1
|
Moradi S, Nowroozi A, Aryaei Nezhad M, Jalali P, Khosravi R, Shahlaei M. A review on description dynamics and conformational changes of proteins using combination of principal component analysis and molecular dynamics simulation. Comput Biol Med 2024; 183:109245. [PMID: 39388840 DOI: 10.1016/j.compbiomed.2024.109245] [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: 07/24/2024] [Revised: 09/22/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Understanding how proteins behave dynamically and undergo conformational changes is essential to comprehending their biological roles. This review article examines the potent tool of using Molecular Dynamics simulations in conjunction with Principal Component Analysis (PCA) to explore protein dynamics. Molecular dynamics data can be made easier to read by removing prominent patterns through the use of PCA, a sophisticated dimensionality reduction approach. Researchers can obtain critical insights into the fundamental principles governing protein function by using PCA on MD simulation data. We provide a systematic approach to PCA that includes data collection, input coordinate selection, and result interpretation. Protein collective movements and fundamental dynamics are made visible by PCA, which makes it possible to identify conformational substates that are crucial to function. By means of principal component analysis, scientists are able to observe and measure large-scale movements, like hinge bending and domain motions, as well as pinpoint areas of protein structural stiffness and flexibility. Moreover, PCA allows temporal separation, distinguishing slower global motions from faster local changes. A strong foundation for researching protein dynamics is provided by the combination of PCA and Molecular Dynamics simulations, which have applications in drug development and enhance our comprehension of intricate biological systems.
Collapse
Affiliation(s)
- Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amin Nowroozi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Aryaei Nezhad
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parvin Jalali
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rasool Khosravi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Shahlaei
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
2
|
Zhu Y, Hu Z, Liu Y, Yan T, Liu L, Wang Y, Bai B. AChE activity self-breathing control mechanisms regulated by H 2S n and GSH: Persulfidation and glutathionylation on sulfhydryl after disulfide bonds cleavage. Int J Biol Macromol 2024; 259:129117. [PMID: 38211930 DOI: 10.1016/j.ijbiomac.2023.129117] [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: 07/13/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/13/2024]
Abstract
Hydrogen sulfide (H2S), or dihydrogen sulfane (H2Sn), acts as a signal molecule through the beneficial mechanism of persulfidation, known as the post-translational transformation of cysteine residues to persulfides. We previously reported that Glutathione (GSH) could regulate enzyme activity through S-desulfurization or glutathionylation of residues to generate protein-SG or protein-SSG, releasing H2S. However, little is known about the mechanisms by which H2Sn and GSH affect the disulfide bonds. In this study, we provide direct evidences that H2Sn and GSH modify the sulfhydryl group on Cys272, which forms disulfide bonds in acetylcholinesterase (AChE), to generate Cys-SSH and Cys-SSG, respectively. Glutathionylation of disulfide is a two-step reaction based on nucleophilic substitution, in which the first CS bond is broken, then the SS bond is broken to release H2S. H2Sn and GSH controlled self-breathing motion in enzyme catalysis by disconnecting specific disulfide bonds and modifying cysteine residues, thereby regulating AChE activity. Here, we elucidated H2Sn and GSH mechanisms on disulfide in the AChE system and proposed a self-breathing control theory induced by H2Sn and GSH. These theoretical findings shed light on the biological functions of H2Sn and GSH on sulfhydryl and disulfide bonds and enrich the theory of enzyme activity regulation.
Collapse
Affiliation(s)
- Yanwen Zhu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Zhaoliang Hu
- Department of Surgical Oncology, First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Yunen Liu
- Shenyang Medical College, Shenyang 110034, China
| | - Tingcai Yan
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Ling Liu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Yanqun Wang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Bing Bai
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| |
Collapse
|
3
|
Monti S, Trouki C, Barcaro G. Disclosing gate-opening/closing events inside a flexible metal-organic framework loaded with CO 2 by reactive and essential dynamics. NANOSCALE 2023; 15:14505-14513. [PMID: 37609787 DOI: 10.1039/d3nr02760k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
We have combined reactive molecular dynamics simulations with principal component analysis to provide a clearer view of the interactions and motion of the CO2 molecules inside a metal-organic framework and the movements of the MOF components that regulate storage, adsorption, and diffusion of the guest species. The tens-of-nanometer size of the simulated model, the capability of the reactive force field tuned to reproduce the inorganic-organic material confidently, and the unconventional use of essential dynamics have effectively disclosed the gate-opening/closing phenomenon, possible coordinations of CO2 at the metal centers, all the diffusion steps inside the MOF channels, the primary motions of the linkers, and the effects of their concerted rearrangements on local CO2 relocations.
Collapse
Affiliation(s)
- Susanna Monti
- CNR-ICCOM, Institute of Chemistry of Organometallic Compounds, Pisa 56124, Italy.
| | - Cheherazade Trouki
- CNR-IPCF, Institute of Chemical and Physical Processes, Pisa 56124, Italy
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
| | - Giovanni Barcaro
- CNR-IPCF, Institute of Chemical and Physical Processes, Pisa 56124, Italy
| |
Collapse
|
4
|
Probing the structural basis of Citrus phytochrome B using computational modelling and molecular dynamics simulation approaches. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
5
|
Bagri K, Kumar A, Manisha, Kumar P. Computational Studies on Acetylcholinesterase Inhibitors: From Biochemistry to Chemistry. Mini Rev Med Chem 2021; 20:1403-1435. [PMID: 31884928 DOI: 10.2174/1389557520666191224144346] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/23/2019] [Accepted: 10/28/2019] [Indexed: 11/22/2022]
Abstract
Acetylcholinesterase inhibitors are the most promising therapeutics for Alzheimer's disease treatment as these prevent the loss of acetylcholine and slows the progression of the disease. The drugs approved for the management of Alzheimer's disease by the FDA are acetylcholinesterase inhibitors but are associated with side effects. Consistent and stringent efforts by the researchers with the help of computational methods opened new ways of developing novel molecules with good acetylcholinesterase inhibitory activity. In this manuscript, we reviewed the studies that identified the essential structural features of acetylcholinesterase inhibitors at the molecular level as well as the techniques like molecular docking, molecular dynamics, quantitative structure-activity relationship, virtual screening, and pharmacophore modelling that were used in designing these inhibitors.
Collapse
Affiliation(s)
- Kiran Bagri
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology, Hisar 125001, India
| | - Ashwani Kumar
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology, Hisar 125001, India
| | - Manisha
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology, Hisar 125001, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| |
Collapse
|
6
|
De Boer D, Nguyen N, Mao J, Moore J, Sorin EJ. A Comprehensive Review of Cholinesterase Modeling and Simulation. Biomolecules 2021; 11:580. [PMID: 33920972 PMCID: PMC8071298 DOI: 10.3390/biom11040580] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 01/18/2023] Open
Abstract
The present article reviews published efforts to study acetylcholinesterase and butyrylcholinesterase structure and function using computer-based modeling and simulation techniques. Structures and models of both enzymes from various organisms, including rays, mice, and humans, are discussed to highlight key structural similarities in the active site gorges of the two enzymes, such as flexibility, binding site location, and function, as well as differences, such as gorge volume and binding site residue composition. Catalytic studies are also described, with an emphasis on the mechanism of acetylcholine hydrolysis by each enzyme and novel mutants that increase catalytic efficiency. The inhibitory activities of myriad compounds have been computationally assessed, primarily through Monte Carlo-based docking calculations and molecular dynamics simulations. Pharmaceutical compounds examined herein include FDA-approved therapeutics and their derivatives, as well as several other prescription drug derivatives. Cholinesterase interactions with both narcotics and organophosphate compounds are discussed, with the latter focusing primarily on molecular recognition studies of potential therapeutic value and on improving our understanding of the reactivation of cholinesterases that are bound to toxins. This review also explores the inhibitory properties of several other organic and biological moieties, as well as advancements in virtual screening methodologies with respect to these enzymes.
Collapse
Affiliation(s)
- Danna De Boer
- Department of Chemistry & Biochemistry, California State University, Long Beach, CA 90840, USA;
| | - Nguyet Nguyen
- Department of Chemical Engineering, California State University, Long Beach, CA 90840, USA; (N.N.); (J.M.)
| | - Jia Mao
- Department of Chemical Engineering, California State University, Long Beach, CA 90840, USA; (N.N.); (J.M.)
| | - Jessica Moore
- Department of Biomedical Engineering, California State University, Long Beach, CA 90840, USA;
| | - Eric J. Sorin
- Department of Chemistry & Biochemistry, California State University, Long Beach, CA 90840, USA;
| |
Collapse
|
7
|
Zueva IV, Lushchekina SV, Pottie IR, Darvesh S, Masson P. 1-(3- Tert-Butylphenyl)-2,2,2-Trifluoroethanone as a Potent Transition-State Analogue Slow-Binding Inhibitor of Human Acetylcholinesterase: Kinetic, MD and QM/MM Studies. Biomolecules 2020; 10:biom10121608. [PMID: 33260981 PMCID: PMC7760592 DOI: 10.3390/biom10121608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/14/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Kinetic studies and molecular modeling of human acetylcholinesterase (AChE) inhibition by a fluorinated acetophenone derivative, 1-(3-tert-butylphenyl)-2,2,2-trifluoroethanone (TFK), were performed. Fast reversible inhibition of AChE by TFK is of competitive type with Ki = 5.15 nM. However, steady state of inhibition is reached slowly. Kinetic analysis showed that TFK is a slow-binding inhibitor (SBI) of type B with Ki* = 0.53 nM. Reversible binding of TFK provides a long residence time, τ = 20 min, on AChE. After binding, TFK acylates the active serine, forming an hemiketal. Then, disruption of hemiketal (deacylation) is slow. AChE recovers full activity in approximately 40 min. Molecular docking and MD simulations depicted the different steps. It was shown that TFK binds first to the peripheral anionic site. Then, subsequent slow induced-fit step enlarged the gorge, allowing tight adjustment into the catalytic active site. Modeling of interactions between TFK and AChE active site by QM/MM showed that the “isomerization” step of enzyme-inhibitor complex leads to a complex similar to substrate tetrahedral intermediate, a so-called “transition state analog”, followed by a labile covalent intermediate. SBIs of AChE show prolonged pharmacological efficacy. Thus, this fluoroalkylketone intended for neuroimaging, could be of interest in palliative therapy of Alzheimer’s disease and protection of central AChE against organophosphorus compounds.
Collapse
Affiliation(s)
- Irina V. Zueva
- Arbuzov Institute of Organic and Physical Chemistry, Federal Research Center “Kazan Scientific Center of the Russian Academy of Sciences”, Arbuzov str., 8, 420088 Kazan, Russia;
| | - Sofya V. Lushchekina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Kosygin str. 4, 119334 Moscow, Russia;
| | - Ian R. Pottie
- Department of Chemistry and Physics, Mount Saint Vincent University, Halifax, NS B3M 2J6, Canada; (I.R.P.); (S.D.)
- Department of Chemistry, Saint Mary’s University, Halifax, NS B3M 2J6, Canada
| | - Sultan Darvesh
- Department of Chemistry and Physics, Mount Saint Vincent University, Halifax, NS B3M 2J6, Canada; (I.R.P.); (S.D.)
- Department of Medicine (Neurology and Geriatric Medicine) & Medical Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Patrick Masson
- Neuropharmacology Laboratory, Kazan Federal University, Kremlevskaya str. 18, 480002 Kazan, Russia
- Correspondence:
| |
Collapse
|
8
|
Modeling Epac1 interactions with the allosteric inhibitor AM-001 by co-solvent molecular dynamics. J Comput Aided Mol Des 2020; 34:1171-1179. [PMID: 32700175 PMCID: PMC7533256 DOI: 10.1007/s10822-020-00332-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
The exchange proteins activated by cAMP (EPAC) are implicated in a large variety of physiological processes and they are considered as promising targets for a wide range of therapeutic applications. Several recent reports provided evidence for the therapeutic effectiveness of the inhibiting EPAC1 activity cardiac diseases. In that context, we recently characterized a selective EPAC1 antagonist named AM-001. This compound was featured by a non-competitive mechanism of action but the localization of its allosteric site to EPAC1 structure has yet to be investigated. Therefore, we performed cosolvent molecular dynamics with the aim to identify a suitable allosteric binding site. Then, the docking and molecular dynamics were used to determine the binding of the AM-001 to the regions highlighted by cosolvent molecular dynamics for EPAC1. These analyses led us to the identification of a suitable allosteric AM-001 binding pocket at EPAC1. As a model validation, we also evaluated the binding poses of the available AM-001 analogues, with a different biological potency. Finally, the complex EPAC1 with AM-001 bound at the putative allosteric site was further refined by molecular dynamics. The principal component analysis led us to identify the protein motion that resulted in an inactive like conformation upon the allosteric inhibitor binding.
Collapse
|
9
|
Ancy I, Sivanandam M, Kalaivani R, Kumaradhas P. Insights of inhibition mechanism of sifuvirtide and MT-sifuvirtide against wild and mutant HIV-1 envelope glycoprotein41: a molecular dynamics simulation and binding free energy study. MOLECULAR SIMULATION 2020. [DOI: 10.1080/08927022.2020.1716978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Iruthayaraj Ancy
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| | - Magudeeswaran Sivanandam
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| | - Raju Kalaivani
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| | - Poomani Kumaradhas
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| |
Collapse
|
10
|
Nair MS, Shukla A. Molecular modeling, simulation and principal component analysis of binding of resveratrol and its analogues with DNA. J Biomol Struct Dyn 2019; 38:3087-3097. [PMID: 31476951 DOI: 10.1080/07391102.2019.1662849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Structure-based drug designing has become a significant subject of research, and several clinically promising DNA binding compounds were evolved using this technique. The interaction of an octamer DNA sequence d(CCAATTGG)2 with a natural stilbene, resveratrol and its analogues have been studied using molecular docking method. Out of the ten compounds studied, seven compounds were found to bind to the minor groove of AATT segment of the sequence. Pterostilbene, a natural analogue of resveratrol, showed the lowest binding energy. Rhaponticin, a natural analogue of resveratrol and digalloylresveratrol, a synthetic ester of resveratrol bind to the major groove of the AATT segment while dihydroresveratrol binds to the minor groove of GC terminal base pair. ADMET (Absorption, distribution, metabolism, excretion and toxicity) study showed that all compounds obey Lipinski rule and are accepted as orally active drugs based on different physicochemical descriptors. Molecular dynamics simulations were performed for the complex with lowest binding energy and trajectory analysis were performed. Principal component analysis has been performed to underline the prominent motions in alone DNA and when it is bound to pterostilbene. AbbreviationsADMETAbsorption, distribution, metabolism, excretion and toxicityDIGDigalloyl resveratrolDNADeoxyribonucleic acidELElectrostatic energyENPOLARNonpolar solvation energyESURFSurface areaGBGeneralized BornHBAHydrogen bond acceptorsHBDHydrogen bond donorsLGALamarckian genetic algorithmMDMolecular dynamicsPBPoisson-BoltzmannPCAPrincipal component analysisPTPterostilbeneRMSDRoot mean square deviationSASimulated annealingTLX3T-cell leukemia homeobox 3VDWvan der WaalsCommunicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Maya S Nair
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, India
| | - Aishwarya Shukla
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, India
| |
Collapse
|
11
|
Sivakumar M, Saravanan K, Saravanan V, Sugarthi S, kumar SM, Alhaji Isa M, Rajakumar P, Aravindhan S. Discovery of new potential triplet acting inhibitor for Alzheimer’s disease via X-ray crystallography, molecular docking and molecular dynamics. J Biomol Struct Dyn 2019; 38:1903-1917. [DOI: 10.1080/07391102.2019.1620128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | - Kandasamy Saravanan
- X-Ray Crystallography and Computational Molecular Biology Lab, Department of Physics, Periyar University, Salem, India
| | | | - Srinivasan Sugarthi
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur, Kancheepuram, Tamil Nadu, India
| | | | - Mustafa Alhaji Isa
- Bioinformatics and Computational Biology Lab, Department of Microbiology, Faculty of Sciences, University of Maiduguri, Maiduguri, Nigeria
| | - Perumal Rajakumar
- Department of Organic Chemistry, University of Madras, Chennai, India
| | | |
Collapse
|
12
|
Khalid RR, Siddiqi AR, Mylonas E, Maryam A, Kokkinidis M. Dynamic Characterization of the Human Heme Nitric Oxide/Oxygen (HNOX) Domain under the Influence of Diatomic Gaseous Ligands. Int J Mol Sci 2019; 20:E698. [PMID: 30736292 PMCID: PMC6387030 DOI: 10.3390/ijms20030698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/29/2019] [Accepted: 02/03/2019] [Indexed: 01/25/2023] Open
Abstract
Soluble guanylate cyclase (sGC) regulates numerous physiological processes. The β subunit Heme Nitric Oxide/Oxygen (HNOX) domain makes this protein sensitive to small gaseous ligands. The structural basis of the activation mechanism of sGC under the influence of ligands (NO, O₂, CO) is poorly understood. We examine the effect of different ligands on the human sGC HNOX domain. HNOX systems with gaseous ligands were generated and explored using Molecular Dynamics (MD). The distance between heme Fe2+ and histidine in the NO-ligated HNOX (NO-HNOX) system is larger compared to the O₂, CO systems. NO-HNOX rapidly adopts the conformation of the five-group metal coordination system. Loops α, β, γ and helix-f exhibit increased mobility and different hydrogen bond networks in NO-HNOX compared to the other systems. The removal of His from the Fe coordination sphere in NO-HNOX is assisted by interaction of the imidazole ring with the surrounding residues which in turn leads to the release of signaling helix-f and activation of the sGC enzyme. Insights into the conformational dynamics of a human sGC HNOX domain, especially for regions which are functionally critical for signal transduction, are valuable in the understanding of cardiovascular diseases.
Collapse
Affiliation(s)
- Rana Rehan Khalid
- Department of Biosciences, COMSATS University, Islamabad 45550, Pakistan.
- Department of Biology, University of Crete, 70013 Heraklion, Greece.
| | - Abdul Rauf Siddiqi
- Department of Biosciences, COMSATS University, Islamabad 45550, Pakistan.
| | - Efstratios Mylonas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), 70013 Heraklion, Greece.
| | - Arooma Maryam
- Department of Biosciences, COMSATS University, Islamabad 45550, Pakistan.
| | - Michael Kokkinidis
- Department of Biology, University of Crete, 70013 Heraklion, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), 70013 Heraklion, Greece.
| |
Collapse
|
13
|
Roca C, Requena C, Sebastián-Pérez V, Malhotra S, Radoux C, Pérez C, Martinez A, Antonio Páez J, Blundell TL, Campillo NE. Identification of new allosteric sites and modulators of AChE through computational and experimental tools. J Enzyme Inhib Med Chem 2018; 33:1034-1047. [PMID: 29873262 PMCID: PMC6010107 DOI: 10.1080/14756366.2018.1476502] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/09/2018] [Accepted: 05/09/2018] [Indexed: 11/15/2022] Open
Abstract
Allosteric sites on proteins are targeted for designing more selective inhibitors of enzyme activity and to discover new functions. Acetylcholinesterase (AChE), which is most widely known for the hydrolysis of the neurotransmitter acetylcholine, has a peripheral allosteric subsite responsible for amyloidosis in Alzheimer's disease through interaction with amyloid β-peptide. However, AChE plays other non-hydrolytic functions. Here, we identify and characterise using computational tools two new allosteric sites in AChE, which have allowed us to identify allosteric inhibitors by virtual screening guided by structure-based and fragment hotspot strategies. The identified compounds were also screened for in vitro inhibition of AChE and three were observed to be active. Further experimental (kinetic) and computational (molecular dynamics) studies have been performed to verify the allosteric activity. These new compounds may be valuable pharmacological tools in the study of non-cholinergic functions of AChE.
Collapse
Affiliation(s)
- Carlos Roca
- a Centro de Investigaciones Biológicas (CIB-CSIC), C/Ramiro de Maeztu , Madrid , Spain
| | - Carlos Requena
- a Centro de Investigaciones Biológicas (CIB-CSIC), C/Ramiro de Maeztu , Madrid , Spain
| | | | - Sony Malhotra
- b Department of Biochemistry , University of Cambridge , Cambridge , UK
| | - Chris Radoux
- b Department of Biochemistry , University of Cambridge , Cambridge , UK
- c Cambridge Crystallographic Data Centre , Cambridge , UK
| | - Concepción Pérez
- d Instituto de Química Médica (IQM-CSIC) , C/Juan de la Cierva , Madrid , Spain
| | - Ana Martinez
- a Centro de Investigaciones Biológicas (CIB-CSIC), C/Ramiro de Maeztu , Madrid , Spain
| | - Juan Antonio Páez
- d Instituto de Química Médica (IQM-CSIC) , C/Juan de la Cierva , Madrid , Spain
| | - Tom L Blundell
- b Department of Biochemistry , University of Cambridge , Cambridge , UK
| | - Nuria E Campillo
- a Centro de Investigaciones Biológicas (CIB-CSIC), C/Ramiro de Maeztu , Madrid , Spain
| |
Collapse
|
14
|
Influence of gauche effect on uncharged oxime reactivators for the reactivation of tabun-inhibited AChE: quantum chemical and steered molecular dynamics studies. J Comput Aided Mol Des 2018; 32:793-807. [DOI: 10.1007/s10822-018-0130-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/04/2018] [Indexed: 02/06/2023]
|
15
|
Design, synthesis and biological evaluation of 1,3-dihydroxyxanthone derivatives: Effective agents against acetylcholinesterase. Bioorg Chem 2017; 75:201-209. [DOI: 10.1016/j.bioorg.2017.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/16/2017] [Accepted: 09/18/2017] [Indexed: 12/31/2022]
|
16
|
Xu Y, Cheng S, Sussman JL, Silman I, Jiang H. Computational Studies on Acetylcholinesterases. Molecules 2017; 22:molecules22081324. [PMID: 28796192 PMCID: PMC6152020 DOI: 10.3390/molecules22081324] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 01/18/2023] Open
Abstract
Functions of biomolecules, in particular enzymes, are usually modulated by structural fluctuations. This is especially the case in a gated diffusion-controlled reaction catalyzed by an enzyme such as acetylcholinesterase. The catalytic triad of acetylcholinesterase is located at the bottom of a long and narrow gorge, but it catalyzes the extremely rapid hydrolysis of the neurotransmitter, acetylcholine, with a reaction rate close to the diffusion-controlled limit. Computational modeling and simulation have produced considerable advances in exploring the dynamical and conformational properties of biomolecules, not only aiding in interpreting the experimental data, but also providing insights into the internal motions of the biomolecule at the atomic level. Given the remarkably high catalytic efficiency and the importance of acetylcholinesterase in drug development, great efforts have been made to understand the dynamics associated with its functions by use of various computational methods. Here, we present a comprehensive overview of recent computational studies on acetylcholinesterase, expanding our views of the enzyme from a microstate of a single structure to conformational ensembles, strengthening our understanding of the integration of structure, dynamics and function associated with the enzyme, and promoting the structure-based and/or mechanism-based design of new inhibitors for it.
Collapse
Affiliation(s)
- Yechun Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China.
| | - Shanmei Cheng
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China.
| | - Joel L Sussman
- Israel Structural Proteomics Center, Weizmann Institute of Science, Rehovot 76100, Israel.
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Israel Silman
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Hualiang Jiang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China.
| |
Collapse
|
17
|
Saravanan K, Kalaiarasi C, Kumaradhas P. Understanding the conformational flexibility and electrostatic properties of curcumin in the active site of rhAChE via molecular docking, molecular dynamics, and charge density analysis. J Biomol Struct Dyn 2017; 35:3627-3647. [DOI: 10.1080/07391102.2016.1264891] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Kandasamy Saravanan
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Chinnasamy Kalaiarasi
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Poomani Kumaradhas
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| |
Collapse
|
18
|
Zobnina V, Lambreva MD, Rea G, Campi G, Antonacci A, Scognamiglio V, Giardi MT, Polticelli F. The plastoquinol-plastoquinone exchange mechanism in photosystem II: insight from molecular dynamics simulations. PHOTOSYNTHESIS RESEARCH 2017; 131:15-30. [PMID: 27376842 DOI: 10.1007/s11120-016-0292-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/22/2016] [Indexed: 05/23/2023]
Abstract
In the photosystem II (PSII) of oxygenic photosynthetic organisms, the reaction center (RC) core mediates the light-induced electron transfer leading to water splitting and production of reduced plastoquinone molecules. The reduction of plastoquinone to plastoquinol lowers PSII affinity for the latter and leads to its release. However, little is known about the role of protein dynamics in this process. Here, molecular dynamics simulations of the complete PSII complex embedded in a lipid bilayer have been used to investigate the plastoquinol release mechanism. A distinct dynamic behavior of PSII in the presence of plastoquinol is observed which, coupled to changes in charge distribution and electrostatic interactions, causes disruption of the interactions seen in the PSII-plastoquinone complex and leads to the "squeezing out" of plastoquinol from the binding pocket. Displacement of plastoquinol closes the second water channel, recently described in a 2.9 Å resolution PSII structure (Guskov et al. in Nat Struct Mol Biol 16:334-342, 2009), allowing to rule out the proposed "alternating" mechanism of plastoquinol-plastoquinone exchange, while giving support to the "single-channel" one. The performed simulations indicated a pivotal role of D1-Ser264 in modulating the dynamics of the plastoquinone binding pocket and plastoquinol-plastoquinone exchange via its interaction with D1-His252 residue. The effects of the disruption of this hydrogen bond network on the PSII redox reactions were experimentally assessed in the D1 site-directed mutant Ser264Lys.
Collapse
Affiliation(s)
- Veranika Zobnina
- Theoretical Biology and Bioinformatics Laboratory, Department of Sciences, Roma Tre University, Viale G. Marconi 446, 00146, Rome, Italy
| | - Maya D Lambreva
- Institute of Crystallography CNR, 00015, Monterotondo Scalo, Rome, Italy
| | - Giuseppina Rea
- Institute of Crystallography CNR, 00015, Monterotondo Scalo, Rome, Italy
| | - Gaetano Campi
- Institute of Crystallography CNR, 00015, Monterotondo Scalo, Rome, Italy
| | - Amina Antonacci
- Institute of Crystallography CNR, 00015, Monterotondo Scalo, Rome, Italy
| | | | | | - Fabio Polticelli
- Theoretical Biology and Bioinformatics Laboratory, Department of Sciences, Roma Tre University, Viale G. Marconi 446, 00146, Rome, Italy.
- National Institute of Nuclear Physics, Roma Tre Section, 00146, Rome, Italy.
| |
Collapse
|
19
|
Molecular investigations of protriptyline as a multi-target directed ligand in Alzheimer's disease. PLoS One 2014; 9:e105196. [PMID: 25141174 PMCID: PMC4139341 DOI: 10.1371/journal.pone.0105196] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/18/2014] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder involving multiple cellular and molecular processes. The discovery of drug molecules capable of targeting multiple factors involved in AD pathogenesis would greatly facilitate in improving therapeutic strategies. The repositioning of existing non-toxic drugs could dramatically reduce the time and costs involved in developmental and clinical trial stages. In this study, preliminary screening of 140 FDA approved nervous system drugs by docking suggested the viability of the tricyclic group of antidepressants against three major AD targets, viz. Acetylcholinesterase (AChE), β-secretase (BACE-1), and amyloid β (Aβ) aggregation, with one member, protriptyline, showing highest inhibitory activity. Detailed biophysical assays, together with isothermal calorimetry, fluorescence quenching experiments, kinetic studies and atomic force microscopy established the strong inhibitory activity of protriptyline against all three major targets. The molecular basis of inhibition was supported with comprehensive molecular dynamics simulations. Further, the drug inhibited glycation induced amyloid aggregation, another important causal factor in AD progression. This study has led to the discovery of protriptyline as a potent multi target directed ligand and established its viability as a promising candidate for AD treatment.
Collapse
|
20
|
Le HT, Dean WL, Buscaglia R, Chaires JB, Trent JO. An investigation of G-quadruplex structural polymorphism in the human telomere using a combined approach of hydrodynamic bead modeling and molecular dynamics simulation. J Phys Chem B 2014; 118:5390-405. [PMID: 24779348 PMCID: PMC4032189 DOI: 10.1021/jp502213y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/21/2014] [Indexed: 01/12/2023]
Abstract
Guanine-rich oligonucleotides can adopt noncanonical tertiary structures known as G-quadruplexes, which can exist in different forms depending on experimental conditions. High-resolution structural methods, such as X-ray crystallography and NMR spectroscopy, have been of limited usefulness in resolving the inherent structural polymorphism associated with G-quadruplex formation. The lack of, or the ambiguous nature of, currently available high-resolution structural data, in turn, has severely hindered investigations into the nature of these structures and their interactions with small-molecule inhibitors. We have used molecular dynamics in conjunction with hydrodynamic bead modeling to study the structures of the human telomeric G-quadruplex-forming sequences at the atomic level. We demonstrated that molecular dynamics can reproduce experimental hydrodynamic measurements and thus can be a powerful tool in the structural study of existing G-quadruplex sequences or in the prediction of new G-quadruplex structures.
Collapse
Affiliation(s)
- Huy T. Le
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
| | - William L. Dean
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
- James
Graham Brown Cancer Center, University of
Louisville, 529 South
Jackson Street, Louisville, Kentucky 40202, United
States
| | - Robert Buscaglia
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
| | - Jonathan B. Chaires
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
- James
Graham Brown Cancer Center, University of
Louisville, 529 South
Jackson Street, Louisville, Kentucky 40202, United
States
- Department
of Medicine, School of Medicine, University
of Louisville, 550 South
Jackson Street, Louisville, Kentucky 40202, United
States
| | - John O. Trent
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
- James
Graham Brown Cancer Center, University of
Louisville, 529 South
Jackson Street, Louisville, Kentucky 40202, United
States
- Department
of Medicine, School of Medicine, University
of Louisville, 550 South
Jackson Street, Louisville, Kentucky 40202, United
States
| |
Collapse
|
21
|
Orhan IE. Nature: a substantial source of auspicious substances with acetylcholinesterase inhibitory action. Curr Neuropharmacol 2013; 11:379-87. [PMID: 24381529 PMCID: PMC3744902 DOI: 10.2174/1570159x11311040003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 12/26/2022] Open
Abstract
Acetylcholinesterase (AChE) (EC 3.1.1.7) is an important enzyme that breaks down of acetylcholine in synaptic cleft in neuronal junctions. Inhibition of AChE is associated with treatment of several diseases such as Alzheimer's disease (AD), myasthenia gravis, and glaucoma as well as the mechanisms of insecticide and anthelmintic drugs. Several AChE inhibitors are available in clinical use currently for the treatment of AD; however, none of them has ability, yet, to seize progress of the disease. Consequently, an extensive research has been going on finding new AChE inhibitors. In this sense, natural inhibitors have gained great attention due to their encouraging effects toward AChE. In this review, promising candidate molecules with marked AChE inhibition from both plant and animal sources will be underlined.
Collapse
Affiliation(s)
- Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Turkey
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Eastern Mediterranean University, Gazimagosa, The Northern Cyprus via Turkey
| |
Collapse
|
22
|
Sgobba M, Olubiyi O, Ke S, Haider S. Molecular dynamics of HIV1-integrase in complex with 93del - a structural perspective on the mechanism of inhibition. J Biomol Struct Dyn 2012; 29:863-77. [PMID: 22292948 DOI: 10.1080/07391102.2012.10507418] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
HIV1 integrase is an important target for the antiviral therapy. Guanine-rich quadruplex, such as 93del, have been shown to be potent inhibitors of this enzyme and thus representing a new class of antiviral agents. Although X-ray and NMR structures of HIV1 integrase and 93del have been reported, there is no structural information of the complex and the mechanism of inhibition still remains unexplored. A number of computational methods including automated protein-DNA docking and molecular dynamics simulation in explicit solvent were used to model the binding of 93del to HIV1 integrase. Analysis of the dynamic behaviour of the complex using principal components analysis and elastic network modelling techniques allow us to understand how the binding of 93del aptamer and its interactions with key residues affect the intrinsic motions of the catalytic loops by stabilising them in catalytically inactive conformations. Such insights into the structural mechanism of inhibition can aid in improving the design of anti-HIV aptamers.
Collapse
Affiliation(s)
- Miriam Sgobba
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, 97 Lisburn Road, BT9 7BL, Belfast, UK.
| | | | | | | |
Collapse
|
23
|
Larif S, Ben Salem C, Soua Z, Hmouda H, Bouraoui K. Insight into TPMT(∗)23 mutation mis-folding using molecular dynamics simulation and protein structure analysis. J Biomol Struct Dyn 2012; 31:1066-76. [PMID: 23025308 DOI: 10.1080/07391102.2012.721495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Thiopurine S-methyltransferase (TPMT) is an important enzyme that metabolizes thiopurine drugs. This enzyme exhibits a large number of interindividual polymorphism. TPMT(∗)23 polymorphism has been reported in a few cases in the world in co-dominance with TPMT(∗)3A. The phenotype has been reported to affect enzyme activity in vivo and in vitro. Its underlying structural basis is not clarified yet. In our study, the wild type (WT) protein structure was analyzed and the amino acids bordering water channels in thiopurine sites were identified. Molecular dynamics of both the WT and TPMT(∗)23 mutation was carried out. In addition, the effects of this mutation, especially on the thiopurine site which is closed with a pincer like mechanism, were investigated. We focused on explaining how a locally occurred A167G substitution propagated through hydrogen bonds alteration to induce structural modification which affects both thiopurine and S-adenosylmethionine receptors. Finally, a genetic prediction of mutation functional consequences has been conducted confirming altered activity. An animated Interactive 3D Complement (I3DC) is available in Proteopedia at http://proteopedia.org/w/Journal:JBSD:20.
Collapse
Affiliation(s)
- Sofiene Larif
- a Faculty of Medicine of Sousse, Metabolic Biophysics and Applied Pharmacology Laboratory, Department of Biophysics , Avenue Mohamed Karoui , Sousse , 4002 , Tunisia
| | | | | | | | | |
Collapse
|
24
|
Abstract
Venoms and toxins are of significant interest due to their ability to cause a wide range of pathophysiological conditions that can potentially result in death. Despite their wide distribution among plants and animals, the biochemical pathways associated with these pathogenic agents remain largely unexplored. Impoverished and underdeveloped regions appear especially susceptible to increased incidence and severity due to poor socioeconomic conditions and lack of appropriate medical treatment infrastructure. To facilitate better management and treatment of envenomation victims, it is essential that the biochemical mechanisms of their action be elucidated. This review aims to characterize downstream envenomation mechanisms by addressing the major neuro-, cardio-, and hemotoxins as well as ion-channel toxins. Because of their use in folk and traditional medicine, the biochemistry behind venom therapy and possible implications on conventional medicine will also be addressed.
Collapse
|
25
|
Ling B, Sun M, Bi S, Jing Z, Liu Y. Molecular dynamics simulations of the coenzyme induced conformational changes of Mycobacterium tuberculosis L-alanine dehydrogenase. J Mol Graph Model 2012; 35:1-10. [PMID: 22459692 DOI: 10.1016/j.jmgm.2012.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 12/31/2011] [Accepted: 01/26/2012] [Indexed: 10/14/2022]
Abstract
Mycobacterium tuberculosis L-alanine dehydrogenase (L-MtAlaDH) catalyzes the NADH-dependent reversible oxidative deamination of L-alanine to pyruvate and ammonia. L-MtAlaDH has been proposed to be a potential target in the treatment of tuberculosis. Based on the crystal structures of this enzyme, molecular dynamics simulations were performed to investigate the conformational changes of L-MtAlaDH induced by coenzyme NADH. The results show that the presence of NADH in the binding domain restricts the motions and conformational distributions of L-MtAlaDH. There are two loops (residues 94-99 and 238-251) playing important roles for the binding of NADH, while another loop (residues 267-293) is responsible for the binding of substrate. The opening/closing and twisting motions of two domains are closely related to the conformational changes of L-MtAlaDH induced by NADH.
Collapse
Affiliation(s)
- Baoping Ling
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, Shandong 273165, China
| | | | | | | | | |
Collapse
|
26
|
Fang L, Pan Y, Muzyka JL, Zhan CG. Active site gating and substrate specificity of butyrylcholinesterase and acetylcholinesterase: insights from molecular dynamics simulations. J Phys Chem B 2011; 115:8797-805. [PMID: 21682268 PMCID: PMC3135420 DOI: 10.1021/jp112030p] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Butyrylcholinesterase (BChE) and acetylcholinesterase (AChE) are highly homologous proteins with distinct substrate preferences. In this study we compared the active sites of monomers and tetramers of human BChE and human AChE after performing molecular dynamics (MD) simulations in water-solvated systems. By comparing the conformational dynamics of gating residues of AChE and BChE, we found that the gating mechanisms of the main door of AChE and BChE are responsible for their different substrate specificities. Our simulation of the tetramers of AChE and BChE indicates that both enzymes could have two dysfunctional active sites due to their restricted accessibility to substrates. The further study on catalytic mechanisms of multiple forms of AChE and BChE would benefit from our comparison of the active sites of the monomers and tetramers of both enzymes.
Collapse
Affiliation(s)
- Lei Fang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536
| | - Yongmei Pan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536
| | | | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536
| |
Collapse
|
27
|
Xu Y, Colletier JP, Weik M, Qin G, Jiang H, Silman I, Sussman JL. Long route or shortcut? A molecular dynamics study of traffic of thiocholine within the active-site gorge of acetylcholinesterase. Biophys J 2011; 99:4003-11. [PMID: 21156143 DOI: 10.1016/j.bpj.2010.10.047] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 10/27/2010] [Accepted: 10/28/2010] [Indexed: 11/17/2022] Open
Abstract
The principal role of acetylcholinesterase is termination of nerve impulse transmission at cholinergic synapses, by rapid hydrolysis of the neurotransmitter acetylcholine to acetate and choline. Its active site is buried at the bottom of a deep and narrow gorge, at the rim of which is found a second anionic site, the peripheral anionic site. The fact that the active site is so deeply buried has raised cogent questions as to how rapid traffic of substrate and products occurs in such a confined environment. Various theoretical and experimental approaches have been used to solve this problem. Here, multiple conventional molecular dynamics simulations have been performed to investigate the clearance of the product, thiocholine, from the active-site gorge of acetylcholinesterase. Our results indicate that thiocholine is released from the peripheral anionic site via random pathways, while three exit routes appear to be favored for its release from the active site, namely, along the axis of the active-site gorge, and through putative back- and side-doors. The back-door pathway is that via which thiocholine exits most frequently. Our results are in good agreement with kinetic and kinetic-crystallography studies. We propose the use of multiple molecular dynamics simulations as a fast yet accurate complementary tool in structural studies of enzymatic trafficking.
Collapse
Affiliation(s)
- Yechun Xu
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | | | | | | | | | | | | |
Collapse
|
28
|
Bar-On D, Nachliel E, Gutman M, Ashery U. Dynamic conformational changes in munc18 prevent syntaxin binding. PLoS Comput Biol 2011; 7:e1001097. [PMID: 21390273 PMCID: PMC3048386 DOI: 10.1371/journal.pcbi.1001097] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 01/28/2011] [Indexed: 11/19/2022] Open
Abstract
The Sec1/munc18 protein family is essential for vesicle fusion in eukaryotic cells via binding to SNARE proteins. Protein kinase C modulates these interactions by phosphorylating munc18a thereby reducing its affinity to one of the central SNARE members, syntaxin-1a. The established hypothesis is that the reduced affinity of the phosphorylated munc18a to syntaxin-1a is a result of local electrostatic repulsion between the two proteins, which interferes with their compatibility. The current study challenges this paradigm and offers a novel mechanistic explanation by revealing a syntaxin-non-binding conformation of munc18a that is induced by the phosphomimetic mutations. In the present study, using molecular dynamics simulations, we explored the dynamics of the wild-type munc18a versus phosphomimetic mutant munc18a. We focused on the structural changes that occur in the cavity between domains 3a and 1, which serves as the main syntaxin-binding site. The results of the simulations suggest that the free wild-type munc18a exhibits a dynamic equilibrium between several conformations differing in the size of its cavity (the main syntaxin-binding site). The flexibility of the cavity's size might facilitate the binding or unbinding of syntaxin. In silico insertion of phosphomimetic mutations into the munc18a structure induces the formation of a conformation where the syntaxin-binding area is rigid and blocked as a result of interactions between residues located on both sides of the cavity. Therefore, we suggest that the reduced affinity of the phosphomimetic mutant/phosphorylated munc18a is a result of the closed-cavity conformation, which makes syntaxin binding energetically and sterically unfavorable. The current study demonstrates the potential of phosphoryalation, an essential biological process, to serve as a driving force for dramatic conformational changes of proteins modulating their affinity to target proteins. Protein phosphorylation plays a significant regulatory role in multi-component systems engaged in signal transduction or coordination of cellular processes, by activating or deactivating proteins. The potential of phosphorylation to induce substantial conformational changes in proteins, thereby changing their affinity to target proteins, has already been shown but the dynamics of the process is not fully elucidated. In the present study, we investigated, by molecular dynamics simulations, the dynamic conformational changes in munc18a, a protein that is crucial for neurotransmitter release and interacts tightly with the SNARE syntaxin-1. We further investigated the conformational changes that occur in munc18a when it is phosphorylated, reducing its affinity to syntaxin-1a. The results of the simulations suggest that there is a conformational flexibility of the syntaxin-unbounded munc18a that allows changes in the shape of the syntaxin-1a binding cavity. In silico insertion of phosphomimetic mutations into munc18a led to a reduction in the flexibility and closure of the syntaxin-binding site. We suggest that the reduced affinity of phosphorylated munc18a to syntaxin-1a stems from the difficulty of syntaxin-1a to bind to the munc18a closed-cavity conformation, induced by the PKC phosphorylation of munc18a.
Collapse
Affiliation(s)
- Dana Bar-On
- Department of Neurobiology, Tel Aviv University, Tel Aviv, Israel
- Laser Laboratory for Fast Reactions in Biology, Department of Biochemistry, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Esther Nachliel
- Laser Laboratory for Fast Reactions in Biology, Department of Biochemistry, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Menachem Gutman
- Laser Laboratory for Fast Reactions in Biology, Department of Biochemistry, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ashery
- Department of Neurobiology, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
29
|
Wiesner J, Kříž Z, Kuča K, Jun D, Koča J. Influence of the acetylcholinesterase active site protonation on omega loop and active site dynamics. J Biomol Struct Dyn 2011; 28:393-403. [PMID: 20919754 DOI: 10.1080/07391102.2010.10507368] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Existence of alternative entrances in acetylcholinesterase (AChE) could explain the contrast between the very high AChE catalytic efficiency and the narrow and long access path to the active site revealed by X-ray crystallography. Alternative entrances could facilitate diffusion of the reaction products or at least water and ions from the active site. Previous molecular dynamics simulations identified side door and back door as the most probable alternative entrances. The simulations of non-inhibited AChE suggested that the back door opening events occur only rarely (0.8% of the time in the 10ns trajectory). Here we present a molecular dynamics simulation of non-inhibited AChE, where the back door opening appears much more often (14% of the time in the 12ns trajectory) and where the side door opening was observed quite frequently (78% of trajectory time). We also present molecular dynamics, where the back door does not open at all, or where large conformational changes of the AChE omega loop occur together with alternative passage opening events. All these differences in AChE dynamical behavior are caused by different protonation states of two glutamate residues located on bottom of the active site gorge (Glu202 and G450 in Mus musculus AChE). Our results confirm the results of previous molecular dynamics simulations, expand the view and suggest the probable reasons for the overall conformational behavior of AChE omega loop.
Collapse
Affiliation(s)
- Jiří Wiesner
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5/A4, 625 00 Brno, Czech Republic
| | | | | | | | | |
Collapse
|
30
|
Yan C, Kaoud T, Lee S, Dalby KN, Ren P. Understanding the specificity of a docking interaction between JNK1 and the scaffolding protein JIP1. J Phys Chem B 2011; 115:1491-502. [PMID: 21261310 DOI: 10.1021/jp1073522] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The up-regulation of JNK activity is associated with a number of disease states. The JNK-JIP1 interaction represents an attractive target for the inhibition of JNK-mediated signaling. In this study, molecular dynamics simulations have been performed on the apo-JNK1 and the JNK1•L-pepJIP1 and JNK1•D-pepJIP1 complexes to investigate the interaction between the JIP1 peptides and JNK1. Dynamic domain studies based on essential dynamics (ED) analysis of apo-JNK1 and the JNK1•L-pepJIP1 complex have been performed to analyze and compare details of conformational changes, hinge axes, and hinge bending regions in both structures. The activation loop, the αC helix, and the G loop are found to be highly flexible and to exhibit significant changes in dynamics upon L-pepJIP1 binding. The conformation of the activation loop for the apo state is similar to that of inactive apo-ERK2, while the activation loop in JNK1•L-pepJIP1 complex resembles that of the inactive ERK2 bound with pepHePTP. ED analysis shows that, after the binding of l-pepJIP1, the N- and C-terminal domains of JNK1 display both a closure and a twisting motion centered around the activation loop, which functions as a hinge. In contrast, no domain motion is detected for the apo state for which an open conformation is favored. The present study suggests that L-pepJIP1 regulates the interdomain motions of JNK1 and potentially the active site via an allosteric mechanism. The binding free energies of L-pepJIP1 and D-pepJIP1 to JNK1 are estimated using the molecular mechanics Poisson-Boltzmann and generalized-Born surface area (MM-PB/GBSA) methods. The contribution of each residue at the interaction interface to the binding affinity of L-pepJIP1 with JNK1 has been analyzed by means of computational alanine-scanning mutagenesis and free energy decomposition. Several critical interactions for binding (e.g., Arg156/L-pepJIP1 and Glu329/JNK1) have been identified. The binding free energy calculation indicates that the electrostatic interaction contributes critically to specificity, rather than to binding affinity between the peptide and JNK1. Notably, the binding free energy calculations predict that D-pepJIP1 binding to JNK1 is significantly weaker than the L form, contradicting the previous suggestion that D-pepJIP1 acts as an inhibitor toward JNK1. We have performed experiments using purified JNK1 to confirm that, indeed, D-pepJIP1 does not inhibit the ability of JNK1 to phosphorylate c-Jun in vitro.
Collapse
Affiliation(s)
- Chunli Yan
- Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA
| | | | | | | | | |
Collapse
|
31
|
Lin YS, Lin JH, Chang CC. Molecular dynamics simulations of the rotary motor F(0) under external electric fields across the membrane. Biophys J 2010; 98:1009-17. [PMID: 20303858 DOI: 10.1016/j.bpj.2009.11.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 10/12/2009] [Accepted: 11/13/2009] [Indexed: 11/28/2022] Open
Abstract
The membrane-bound component F(0), which is a major component of the F(0)F(1)-ATP synthase, works as a rotary motor and plays a central role in driving the F(1) component to transform chemiosmotic energy into ATP synthesis. We conducted molecular dynamics simulations of b(2)-free F(0) in a 1-palmitoyl-2-oleoyl-phosphatidylcholine lipid bilayer for tens of nanoseconds with two different protonation states of the cAsp-61 residue at the interface of the a-c complex in the absence of electric fields and under electric fields of +/-0.03 V/nm across the membrane. To our surprise, we observed that the upper half of the N-terminal helix of the c(1) subunit rotated about its axis clockwise by 30 degrees . An energetic analysis revealed that the electrostatic repulsion between this N-terminal helix and subunit c(12) was a major contributor to the observed rotation. A correlation map analysis indicated that the correlated motions of residues in the interface of the a-c complex were significantly reduced by external electric fields. The deuterium order parameter (S(CD)) profile calculated by averaging all the lipids in the F(0)-bound bilayer was not very different from that of the pure bilayer system, in agreement with recent (2)H solid-state NMR experiments. However, by delineating the lipid properties according to their vicinity to F(0), we found that the S(CD) profiles of different lipid shells were prominently different. Lipids close to F(0) formed a more ordered structure. Similarly, the lateral diffusion of lipids on the membrane surface also followed a shell-dependent behavior. The lipids in the proximity of F(0) exhibited very significantly reduced diffusional motion. The numerical value of S(CD) was anticorrelated with that of the diffusion coefficient, i.e., the more ordered lipid structures led to slower lipid diffusion. Our findings will help elucidate the dynamics of F(0) depending on the protonation state and electric field, and may also shed some light on the interactions between the motor F(0) and its surrounding lipids under physiological conditions, which could help to rationalize its extraordinary energy conversion efficiency.
Collapse
Affiliation(s)
- Yang-Shan Lin
- Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
32
|
Differential binding of bispyridinium oxime drugs with acetylcholinesterase. Acta Pharmacol Sin 2010; 31:313-28. [PMID: 20140002 DOI: 10.1038/aps.2009.193] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
AIM To performe a time-dependent topographical delineation of protein-drug interactions to gain molecular insight into the supremacy of Ortho-7 over HI-6 in reactivating tabun-conjugated mouse acetylcholinesterase (mAChE). METHODS We conducted all-atom steered molecular dynamics simulations of the two protein-drug complexes. Through a host of protein-drug interaction parameters (rupture force profiles, hydrogen bonds, water bridges, hydrophobic interactions), geometrical, and orientation ordering of the drugs, we monitored the enzyme's response during the release of the drugs from its active-site. RESULTS The results show the preferential binding of the drugs with the enzyme. The pyridinium ring of HI-6 shows excellent complementary binding with the peripheral anionic site, whereas one of two identical pyridinium rings of Ortho-7 has excellent binding compatibility in the enzyme active-site where it can orchestrate the reactivation process. We found that the active pyridinium ring of HI-6 undergoes a complete turn along the active site axis, directed away from the active-site region during the course of the simulation. CONCLUSION Due to excellent cooperative binding of Ortho-7, as rendered by several cation-pi interactions with the active-site gorge of the enzyme, Ortho-7 may be a more efficient reactivator than HI-6. Our work supports the growing body of evidence that the efficacy of the drugs is due to the differential bindings of the oximes with AChE and can aid to the rational design of oxime drugs.
Collapse
|
33
|
Butini S, Campiani G, Borriello M, Gemma S, Panico A, Persico M, Catalanotti B, Ros S, Brindisi M, Agnusdei M, Fiorini I, Nacci V, Novellino E, Belinskaya T, Saxena A, Fattorusso C. Exploiting Protein Fluctuations at the Active-Site Gorge of Human Cholinesterases: Further Optimization of the Design Strategy to Develop Extremely Potent Inhibitors. J Med Chem 2008; 51:3154-70. [DOI: 10.1021/jm701253t] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Stefania Butini
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Giuseppe Campiani
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Marianna Borriello
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Sandra Gemma
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Alessandro Panico
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Marco Persico
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Bruno Catalanotti
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Sindu Ros
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Margherita Brindisi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Marianna Agnusdei
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Isabella Fiorini
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Vito Nacci
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Ettore Novellino
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Tatyana Belinskaya
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Ashima Saxena
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| | - Caterina Fattorusso
- European Research Centre for Drug Discovery and Development (NatSynDrugs), Università di Siena, 53100 Siena, Italy, Dipartimento Farmaco Chimico Tecnologico, via Aldo Moro, Università di Siena, 53100 Siena, Italy Dipartimento di Chimica delle Sostanze Naturali e Dipartimento di Chimica Farmaceutica e Tossicologica Università di Napoli Federico II, via D. Montesano 49, 80131 Napoli, Italy, and Division of Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910
| |
Collapse
|
34
|
Molecular dynamics and principal components analysis of human telomeric quadruplex multimers. Biophys J 2008; 95:296-311. [PMID: 18375510 DOI: 10.1529/biophysj.107.120501] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Guanine-rich DNA repeat sequences located at the terminal ends of chromosomal DNA can fold in a sequence-dependent manner into G-quadruplex structures, notably the terminal 150-200 nucleotides at the 3' end, which occur as a single-stranded DNA overhang. The crystal structures of quadruplexes with two and four human telomeric repeats show an all-parallel-stranded topology that is readily capable of forming extended stacks of such quadruplex structures, with external TTA loops positioned to potentially interact with other macromolecules. This study reports on possible arrangements for these quadruplex dimers and tetramers, which can be formed from 8 or 16 telomeric DNA repeats, and on a methodology for modeling their interactions with small molecules. A series of computational methods including molecular dynamics, free energy calculations, and principal components analysis have been used to characterize the properties of these higher-order G-quadruplex dimers and tetramers with parallel-stranded topology. The results confirm the stability of the central G-tetrads, the individual quadruplexes, and the resulting multimers. Principal components analysis has been carried out to highlight the dominant motions in these G-quadruplex dimer and multimer structures. The TTA loop is the most flexible part of the model and the overall multimer quadruplex becoming more stable with the addition of further G-tetrads. The addition of a ligand to the model confirms the hypothesis that flat planar chromophores stabilize G-quadruplex structures by making them less flexible.
Collapse
|
35
|
Dalhaimer P, Pollard TD, Nolen BJ. Nucleotide-mediated conformational changes of monomeric actin and Arp3 studied by molecular dynamics simulations. J Mol Biol 2007; 376:166-83. [PMID: 18155236 DOI: 10.1016/j.jmb.2007.11.068] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 11/20/2007] [Accepted: 11/21/2007] [Indexed: 10/22/2022]
Abstract
Members of the actin family of proteins exhibit different biochemical properties when ATP, ADP-P(i), ADP, or no nucleotide is bound. We used molecular dynamics simulations to study the effect of nucleotides on the behavior of actin and actin-related protein 3 (Arp3). In all of the actin simulations, the nucleotide cleft stayed closed, as in most crystal structures. ADP was much more mobile within the cleft than ATP, despite the fact that both nucleotides adopt identical conformations in actin crystal structures. The nucleotide cleft of Arp3 opened in most simulations with ATP, ADP, and no bound nucleotide. Deletion of a C-terminal region of Arp3 that extends beyond the conserved actin sequence reduced the tendency of the Arp3 cleft to open. When the Arp3 cleft opened, we observed multiple instances of partial release of the nucleotide. Cleft opening in Arp3 also allowed us to observe correlated movements of the phosphate clamp, cleft mouth, and barbed-end groove, providing a way for changes in the nucleotide state to be relayed to other parts of Arp3. The DNase binding loop of actin was highly flexible regardless of the nucleotide state. The conformation of Ser14/Thr14 in the P1 loop was sensitive to the presence of the gamma-phosphate, but other changes observed in crystal structures were not correlated with the nucleotide state on nanosecond timescales. The divalent cation occupied three positions in the nucleotide cleft, one of which was not previously observed in actin or Arp2/3 complex structures. In sum, these simulations show that subtle differences in structures of actin family proteins have profound effects on their nucleotide-driven behavior.
Collapse
Affiliation(s)
- Paul Dalhaimer
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | | | | |
Collapse
|
36
|
Colletier JP, Weik M. Exploration structurale du paysage conformationnel de l’acétylcholinestérase par cristallographie cinétique. ANNALES PHARMACEUTIQUES FRANÇAISES 2007; 65:108-18. [PMID: 17404544 DOI: 10.1016/s0003-4509(07)90024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Acetylcholinesterase is a very rapid enzyme, essential in the process of nerve impulse transmission at cholinergic synapses. It is the target of all currently approved anti-Alzheimer drugs and further progress in the modulation of its activity requires structural as well as dynamical information. Exploration of the conformational energy landscape of a protein by means of X-ray crystallography requires the use of experimental tricks, to overcome the inherently static nature of crystallographic structures. Here we report three experimental approaches that allowed to gain structural insight into the dynamics of acetylcholinesterase, which is relevant for structure-based drug design.
Collapse
Affiliation(s)
- J-P Colletier
- CEA-CNRS-UJF, Institut de biologie structurale J.-P. Ebel, Umr 5075, Laboratoire de biophysique moléculaire, F 38027 Grenoble.
| | | |
Collapse
|
37
|
Wang J, Gu J, Leszczynski J. A model study of interactions between TcAChE peripheral site segment Tyr70Val71 and loop 1 of Fasciculin 2. J Biomol Struct Dyn 2006; 24:139-48. [PMID: 16928137 DOI: 10.1080/07391102.2006.10507107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The continuous chain of residues (Thr7 to Ala12) of Loop1 of Fas2 (F1) and its interaction with the peripheral binding sites (Tyr70-Val71) of AChE (P1) has been studied. Our results suggest that the flexibility of Loop1 might be caused by either the partially protonated guanidine group of Arg11 under experimental conditions or by the interaction with the negatively charged center of substrates. The binding energy of F1-P1 is predicted to be -16.6 kcal/mol at the B3LYP/6-311G(d,p) level, which is assumed to originate from one isolated O7...HN10 H-bond, one possible O10...HC71 unconventional O...HC type H-bonding, and the improved pi-bonding cooperativity around the peptide group of the AChE segment Tyr70-Val71. The classical Kitaura-Morokuma energy decomposition analysis, the NPA charge analysis, and the AIM analysis consistently reveal that the peptide group in segment P1 is more polarizable, which might play the key role in the interactions between F1 and P1. The PCM solvent effect corrected results reveal decrease of the interaction energy of the considered model. The importance of Thr8 of Fas2 in the P-site binding of AChE is also concluded. Site-directed mutations on either the Fas2 residue of Thr8 or the AChE residue of Tyr70 are expected to alter the binding behavior of the Loop1 of Fas2 with AChE.
Collapse
Affiliation(s)
- Jing Wang
- Computational Center for Molecular Structure and Interactions, Department of Chemistry, Jackson State University, Jackson, MS 39217, USA
| | | | | |
Collapse
|
38
|
Modelling interactions between Loop1 of Fasciculin2 (Fas2) and Torpedo californica acetylcholinesterase (TcAChE). Chem Phys Lett 2006. [DOI: 10.1016/j.cplett.2006.09.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
39
|
Bui JM, McCammon JA. Protein complex formation by acetylcholinesterase and the neurotoxin fasciculin-2 appears to involve an induced-fit mechanism. Proc Natl Acad Sci U S A 2006; 103:15451-6. [PMID: 17021015 PMCID: PMC1591298 DOI: 10.1073/pnas.0605355103] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Specific, rapid association of protein complexes is essential for all forms of cellular existence. The initial association of two molecules in diffusion-controlled reactions is often influenced by the electrostatic potential. Yet, the detailed binding mechanisms of proteins highly depend on the particular system. A complete protein complex formation pathway has been delineated by using structural information sampled over the course of the transformation reaction. The pathway begins at an encounter complex that is formed by one of the apo forms of neurotoxin fasciculin-2 (FAS2) and its high-affinity binding protein, acetylcholinesterase (AChE), followed by rapid conformational rearrangements into an intermediate complex that subsequently converts to the final complex as observed in crystal structures. Formation of the intermediate complex has also been independently captured in a separate 20-ns molecular dynamics simulation of the encounter complex. Conformational transitions between the apo and liganded states of FAS2 in the presence and absence of AChE are described in terms of their relative free energy profiles that link these two states. The transitions of FAS2 after binding to AChE are significantly faster than in the absence of AChE; the energy barrier between the two conformational states is reduced by half. Conformational rearrangements of FAS2 to the final liganded form not only bring the FAS2/AChE complex to lower energy states, but by controlling transient motions that lead to opening or closing one of the alternative passages to the active site of the enzyme also maximize the ligand's inhibition of the enzyme.
Collapse
Affiliation(s)
- Jennifer M Bui
- Department of Chemistry and Biochemistry, Howard Hughes Medical Institute, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0365, USA.
| | | |
Collapse
|
40
|
Tai K, Baaden M, Murdock S, Wu B, Ng MH, Johnston S, Boardman R, Fangohr H, Cox K, Essex JW, Sansom MSP. Three hydrolases and a transferase: comparative analysis of active-site dynamics via the BioSimGrid database. J Mol Graph Model 2006; 25:896-902. [PMID: 17011806 DOI: 10.1016/j.jmgm.2006.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Revised: 08/25/2006] [Accepted: 08/29/2006] [Indexed: 11/25/2022]
Abstract
Comparative molecular dynamics (MD) simulations enable us to explore the conformational dynamics of the active sites of distantly related enzymes. We have used the BioSimGrid (http://www.biosimgrid.org) database to facilitate such a comparison. Simulations of four enzymes were analyzed. These included three hydrolases and a transferase, namely acetylcholinesterase, outer-membrane phospholipase A, outer-membrane protease T, and PagP (an outer-membrane enzyme which transfers a palmitate chain from a phospholipid to lipid A). A set of 17 simulations were analyzed corresponding to a total of approximately 0.1 micros simulation time. A simple metric for active-site integrity was used to demonstrate the existence of clusters of dynamic conformational behaviour of the active sites. Small (i.e. within a cluster) fluctuations appear to be related to the function of an enzymatically active site. Larger fluctuations (i.e. between clusters) correlate with transitions between catalytically active and inactive states. Overall, these results demonstrate the potential of a comparative MD approach to analysis of enzyme function. This approach could be extended to a wider range of enzymes using current high throughput MD simulation and database methods.
Collapse
Affiliation(s)
- Kaihsu Tai
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Herein, we present results from molecular dynamics (MD) simulations of the human butyrylcholinesterase (BuChE) enzyme in aqueous solution. Two configurations of the unbound form of BuChE differing in the presence or absence of a sodium ion inside the protein gorge were simulated for 10 and 5 ns, respectively. Besides complementing the structural information provided by X-ray data, the MD simulations give insight into the structure of the native BuChE enzyme. For example, it is shown that: the nucleophilic Ser(198) residue and the various binding subsites in the BuChE catalytic cavity are readily accessible from the exterior of the protein; the presence of the sodium ion dynamically explores two different binding sites in the gorge leading to the active site and stabilizes the productive conformation of the Glu(325)/His(438)/Ser(198) catalytic triad; several long-lived water bridges are fully integrated into the architecture of the active site; the positions of the residues at the rim of the gorge region display large deviations with respect to the crystal structure; and two side doors, constituted by residues situated at the tip of the acyl- and Omega-loops, respectively, open wide enough to allow the passage of water molecules. In conclusion, we compare our theoretical results with those from previous work on mouse acetylcholinesterase and discuss their implications for substrate binding and catalysis in BuChE.
Collapse
|
42
|
Alisaraie L, Fels G. Molecular docking study on the “back door” hypothesis for product clearance in acetylcholinesterase. J Mol Model 2005; 12:348-54. [PMID: 16341717 DOI: 10.1007/s00894-005-0051-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2005] [Accepted: 09/23/2005] [Indexed: 11/26/2022]
Abstract
Acetylcholinesterase (AChE) is one of the fastest enzymes known, even though the active site is buried inside the protein at the end of a 20-A deep narrow gorge. Among the great variety of crystal structures of this enzyme, both in the absence and presence of various ligands and proteins, the structure of a complex of AChE with the pseudo-irreversible inhibitor Mf268 is of particular interest, as it assists in the proposal of a back door for product clearance from the active site. Binding of Mf268 to AChE results in the carbamoylation of Ser200 and liberation of an eseroline-fragment as the leaving group. The crystal structure of the AChE-Mf268 complex, however, proves that eseroline has escaped from the enzyme, despite the fact that the Ser-bound inhibitor fragment blocks the gorge entrance. The existence of alternative routes other than through the gorge for product clearance has been postulated but is still controversially discussed in the literature, as an experimental proof for such a back door is still missing. We have used Monte Carlo-based molecular docking methods in order to examine possible alternative pathways that could allow eseroline to be released from the protein after being cleaved from the substrate by Ser200. Based on our results, a short channel at the bottom of the gorge seems to be the most probable back-door site, which begins at amino acid Trp84 and ends at the enzyme surface in a cavity close to amino acid Glu445. [Figure: see text].
Collapse
Affiliation(s)
- Laleh Alisaraie
- Department of Chemistry, University of Paderborn, Warburgerstr. 100, D-33098, Paderborn, Germany
| | | |
Collapse
|
43
|
Minh DDL, Bui JM, Chang CE, Jain T, Swanson JMJ, McCammon JA. The entropic cost of protein-protein association: a case study on acetylcholinesterase binding to fasciculin-2. Biophys J 2005; 89:L25-7. [PMID: 16100267 PMCID: PMC1366787 DOI: 10.1529/biophysj.105.069336] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
44
|
Gabel F, Weik M, Masson P, Renault F, Fournier D, Brochier L, Doctor BP, Saxena A, Silman I, Zaccai G. Effects of soman inhibition and of structural differences on cholinesterase molecular dynamics: a neutron scattering study. Biophys J 2005; 89:3303-11. [PMID: 16100272 PMCID: PMC1366826 DOI: 10.1529/biophysj.105.061028] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Incoherent elastic neutron scattering experiments on members of the cholinesterase family were carried out to investigate how molecular dynamics is affected by covalent inhibitor binding and by differences in primary and quaternary structure. Tetrameric native and soman-inhibited human butyrylcholinesterase (HuBChE) as well as native dimeric Drosophila melanogaster acetylcholinesterase (DmAChE) hydrated protein powders were examined. Atomic mean-square displacements (MSDs) were found to be identical for native HuBChE and for DmAChE in the whole temperature range examined, leading to the conclusion that differences in activity and substrate specificity are not reflected by a global modification of subnanosecond molecular dynamics. MSDs of native and soman-inhibited HuBChE were identical below the thermal denaturation temperature of the native enzyme, indicating a common mean free-energy surface. Denaturation of the native enzyme is reflected by a relative increase of MSDs consistent with entropic stabilization of the unfolded state. The results suggest that the stabilization of HuBChE phosphorylated by soman is due to an increase in free energy of the unfolded state due to a decrease in entropy.
Collapse
Affiliation(s)
- F Gabel
- Laboratoire de Biophysique Moléculaire, Institut de Biologie Structurale, 41 rue Jules Horowitz, F-38027 Grenoble Cedex 1, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Haider S, Grottesi A, Hall BA, Ashcroft FM, Sansom MSP. Conformational dynamics of the ligand-binding domain of inward rectifier K channels as revealed by molecular dynamics simulations: toward an understanding of Kir channel gating. Biophys J 2005; 88:3310-20. [PMID: 15749783 PMCID: PMC1305479 DOI: 10.1529/biophysj.104.052019] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Inward rectifier (Kir) potassium channels are characterized by two transmembrane helices per subunit, plus an intracellular C-terminal domain that controls channel gating in response to changes in concentration of various ligands. Based on the crystal structure of the tetrameric C-terminal domain of Kir3.1, it is possible to build a homology model of the ATP-binding C-terminal domain of Kir6.2. Molecular dynamics simulations have been used to probe the dynamics of Kir C-terminal domains and to explore the relationship between their dynamics and possible mechanisms of channel gating. Multiple simulations, each of 10 ns duration, have been performed for Kir3.1 (crystal structure) and Kir6.2 (homology model), in both their monomeric and tetrameric forms. The Kir6.2 simulations were performed with and without bound ATP. The results of the simulations reveal comparable conformational stability for the crystal structure and the homology model. There is some decrease in conformational flexibility when comparing the monomers with the tetramers, corresponding mainly to the subunit interfaces in the tetramer. The beta-phosphate of ATP interacts with the side chain of K185 in the Kir6.2 model and simulations. The flexibility of the Kir6.2 tetramer is not changed greatly by the presence of bound ATP, other than in two loop regions. Principal components analysis of the simulated dynamics suggests loss of symmetry in both the Kir3.1 and Kir6.2 tetramers, consistent with "dimer-of-dimers" motion of subunits in C-terminal domains of the corresponding Kir channels. This is suggestive of a gating model in which a transition between exact tetrameric symmetry and dimer-of-dimers symmetry is associated with a change in transmembrane helix packing coupled to gating of the channel. Dimer-of-dimers motion of the C-terminal domain tetramer is also supported by coarse-grained (anisotropic network model) calculations. It is of interest that loss of exact rotational symmetry has also been suggested to play a role in gating in the bacterial Kir homolog, KirBac1.1, and in the nicotinic acetylcholine receptor channel.
Collapse
Affiliation(s)
- Shozeb Haider
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | | | | | | | | |
Collapse
|
46
|
Hung A, Tai K, Sansom MSP. Molecular dynamics simulation of the M2 helices within the nicotinic acetylcholine receptor transmembrane domain: structure and collective motions. Biophys J 2005; 88:3321-33. [PMID: 15722430 PMCID: PMC1305480 DOI: 10.1529/biophysj.104.052878] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multiple nanosecond duration molecular dynamics simulations were performed on the transmembrane region of the Torpedo nicotinic acetylcholine receptor embedded within a bilayer mimetic octane slab. The M2 helices and M2-M3 loop regions were free to move, whereas the outer (M1, M3, M4) helix bundle was backbone restrained. The M2 helices largely retain their hydrogen-bonding pattern throughout the simulation, with some distortions in the helical end and loop regions. All of the M2 helices exhibit bending motions, with the hinge point in the vicinity of the central hydrophobic gate region (corresponding to residues alphaL251 and alphaV255). The bending motions of the M2 helices lead to a degree of dynamic narrowing of the pore in the region of the proposed hydrophobic gate. Calculations of Born energy profiles for various structures along the simulation trajectory suggest that the conformations of the M2 bundle sampled correspond to a closed conformation of the channel. Principal components analyses of each of the M2 helices, and of the five-helix M2 bundle, reveal concerted motions that may be relevant to channel function. Normal mode analyses using the anisotropic network model reveal collective motions similar to those identified by principal components analyses.
Collapse
Affiliation(s)
- Andrew Hung
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | | | | |
Collapse
|
47
|
Kawakubo T, Okada O, Minami T. Molecular dynamics simulations of evolved collective motions of atoms in the myosin motor domain upon perturbation of the ATPase pocket. Biophys Chem 2005; 115:77-85. [PMID: 15848287 DOI: 10.1016/j.bpc.2004.12.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2004] [Revised: 12/24/2004] [Accepted: 12/24/2004] [Indexed: 10/25/2022]
Abstract
A crucial point for mechanical force generation in actomyosin systems is how the energy released by ATP hydrolysis in the myosin motor domain gives rise to the movement of the myosin head along the actin filament. We assumed the signal of the ATP hydrolysis to be transmitted as modulated atomic vibrations from the nucleotide-binding site throughout the myosin head, and carried out 1-ns all-atom molecular dynamics simulations for that signal transmission. We distributed the released energy to atoms located around the ATPase pocket as kinetic energies and examined how the effect of disturbance extended throughout the motor domain. The result showed that the disturbance signal extended over the motor domain in 150 ps and induced slowly varying collective motions of atoms at the actin-binding site and the junction with the neck, both of which are relevant to the movement of the myosin head along the actin filament. We also performed a principal component analysis of thermal atomic motions for the motor domain, and the first principal component was consistent with the response to the disturbance given to the ATPase pocket.
Collapse
|
48
|
Bui JM, Henchman RH, McCammon JA. The dynamics of ligand barrier crossing inside the acetylcholinesterase gorge. Biophys J 2004; 85:2267-72. [PMID: 14507691 PMCID: PMC1303452 DOI: 10.1016/s0006-3495(03)74651-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The dynamics of ligand movement through the constricted region of the acetylcholinesterase gorge is important in understanding how the ligand gains access to and is released from the active site of the enzyme. Molecular dynamics simulations of the simple ligand, tetramethylammonium, crossing this bottleneck region are conducted using umbrella potential sampling and activated flux techniques. The low potential of mean force obtained is consistent with the fast reaction rate of acetylcholinesterase observed experimentally. From the results of the activated dynamics simulations, local conformational fluctuations of the gorge residues and larger scale collective motions of the protein are found to correlate highly with the ligand crossing.
Collapse
Affiliation(s)
- Jennifer M Bui
- Howard Hughes Medical Institute, Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093-0365, USA.
| | | | | |
Collapse
|
49
|
Zeev-Ben-Mordehai T, Silman I, Sussman JL. Acetylcholinesterase in motion: visualizing conformational changes in crystal structures by a morphing procedure. Biopolymers 2003; 68:395-406. [PMID: 12601798 DOI: 10.1002/bip.10287] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In order to visualize and appreciate conformational changes between homologous three-dimensional (3D) protein structures or protein/inhibitor complexes, we have developed a user-friendly morphing procedure. It enabled us to detect coordinated conformational changes not easily discernible by analytic methods or by comparison of static images. This procedure was applied to comparison of native Torpedo californica acetylcholinesterase and of complexes with reversible inhibitors and conjugates with covalent inhibitors. It was likewise shown to be valuable for the visualization of conformational differences between acetylcholinesterases from different species. The procedure involves generation, in Cartesian space, of 25 interpolated intermediate structures between the initial and final 3D structures, which then serve as the individual frames in a QuickTime movie.
Collapse
Affiliation(s)
- T Zeev-Ben-Mordehai
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | |
Collapse
|
50
|
Canino LS, Shen T, McCammon JA. Changes in flexibility upon binding: Application of the self-consistent pair contact probability method to protein-protein interactions. J Chem Phys 2002. [DOI: 10.1063/1.1517605] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|