chicago, Illinois 60637, USA
| | - Jonathan Weare
- Courant Institute of Mathematical Sciences, New York University, New York, New York 10012, USA
| | - Aaron R. Dinner
- Department of Chemistry and James Franck Institute, University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
37
|
Nuqui X, Casalino L, Zhou L, Shehata M, Wang A, Tse AL, Ojha AA, Kearns FL, Rosenfeld MA, Miller EH, Acreman CM, Ahn SH, Chandran K, McLellan JS, Amaro RE. Simulation-driven design of stabilized SARS-CoV-2 spike S2 immunogens. Nat Commun 2024; 15:7370. [PMID: 39191724 PMCID: PMC11350062 DOI: 10.1038/s41467-024-50976-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
The full-length prefusion-stabilized SARS-CoV-2 spike (S) is the principal antigen of COVID-19 vaccines. Vaccine efficacy has been impacted by emerging variants of concern that accumulate most of the sequence modifications in the immunodominant S1 subunit. S2, in contrast, is the most evolutionarily conserved region of the spike and can elicit broadly neutralizing and protective antibodies. Yet, S2's usage as an alternative vaccine strategy is hampered by its general instability. Here, we use a simulation-driven approach to design S2-only immunogens stabilized in a closed prefusion conformation. Molecular simulations provide a mechanistic characterization of the S2 trimer's opening, informing the design of tryptophan substitutions that impart kinetic and thermodynamic stabilization. Structural characterization via cryo-EM shows the molecular basis of S2 stabilization in the closed prefusion conformation. Informed by molecular simulations and corroborated by experiments, we report an engineered S2 immunogen that exhibits increased protein expression, superior thermostability, and preserved immunogenicity against sarbecoviruses.
Collapse
Affiliation(s)
- Xandra Nuqui
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Lorenzo Casalino
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Ling Zhou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Mohamed Shehata
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Albert Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexandra L Tse
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anupam A Ojha
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Fiona L Kearns
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Mia A Rosenfeld
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
- Laboratory of Computational Biology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emily Happy Miller
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Cory M Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Surl-Hee Ahn
- Department of Chemical Engineering, University of California Davis, Davis, CA, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
38
|
Yang D, Chong LT. WEDAP: A Python Package for Streamlined Plotting of Molecular Simulation Data. J Chem Inf Model 2024; 64:5749-5755. [PMID: 39013164 PMCID: PMC11323263 DOI: 10.1021/acs.jcim.4c00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/18/2024]
Abstract
Given the growing interest in path sampling methods for extending the time scales of molecular dynamics (MD) simulations, there has been great interest in software tools that streamline the generation of plots for monitoring the progress of large-scale simulations. Here, we present the WEDAP Python package for simplifying the analysis of data generated from either conventional MD simulations or the weighted ensemble (WE) path sampling method, as implemented in the widely used WESTPA software package. WEDAP facilitates (i) the parsing of WE simulation data stored in highly compressed, hierarchical HDF5 files and (ii) incorporates trajectory weights from WE simulations into all generated plots. Our Python package consists of multiple user-friendly interfaces: a command-line interface, a graphical user interface, and a Python application programming interface. We demonstrate the plotting features of WEDAP through a series of examples using data from WE and conventional MD simulations that focus on the HIV-1 capsid protein's C-terminal domain dimer as a showcase system. The source code for WEDAP is freely available on GitHub at https://github.com/chonglab-pitt/wedap.
Collapse
Affiliation(s)
- Darian
T. Yang
- Molecular
Biophysics and Structural Biology Graduate Program, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, Pennsylvania 15260, United States
- Department
of Structural Biology, University of Pittsburgh
School of Medicine, Pittsburgh, Pennsylvania 15260, United States
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Lillian T. Chong
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
39
|
Rosa-Raíces JL, Limmer DT. Variational time reversal for free-energy estimation in nonequilibrium steady states. Phys Rev E 2024; 110:024120. [PMID: 39295045 DOI: 10.1103/physreve.110.024120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/22/2024] [Indexed: 09/21/2024]
Abstract
Studying the structure of systems in nonequilibrium steady states necessitates tools that quantify population shifts and associated deformations of equilibrium free-energy landscapes under persistent currents. Within the framework of stochastic thermodynamics, we establish a variant of the Kawasaki-Crooks equality that relates nonequilibrium free-energy corrections in overdamped Langevin systems to heat dissipation statistics along time-reversed relaxation trajectories computable with molecular simulation. Using stochastic control theory, we arrive at a general variational approach to evaluate the Kawasaki-Crooks equality and use it to estimate distribution functions of order parameters in specific models of driven and active matter, attaining substantial improvement in accuracy over simple perturbative methods.
Collapse
Affiliation(s)
| | - David T Limmer
- Department of Chemistry, University of California, Berkeley, California 94720, USA
- Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
- Chemical Science Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
- Kavli Energy NanoScience Institute, Berkeley, California 94720, USA
| |
Collapse
|
40
|
Plotnikov D, Ahn SH. Optimization of the resampling method in the weighted ensemble simulation toolkit with parallelization and analysis (WESTPA). J Chem Phys 2024; 161:046101. [PMID: 39037142 DOI: 10.1063/5.0197141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Affiliation(s)
- Dennis Plotnikov
- Department of Chemical Engineering, University of California, Davis, Davis, California 95616, USA
| | - Surl-Hee Ahn
- Department of Chemical Engineering, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
41
|
Teng D, Mironenko AV, Voth GA. QM/CG-MM: Systematic Embedding of Quantum Mechanical Systems in a Coarse-Grained Environment with Accurate Electrostatics. J Phys Chem A 2024; 128:6061-6071. [PMID: 39016145 DOI: 10.1021/acs.jpca.4c02906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Quantum Mechanics/Molecular Mechanics (QM/MM) can describe chemical reactions in molecular dynamics (MD) simulations at a much lower cost than ab initio MD. Still, it is prohibitively expensive for many systems of interest because such systems usually require long simulations for sufficient statistical sampling. Additional MM degrees of freedom are often slow and numerous but secondary in interest. Coarse-graining (CG) is well-known to be able to speed up sampling through both reduction in simulation cost and the ability to accelerate the dynamics. Therefore, embedding a QM system in a CG environment can be a promising way of expediting sampling without compromising the information about the QM subsystem. Sinitskiy and Voth first proposed the theory of Quantum Mechanics/Coarse-grained Molecular Mechanics (QM/CG-MM) with a bottom-up CG mapping. Mironenko and Voth subsequently introduced the DFT-QM/CG-MM formalism to couple a Density Functional Theory (DFT) treated QM system and to an apolar environment. Here, we present a more complete theory that addresses MM environments with significant polarity by explicitly accounting for the electrostatic coupling. We demonstrate our QM/CG-MM method with a chloride-methyl chloride SN2 reaction system in acetone, which is sensitive to solvent polarity. The method accurately recapitulates the potential of mean force for the substitution reaction, and the reaction barrier from the best model agrees with the atomistic simulations within sampling error. These models also have generalizability. In two other reactive systems that they have not been trained on, the QM/CG-MM model still achieves the same level of agreement with the atomistic QM/MM models. Finally, we show that in these examples the speed-up in the sampling is proportional to the acceleration of the rotational dynamics of the solvent in the CG system.
Collapse
Affiliation(s)
- Da Teng
- Department of Chemistry, Chicago Center for Theoretical Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| | - Alexander V Mironenko
- Department of Chemistry, Chicago Center for Theoretical Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Gregory A Voth
- Department of Chemistry, Chicago Center for Theoretical Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
42
|
Lee S, Wang D, Seeliger MA, Tiwary P. Calculating Protein-Ligand Residence Times through State Predictive Information Bottleneck Based Enhanced Sampling. J Chem Theory Comput 2024; 20:6341-6349. [PMID: 38991145 PMCID: PMC11990086 DOI: 10.1021/acs.jctc.4c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
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.
Collapse
Affiliation(s)
- Suemin Lee
- Biophysics Program and Institute for Physical Science and Technology, University of Maryland, College Park 20742, USA
| | - Dedi Wang
- Biophysics Program and Institute for Physical Science and Technology, University of Maryland, College Park 20742, USA
| | - Markus A. Seeliger
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Pratyush Tiwary
- Biophysics Program and Institute for Physical Science and Technology, University of Maryland, College Park 20742, USA
- Department of Chemistry and Biochemistry and Institute for Physical Science and Technology, University of Maryland, College Park 20742, USA
- University of Maryland Institute for Health Computing, Bethesda, Maryland 20852, USA
| |
Collapse
|
43
|
Mazzaferro N, Sasmal S, Cossio P, Hocky GM. Good Rates From Bad Coordinates: The Exponential Average Time-dependent Rate Approach. J Chem Theory Comput 2024; 20:5901-5912. [PMID: 38954555 PMCID: PMC11270837 DOI: 10.1021/acs.jctc.4c00425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024]
Abstract
Our ability to calculate rate constants of biochemical processes using molecular dynamics simulations is severely limited by the fact that the time scales for reactions, or changes in conformational state, scale exponentially with the relevant free-energy barrier heights. In this work, we improve upon a recently proposed rate estimator that allows us to predict transition times with molecular dynamics simulations biased to rapidly explore one or several collective variables (CVs). This approach relies on the idea that not all bias goes into promoting transitions, and along with the rate, it estimates a concomitant scale factor for the bias termed the "CV biasing efficiency" γ. First, we demonstrate mathematically that our new formulation allows us to derive the commonly used Infrequent Metadynamics (iMetaD) estimator when using a perfect CV, where γ = 1. After testing it on a model potential, we then study the unfolding behavior of a previously well characterized coarse-grained protein, which is sufficiently complex that we can choose many different CVs to bias, but which is sufficiently simple that we are able to compute the unbiased rate directly. For this system, we demonstrate that predictions from our new Exponential Average Time-Dependent Rate (EATR) estimator converge to the true rate constant more rapidly as a function of bias deposition time than does the previous iMetaD approach, even for bias deposition times that are short. We also show that the γ parameter can serve as a good metric for assessing the quality of the biasing coordinate. We demonstrate that these results hold when applying the methods to an atomistic protein folding example. Finally, we demonstrate that our approach works when combining multiple less-than-optimal bias coordinates, and adapt our method to the related "OPES flooding" approach. Overall, our time-dependent rate approach offers a powerful framework for predicting rate constants from biased simulations.
Collapse
Affiliation(s)
- Nicodemo Mazzaferro
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Subarna Sasmal
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Pilar Cossio
- Center
for Computational Mathematics, Flatiron
Institute, New York, New York 10010, United States
- Center
for Computational Biology, Flatiron Institute, New York, New York 10010, United States
| | - Glen M. Hocky
- Department
of Chemistry, New York University, New York, New York 10003, United States
- Simons
Center for Computational Physical Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
44
|
Cerutti DS, Wiewiora R, Boothroyd S, Sherman W. STORMM: Structure and topology replica molecular mechanics for chemical simulations. J Chem Phys 2024; 161:032501. [PMID: 39007368 DOI: 10.1063/5.0211032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The Structure and TOpology Replica Molecular Mechanics (STORMM) code is a next-generation molecular simulation engine and associated libraries optimized for performance on fast, vectorized central processor units and graphics processing units (GPUs) with independent memory and tens of thousands of threads. STORMM is built to run thousands of independent molecular mechanical calculations on a single GPU with novel implementations that tune numerical precision, mathematical operations, and scarce on-chip memory resources to optimize throughput. The libraries are built around accessible classes with detailed documentation, supporting fine-grained parallelism and algorithm development as well as copying or swapping groups of systems on and off of the GPU. A primary intention of the STORMM libraries is to provide developers of atomic simulation methods with access to a high-performance molecular mechanics engine with extensive facilities to prototype and develop bespoke tools aimed toward drug discovery applications. In its present state, STORMM delivers molecular dynamics simulations of small molecules and small proteins in implicit solvent with tens to hundreds of times the throughput of conventional codes. The engineering paradigm transforms two of the most memory bandwidth-intensive aspects of condensed-phase dynamics, particle-mesh mapping, and valence interactions, into compute-bound problems for several times the scalability of existing programs. Numerical methods for compressing and streamlining the information present in stored coordinates and lookup tables are also presented, delivering improved accuracy over methods implemented in other molecular dynamics engines. The open-source code is released under the MIT license.
Collapse
Affiliation(s)
| | | | | | - Woody Sherman
- Psivant Therapeutics, Boston, Massachusetts 02210, USA
| |
Collapse
|
45
|
Wehrhan L, Keller BG. Prebound State Discovered in the Unbinding Pathway of Fluorinated Variants of the Trypsin-BPTI Complex Using Random Acceleration Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:5194-5206. [PMID: 38870039 PMCID: PMC11234359 DOI: 10.1021/acs.jcim.4c00338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The serine protease trypsin forms a tightly bound inhibitor complex with the bovine pancreatic trypsin inhibitor (BPTI). The complex is stabilized by the P1 residue Lys15, which interacts with negatively charged amino acids at the bottom of the S1 pocket. Truncating the P1 residue of wildtype BPTI to α-aminobutyric acid (Abu) leaves a complex with moderate inhibitor strength, which is held in place by additional hydrogen bonds at the protein-protein interface. Fluorination of the Abu residue partially restores the inhibitor strength. The mechanism with which fluorination can restore the inhibitor strength is unknown, and accurate computational investigation requires knowledge of the binding and unbinding pathways. The preferred unbinding pathway is likely to be complex, as encounter states have been described before, and unrestrained umbrella sampling simulations of these complexes suggest additional energetic minima. Here, we use random acceleration molecular dynamics to find a new metastable state in the unbinding pathway of Abu-BPTI variants and wildtype BPTI from trypsin, which we call the prebound state. The prebound state and the fully bound state differ by a substantial shift in the position, a slight shift in the orientation of the BPTI variants, and changes in the interaction pattern. Particularly important is the breaking of three hydrogen bonds around Arg17. Fluorination of the P1 residue lowers the energy barrier of the transition between the fully bound state and prebound state and also lowers the energy minimum of the prebound state. While the effect of fluorination is in general difficult to quantify, here, it is in part caused by favorable stabilization of a hydrogen bond between Gln194 and Cys14. The interaction pattern of the prebound state offers insights into the inhibitory mechanism of BPTI and might add valuable information for the design of serine protease inhibitors.
Collapse
Affiliation(s)
- Leon Wehrhan
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Arnimallee 22, Berlin 14195, Germany
| | - Bettina G Keller
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Arnimallee 22, Berlin 14195, Germany
| |
Collapse
|
46
|
Schäfer JL, Keller BG. Implementation of Girsanov Reweighting in OpenMM and Deeptime. J Phys Chem B 2024; 128:6014-6027. [PMID: 38865491 PMCID: PMC11215775 DOI: 10.1021/acs.jpcb.4c01702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024]
Abstract
Classical molecular dynamics (MD) simulations provide invaluable insights into complex molecular systems but face limitations in capturing phenomena occurring on time scales beyond their reach. To bridge this gap, various enhanced sampling techniques have been developed, which are complemented by reweighting techniques to recover the unbiased dynamics. Girsanov reweighting is a reweighting technique that reweights simulation paths, generated by a stochastic MD integrator, without evoking an effective model of the dynamics. Instead, it calculates the relative path probability density at the time resolution of the MD integrator. Efficient implementation of Girsanov reweighting requires that the reweighting factors are calculated on-the-fly during the simulations and thus needs to be implemented within the MD integrator. Here, we present a comprehensive guide for implementing Girsanov reweighting into MD simulations. We demonstrate the implementation in the MD simulation package OpenMM by extending the library openmmtools. Additionally, we implemented a reweighted Markov state model estimator within the time series analysis package Deeptime.
Collapse
Affiliation(s)
- Joana-Lysiane Schäfer
- Department of Biology, Chemistry, and
Pharmacy, Freie Universität Berlin, Berlin 14195, Germany
| | - Bettina G. Keller
- Department of Biology, Chemistry, and
Pharmacy, Freie Universität Berlin, Berlin 14195, Germany
| |
Collapse
|
47
|
Ghysbrecht S, Keller BG. Thermal isomerization rates in retinal analogues using Ab-Initio molecular dynamics. J Comput Chem 2024; 45:1390-1403. [PMID: 38414274 DOI: 10.1002/jcc.27332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/29/2024]
Abstract
For a detailed understanding of chemical processes in nature and industry, we need accurate models of chemical reactions in complex environments. While Eyring transition state theory is commonly used for modeling chemical reactions, it is most accurate for small molecules in the gas phase. A wide range of alternative rate theories exist that can better capture reactions involving complex molecules and environmental effects. However, they require that the chemical reaction is sampled by molecular dynamics simulations. This is a formidable challenge since the accessible simulation timescales are many orders of magnitude smaller than typical timescales of chemical reactions. To overcome these limitations, rare event methods involving enhanced molecular dynamics sampling are employed. In this work, thermal isomerization of retinal is studied using tight-binding density functional theory. Results from transition state theory are compared to those obtained from enhanced sampling. Rates obtained from dynamical reweighting using infrequent metadynamics simulations were in close agreement with those from transition state theory. Meanwhile, rates obtained from application of Kramers' rate equation to a sampled free energy profile along a torsional dihedral reaction coordinate were found to be up to three orders of magnitude higher. This discrepancy raises concerns about applying rate methods to one-dimensional reaction coordinates in chemical reactions.
Collapse
Affiliation(s)
- Simon Ghysbrecht
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Bettina G Keller
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
48
|
Xu X, Closson J, Marcelino LP, Favaro DC, Silvestrini ML, Solazzo R, Chong LT, Gardner KH. Identification of Small Molecule Ligand Binding Sites On and In the ARNT PAS-B Domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.03.565595. [PMID: 37961463 PMCID: PMC10635134 DOI: 10.1101/2023.11.03.565595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Transcription factors are generally challenging to target with small molecule inhibitors due to their structural plasticity and lack of catalytic sites. Notable exceptions include several naturally ligand-regulated transcription factors, including our prior work with the heterodimeric HIF-2 transcription factor which showed that small molecule binding within an internal pocket of the HIF-2α PAS-B domain can disrupt its interactions with its dimerization partner, ARNT. Here, we explore the feasibility of similarly targeting small molecules to the analogous ARNT PAS-B domain itself, potentially opening a promising route to simultaneously modulate several ARNT-mediated signaling pathways. Using solution NMR screening of an in-house fragment library, we previously identified several compounds that bind ARNT PAS-B and, in certain cases, antagonize ARNT association with the TACC3 transcriptional coactivator. However, these ligands have only modest binding affinities, complicating characterization of their binding sites. We address this challenge by combining NMR, MD simulations, and ensemble docking to identify ligand-binding 'hotspots' on and within the ARNT PAS-B domain. Our data indicate that the two ARNT/TACC3 inhibitors, KG-548 and KG-655, bind to a β-sheet surface implicated in both HIF-2 dimerization and coactivator recruitment. Furthermore, while KG-548 binds exclusively to the β-sheet surface, KG-655 can additionally bind within a water-accessible internal cavity in ARNT PAS-B. Finally, KG-279, while not a coactivator inhibitor, exemplifies ligands that preferentially bind only to the internal cavity. All three ligands promoted ARNT PAS-B homodimerization, albeit to varying degrees. Taken together, our findings provide a comprehensive overview of ARNT PAS-B ligand-binding sites and may guide the development of more potent coactivator inhibitors for cellular and functional studies.
Collapse
|
49
|
Abstract
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.
Collapse
Affiliation(s)
- Shams Mehdi
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, USA;
- Biophysics Program, University of Maryland, College Park, Maryland, USA
| | - Zachary Smith
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, USA;
- Biophysics Program, University of Maryland, College Park, Maryland, USA
| | - Lukas Herron
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, USA;
- Biophysics Program, University of Maryland, College Park, Maryland, USA
| | - Ziyue Zou
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Pratyush Tiwary
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, USA;
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
50
|
Brossard EE, Corcelli SA. Mechanism of Daunomycin Intercalation into DNA from Enhanced Sampling Simulations. J Phys Chem Lett 2024; 15:5770-5778. [PMID: 38776167 DOI: 10.1021/acs.jpclett.4c00961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Daunomycin is a widely used anticancer drug, yet the mechanism underlying how it binds to DNA remains contested. 469 all-atom trajectories of daunomycin binding to the DNA oligonucleotide d(GCG CAC GTG CGC) were collected using weighted ensemble (WE)-enhanced sampling. Mechanistic insights were revealed through analysis of the ensemble of trajectories. Initially, the binding process involves a ubiquitous hydrogen bond between the DNA backbone and the NH3+ group on daunomycin. During the binding process, most trajectories exhibited similar structural changes to DNA, including DNA base pair rise, bending, and minor groove width changes. Variability within the ensemble of binding trajectories illuminates differences in the orientation of daunomycin as it initially intercalates; around 10% of trajectories needed minimal rearrangement from intercalation to reaching the fully bound configuration, whereas most needed an additional 1-5 ns to rearrange. The results here emphasize the utility of generating an ensemble of trajectories to discern biomolecular binding mechanisms.
Collapse
Affiliation(s)
- E E Brossard
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - S A Corcelli
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|