1
|
Marasco M, Kirkpatrick J, Carlomagno T, Hub JS, Anselmi M. Phosphopeptide binding to the N-SH2 domain of tyrosine phosphatase SHP2 correlates with the unzipping of its central β-sheet. Comput Struct Biotechnol J 2024; 23:1169-1180. [PMID: 38510972 PMCID: PMC10951427 DOI: 10.1016/j.csbj.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
SHP2 is a tyrosine phosphatase that plays a regulatory role in multiple intracellular signaling cascades and is known to be oncogenic in certain contexts. In the absence of effectors, SHP2 adopts an autoinhibited conformation with its N-SH2 domain blocking the active site. Given the key role of N-SH2 in regulating SHP2, this domain has been extensively studied, often by X-ray crystallography. Using a combination of structural analyses and molecular dynamics (MD) simulations we show that the crystallographic environment can significantly influence the structure of the isolated N-SH2 domain, resulting in misleading interpretations. As an orthogonal method to X-ray crystallography, we use a combination of NMR spectroscopy and MD simulations to accurately determine the conformation of apo N-SH2 in solution. In contrast to earlier reports based on crystallographic data, our results indicate that apo N-SH2 in solution primarily adopts a conformation with a fully zipped central β-sheet, and that partial unzipping of this β-sheet is promoted by binding of either phosphopeptides or even phosphate/sulfate ions.
Collapse
Affiliation(s)
- Michelangelo Marasco
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Kirkpatrick
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK
| | - Teresa Carlomagno
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK
| | - Jochen S. Hub
- Theoretical Physics and Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | - Massimiliano Anselmi
- Theoretical Physics and Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
2
|
Versini R, Baaden M, Cavellini L, Cohen MM, Taly A, Fuchs PFJ. Lys716 in the transmembrane domain of yeast mitofusin Fzo1 modulates anchoring and fusion. Structure 2024; 32:1997-2012.e7. [PMID: 39299234 DOI: 10.1016/j.str.2024.08.017] [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: 12/22/2023] [Revised: 05/06/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
Outer mitochondrial membrane fusion, a vital cellular process, is mediated by mitofusins. However, the underlying molecular mechanism remains elusive. We have performed extensive multiscale molecular dynamics simulations to predict a model of the transmembrane (TM) domain of the yeast mitofusin Fzo1. Coarse-grained simulations of the two TM domain helices, TM1 and TM2, reveal a stable interface, which is controlled by the charge status of residue Lys716. Atomistic replica-exchange simulations further tune our model, which is confirmed by a remarkable agreement with an independent AlphaFold2 (AF2) prediction of Fzo1 in complex with its fusion partner Ugo1. Furthermore, the presence of the TM domain destabilizes the membrane, even more if Lys716 is charged, which can be an asset for initiating fusion. The functional role of Lys716 was confirmed with yeast experiments, which show that mutating Lys716 to a hydrophobic residue prevents mitochondrial fusion.
Collapse
Affiliation(s)
- Raphaëlle Versini
- Laboratoire de Biochimie Théorique, CNRS, Université Paris Cité, 75005 Paris, France; Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Marc Baaden
- Laboratoire de Biochimie Théorique, CNRS, Université Paris Cité, 75005 Paris, France
| | - Laetitia Cavellini
- Laboratoire de Biologie Cellulaire et Moléculaire des Eucaryotes, Institut de Biologie Physico-Chimique, UMR 8226, CNRS, Sorbonne Université, Paris, France
| | - Mickaël M Cohen
- Laboratoire de Biologie Cellulaire et Moléculaire des Eucaryotes, Institut de Biologie Physico-Chimique, UMR 8226, CNRS, Sorbonne Université, Paris, France
| | - Antoine Taly
- Laboratoire de Biochimie Théorique, CNRS, Université Paris Cité, 75005 Paris, France.
| | - Patrick F J Fuchs
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France; Université Paris Cité, 75006 Paris, France.
| |
Collapse
|
3
|
Tang Z, Fang Z, Wu X, Liu J, Tian L, Li X, Diao J, Ji B, Li D. Folding of N-terminally acetylated α-synuclein upon interaction with lipid membranes. Biophys J 2024; 123:3698-3720. [PMID: 39306670 DOI: 10.1016/j.bpj.2024.09.019] [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: 05/29/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
α-Synuclein (α-syn) is an abundant presynaptic neuronal protein whose aggregation is strongly associated with Parkinson's disease. It has been proposed that lipid membranes significantly affect α-syn's aggregation process. Extensive studies have been conducted to understand the interactions between α-syn and lipid membranes and have demonstrated that the N-terminus plays a critical role. However, the dynamics of the interactions and the conformational transitions of the N-terminus of α-syn at the atomistic scale details are still highly desired. In this study, we performed extensive enhanced sampling molecular dynamics simulations to quantify the folding and interactions of wild-type and N-terminally acetylated α-syn when interacting with lipid structures. We found that N-terminal acetylation significantly increases the helicity of the first few residues in solution or when interacting with lipid membranes. The observations in simulations showed that the binding of α-syn with lipid membranes mainly follows the induced-fit model, where the disordered α-syn binds with the lipid membrane through the electrostatic interactions and hydrophobic contacts with the packing defects; after stable insertion, N-terminal acetylation promotes the helical folding of the N-terminus to enhance the anchoring, thus increasing the binding affinity. We have shown the critical role of the first N-terminal residue methionine for recognition and anchoring to the negatively charged membrane. Although N-terminal acetylation neutralizes the positive charge of Met1 that may affect the electrostatic interactions of α-syn with membranes, the increase in helicity of the N-terminus should compensate for the binding affinity. This study provides detailed insight into the folding dynamics of α-syn's N-terminus with or without acetylation in solution and upon interaction with lipids, which clarifies how the N-terminal acetylation regulates the affinity of α-syn binding to lipid membranes. It also shows how packing defects and electrostatic effects coregulate the N-terminus of α-syn folding and its interaction with membranes.
Collapse
Affiliation(s)
- Zihan Tang
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Zhou Fang
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Xuwei Wu
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Jie Liu
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Liangfei Tian
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Xuejin Li
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Baohua Ji
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) and Wenzhou Institute of University of Chinese Academy of Science, Wenzhou, China
| | - Dechang Li
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Song Z, Tang H, Gatch A, Sun Y, Ding F. Islet amyloid polypeptide fibril catalyzes amyloid-β aggregation by promoting fibril nucleation rather than direct axial growth. Int J Biol Macromol 2024; 279:135137. [PMID: 39208885 PMCID: PMC11469950 DOI: 10.1016/j.ijbiomac.2024.135137] [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: 05/29/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Aberrant aggregation of amyloid-β (Aβ) and islet amyloid polypeptide (IAPP) into amyloid fibrils underlies the pathogenesis of Alzheimer's disease (AD) and type 2 diabetes (T2D), respectively. T2D significantly increases AD risk, with evidence suggesting that IAPP and Aβ co-aggregation and cross-seeding might contribute to the cross-talk between two diseases. Experimentally, preformed IAPP fibril seeds can accelerate Aβ aggregation, though the cross-seeding mechanism remains elusive. Here, we computationally demonstrated that Aβ monomer preferred to bind to the elongation ends of preformed IAPP fibrils. However, due to sequence mismatch, the Aβ monomer could not directly grow onto IAPP fibrils by forming multiple stable β-sheets with the exposed IAPP peptides. Conversely, in our control simulations of self-seeding, the Aβ monomer could axially grow on the Aβ fibril, forming parallel in-register β-sheets. Additionally, we showed that the IAPP fibril could catalyze Aβ fibril nucleation by promoting the formation of parallel in-register β-sheets in the C-terminus between bound Aβ peptides. This study enhances our understanding of the molecular interplay between Aβ and IAPP, shedding light on the cross-seeding mechanisms potentially linking T2D and AD. Our findings also underscore the importance of clearing IAPP deposits in T2D patients to mitigate AD risk.
Collapse
Affiliation(s)
- Zhiyuan Song
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Huayuan Tang
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States; Department of Engineering Mechanics, Hohai University, Nanjing 210098, China
| | - Adam Gatch
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Yunxiang Sun
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States; School of Physical Science and Technology, Ningbo University, Ningbo 315211, China
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States.
| |
Collapse
|
5
|
Zhou X, An L, Yang Y, Liu Z, Wang Y, Yao L. Positive activation entropy of Bacillus circulans xylanase catalyzed ONPX 2 hydrolysis: A mechanistic and engineering study. Int J Biol Macromol 2024; 282:137087. [PMID: 39489233 DOI: 10.1016/j.ijbiomac.2024.137087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Transition state (TS) stabilization by enzymes greatly accelerates catalytic reactions. For some enzymes, the TS complex has entropy higher than enzyme substrate (ES) complex. But the origin of favorable entropy remains unclear. In this work, we studied the mechanism of Bacillus Circulans xylanase (BCX) 11 catalyzed o-nitrophenyl β-xylobioside (ONPX2) glycoside hydrolysis. The catalytic reaction exhibits a positive activation entropy, and an increase in ionic strength leads to a decrease in entropy without affecting the activation free energy, indicating that the entropy is predominantly influenced by electrostatic forces. Moreover, NMR measurements of electrostatic attractions within the active site demonstrate a positive entropy, aligning with molecular dynamics (MD) simulations showing that electrostatic interactions contribute to the entropic stabilization of the TS complex. These findings suggest that the positive entropy primarily originates from alterations in electrostatic interactions due to the formation of the oxocarbenium ion at C1 in the TS. Differences of electrostatic interactions in ES and TS modify hydrogen bonding of surrounding residues in the active site which causes their side chain dynamics and thus conformational entropy changes. Residues critical for the positive activation entropy are identified. A new BCX mutant with an increased activation entropy and catalytic activity is found.
Collapse
Affiliation(s)
- Xuchen Zhou
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liaoyuan An
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; Shandong Energy Institute, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, Qingdao 266101, China
| | - Ying Yang
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; Shandong Energy Institute, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, Qingdao 266101, China
| | - Zhijun Liu
- National Facility for Protein Science, Zhangjiang Lab, Shanghai Advanced Research Institute, CAS, 201210, China
| | - Yefei Wang
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; Shandong Energy Institute, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, Qingdao 266101, China.
| | - Lishan Yao
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; Shandong Energy Institute, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, Qingdao 266101, China.
| |
Collapse
|
6
|
Quarta N, Bhandari TR, Girard M, Hellmann N, Schneider D. Monomer unfolding of a bacterial ESCRT-III superfamily member is coupled to oligomer disassembly. Protein Sci 2024; 33:e5187. [PMID: 39470325 PMCID: PMC11520248 DOI: 10.1002/pro.5187] [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: 05/17/2024] [Revised: 08/22/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024]
Abstract
The inner membrane associated protein of 30 kDa (IM30), a member of the endosomal sorting complex required for transport (ESCRT-III) superfamily, is crucially involved in the biogenesis and maintenance of thylakoid membranes in cyanobacteria and chloroplasts. In solution, IM30 assembles into various large oligomeric barrel- or tube-like structures, whereas upon membrane binding it forms large, flat carpet structures. Dynamic localization of the protein in solution, to membranes and changes of the oligomeric states are crucial for its in vivo function. ESCRT-III proteins are known to form oligomeric structures that are dynamically assembled from monomeric/smaller oligomeric proteins, and thus these smaller building blocks must be assembled sequentially in a highly orchestrated manner, a still poorly understood process. The impact of IM30 oligomerization on function remains difficult to study due to its high intrinsic tendency to homo-oligomerize. Here, we used molecular dynamics simulations to investigate the stability of individual helices in IM30 and identified unstable regions that may provide structural flexibility. Urea-mediated disassembly of the IM30 barrel structures was spectroscopically monitored, as well as changes in the protein's tertiary and secondary structure. The experimental data were finally compared to a three-state model that describes oligomer disassembly and monomer unfolding. In this study, we identified a highly stable conserved structural core of ESCRT-III proteins and discuss the advantages of having flexible intermediate structures and their putative relevance for ESCRT-III proteins.
Collapse
Affiliation(s)
- Ndjali Quarta
- Department of Chemistry – BiochemistryJohannes Gutenberg UniversityMainzGermany
| | | | - Martin Girard
- Max Planck Institute for Polymer ResearchMainzGermany
| | - Nadja Hellmann
- Department of Chemistry – BiochemistryJohannes Gutenberg UniversityMainzGermany
| | - Dirk Schneider
- Department of Chemistry – BiochemistryJohannes Gutenberg UniversityMainzGermany
- Institute of Molecular PhysiologyJohannes Gutenberg UniversityMainzGermany
| |
Collapse
|
7
|
Sarkar S, Gupta S, Mahato C, Das D, Mondal J. Elucidating ATP's role as solubilizer of biomolecular aggregate. eLife 2024; 13:RP99150. [PMID: 39475790 PMCID: PMC11524580 DOI: 10.7554/elife.99150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Proteins occurring in significantly high concentrations in cellular environments (over 100 mg/ml) and functioning in crowded cytoplasm, often face the prodigious challenges of aggregation which are the pathological hallmark of aging and are critically responsible for a wide spectrum of rising human diseases. Here, we combine a joint-venture of complementary wet-lab experiment and molecular simulation to discern the potential ability of adenosine triphosphate (ATP) as solubilizer of protein aggregates. We show that ATP prevents both condensation of aggregation-prone intrinsically disordered protein Aβ40 and promotes dissolution of preformed aggregates. Computer simulation links ATP's solubilizing role to its ability to modulate protein's structural plasticity by unwinding protein conformation. We show that ATP is positioned as a superior biological solubilizer of protein aggregates over traditional chemical hydrotropes, potentially holding promises in therapeutic interventions in protein-aggregation-related diseases. Going beyond its conventional activity as energy currency, the amphiphilic nature of ATP enables its protein-specific interaction that would enhance ATP's efficiency in cellular processes.
Collapse
Affiliation(s)
- Susmita Sarkar
- Tata Institute of Fundamental Research HyderabadHyderabadIndia
| | - Saurabh Gupta
- Indian Institute of Science Education and Research KolkataKolkataIndia
| | - Chiranjit Mahato
- Indian Institute of Science Education and Research KolkataKolkataIndia
| | - Dibyendu Das
- Indian Institute of Science Education and Research KolkataKolkataIndia
| | | |
Collapse
|
8
|
Waibl F, Casagrande F, Dey F, Riniker S. Validating Small-Molecule Force Fields for Macrocyclic Compounds Using NMR Data in Different Solvents. J Chem Inf Model 2024; 64:7938-7948. [PMID: 39405498 PMCID: PMC11523072 DOI: 10.1021/acs.jcim.4c01120] [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: 06/26/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024]
Abstract
Macrocycles are a promising class of compounds as therapeutics for difficult drug targets due to a favorable combination of properties: They often exhibit improved binding affinity compared to their linear counterparts due to their reduced conformational flexibility, while still being able to adapt to environments of different polarity. To assist in the rational design of macrocyclic drugs, there is need for computational methods that can accurately predict conformational ensembles of macrocycles in different environments. Molecular dynamics (MD) simulations remain one of the most accurate methods to predict ensembles quantitatively, although the accuracy is governed by the underlying force field. In this work, we benchmark four different force fields for their application to macrocycles by performing replica exchange with solute tempering (REST2) simulations of 11 macrocyclic compounds and comparing the obtained conformational ensembles to nuclear Overhauser effect (NOE) upper distance bounds from NMR experiments. Especially, the modern force fields OpenFF 2.0 and XFF yield good results, outperforming force fields like GAFF2 and OPLS/AA. We conclude that REST2 in combination with modern force fields can often produce accurate ensembles of macrocyclic compounds. However, we also highlight examples for which all examined force fields fail to produce ensembles that fulfill the experimental constraints.
Collapse
Affiliation(s)
- Franz Waibl
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| | - Fabio Casagrande
- Roche
Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Fabian Dey
- Roche
Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Sereina Riniker
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 2, 8093 Zürich, Switzerland
| |
Collapse
|
9
|
Amith W, Chen VT, Dutagaci B. Clustering of RNA Polymerase II C-Terminal Domain Models upon Phosphorylation. J Phys Chem B 2024; 128:10385-10396. [PMID: 39395159 PMCID: PMC11514005 DOI: 10.1021/acs.jpcb.4c04457] [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: 07/03/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
RNA polymerase II (Pol II) C-terminal domain (CTD) is known to have crucial roles in regulating transcription. CTD has also been highly recognized for undergoing phase separation, which is further associated with its regulatory functions. However, the molecular interactions that the CTD forms to induce clustering to drive phase separations and how the phosphorylation of the CTD affects clustering are not entirely known. In this work, we studied the concentrated solutions of two heptapeptide repeat (2CTD) models at different phosphorylation patterns and protein and ion concentrations using all-atom molecular dynamics simulations to investigate clustering behavior and molecular interactions driving the cluster formation. Our results show that salt concentration and phosphorylation patterns play an important role in determining the clustering pattern, specifically at low protein concentrations. The balance between inter- and intrapeptide interactions and counterion coordination together impact the clustering behavior upon phosphorylation.
Collapse
Affiliation(s)
- Weththasinghage
D. Amith
- Department
of Molecular and Cell Biology, University
of California, Merced, California 95343, United States
| | - Vincent T. Chen
- Department
of Molecular and Cell Biology, University
of California, Merced, California 95343, United States
| | - Bercem Dutagaci
- Department
of Molecular and Cell Biology, University
of California, Merced, California 95343, United States
- Health
Sciences Research Institute, University
of California, Merced, California 95343, United States
| |
Collapse
|
10
|
Singh B, Mondal A, Gaalswyk K, MacCallum JL, Perez A. MELD-Adapt: On-the-Fly Belief Updating in Integrative Molecular Dynamics. J Chem Theory Comput 2024; 20:9230-9242. [PMID: 39356805 DOI: 10.1021/acs.jctc.4c00690] [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: 10/04/2024]
Abstract
Integrative structural biology synergizes experimental data with computational methods to elucidate the structures and interactions within biomolecules, a task that becomes critical in the absence of high-resolution structural data. A challenging step for integrating the data is knowing the expected accuracy or belief in the dataset. We previously showed that the Modeling Employing Limited Data (MELD) approach succeeds at predicting structures and finding the best interpretation of the data when the initial belief is equal to or slightly lower than the real value. However, the initial belief might be unknown to the user, as it depends on both the technique and the system of study. Here we introduce MELD-Adapt, designed to dynamically evaluate and infer the reliability of input data while at the same time finding the best interpretation of the data and the structures compatible with it. We demonstrate the utility of this method across different systems, particularly emphasizing its capability to correct initial assumptions and identify the correct fraction of data to produce reliable structural models. The approach is tested with two benchmark sets: the folding of 12 proteins with coarse physical insights and the binding of peptides with varying affinities to the extraterminal domain using chemical shift perturbation data. We find that subtle differences in data structure (e.g., locally clustered or globally distributed), starting belief, and force field preferences can have an impact on the predictions, limiting the possibility of a transferable protocol across all systems and data types. Nonetheless, we find a wide range of initial setup conditions that will lead to successful sampling and identification of native states, leading to a robust pipeline. Furthermore, disagreements about how much data is enforced and satisfied rapidly serve to identify incorrect setup conditions.
Collapse
Affiliation(s)
- Bhumika Singh
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32611-7011, United States
| | - Arup Mondal
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32611-7011, United States
| | - Kari Gaalswyk
- Department of Chemistry, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Justin L MacCallum
- Department of Chemistry, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Alberto Perez
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, Florida 32611-7011, United States
| |
Collapse
|
11
|
Brough HDA, Cheneler D, Hardy JG. Progress in Multiscale Modeling of Silk Materials. Biomacromolecules 2024. [PMID: 39438248 DOI: 10.1021/acs.biomac.4c01122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
As a result of their hierarchical structure and biological processing, silk fibers rank among nature's most remarkable materials. The biocompatibility of silk-based materials and the exceptional mechanical properties of certain fibers has inspired the use of silk in numerous technical and medical applications. In recent years, computational modeling has clarified the relationship between the molecular architecture and emergent properties of silk fibers and has demonstrated predictive power in studies on novel biomaterials. Here, we review advances in modeling the structure and properties of natural and synthetic silk-based materials, from early structural studies of silkworm cocoon fibers to cutting-edge atomistic simulations of spider silk nanofibrils and the recent use of machine learning models. We explore applications of modeling across length scales: from quantum mechanical studies on model peptides, to atomistic and coarse-grained molecular dynamics simulations of silk proteins, to finite element analysis of spider webs. As computational power and algorithmic efficiency continue to advance, we expect multiscale modeling to become an indispensable tool for understanding nature's most impressive fibers and developing bioinspired functional materials.
Collapse
Affiliation(s)
- Harry D A Brough
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - David Cheneler
- School of Engineering, Lancaster University, Lancaster LA1 4YW, United Kingdom
- Materials Science Lancaster, Lancaster University, Lancaster, LA1 4YW, United Kingdom
| | - John G Hardy
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
- Materials Science Lancaster, Lancaster University, Lancaster, LA1 4YW, United Kingdom
| |
Collapse
|
12
|
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
|
13
|
Hwang W, Austin SL, Blondel A, Boittier ED, Boresch S, Buck M, Buckner J, Caflisch A, Chang HT, Cheng X, Choi YK, Chu JW, Crowley MF, Cui Q, Damjanovic A, Deng Y, Devereux M, Ding X, Feig MF, Gao J, Glowacki DR, Gonzales JE, Hamaneh MB, Harder ED, Hayes RL, Huang J, Huang Y, Hudson PS, Im W, Islam SM, Jiang W, Jones MR, Käser S, Kearns FL, Kern NR, Klauda JB, Lazaridis T, Lee J, Lemkul JA, Liu X, Luo Y, MacKerell AD, Major DT, Meuwly M, Nam K, Nilsson L, Ovchinnikov V, Paci E, Park S, Pastor RW, Pittman AR, Post CB, Prasad S, Pu J, Qi Y, Rathinavelan T, Roe DR, Roux B, Rowley CN, Shen J, Simmonett AC, Sodt AJ, Töpfer K, Upadhyay M, van der Vaart A, Vazquez-Salazar LI, Venable RM, Warrensford LC, Woodcock HL, Wu Y, Brooks CL, Brooks BR, Karplus M. CHARMM at 45: Enhancements in Accessibility, Functionality, and Speed. J Phys Chem B 2024; 128:9976-10042. [PMID: 39303207 PMCID: PMC11492285 DOI: 10.1021/acs.jpcb.4c04100] [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: 06/20/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
Since its inception nearly a half century ago, CHARMM has been playing a central role in computational biochemistry and biophysics. Commensurate with the developments in experimental research and advances in computer hardware, the range of methods and applicability of CHARMM have also grown. This review summarizes major developments that occurred after 2009 when the last review of CHARMM was published. They include the following: new faster simulation engines, accessible user interfaces for convenient workflows, and a vast array of simulation and analysis methods that encompass quantum mechanical, atomistic, and coarse-grained levels, as well as extensive coverage of force fields. In addition to providing the current snapshot of the CHARMM development, this review may serve as a starting point for exploring relevant theories and computational methods for tackling contemporary and emerging problems in biomolecular systems. CHARMM is freely available for academic and nonprofit research at https://academiccharmm.org/program.
Collapse
Affiliation(s)
- Wonmuk Hwang
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
- Department
of Materials Science and Engineering, Texas
A&M University, College Station, Texas 77843, United States
- Department
of Physics and Astronomy, Texas A&M
University, College Station, Texas 77843, United States
- Center for
AI and Natural Sciences, Korea Institute
for Advanced Study, Seoul 02455, Republic
of Korea
| | - Steven L. Austin
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Arnaud Blondel
- Institut
Pasteur, Université Paris Cité, CNRS UMR3825, Structural
Bioinformatics Unit, 28 rue du Dr. Roux F-75015 Paris, France
| | - Eric D. Boittier
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Stefan Boresch
- Faculty of
Chemistry, Department of Computational Biological Chemistry, University of Vienna, Wahringerstrasse 17, 1090 Vienna, Austria
| | - Matthias Buck
- Department
of Physiology and Biophysics, Case Western
Reserve University, School of Medicine, Cleveland, Ohio 44106, United States
| | - Joshua Buckner
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Amedeo Caflisch
- Department
of Biochemistry, University of Zürich, CH-8057 Zürich, Switzerland
| | - Hao-Ting Chang
- Institute
of Bioinformatics and Systems Biology, National
Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan, ROC
| | - Xi Cheng
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yeol Kyo Choi
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Jhih-Wei Chu
- Institute
of Bioinformatics and Systems Biology, Department of Biological Science
and Technology, Institute of Molecular Medicine and Bioengineering,
and Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung
University, Hsinchu 30010, Taiwan,
ROC
| | - Michael F. Crowley
- Renewable
Resources and Enabling Sciences Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| | - Qiang Cui
- Department
of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
- Department
of Physics, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
- Department
of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, Massachusetts 02215, United States
| | - Ana Damjanovic
- Department
of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department
of Physics and Astronomy, Johns Hopkins
University, Baltimore, Maryland 21218, United States
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yuqing Deng
- Shanghai
R&D Center, DP Technology, Ltd., Shanghai 201210, China
| | - Mike Devereux
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Xinqiang Ding
- Department
of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Michael F. Feig
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| | - Jiali Gao
- School
of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, China
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen, Guangdong 518055, China
- Department
of Chemistry and Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - David R. Glowacki
- CiTIUS
Centro Singular de Investigación en Tecnoloxías Intelixentes
da USC, 15705 Santiago de Compostela, Spain
| | - James E. Gonzales
- Department
of Biomedical Engineering, Texas A&M
University, College
Station, Texas 77843, United States
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Mehdi Bagerhi Hamaneh
- Department
of Physiology and Biophysics, Case Western
Reserve University, School of Medicine, Cleveland, Ohio 44106, United States
| | | | - Ryan L. Hayes
- Department
of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, Irvine, California 92697, United States
| | - Jing Huang
- Key Laboratory
of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Yandong Huang
- College
of Computer Engineering, Jimei University, Xiamen 361021, China
| | - Phillip S. Hudson
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
- Medicine
Design, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Wonpil Im
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Shahidul M. Islam
- Department
of Chemistry, Delaware State University, Dover, Delaware 19901, United States
| | - Wei Jiang
- Computational
Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Michael R. Jones
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Silvan Käser
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Fiona L. Kearns
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Nathan R. Kern
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Jeffery B. Klauda
- Department
of Chemical and Biomolecular Engineering, Institute for Physical Science
and Technology, Biophysics Program, University
of Maryland, College Park, Maryland 20742, United States
| | - Themis Lazaridis
- Department
of Chemistry, City College of New York, New York, New York 10031, United States
| | - Jinhyuk Lee
- Disease
Target Structure Research Center, Korea
Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
- Department
of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology, Daejeon 34141, Republic of Korea
| | - Justin A. Lemkul
- Department
of Biochemistry, Virginia Polytechnic Institute
and State University, Blacksburg, Virginia 24061, United States
| | - Xiaorong Liu
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yun Luo
- Department
of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California 91766, United States
| | - Alexander D. MacKerell
- Department
of Pharmaceutical Sciences, University of
Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Dan T. Major
- Department
of Chemistry and Institute for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Markus Meuwly
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
- Department
of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Kwangho Nam
- Department
of Chemistry and Biochemistry, University
of Texas at Arlington, Arlington, Texas 76019, United States
| | - Lennart Nilsson
- Karolinska
Institutet, Department of Biosciences and
Nutrition, SE-14183 Huddinge, Sweden
| | - Victor Ovchinnikov
- Harvard
University, Department of Chemistry
and Chemical Biology, Cambridge, Massachusetts 02138, United States
| | - Emanuele Paci
- Dipartimento
di Fisica e Astronomia, Universitá
di Bologna, Bologna 40127, Italy
| | - Soohyung Park
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Richard W. Pastor
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Amanda R. Pittman
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Carol Beth Post
- Borch Department
of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Samarjeet Prasad
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jingzhi Pu
- Department
of Chemistry and Chemical Biology, Indiana
University Indianapolis, Indianapolis, Indiana 46202, United States
| | - Yifei Qi
- School
of Pharmacy, Fudan University, Shanghai 201203, China
| | | | - Daniel R. Roe
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Benoit Roux
- Department
of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | | | - Jana Shen
- Department
of Pharmaceutical Sciences, University of
Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Andrew C. Simmonett
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Alexander J. Sodt
- Eunice
Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kai Töpfer
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Meenu Upadhyay
- Department
of Chemistry, University of Basel, Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Arjan van der Vaart
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | | | - Richard M. Venable
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Luke C. Warrensford
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - H. Lee Woodcock
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Yujin Wu
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Charles L. Brooks
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bernard R. Brooks
- Laboratory
of Computational Biology, National Heart
Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Martin Karplus
- Harvard
University, Department of Chemistry
and Chemical Biology, Cambridge, Massachusetts 02138, United States
- Laboratoire
de Chimie Biophysique, ISIS, Université
de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
14
|
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
|
15
|
Wang K, Huang Y, Wang Y, You Q, Wang L. Recent advances from computer-aided drug design to artificial intelligence drug design. RSC Med Chem 2024:d4md00522h. [PMID: 39493228 PMCID: PMC11523840 DOI: 10.1039/d4md00522h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Computer-aided drug design (CADD), a cornerstone of modern drug discovery, can predict how a molecular structure relates to its activity and interacts with its target using structure-based and ligand-based methods. Fueled by ever-increasing data availability and continuous model optimization, artificial intelligence drug design (AIDD), as an enhanced iteration of CADD, has thrived in the past decade. AIDD demonstrates unprecedented opportunities in protein folding, property prediction, and molecular generation. It can also facilitate target identification, high-throughput screening (HTS), and synthetic route prediction. With AIDD involved, the process of drug discovery is greatly accelerated. Notably, AIDD offers the potential to explore uncharted territories of chemical space beyond current knowledge. In this perspective, we began by briefly outlining the main workflows and components of CADD. Then through showcasing exemplary cases driven by AIDD in recent years, we describe the evolving role of artificial intelligence (AI) in drug discovery from three distinct stages, that is, chemical library screening, linker generation, and de novo molecular generation. In this process, we attempted to draw comparisons between the features of CADD and AIDD.
Collapse
Affiliation(s)
- Keran Wang
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| | - Yanwen Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Beijing 100191 China
| | - Yan Wang
- Department of Urology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine Shanghai 201203 China +86 13122152007
| | - Qidong You
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 210009 China +86 025 83271351 +86 15261483858
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University Nanjing 210009 China
| |
Collapse
|
16
|
Karrenbrock M, Borsatto A, Rizzi V, Lukauskis D, Aureli S, Luigi Gervasio F. Absolute Binding Free Energies with OneOPES. J Phys Chem Lett 2024; 15:9871-9880. [PMID: 39302888 PMCID: PMC11457222 DOI: 10.1021/acs.jpclett.4c02352] [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: 08/09/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
The calculation of absolute binding free energies (ABFEs) for protein-ligand systems has long been a challenge. Recently, refined force fields and algorithms have improved the quality of the ABFE calculations. However, achieving the level of accuracy required to inform drug discovery efforts remains difficult. Here, we present a transferable enhanced sampling strategy to accurately calculate absolute binding free energies using OneOPES with simple geometric collective variables. We tested the strategy on two protein targets, BRD4 and Hsp90, complexed with a total of 17 chemically diverse ligands, including both molecular fragments and drug-like molecules. Our results show that OneOPES accurately predicts protein-ligand binding affinities with a mean unsigned error within 1 kcal mol-1 of experimentally determined free energies, without the need to tailor the collective variables to each system. Furthermore, our strategy effectively samples different ligand binding modes and consistently matches the experimentally determined structures regardless of the initial protein-ligand configuration. Our results suggest that the proposed OneOPES strategy can be used to inform lead optimization campaigns in drug discovery and to study protein-ligand binding and unbinding mechanisms.
Collapse
Affiliation(s)
- Maurice Karrenbrock
- School
of Pharmaceutical Sciences, University of
Geneva, Rue Michel-Servet 1, CH-1206 Geneva, CH
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, CH
- Swiss
Bioinformatics Institute, University of
Geneva, CH-1206 Geneva, CH
| | - Alberto Borsatto
- School
of Pharmaceutical Sciences, University of
Geneva, Rue Michel-Servet 1, CH-1206 Geneva, CH
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, CH
- Swiss
Bioinformatics Institute, University of
Geneva, CH-1206 Geneva, CH
| | - Valerio Rizzi
- School
of Pharmaceutical Sciences, University of
Geneva, Rue Michel-Servet 1, CH-1206 Geneva, CH
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, CH
- Swiss
Bioinformatics Institute, University of
Geneva, CH-1206 Geneva, CH
| | - Dominykas Lukauskis
- Chemistry
Department, University College London (UCL), WC1E 6BT London, U.K.
| | - Simone Aureli
- School
of Pharmaceutical Sciences, University of
Geneva, Rue Michel-Servet 1, CH-1206 Geneva, CH
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, CH
- Swiss
Bioinformatics Institute, University of
Geneva, CH-1206 Geneva, CH
| | - Francesco Luigi Gervasio
- School
of Pharmaceutical Sciences, University of
Geneva, Rue Michel-Servet 1, CH-1206 Geneva, CH
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, CH
- Swiss
Bioinformatics Institute, University of
Geneva, CH-1206 Geneva, CH
- Chemistry
Department, University College London (UCL), WC1E 6BT London, U.K.
| |
Collapse
|
17
|
Sanbonmatsu K. Supercomputing in the biological sciences: Toward Zettascale and Yottascale simulations. Curr Opin Struct Biol 2024; 88:102889. [PMID: 39163795 DOI: 10.1016/j.sbi.2024.102889] [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: 05/04/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 08/22/2024]
Abstract
Molecular simulations of biological systems tend to be significantly more compute-intensive than those in materials science and astrophysics, due to important contributions of long-range electrostatic forces and large numbers of time steps (>1E9) required. Simulations of biomolecular complexes of microseconds to milliseconds are considered state-of-the-art today. However, these time scales are miniscule in comparison to physiological time scales relevant to molecular machine activity, drug action, and elongation cycles for protein synthesis, RNA synthesis, and DNA synthesis (seconds to days). While an exascale supercomputer has simulated an entire virus for nanoseconds, this supercomputer would need to be 10 billion times faster to simulate that virus for 3 hours of physiological time, demonstrating the insatiable need for computing power. With growing interest in computational drug design from the pharmaceutical sector, the biological sciences are positioned to be an industry driver in computing.
Collapse
Affiliation(s)
- Karissa Sanbonmatsu
- Los Alamos National Laboratory, United States; New Mexico Consortium, New Mexico.
| |
Collapse
|
18
|
Vollmers L, Zacharias M. Advanced sampling simulations of coupled folding and binding of phage P22 N-peptide to boxB RNA. Biophys J 2024; 123:3463-3477. [PMID: 39210596 PMCID: PMC11480772 DOI: 10.1016/j.bpj.2024.08.022] [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: 05/18/2024] [Revised: 07/08/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Protein-RNA interactions are crucially important for numerous cellular processes and often involve coupled folding and binding of peptide segments upon association. The Nut-utilization site (N)-protein of bacteriophages contains an N-terminal arginine-rich motif that undergoes such a folding transition upon binding to the boxB RNA hairpin loop target structure. Molecular dynamics free energy simulations were used to calculate the absolute binding free energy of the N-peptide of bacteriophage P22 in complex with the boxB RNA hairpin motif at different salt concentrations and using two different water force field models. We obtained good agreement with experiment also at different salt concentrations for the TIP4P-D water model that has a stabilizing effect on unfolded protein structures. It allowed us to estimate the free energy contribution resulting from restricting the molecules' spatial and conformational freedom upon binding, which makes a large opposing contribution to binding. In a second set of umbrella sampling simulations to dissociate/associate the complex along a separation coordinate, we analyzed the onset of preorientation of the N-peptide and onset of structure formation relative to the RNA and its dependence on the salt concentration. Peptide orientation and conformational transitions are significantly coupled to the first contact formation between peptide and RNA. The initial contacts are mostly formed between peptide residues and the boxB hairpin loop nucleotides. A complete transition to an α-helical bound peptide conformation occurs only at a late stage of the binding process a few angstroms before the complexed state has been reached. However, the N-peptide orients also at distances beyond the contact distance such that the sizable positive charge points toward the RNA's center-of-mass. Our result may have important implications for understanding protein- and peptide-RNA complex formation frequently involving coupled folding and association processes.
Collapse
Affiliation(s)
- Luis Vollmers
- Physics Department and Center of Protein Assemblies, Technical University Munich, Garching, Germany
| | - Martin Zacharias
- Physics Department and Center of Protein Assemblies, Technical University Munich, Garching, Germany.
| |
Collapse
|
19
|
Fan J, Li Z, Alcaide E, Ke G, Huang H, E W. Accurate Conformation Sampling via Protein Structural Diffusion. J Chem Inf Model 2024. [PMID: 39340358 DOI: 10.1021/acs.jcim.4c00928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Accurate sampling of protein conformations is pivotal for advances in biology and medicine. Although there has been tremendous progress in protein structure prediction in recent years due to deep learning, models that can predict the different stable conformations of proteins with high accuracy and structural validity are still lacking. Here, we introduce UFConf, a cutting-edge approach designed for robust sampling of diverse protein conformations based solely on amino acid sequences. This method transforms AlphaFold2 into a diffusion model by implementing a conformation-based diffusion process and adapting the architecture to process diffused inputs effectively. To counteract the inherent conformational bias in the Protein Data Bank, we developed a novel hierarchical reweighting protocol based on structural clustering. Our evaluations demonstrate that UFConf outperforms existing methods in terms of successful sampling and structural validity. The comparisons with long-time molecular dynamics show that UFConf can overcome the energy barrier existing in molecular dynamics simulations and perform more efficient sampling. Furthermore, We showcase UFConf's utility in drug discovery through its application in neural protein-ligand docking. In a blind test, it accurately predicted a novel protein-ligand complex, underscoring its potential to impact real-world biological research. Additionally, we present other modes of sampling using UFConf, including partial sampling with fixed motif, Langevin dynamics, and structural interpolation.
Collapse
Affiliation(s)
- Jiahao Fan
- School of Physics, Peking University, Beijing 100871, China
- DP Technology, Beijing 100080, China
| | - Ziyao Li
- DP Technology, Beijing 100080, China
- Center for Data Science, Peking University, Beijing 100871, China
| | - Eric Alcaide
- DP Technology, Beijing 100080, China
- University of Barcelona, Barcelona 08007, Spain
| | - Guolin Ke
- DP Technology, Beijing 100080, China
| | - Huaqing Huang
- School of Physics, Peking University, Beijing 100871, China
| | - Weinan E
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
20
|
Tsai HC, Xu J, Guo Z, Yi Y, Tian C, Que X, Giese T, Lee TS, York DM, Ganguly A, Pan A. Improvements in Precision of Relative Binding Free Energy Calculations Afforded by the Alchemical Enhanced Sampling (ACES) Approach. J Chem Inf Model 2024; 64:7046-7055. [PMID: 39225694 PMCID: PMC11542680 DOI: 10.1021/acs.jcim.4c00464] [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: 09/04/2024]
Abstract
Accurate in silico predictions of how strongly small molecules bind to proteins, such as those afforded by relative binding free energy (RBFE) calculations, can greatly increase the efficiency of the hit-to-lead and lead optimization stages of the drug discovery process. The success of such calculations, however, relies heavily on their precision. Here, we show that a recently developed alchemical enhanced sampling (ACES) approach can consistently improve the precision of RBFE calculations on a large and diverse set of proteins and small molecule ligands. The addition of ACES to conventional RBFE calculations lowered the average hysteresis by over 35% (0.3-0.4 kcal/mol) and the average replicate spread by over 25% (0.2-0.3 kcal/mol) across a set of 10 protein targets and 213 small molecules while maintaining similar or improved accuracy. We show in atomic detail how ACES improved convergence of several representative RBFE calculations through enhancing the sampling of important slowly transitioning ligand degrees of freedom.
Collapse
Affiliation(s)
- Hsu-Chun Tsai
- TandemAI, New York, NY 10036, United States
- Laboratory for Biomolecular Simulation Research, Institute for Quantitative Biomedicine, and Department of Chemistry and Chemical Biology, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - James Xu
- TandemAI, New York, NY 10036, United States
| | - Zhenyu Guo
- TandemAI, New York, NY 10036, United States
| | - Yinhui Yi
- TandemAI, New York, NY 10036, United States
| | - Chuan Tian
- TandemAI, New York, NY 10036, United States
| | - Xinyu Que
- TandemAI, New York, NY 10036, United States
- The work was done while he was working at TandemAI
| | - Timothy Giese
- Laboratory for Biomolecular Simulation Research, Institute for Quantitative Biomedicine, and Department of Chemistry and Chemical Biology, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Tai-Sung Lee
- Laboratory for Biomolecular Simulation Research, Institute for Quantitative Biomedicine, and Department of Chemistry and Chemical Biology, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Darrin M. York
- Laboratory for Biomolecular Simulation Research, Institute for Quantitative Biomedicine, and Department of Chemistry and Chemical Biology, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | | | - Albert Pan
- TandemAI, New York, NY 10036, United States
| |
Collapse
|
21
|
Kamboukos A, Williams-Noonan BJ, Charchar P, Yarovsky I, Todorova N. Graphitic nanoflakes modulate the structure and binding of human amylin. NANOSCALE 2024; 16:16870-16886. [PMID: 39219407 DOI: 10.1039/d4nr01315h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Human amylin is an inherently disordered protein whose ability to form amyloid fibrils is linked to the onset of type II diabetes. Graphitic nanomaterials have potential in managing amyloid diseases as they can disrupt protein aggregation processes in biological settings, but optimising these materials to prevent fibrillation is challenging. Here, we employ bias-exchange molecular dynamics simulations to systematically study the structure and adsorption preferences of amylin on graphitic nanoflakes that vary in their physical dimensions and surface functionalisation. Our findings reveal that nanoflake size and surface oxidation both influence the structure and adsorption preferences of amylin. The purely hydrophobic substrate of pristine graphene (PG) nanoflakes encourages non-specific protein adsorption, leading to unrestricted lateral mobility once amylin adheres to the surface. Particularly on larger PG nanoflakes, this induces structural changes in amylin that may promote fibril formation, such as the loss of native helical content and an increase in β-sheet character. In contrast, oxidised graphene nanoflakes form hydrogen bonds between surface oxygen sites and amylin, and as such restricting protein mobility. Reduced graphene oxide (rGO) flakes, featuring lower amounts of surface oxidation, are amphiphilic and exhibit substantial regions of bare carbon which promote protein binding and reduced conformational flexibility, leading to conservation of the native structure of amylin. In comparison, graphene oxide (GO) nanoflakes, which are predominantly hydrophilic and have a high degree of surface oxidation, facilitate considerable protein structural variability, resulting in substantial contact area between the protein and GO, and subsequent protein unfolding. Our results indicate that tailoring the size, oxygen concentration and surface patterning of graphitic nanoflakes can lead to specific and robust protein binding, ultimately influencing the likelihood of fibril formation. These atomistic insights provide key design considerations for the development of graphitic nanoflakes that can modulate protein aggregation by sequestering protein monomers in the biological environment and inhibit conformational changes linked to amyloid fibril formation.
Collapse
Affiliation(s)
- Alexa Kamboukos
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia.
| | - Billy J Williams-Noonan
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia.
- School of Science, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Patrick Charchar
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia.
| | - Irene Yarovsky
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia.
| | - Nevena Todorova
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia.
| |
Collapse
|
22
|
Zhang H, Fichthorn KA. Structural classification of Ag and Cu nanocrystals with machine learning. NANOSCALE 2024; 16:17154-17164. [PMID: 39192812 DOI: 10.1039/d4nr02531h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
We use machine learning (ML) to classify the structures of mono-metallic Cu and Ag nanoparticles. Our datasets comprise a broad range of structures - both crystalline and amorphous - derived from parallel-tempering molecular dynamics simulations of nanoparticles in the 100-200 atom size range. We construct nanoparticle features using common neighbor analysis (CNA) signatures, and we utilize principal component analysis to reduce the dimensionality of the CNA feature set. To sort the nanoparticles into structural classes, we employed both K-means clustering and the Gaussian mixture model (GMM). We evaluated the performance of the clustering algorithms through the gap statistic and silhouette score, as well as by analysis of the CNA signatures. For Ag, we found five structural classes, with 14 detailed sub-classes, while for Cu, we found two broad classes (crystalline and amorphous), with the same five classes as for Ag, and 15 detailed sub-classes. Our results demonstrate that these ML methods are effective in identifying and categorizing nanoparticle structures to different levels of complexity, enabling us to classify nanoparticles into distinct and physically relevant structural classes with high accuracy. This capability is important for understanding nanoparticle properties and potential applications.
Collapse
Affiliation(s)
- Huaizhong Zhang
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Kristen A Fichthorn
- Department of Chemical Engineering and Department of Physics, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
23
|
Sun Y. Efficient acceleration of the convergence of the minimum free energy path via a path-planning generated initial guess. J Comput Chem 2024. [PMID: 39291721 DOI: 10.1002/jcc.27504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/25/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
We demonstrate that combining a shifted clustering algorithm with a fast-marching-based algorithm can generate accurate approximations of the minimum energy path (MEP) given a free energy landscape (FEL). Using this approximation as the initial guess for the MEP, followed by further refinement with the string method (referred to as the fast marching tree (FMT)-string combined approach), significantly reduces the number of iterations required for MEP convergence. This approach saves substantial time compared to using linear interpolation (LI) for the initial guess. Our method offers a viable solution for obtaining an effective initial guess of the MEP when an approximate or converged FEL is available. This work highlights the potential of applying FMT-based approaches to extract the MEP in chemical reactions.
Collapse
Affiliation(s)
- Yi Sun
- Department of Chemistry, Chicago Center for Theoretical Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
24
|
Bastida A, Zúñiga J, Fogolari F, Soler MA. Statistical accuracy of molecular dynamics-based methods for sampling conformational ensembles of disordered proteins. Phys Chem Chem Phys 2024; 26:23213-23227. [PMID: 39190324 DOI: 10.1039/d4cp02564d] [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: 08/28/2024]
Abstract
The characterization of the statistical ensemble of conformations of intrinsically disordered regions (IDRs) is a great challenge both from experimental and computational points of view. In this respect, a number of protocols have been developed using molecular dynamics (MD) simulations to sample the huge conformational space of the molecule. In this work, we consider one of the best methods available, replica exchange solute tempering (REST), as a reference to compare the results obtained using this method with the results obtained using other methods, in terms of experimentally measurable quantities. Along with the methods assessed, we propose here a novel protocol called probabilistic MD chain growth (PMD-CG), which combines the flexible-meccano and hierarchical chain growth methods with the statistical data obtained from tripeptide MD trajectories as the starting point. The system chosen for testing is a 20-residue region from the C-terminal domain of the p53 tumor suppressor protein (p53-CTD). Our results show that PMD-CG provides an ensemble of conformations extremely quickly, after suitable computation of the conformational pool for all peptide triplets of the IDR sequence. The measurable quantities computed on the ensemble of conformations agree well with those based on the REST conformational ensemble.
Collapse
Affiliation(s)
- Adolfo Bastida
- Departamento de Química Física, Universidad de Murcia, 30100 Murcia, Spain.
| | - José Zúñiga
- Departamento de Química Física, Universidad de Murcia, 30100 Murcia, Spain.
| | - Federico Fogolari
- Dipartimento di Scienze Matematiche, Informatiche e Fisiche, Università di Udine, 33100 Udine, Italy.
| | - Miguel A Soler
- Dipartimento di Scienze Matematiche, Informatiche e Fisiche, Università di Udine, 33100 Udine, Italy.
| |
Collapse
|
25
|
Yao Y, Liu R, Li W, Huang W, Lai Y, Luo HB, Li Z. Convergence-Adaptive Roundtrip Method Enables Rapid and Accurate FEP Calculations. J Chem Theory Comput 2024. [PMID: 39236257 DOI: 10.1021/acs.jctc.4c00939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The free energy perturbation (FEP) method is a powerful technique for accurate binding free energy calculations, which is crucial for identifying potent ligands with a high affinity in drug discovery. However, the widespread application of FEP is limited by the high computational cost required to achieve equilibrium sampling and the challenges in obtaining converged predictions. In this study, we present the convergence-adaptive roundtrip (CAR) method, which is an enhanced adaptive sampling approach, to address the key challenges in FEP calculations, including the precision-efficiency tradeoff, sampling efficiency, and convergence assessment. By employing on-the-fly convergence analysis to automatically adjust simulation times, enabling efficient traversal of the important phase space through rapid propagation of conformations between different states and eliminating the need for multiple parallel simulations, the CAR method increases convergence and minimizes computational overhead while maintaining calculation accuracy. The performance of the CAR method was evaluated through relative binding free energy (RBFE) calculations on benchmarks comprising four diverse protein-ligand systems. The results demonstrated a significant speedup of over 8-fold compared to conventional FEP methods while maintaining high accuracy. The overall R2 values of 0.65 and 0.56 were obtained using the combined-structure FEP approach and the single-step FEP approach, respectively, in conjunction with the CAR method. In-depth case studies further highlighted the superior performance of the CAR method in terms of convergence acceleration, improved predicted correlations, and reduced computational costs. The advancement of the CAR method makes it a highly effective approach, enhancing the applicability of FEP in drug discovery.
Collapse
Affiliation(s)
- Yufen Yao
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Runduo Liu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Wenchao Li
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Wanyi Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yijun Lai
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya 572000, China
| | - Zhe Li
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
26
|
J AR, D SP, Arumainathan S. Digital nets conformational sampling (DNCS) - an enhanced sampling technique to explore the conformational space of intrinsically disordered peptides. Phys Chem Chem Phys 2024; 26:22640-22655. [PMID: 39158517 DOI: 10.1039/d4cp01891e] [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: 08/20/2024]
Abstract
We propose digital nets conformational sampling (DNCS) - an enhanced sampling technique to explore the conformational ensembles of peptides, especially intrinsically disordered peptides (IDPs). The DNCS algorithm relies on generating history-dependent samples of dihedral variables using bitwise XOR operations and binary angle measurements (BAM). The algorithm was initially studied using met-enkephalin, a highly elusive neuropeptide. The DNCS method predicted near-native structures and the energy landscape of met-enkephalin was observed to be in direct correlation with earlier studies on the neuropeptide. Clustering analysis revealed that there are only 24 low-lying conformations of the molecule. The DNCS method has then been tested for predicting optimal conformations of 42 oligopeptides of length varying from 3 to 8 residues. The closest-to-native structures of 86% of cases are near-native and 24% of them have a root mean square deviation of less than 1.00 Å with respect to their crystal structures. The results obtained reveal that the DNCS method performs well, that too in less computational time.
Collapse
Affiliation(s)
- Abraham Rebairo J
- Department of Nuclear Physics, University of Madras, Chennai, Tamil Nadu, India.
| | - Sam Paul D
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India
| | - Stephen Arumainathan
- Department of Nuclear Physics, University of Madras, Chennai, Tamil Nadu, India.
- Department of Materials Science, University of Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
27
|
Xing G, Zheng Q. Insights into the specific feature of the electrostatic recognition binding mechanism between BM2 and BM1: a molecular dynamics simulation study. Phys Chem Chem Phys 2024; 26:22726-22738. [PMID: 39161312 DOI: 10.1039/d4cp01936a] [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: 08/21/2024]
Abstract
Matrix protein 2 (M2) and matrix protein 1 (M1) of the influenza B virus are two important proteins, and the interactions between BM2 and BM1 play an important role in the process of virus assembly and replication. However, the interaction details between BM2 and BM1 are still unclear at the atomic level. Here, we constructed the BM2-BM1 complex system using homology modelling and molecular docking methods. Molecular dynamics (MD) simulations were used to illustrate the binding mechanism between BM2 and BM1. The results identify that the eight polar residues (E88B, E89B, H119BM1, E94B, R101BM1, K102BM1, R105BM1, and E104B) play an important role in stabilizing the binding through the formation of hydrogen bond networks and salt-bridge interactions at the binding interface. Furthermore, based on the simulation results and the experimental facts, the mutation experiments were designed to verify the influence of the mutation of residues both within and outside the effector domain. The mutations directly or indirectly disrupt interactions between polar residues, thus affecting viral assembly and replication. The results could help us understand the details of the interactions between BM2 and BM1 and provide useful information for the anti-influenza drug design.
Collapse
Affiliation(s)
- Guixuan Xing
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| | - Qingchuan Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| |
Collapse
|
28
|
van Gunsteren WF, Oostenbrink C. Methods for Classical-Mechanical Molecular Simulation in Chemistry: Achievements, Limitations, Perspectives. J Chem Inf Model 2024; 64:6281-6304. [PMID: 39136351 DOI: 10.1021/acs.jcim.4c00823] [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] [Indexed: 08/27/2024]
Abstract
More than a half century ago it became feasible to simulate, using classical-mechanical equations of motion, the dynamics of molecular systems on a computer. Since then classical-physical molecular simulation has become an integral part of chemical research. It is widely applied in a variety of branches of chemistry and has significantly contributed to the development of chemical knowledge. It offers understanding and interpretation of experimental results, semiquantitative predictions for measurable and nonmeasurable properties of substances, and allows the calculation of properties of molecular systems under conditions that are experimentally inaccessible. Yet, molecular simulation is built on a number of assumptions, approximations, and simplifications which limit its range of applicability and its accuracy. These concern the potential-energy function used, adequate sampling of the vast statistical-mechanical configurational space of a molecular system and the methods used to compute particular properties of chemical systems from statistical-mechanical ensembles. During the past half century various methodological ideas to improve the efficiency and accuracy of classical-physical molecular simulation have been proposed, investigated, evaluated, implemented in general simulation software or were abandoned. The latter because of fundamental flaws or, while being physically sound, computational inefficiency. Some of these methodological ideas are briefly reviewed and the most effective methods are highlighted. Limitations of classical-physical simulation are discussed and perspectives are sketched.
Collapse
Affiliation(s)
- Wilfred F van Gunsteren
- Institute for Molecular Physical Science, Swiss Federal Institute of Technology, ETH, CH-8093 Zurich, Switzerland
| | - Chris Oostenbrink
- Institute of Molecular Modelling and Simulation, BOKU University, 1190 Vienna, Austria
- Christian Doppler Laboratory for Molecular Informatics in the Biosciences, BOKU University, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
29
|
Sarkar D, Stelmakh A, Karmakar A, Aebli M, Krieg F, Bhattacharya A, Pawsey S, Kovalenko MV, Michaelis VK. Surface Structure of Lecithin-Capped Cesium Lead Halide Perovskite Nanocrystals Using Solid-State and Dynamic Nuclear Polarization NMR Spectroscopy. ACS NANO 2024; 18:21894-21910. [PMID: 39110153 DOI: 10.1021/acsnano.4c02057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Inorganic colloidal cesium lead halide perovskite nanocrystals (NCs) encapsulated by surface capping ligands exhibit tremendous potential in optoelectronic applications, with their surface structure playing a pivotal role in enhancing their photophysical properties. Soy lecithin, a tightly binding zwitterionic surface-capping ligand, has recently facilitated the high-yield synthesis of stable ultraconcentrated and ultradilute colloids of CsPbX3 NCs, unlocking a myriad of potential device applications. However, the atomic-level understanding of the ligand-terminated surface structure remains uncertain. Herein, we use a versatile solid-state nuclear magnetic resonance (NMR) spectroscopic approach, in combination with dynamic nuclear polarization (DNP) and atomistic molecular dynamics (MD) simulations, to explore the effect of lecithin on the core-to-surface structures of CsPbX3 (X = Cl or Br) perovskites, sized from micron to nanoscale. Surface-selective (cross-polarization, CP) solid-state and DNP NMR (133Cs and 207Pb) methods were used to differentiate the unique surface and core chemical environments, while the head-groups {trimethylammonium [-N(CH3)3+] and phosphate (-PO4-)} of lecithin were assigned via 1H, 13C, and 31P NMR spectroscopy. A direct approach to determining the surface structure by capitalizing on the unique heteronuclear dipolar couplings between the lecithin ligand (1H and 31P) and the surface of the CsPbCl3 NCs (133Cs and 207Pb) is demonstrated. The 1H-133Cs heteronuclear correlation (HETCOR) DNP NMR indicates an abundance of Cs on the NC surface and an intimate proximity of the -N(CH3)3+ groups to the surface and subsurface 133Cs atoms, supported by 1H{133Cs} rotational-echo double-resonance (REDOR) NMR spectroscopy. Moreover, the 1H-31P{207Pb} CP REDOR dephasing curve provides average internuclear distance information that allows assessment of -PO4- groups binding to the subsurface Pb atoms. Atomistic MD simulations of ligand-capped CsPbCl3 surfaces aid in the interpretation of this information and suggest that ligand -N(CH3)3+ and -PO4- head-groups substitute Cs+ and Cl- ions, respectively, at the CsCl-terminated surface of the NCs. These detailed atomistic insights into surface structures can further guide the engineering of various relevant surface-capping zwitterionic ligands for diverse metal halide perovskite NCs.
Collapse
Affiliation(s)
- Diganta Sarkar
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Andriy Stelmakh
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 1-5, Zurich CH-8093, Switzerland
- Empa-Swiss Federal Laboratories for Materials Science and Technology, Überlandstrasse 129, Dübendorf CH-8600, Switzerland
| | - Abhoy Karmakar
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Marcel Aebli
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 1-5, Zurich CH-8093, Switzerland
- Empa-Swiss Federal Laboratories for Materials Science and Technology, Überlandstrasse 129, Dübendorf CH-8600, Switzerland
| | - Franziska Krieg
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 1-5, Zurich CH-8093, Switzerland
- Empa-Swiss Federal Laboratories for Materials Science and Technology, Überlandstrasse 129, Dübendorf CH-8600, Switzerland
| | - Amit Bhattacharya
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Shane Pawsey
- Bruker BioSpin Corporation, Billerica, Massachusetts 01821, United States
| | - Maksym V Kovalenko
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 1-5, Zurich CH-8093, Switzerland
- Empa-Swiss Federal Laboratories for Materials Science and Technology, Überlandstrasse 129, Dübendorf CH-8600, Switzerland
| | - Vladimir K Michaelis
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
30
|
Ngo VA. Insight into molecular basis and dynamics of full-length CRaf kinase in cellular signaling mechanisms. Biophys J 2024; 123:2623-2637. [PMID: 38946141 PMCID: PMC11365224 DOI: 10.1016/j.bpj.2024.06.028] [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: 02/19/2024] [Revised: 05/15/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024] Open
Abstract
Raf kinases play key roles in signal transduction in cells for regulating proliferation, differentiation, and survival. Despite decades of research into functions and dynamics of Raf kinases with respect to other cytosolic proteins, understanding Raf kinases is limited by the lack of their full-length structures at the atomic resolution. Here, we present the first model of the full-length CRaf kinase obtained from artificial intelligence/machine learning algorithms with a converging ensemble of structures simulated by large-scale temperature replica exchange simulations. Our model is validated by comparing simulated structures with the latest cryo-EM structure detailing close contacts among three key domains and regions of the CRaf. Our simulations identify potentially new epitopes of intramolecule interactions within the CRaf and reveal a dynamical nature of CRaf kinases, in which the three domains can move back and forth relative to each other for regulatory dynamics. The dynamic conformations are then used in a docking algorithm to shed insight into the paradoxical effect caused by vemurafenib in comparison with a paradox breaker PLX7904. We propose a model of Raf-heterodimer/KRas-dimer as a signalosome based on the dynamics of the full-length CRaf.
Collapse
Affiliation(s)
- Van A Ngo
- Advanced Computing for Life Sciences and Engineering, Science Engagement Section, Computing and Computational Sciences, National Center for Computational Sciences, Oak Ridge National Laboratory, Oak Ridge, Tennessee.
| |
Collapse
|
31
|
Aupič J, Pokorná P, Ruthstein S, Magistrato A. Predicting Conformational Ensembles of Intrinsically Disordered Proteins: From Molecular Dynamics to Machine Learning. J Phys Chem Lett 2024; 15:8177-8186. [PMID: 39093570 DOI: 10.1021/acs.jpclett.4c01544] [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: 08/04/2024]
Abstract
Intrinsically disordered proteins and regions (IDP/IDRs) are ubiquitous across all domains of life. Characterized by a lack of a stable tertiary structure, IDP/IDRs populate a diverse set of transiently formed structural states that can promiscuously adapt upon binding with specific interaction partners and/or certain alterations in environmental conditions. This malleability is foundational for their role as tunable interaction hubs in core cellular processes such as signaling, transcription, and translation. Tracing the conformational ensemble of an IDP/IDR and its perturbation in response to regulatory cues is thus paramount for illuminating its function. However, the conformational heterogeneity of IDP/IDRs poses several challenges. Here, we review experimental and computational methods devised to disentangle the conformational landscape of IDP/IDRs, highlighting recent computational advances that permit proteome-wide scans of IDP/IDRs conformations. We briefly evaluate selected computational methods using the disordered N-terminal of the human copper transporter 1 as a test case and outline further challenges in IDP/IDRs ensemble prediction.
Collapse
Affiliation(s)
- Jana Aupič
- CNR-IOM at International School for Advanced Studies (SISSA/ISAS), via Bonomea 265, 34136 Trieste, Italy
| | - Pavlína Pokorná
- CNR-IOM at International School for Advanced Studies (SISSA/ISAS), via Bonomea 265, 34136 Trieste, Italy
| | - Sharon Ruthstein
- Department of Chemistry, Faculty of Exact Sciences and the Institute for Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, 5290002 Ramat-Gan, Israel
| | - Alessandra Magistrato
- CNR-IOM at International School for Advanced Studies (SISSA/ISAS), via Bonomea 265, 34136 Trieste, Italy
| |
Collapse
|
32
|
Williams CD, Kalayan J, Burton NA, Bryce RA. Stable and accurate atomistic simulations of flexible molecules using conformationally generalisable machine learned potentials. Chem Sci 2024; 15:12780-12795. [PMID: 39148799 PMCID: PMC11323334 DOI: 10.1039/d4sc01109k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/07/2024] [Indexed: 08/17/2024] Open
Abstract
Computational simulation methods based on machine learned potentials (MLPs) promise to revolutionise shape prediction of flexible molecules in solution, but their widespread adoption has been limited by the way in which training data is generated. Here, we present an approach which allows the key conformational degrees of freedom to be properly represented in reference molecular datasets. MLPs trained on these datasets using a global descriptor scheme are generalisable in conformational space, providing quantum chemical accuracy for all conformers. These MLPs are capable of propagating long, stable molecular dynamics trajectories, an attribute that has remained a challenge. We deploy the MLPs in obtaining converged conformational free energy surfaces for flexible molecules via well-tempered metadynamics simulations; this approach provides a hitherto inaccessible route to accurately computing the structural, dynamical and thermodynamical properties of a wide variety of flexible molecular systems. It is further demonstrated that MLPs must be trained on reference datasets with complete coverage of conformational space, including in barrier regions, to achieve stable molecular dynamics trajectories.
Collapse
Affiliation(s)
- Christopher D Williams
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester Oxford Road Manchester M13 9PL UK
| | - Jas Kalayan
- Science and Technologies Facilities Council (STFC), Daresbury Laboratory Keckwick Lane, Daresbury Warrington WA4 4AD UK
| | - Neil A Burton
- Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester Oxford Road Manchester M13 9PL UK
| | - Richard A Bryce
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester Oxford Road Manchester M13 9PL UK
| |
Collapse
|
33
|
Martins G, Galamba N. Wild-Type α-Synuclein Structure and Aggregation: A Comprehensive Coarse-Grained and All-Atom Molecular Dynamics Study. J Chem Inf Model 2024; 64:6115-6131. [PMID: 39046235 PMCID: PMC11323248 DOI: 10.1021/acs.jcim.4c00965] [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: 06/05/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024]
Abstract
α-Synuclein (α-syn) is a 140 amino acid intrinsically disordered protein (IDP) and the primary component of cytotoxic oligomers implicated in the etiology of Parkinson's disease (PD). While IDPs lack a stable three-dimensional structure, they sample a heterogeneous ensemble of conformations that can, in principle, be assessed through molecular dynamics simulations. However, describing the structure and aggregation of large IDPs is challenging due to force field (FF) accuracy and sampling limitations. To cope with the latter, coarse-grained (CG) FFs emerge as a potential alternative at the expense of atomic detail loss. Whereas CG models can accurately describe the structure of the monomer, less is known about aggregation. The latter is key for assessing aggregation pathways and designing aggregation inhibitor drugs. Herein, we investigate the structure and dynamics of α-syn using different resolution CG (Martini3 and Sirah2) and all-atom (Amber99sb and Charmm36m) FFs to gain insight into the differences and resemblances between these models. The dependence of the magnitude of protein-water interactions and the putative need for enhanced sampling (replica exchange) methods in CG simulations are analyzed to distinguish between force field accuracy and sampling limitations. The stability of the CG models of an α-syn fibril was also investigated. Additionally, α-syn aggregation was studied through umbrella sampling for the CG models and CG/all-atom models for an 11-mer peptide (NACore) from an amyloidogenic domain of α-syn. Our results show that despite the α-syn structures of Martini3 and Sirah2 with enhanced protein-water interactions being similar, major differences exist concerning aggregation. The Martini3 fibril is not stable, and the binding free energy of α-syn and NACore is positive, opposite to Sirah2. Sirah2 peptides in a zwitterionic form, in turn, display termini interactions that are too strong, resulting in end-to-end orientation. Sirah2, with enhanced protein-water interactions and neutral termini, provides, however, a peptide aggregation free energy profile similar to that found with all-atom models. Overall, we find that Sirah2 with enhanced protein-water interactions is suitable for studying protein-protein and protein-drug aggregation.
Collapse
Affiliation(s)
- Gabriel
F. Martins
- BioISI—Biosystems
and Integrative Sciences Institute, Faculty
of Sciences of the University of Lisbon, C8, Campo Grande, 1749-016 Lisbon, Portugal
| | - Nuno Galamba
- BioISI—Biosystems
and Integrative Sciences Institute, Faculty
of Sciences of the University of Lisbon, C8, Campo Grande, 1749-016 Lisbon, Portugal
| |
Collapse
|
34
|
Zhou J, Huang M. Navigating the landscape of enzyme design: from molecular simulations to machine learning. Chem Soc Rev 2024; 53:8202-8239. [PMID: 38990263 DOI: 10.1039/d4cs00196f] [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: 07/12/2024]
Abstract
Global environmental issues and sustainable development call for new technologies for fine chemical synthesis and waste valorization. Biocatalysis has attracted great attention as the alternative to the traditional organic synthesis. However, it is challenging to navigate the vast sequence space to identify those proteins with admirable biocatalytic functions. The recent development of deep-learning based structure prediction methods such as AlphaFold2 reinforced by different computational simulations or multiscale calculations has largely expanded the 3D structure databases and enabled structure-based design. While structure-based approaches shed light on site-specific enzyme engineering, they are not suitable for large-scale screening of potential biocatalysts. Effective utilization of big data using machine learning techniques opens up a new era for accelerated predictions. Here, we review the approaches and applications of structure-based and machine-learning guided enzyme design. We also provide our view on the challenges and perspectives on effectively employing enzyme design approaches integrating traditional molecular simulations and machine learning, and the importance of database construction and algorithm development in attaining predictive ML models to explore the sequence fitness landscape for the design of admirable biocatalysts.
Collapse
Affiliation(s)
- Jiahui Zhou
- School of Chemistry and Chemical Engineering, Queen's University, David Keir Building, Stranmillis Road, Belfast BT9 5AG, Northern Ireland, UK.
| | - Meilan Huang
- School of Chemistry and Chemical Engineering, Queen's University, David Keir Building, Stranmillis Road, Belfast BT9 5AG, Northern Ireland, UK.
| |
Collapse
|
35
|
Nicy, Morgan JWR, Wales DJ. Energy landscapes for clusters of hexapeptides. J Chem Phys 2024; 161:054112. [PMID: 39092941 DOI: 10.1063/5.0220652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
We present the results for energy landscapes of hexapeptides obtained using interfaces to the Large-scale Atomic/Molecular Massively Parallel Simulator (LAMMPS) program. We have used basin-hopping global optimization and discrete path sampling to explore the landscapes of hexapeptide monomers, dimers, and oligomers containing 10, 100, and 200 monomers modeled using a residue-level coarse-grained potential, Mpipi, implemented in LAMMPS. We find that the dimers of peptides containing amino acid residues that are better at promoting phase separation, such as tyrosine and arginine, have melting peaks at higher temperature in their heat capacity compared to phenylalanine and lysine, respectively. This observation correlates with previous work on the same uncapped hexapeptide monomers modeled using atomistic potential. For oligomers, we compare the variation in monomer conformations with radial distance and observe trends for selected angles calculated for each monomer. The LAMMPS interfaces to the GMIN and OPTIM programs for landscape exploration offer new opportunities to investigate larger systems and provide access to the coarse-grained potentials implemented within LAMMPS.
Collapse
Affiliation(s)
- Nicy
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - John W R Morgan
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - David J Wales
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
36
|
Schön JC. Energy landscapes-Past, present, and future: A perspective. J Chem Phys 2024; 161:050901. [PMID: 39101536 DOI: 10.1063/5.0212867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/17/2024] [Indexed: 08/06/2024] Open
Abstract
Energy landscapes and the closely related cost function landscapes have been recognized in science, mathematics, and various other fields such as economics as being highly useful paradigms and tools for the description and analysis of the properties of many systems, ranging from glasses, proteins, and abstract global optimization problems to business models. A multitude of algorithms for the exploration and exploitation of such landscapes have been developed over the past five decades in the various fields of applications, where many re-inventions but also much cross-fertilization have occurred. Twenty-five years ago, trying to increase the fruitful interactions between workers in different fields led to the creation of workshops and small conferences dedicated to the study of energy landscapes in general instead of only focusing on specific applications. In this perspective, I will present some history of the development of energy landscape studies and try to provide an outlook on in what directions the field might evolve in the future and what larger challenges are going to lie ahead, both from a conceptual and a practical point of view, with the main focus on applications of energy landscapes in chemistry and physics.
Collapse
Affiliation(s)
- J C Schön
- Max-Planck-Institute for Solid State Research, Heisenbergstr. 1, D-70569 Stuttgart, Germany
| |
Collapse
|
37
|
Tanimoto S, Okumura H. Why Is Arginine the Only Amino Acid That Inhibits Polyglutamine Monomers from Taking on Toxic Conformations? ACS Chem Neurosci 2024; 15:2925-2935. [PMID: 39009034 PMCID: PMC11311134 DOI: 10.1021/acschemneuro.4c00276] [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: 05/10/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Polyglutamine (polyQ) diseases are devastating neurodegenerative disorders characterized by abnormal expansion of glutamine repeats within specific proteins. The aggregation of polyQ proteins is a critical pathological hallmark of these diseases. Arginine was identified as a promising inhibitory compound because it prevents polyQ-protein monomers from forming intra- and intermolecular β-sheet structures and hinders polyQ proteins from aggregating to form oligomers. Such an aggregation inhibitory effect was not observed in other amino acids. However, the underlying molecular mechanism of the aggregation inhibition and the factors that differentiate arginine from other amino acids, in terms of the inhibition of the polyQ-protein aggregation, remain poorly understood. Here, we performed replica-permutation molecular dynamics simulations to elucidate the molecular mechanism by which arginine inhibits the formation of the intramolecular β-sheet structure of a polyQ monomer. We found that the intramolecular β-sheet structure with more than four β-bridges of the polyQ monomer with arginine is more unstable than without any ligand and with lysine. We also found that arginine has 1.6-2.1 times more contact with polyQ than lysine. In addition, we revealed that arginine forms more hydrogen bonds with the main chain of the polyQ monomer than lysine. More hydrogen bonds formed between arginine and polyQ inhibit polyQ from forming the long intramolecular β-sheet structure. It is known that intramolecular β-sheet structure enhances intermolecular β-sheet structure between proteins. These effects are thought to be the reason for the inhibition of polyQ aggregation. This study provides insights into the molecular events underlying arginine's inhibition of polyQ-protein aggregation.
Collapse
Affiliation(s)
- Shoichi Tanimoto
- Exploratory
Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
| | - Hisashi Okumura
- Exploratory
Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
- National
Institutes of Natural Sciences, Institute
for Molecular Science, Okazaki 444-8787, Aichi, Japan
- Graduate
Institute for Advanced Studies, SOKENDAI, Okazaki 444-8787, Aichi, Japan
| |
Collapse
|
38
|
Verma S, Nair NN. A Comprehensive Study of Factors Affecting the Prediction of the p Ka Shift of Asp 26 in Thioredoxin Protein. J Phys Chem B 2024; 128:7304-7312. [PMID: 39023356 DOI: 10.1021/acs.jpcb.4c01516] [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: 07/20/2024]
Abstract
The stable protonation state of ionizable amino acids in a protein can be predicted by computing the pKa shift of that residue within the protein environment. Thermodynamic Integration (TI) is an ideal molecular dynamics-based approach for predicting the pKa shift of ionizable protein residues. Here, we probe TI-based simulation protocols for their ability to accurately predict the pKa shift of Asp26 in thioredoxin. While implicit solvent models can predict the pKa shift accurately, explicit solvent models result in substantial errors. To understand the underlying reason for this surprising discrepancy, we investigate the role of various factors such as solvent models, conformational sampling, background charges, and polarization.
Collapse
Affiliation(s)
- Shivani Verma
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur - 208016, India
| | - Nisanth N Nair
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur - 208016, India
| |
Collapse
|
39
|
Mukadum F, Ccoa WJP, Hocky GM. Molecular simulation approaches to probing the effects of mechanical forces in the actin cytoskeleton. Cytoskeleton (Hoboken) 2024; 81:318-327. [PMID: 38334204 PMCID: PMC11310368 DOI: 10.1002/cm.21837] [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: 11/21/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
In this article we give our perspective on the successes and promise of various molecular and coarse-grained simulation approaches to probing the effect of mechanical forces in the actin cytoskeleton.
Collapse
Affiliation(s)
- Fatemah Mukadum
- Department of Chemistry, New York University, New York, NY 10003, USA
| | | | - Glen M. Hocky
- Department of Chemistry, New York University, New York, NY 10003, USA
- Simons Center for Computational Physical Chemistry, New York, NY 10003, USA
| |
Collapse
|
40
|
Das B, Mathew AT, Baidya ATK, Devi B, Salmon RR, Kumar R. Artificial intelligence assisted identification of potential tau aggregation inhibitors: ligand- and structure-based virtual screening, in silico ADME, and molecular dynamics study. Mol Divers 2024; 28:2013-2031. [PMID: 37022608 DOI: 10.1007/s11030-023-10645-3] [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: 10/29/2022] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
Alzheimer's disease (AD) is a severe, growing, multifactorial disorder affecting millions of people worldwide characterized by cognitive decline and neurodegeneration. The accumulation of tau protein into paired helical filaments is one of the major pathological hallmarks of AD and has gained the interest of researchers as a potential drug target to treat AD. Lately, Artificial Intelligence (AI) has revolutionized the drug discovery process by speeding it up and reducing the overall cost. As a part of our continuous effort to identify potential tau aggregation inhibitors, and leveraging the power of AI, in this study, we used a fully automated AI-assisted ligand-based virtual screening tool, PyRMD to screen a library of 12 million compounds from the ZINC database to identify potential tau aggregation inhibitors. The preliminary hits from virtual screening were filtered for similar compounds and pan-assay interference compounds (the compounds containing reactive functional groups which can interfere with the assays) using RDKit. Further, the selected compounds were prioritized based on their molecular docking score with the binding pocket of tau where the binding pockets were identified using replica exchange molecular dynamics simulation. Thirty-three compounds showing good docking scores for all the tau clusters were selected and were further subjected to in silico pharmacokinetic prediction. Finally, top 10 compounds were selected for molecular dynamics simulation and MMPBSA binding free energy calculations resulting in the identification of UNK_175, UNK_1027, UNK_1172, UNK_1173, UNK_1237, UNK_1518, and UNK_2181 as potential tau aggregation inhibitors.
Collapse
Affiliation(s)
- Bhanuranjan Das
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, UP, India
| | - Alen T Mathew
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, UP, India
| | - Anurag T K Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, UP, India
| | - Bharti Devi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, UP, India
| | - Rahul Rampa Salmon
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, UP, India
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, UP, India.
| |
Collapse
|
41
|
Dai B, Chen JN, Zeng Q, Geng H, Wu YD. Accurate Structure Prediction for Cyclic Peptides Containing Proline Residues with High-Temperature Molecular Dynamics. J Phys Chem B 2024; 128:7322-7331. [PMID: 39028892 DOI: 10.1021/acs.jpcb.4c02004] [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: 07/21/2024]
Abstract
Cyclic peptides (CPs) are emerging as promising drug candidates. Numerous natural CPs and their analogs are effective therapeutics against various diseases. Notably, many of them contain peptidyl cis-prolyl bonds. Due to the high rotational barrier of peptide bonds, conventional molecular dynamics simulations struggle to effectively sample the cis/trans-isomerization of peptide bonds. Previous studies have highlighted the high accuracy of the residue-specific force field (RSFF) and the high sampling efficiency of high-temperature molecular dynamics (high-T MD). Herein, we propose a protocol that combines high-T MD with RSFF2C and a recently developed reweighting method based on probability densities for accurate structure prediction of proline-containing CPs. Our method successfully predicted 19 out of 23 CPs with the backbone rmsd < 1.0 Å compared to X-ray structures. Furthermore, we performed high-T MD and density reweighting on the sunflower trypsin inhibitor (SFTI-1)/trypsin complex to demonstrate its applicability in studying CP-complexes containing cis-prolines. Our results show that the conformation of SFTI-1 in aqueous solution is consistent with its bound conformation, potentially facilitating its binding.
Collapse
Affiliation(s)
- Botao Dai
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Jia-Nan Chen
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Qing Zeng
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Hao Geng
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yun-Dong Wu
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518132, China
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
42
|
Hasse T, Huang YMM. Multiple Parameter Replica Exchange Gaussian Accelerated Molecular Dynamics for Enhanced Sampling and Free Energy Calculation of Biomolecular Systems. J Chem Theory Comput 2024. [PMID: 39085770 DOI: 10.1021/acs.jctc.4c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
This study introduces a novel method named multiple parameter replica exchange Gaussian accelerated molecular dynamics (MP-Rex-GaMD), building on the Gaussian accelerated molecular dynamics (GaMD) algorithm. GaMD enhances sampling and retrieves free energy information for biomolecular systems by adding a harmonic boost potential to smooth the potential energy surface without the need for predefined reaction coordinates. Our innovative approach advances the acceleration power and energetic reweighting accuracy of GaMD by incorporating a replica exchange algorithm that enables the exchange of multiple parameters, including the GaMD boost parameters of force constant and energy threshold, as well as temperature. Applying MP-Rex-GaMD to the three model systems of dialanine, chignolin, and HIV protease, we demonstrate its superior capability over conventional molecular dynamics and GaMD simulations in exploring protein conformations and effectively navigating various biomolecular states across energy barriers. MP-Rex-GaMD allows users to accurately map free energy landscapes through energetic reweighting, capturing the ensemble of biomolecular states from low-energy conformations to rare high-energy transitions within practical computational time scales.
Collapse
Affiliation(s)
- Timothy Hasse
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan 48201, United States
| | - Yu-Ming M Huang
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan 48201, United States
| |
Collapse
|
43
|
Cheung DL. Surface Hydrophobicity Strongly Influences Adsorption and Conformation of Amyloid Beta Derived Peptides. Molecules 2024; 29:3634. [PMID: 39125038 PMCID: PMC11314246 DOI: 10.3390/molecules29153634] [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: 06/14/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The formation of amyloid fibrils is a common feature of many protein systems. It has implications in both health, as amyloid fibrils are implicated in over 30 degenerative diseases, and in the biological functions of proteins. Surfaces have long been known to affect the formation of fibrils but the specific effect depends on the details of both the surface and protein. Fully understanding the role of surfaces in fibrillization requires microscopic information on protein conformation on surfaces. In this paper replica exchange molecular dynamics simulation is used to investigate the model fibril forming protein, Aβ(10-40) (a 31-residue segment of the amyloid-beta protein) on surfaces of different hydrophobicity. Similar to other proteins Aβ(10-40) is found to adsorb strongly onto hydrophobic surfaces. It also adopts significantly different sets of conformations on hydrophobic and polar surfaces, as well as in bulk solution. On hydrophobic surfaces, it adopts partially helical structures, with the helices overlapping with beta-strand regions in the mature fibril. These may be helical intermediates on the fibril formation pathway, suggesting a mechanism for the enhanced fibril formation seen on hydrophobic surfaces.
Collapse
Affiliation(s)
- David L Cheung
- School of Biological and Chemical Sciences, University of Galway, University Road, H91 TK33 Galway, Ireland
| |
Collapse
|
44
|
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
|
45
|
Strohkendl I, Saha A, Moy C, Nguyen AH, Ahsan M, Russell R, Palermo G, Taylor DW. Cas12a domain flexibility guides R-loop formation and forces RuvC resetting. Mol Cell 2024; 84:2717-2731.e6. [PMID: 38955179 PMCID: PMC11283365 DOI: 10.1016/j.molcel.2024.06.007] [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: 09/11/2023] [Revised: 05/17/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
The specific nature of CRISPR-Cas12a makes it a desirable RNA-guided endonuclease for biotechnology and therapeutic applications. To understand how R-loop formation within the compact Cas12a enables target recognition and nuclease activation, we used cryo-electron microscopy to capture wild-type Acidaminococcus sp. Cas12a R-loop intermediates and DNA delivery into the RuvC active site. Stages of Cas12a R-loop formation-starting from a 5-bp seed-are marked by distinct REC domain arrangements. Dramatic domain flexibility limits contacts until nearly complete R-loop formation, when the non-target strand is pulled across the RuvC nuclease and coordinated domain docking promotes efficient cleavage. Next, substantial domain movements enable target strand repositioning into the RuvC active site. Between cleavage events, the RuvC lid conformationally resets to occlude the active site, requiring re-activation. These snapshots build a structural model depicting Cas12a DNA targeting that rationalizes observed specificity and highlights mechanistic comparisons to other class 2 effectors.
Collapse
Affiliation(s)
- Isabel Strohkendl
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Aakash Saha
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA
| | - Catherine Moy
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Alexander-Hoi Nguyen
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Mohd Ahsan
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA
| | - Rick Russell
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Life Sciences Graduate Programs, University of Texas at Austin, Austin, TX 78712, USA
| | - Giulia Palermo
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA; Department of Chemistry, University of California, Riverside, Riverside, CA 92521, USA
| | - David W Taylor
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Interdisciplinary Life Sciences Graduate Programs, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; LIVESTRONG Cancer Institute, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
46
|
Mondal S, Sauer MA, Heyden M. Exploring Conformational Landscapes Along Anharmonic Low-Frequency Vibrations. J Phys Chem B 2024; 128:7112-7120. [PMID: 38986052 DOI: 10.1021/acs.jpcb.4c02743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
We aim to automatize the identification of collective variables to simplify and speed up enhanced sampling simulations of conformational dynamics in biomolecules. We focus on anharmonic low-frequency vibrations that exhibit fluctuations on time scales faster than conformational transitions but describe a path of least resistance toward structural change. A key challenge is that harmonic approximations are ill-suited to characterize these vibrations, which are observed at far-infrared frequencies and are easily excited by thermal collisions at room temperature. Here, we approached this problem with a frequency-selective anharmonic (FRESEAN) mode analysis that does not rely on harmonic approximations and successfully isolates anharmonic low-frequency vibrations from short molecular dynamics simulation trajectories. We applied FRESEAN mode analysis to simulations of alanine dipeptide, a common test system for enhanced sampling simulation protocols, and compared the performance of isolated low-frequency vibrations to conventional user-defined collective variables (here backbone dihedral angles) in enhanced sampling simulations. The comparison shows that enhanced sampling along anharmonic low-frequency vibrations not only reproduces known conformational dynamics but can even further improve the sampling of slow transitions compared to user-defined collective variables. Notably, free energy surfaces spanned by low-frequency anharmonic vibrational modes exhibit lower barriers associated with conformational transitions relative to representations in backbone dihedral space. We thus conclude that anharmonic low-frequency vibrations provide a promising path for highly effective and fully automated enhanced sampling simulations of conformational dynamics in biomolecules.
Collapse
Affiliation(s)
- Souvik Mondal
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Michael A Sauer
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Matthias Heyden
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
47
|
Laplaza R, Wodrich MD, Corminboeuf C. Overcoming the Pitfalls of Computing Reaction Selectivity from Ensembles of Transition States. J Phys Chem Lett 2024; 15:7363-7370. [PMID: 38990895 DOI: 10.1021/acs.jpclett.4c01657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
The prediction of reaction selectivity is a challenging task for computational chemistry, not only because many molecules adopt multiple conformations but also due to the exponential relationship between effective activation energies and rate constants. To account for molecular flexibility, an increasing number of methods exist that generate conformational ensembles of transition state (TS) structures. Typically, these TS ensembles are Boltzmann weighted and used to compute selectivity assuming Curtin-Hammett conditions. This strategy, however, can lead to erroneous predictions if the appropriate filtering of the conformer ensembles is not conducted. Here, we demonstrate how any possible selectivity can be obtained by processing the same sets of TS ensembles for a model reaction. To address the burdensome filtering task in a consistent and automated way, we introduce marc, a tool for the modular analysis of representative conformers that aids in avoiding human errors while minimizing the number of reoptimization computations needed to obtain correct reaction selectivity.
Collapse
Affiliation(s)
- Ruben Laplaza
- Laboratory for Computational Molecular Design, Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- National Center for Competence in Research-Catalysis (NCCR-Catalysis), École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Matthew D Wodrich
- Laboratory for Computational Molecular Design, Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- National Center for Competence in Research-Catalysis (NCCR-Catalysis), École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Clemence Corminboeuf
- Laboratory for Computational Molecular Design, Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- National Center for Competence in Research-Catalysis (NCCR-Catalysis), École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
48
|
Wang J, Miao Y. Ligand Gaussian Accelerated Molecular Dynamics 3 (LiGaMD3): Improved Calculations of Binding Thermodynamics and Kinetics of Both Small Molecules and Flexible Peptides. J Chem Theory Comput 2024; 20:5829-5841. [PMID: 39002136 DOI: 10.1021/acs.jctc.4c00502] [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: 07/15/2024]
Abstract
Binding thermodynamics and kinetics play critical roles in drug design. However, it has proven challenging to efficiently predict ligand binding thermodynamics and kinetics of small molecules and flexible peptides using conventional molecular dynamics (cMD), due to limited simulation time scales. Based on our previously developed ligand Gaussian accelerated molecular dynamics (LiGaMD) method, we present a new approach, termed "LiGaMD3″, in which we introduce triple boosts into three individual energy terms that play important roles in small-molecule/peptide dissociation, rebinding, and system conformational changes to improve the sampling efficiency of small-molecule/peptide interactions with target proteins. To validate the performance of LiGaMD3, MDM2 bound by a small molecule (Nutlin 3) and two highly flexible peptides (PMI and P53) were chosen as the model systems. LiGaMD3 could efficiently capture repetitive small-molecule/peptide dissociation and binding events within 2 μs simulations. The predicted binding kinetic constant rates and free energies from LiGaMD3 were in agreement with the available experimental values and previous simulation results. Therefore, LiGaMD3 provides a more general and efficient approach to capture dissociation and binding of both small-molecule ligands and flexible peptides, allowing for accurate prediction of their binding thermodynamics and kinetics.
Collapse
Affiliation(s)
- Jinan Wang
- Computational Medicine Program and Department of Pharmacology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yinglong Miao
- Computational Medicine Program and Department of Pharmacology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
49
|
Lutsyk V, Wolski P, Plazinski W. The Conformation of Glycosidic Linkages According to Various Force Fields: Monte Carlo Modeling of Polysaccharides Based on Extrapolation of Short-Chain Properties. J Chem Theory Comput 2024; 20:6350-6368. [PMID: 38985993 PMCID: PMC11270825 DOI: 10.1021/acs.jctc.4c00543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024]
Abstract
The conformational features of the glycosidic linkage are the most important variable to consider when studying di-, oligo-, and polysaccharide molecules using molecular dynamics (MD) simulations. The accuracy of the theoretical model describing this degree of freedom influences the quality of the results obtained from MD calculations based on this model. This article focuses on the following two issues related to the conformation of the glycosidic linkage. First, we describe the results of a comparative analysis of the predictions of three carbohydrate-dedicated classical force fields for MD simulations, namely, CHARMM, GLYCAM, and GROMOS, in the context of different parameters of structural and energetic nature related to the conformation of selected types of glycosidic linkages, α(1 → 4), β(1 → 3), and β(1 → 4), connecting glucopyranose units. This analysis revealed several differences, mainly concerning the energy levels of the secondary and tertiary conformers and the linkage flexibility within the dominant exo-syn conformation for α(1 → 4) and β(1 → 3) linkages. Some aspects of the comparative analysis also included the newly developed, carbohydrate-dedicated Martini 3 coarse-grained force field. Second, to overcome the time-scale problem associated with sampling slow degrees of freedom in polysaccharide chains during MD simulations, we developed a coarse-grained (CG) model based on the data from MD simulations and designed for Monte Carlo modeling. This model (CG MC) is based on information from simulations of short saccharide chains, effectively sampled in atomistic MD simulations, and is capable of extrapolating local conformational properties to the case of polysaccharides of arbitrary length. The CG MC model has the potential to estimate the conformations of very long polysaccharide chains, taking into account the influence of secondary and tertiary conformations of glycosidic linkages. With respect to the comparative analysis of force fields, the application of CG MC modeling showed that relatively small differences in the predictions of individual force fields with respect to a single glycosidic linkage accumulate when considering their effect on the structure of longer chains, leading to drastically different predictions with respect to parameters describing the polymer conformation, such as the persistence length.
Collapse
Affiliation(s)
- Valery Lutsyk
- Jerzy
Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland
| | - Pawel Wolski
- Jerzy
Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland
| | - Wojciech Plazinski
- Jerzy
Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland
- Department
of Biopharmacy, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland
| |
Collapse
|
50
|
Chen L, Mondal A, Perez A, Miranda-Quintana RA. Protein Retrieval via Integrative Molecular Ensembles (PRIME) through Extended Similarity Indices. J Chem Theory Comput 2024; 20:6303-6315. [PMID: 38978294 DOI: 10.1021/acs.jctc.4c00362] [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: 07/10/2024]
Abstract
Molecular dynamics (MD) simulations are ideally suited to describe conformational ensembles of biomolecules such as proteins and nucleic acids. Microsecond-long simulations are now routine, facilitated by the emergence of graphical processing units. Clustering, which groups objects based on structural similarity, is typically used to process ensembles, leading to different states, their populations, and the identification of representative structures. A popular pipeline combines hierarchical clustering for clustering and selecting the cluster centroid as representative of the cluster. Here, we propose to improve on this approach, by developing a module-Protein Retrieval via Integrative Molecular Ensembles (PRIME), that consists of tools to improve the prediction of the representative in the most populated cluster using extended continuous similarity. PRIME is integrated with our Molecular Dynamics Analysis with N-ary Clustering Ensembles (MDANCE) package and can be used as a postprocessing tool for arbitrary clustering algorithms, compatible with several MD suites. PRIME predictions produced structures that when aligned to the experimental structure were better superposed (lower RMSD). A further benefit of PRIME is its linear scaling─rather than the traditional O(N2) traditionally associated with comparisons of elements in a set.
Collapse
Affiliation(s)
- Lexin Chen
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
- Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| | - Arup Mondal
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
- Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| | - Alberto Perez
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
- Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| | - Ramón Alain Miranda-Quintana
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
- Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|