Scalable Constant pH Molecular Dynamics in GROMACSClick to copy article linkArticle link copied!
- Noora Aho*Noora Aho*Email: [email protected]Nanoscience Center and Department of Chemistry, University of Jyväskylä, 40014Jyväskylä, FinlandMore by Noora Aho
- Pavel Buslaev*Pavel Buslaev*Email: [email protected]Nanoscience Center and Department of Chemistry, University of Jyväskylä, 40014Jyväskylä, FinlandMore by Pavel Buslaev
- Anton JansenAnton JansenDepartment of Applied Physics and Swedish e-Science Research Center, Science for Life Laboratory, KTH Royal Institute of Technology, 100 44Stockholm, SwedenMore by Anton Jansen
- Paul BauerPaul BauerDepartment of Applied Physics and Swedish e-Science Research Center, Science for Life Laboratory, KTH Royal Institute of Technology, 100 44Stockholm, SwedenMore by Paul Bauer
- Gerrit Groenhof*Gerrit Groenhof*Email: [email protected]Nanoscience Center and Department of Chemistry, University of Jyväskylä, 40014Jyväskylä, FinlandMore by Gerrit Groenhof
- Berk Hess*Berk Hess*Email: [email protected]Department of Applied Physics and Swedish e-Science Research Center, Science for Life Laboratory, KTH Royal Institute of Technology, 100 44Stockholm, SwedenMore by Berk Hess
Abstract
Molecular dynamics (MD) computer simulations are used routinely to compute atomistic trajectories of complex systems. Systems are simulated in various ensembles, depending on the experimental conditions one aims to mimic. While constant energy, temperature, volume, and pressure are rather straightforward to model, pH, which is an equally important parameter in experiments, is more difficult to account for in simulations. Although a constant pH algorithm based on the λ-dynamics approach by Brooks and co-workers [Kong, X.; Brooks III, C. L. J. Chem. Phys.1996, 105, 2414–2423] was implemented in a fork of the GROMACS molecular dynamics program, uptake has been rather limited, presumably due to the poor scaling of that code with respect to the number of titratable sites. To overcome this limitation, we implemented an alternative scheme for interpolating the Hamiltonians of the protonation states that makes the constant pH molecular dynamics simulations almost as fast as a normal MD simulation with GROMACS. In addition, we implemented a simpler scheme, called multisite representation, for modeling side chains with multiple titratable sites, such as imidazole rings. This scheme, which is based on constraining the sum of the λ-coordinates, not only reduces the complexity associated with parametrizing the intramolecular interactions between the sites but also is easily extendable to other molecules with multiple titratable sites. With the combination of a more efficient interpolation scheme and multisite representation of titratable groups, we anticipate a rapid uptake of constant pH molecular dynamics simulations within the GROMACS user community.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Theory
λ-Dynamics-Based Constant pH MD Simulations
λ-Dependent Potential Energy Function
Figure 1
Figure 1. Illustration of the additional λ-dependent potential energy terms (B–D). Panel A shows the protonation free energy of a titratable residue in its reference state obtained at the force field level, ΔGiMM(λi) . To compensate for the shortcomings of the force field and obtain a zero free energy difference between the two protonation states (A + B), we add the correction potential, ViMM(λ), shown in panel B. A biasing potential, Vibias(λ), (42) is introduced to avoid sampling of nonphysical states (panel C). To model the proton chemical potential (pH), we add a pH-dependent term, VpH(λi) (panel D). For a titratable residue at pH ≠ pKa, the total λ-dependent potential, including the interpolated force field functions and the three additional terms, is illustrated in panel (A + B + C + D).
Linear Interpolation of Potential Energy Functions

1. | Interactions between atoms that are not part of any λ-group, and hence independent of the λi’s: (10) | ||||
2. | Interpolated interactions between atoms of each λ-group with atoms that are not part of any λ-group: (11) | ||||
3. | Interpolated interactions between atoms belonging to two different λ-groups: (12) | ||||
4. | Interpolated interactions within each of the λ-groups: (13) |
Linear Interpolation of Partial Charges
Multisite Representation of Chemically Coupled Titratable Sites
Figure 2
Figure 2. Multisite representation illustrated for a histidine side chain. Each possible protonation state is represented by its own λ-coordinate. HSP refers to doubly protonated histidine, HSD, and HSE to histidine singly protonated at the Nδ or Nϵ, respectively. HS0 is a common, nonphysical state of the residue. To restrict the sampling to the plane connecting the physical states, a constraint λ1 + λ2 + λ3 = 1 is applied (gray plane). A biasing potential is also applied to enhance sampling at the end states, where one of the λ-coordinates is close to one, while the other coordinates are close to zero.
Methods
Simulated Systems
Constant pH MD Simulation Setups
Reference States and Force Field Correction Potentials
tripeptide simulations (69,70) | |||
---|---|---|---|
pKa values | |||
amino acid | CHARMM36 | MARTINI | exp. |
Asp | 3.61 ± 0.03 | 3.69 ± 0.02 | 3.65 |
Glu | 4.26 ± 0.04 | 4.30 ± 0.03 | 4.25 |
His macroscopic | 6.34 ± 0.08 | 6.40 ± 0.03 | 6.42 |
His HSD | 6.56 ± 0.06 | 6.53 | |
His HSE | 6.90 ± 0.05 | 6.94 |
simulation of cardiotoxin V (71,72) | |||
---|---|---|---|
pKa values | |||
amino acid | CHARMM36 | MARTINI | exp. |
His-4 | 5.14 ± 0.16 | 4.54 ± 0.09 | 5.5 |
Glu-17 | 4.08 ± 0.08 | 4.36 ± 0.04 | 4 |
Asp-42 | 4.02 ± 0.10 | 4.30 ± 0.05 | 3.2 |
Asp-59 | 2.41 ± 0.07 | 1.45 ± 0.03 | <2 |
r = 0.96 | r = 0.80 | ||
MSE = 0.24 | MSE = 0.64 | ||
RMSE = 0.49 | RMSE = 0.80 |
simulation of HEWL (73) | |||
---|---|---|---|
pKa values | |||
amino acid | CHARMM36 | MARTINI | exp. |
Glu-7 | 2.82 ± 0.07 | 4.86 ± 0.05 | 2.6 |
His-15 | 4.84 ± 0.05 | 5.42 ± 0.05 | 5.5 |
Asp-18 | 3.35 ± 0.05 | 3.31 ± 0.03 | 2.8 |
Glu-35 | 7.64 ± 0.13 | 6.36 ± 0.05 | 6.1 |
Asp-48 | 0.99 ± 0.07 | 3.36 ± 0.05 | 1.4 |
Asp-52 | 5.69 ± 0.12 | 7.18 ± 0.10 | 3.6 |
Asp-66 | 1.70 ± 0.10 | 5.22 ± 0.05 | 1.2 |
Asp-87 | 1.73 ± 0.03 | 3.47 ± 0.05 | 2.2 |
Asp-101 | 5.43 ± 0.11 | 4.20 ± 0.06 | 4.5 |
Asp-119 | 2.77 ± 0.05 | 3.80 ± 0.05 | 3.5 |
r = 0.90 | r = 0.49 | ||
MSE = 0.96 | MSE = 4.01 | ||
RMSE = 0.98 | RMSE = 2.00 |
The reference pKa values for tripeptides are given in the last column that contain the experimental pKa values. The values for Asp and Glu are taken from ref (69), while the microscopic and macroscopic pKa values for His are taken from ref (70). Experimental pKa values for cardiotoxin V are from refs (71and72) and for HEWL from ref (73). For both proteins Pearson correlation (r), MSE and RMSE errors are provided.
Buffer Particles
Analysis of the Constant pH Trajectories
Results and Discussion
Titration of Single Amino Acids
Figure 3
Figure 3. Titrations of tripeptide amino acids (Glu, Asp, and His) in water. The top and bottom rows show titrations performed with the modified AA CHARMM36 (55) and CG Martini (41) force fields, respectively. In all simulations, neutrality was maintained by including ten buffer particles in combination with the charge constraint. Dots show the fraction of frames in which the residue is deprotonated, and the dashed lines represent the fits to the Henderson–Hasselbalch equation. For His, the blue color represents the macroscopic pKa, while yellow and red represent the microscopic pKa values for HSD (proton on Nδ) and HSE (proton on Nϵ), respectively. In the Martini 2.0 model, HSD and HSE are indistinguishable and hence only the macroscopic titration curve is shown. Errors of Sdeprot were estimated from the standard error of the mean for the different replicas. From the fits, the pKa values were estimated and listed in Table 1.
Titration of Proteins
Figure 4
Figure 4. Titration curves of the cardiotoxin V protein obtained from constant pH MD simulations with the CHARMM36 (top) and Martini 2.0 force fields (bottom). For each of the four titratable residues in this protein, the dots show the fraction of frames in which the residue is deprotonated. Errors of Sdeprot were estimated from the standard error of the mean for the different replicas. The lines show the best fits to the Henderson–Hasselbalch equation. The pKa values for each titratable residue were estimated from these fits and listed in Table 1. The right panel shows the protein structure with the four titratable residues highlighted in stick representation.
Figure 5
Figure 5. Titration curves of the HEWL protein obtained from constant pH MD simulations with the CHARMM36 (top) and Martini 2.0 force fields (bottom). For each of the ten titratable residues, the dots show the fraction of frames in which that residue is deprotonated. Errors of Sdeprot were estimated from the standard error of the mean for the different replicas. The lines show the best fit to the Henderson-Hasselbach equation. The pKa values for each titratable residue were estimated from these fits and listed in Table 1. The right panel shows the protein structure with the ten titratable residues highlighted in stick representation.
Efficiency of the Implementation
Figure 6
Figure 6. (A) Relative performance of interpolating potentials in a previous implementation of CpHMD into a fork of GROMACS 3.3 release (blue) and of charge interpolation in our new implementation (red) as a function of the number of titratable sites. The simulations were performed for the turkey ovomucoid inhibitor protein (PDB ID: 2GKR (53)), shown in the inset, where the titratable sites are highlighted in stick representation. (B) Comparison of the performance between CPU-only and CPU+GPU implementations for the ligand-gated ion channel GLIC (PDB ID: 4HFI (52)) with 185 titratable sites. In total, the GLIC system contained 292135 atoms.
Conclusions
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jctc.2c00516.
The input files and parameters used for MD simulations in the presented work (ZIP)
A Mathematica notebook with instructions and routines to fit VMM (ZIP)
Description of λ-potentials, the effect of neglecting the interpolation of Lennard-Jones interactions, titration results for Asp and Glu within the single-site representation, a comparison of pKa values for HEWL obtained with various λ-dynamics-based constant pH methods, demonstration that charge interpolation requires a single evaluation of the electrostatic potential for both single- and multisite representations (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This research was supported by the Swedish Research Council (Grant 2019-04477), Academy of Finland (Grants 311031 and 332743), and the BioExcel CoE (Grant H2020-INFRAEDI-02-2018-823830). The simulations were performed on resources provided by the CSC ─ IT Center for Science, Finland, and the Swedish National Infrastructure for Computing (SNIC 2021/1-38). We also thank Erik Lindahl, Helmut Grubmüller, Dmitry Morozov, Serena Donnini, and Plamen Dobrev for their support during the project.
Appendix: Constraint Algorithm
References
This article references 76 other publications.
- 1Hollingsworth, S. A.; Dror, R. O. Molecular Dynamics Simulation for All. Neuron 2018, 99, 1129– 1143, DOI: 10.1016/j.neuron.2018.08.011Google Scholar1Molecular Dynamics Simulation for AllHollingsworth, Scott A.; Dror, Ron O.Neuron (2018), 99 (6), 1129-1143CODEN: NERNET; ISSN:0896-6273. (Cell Press)A review. The impact of mol. dynamics (MD) simulations in mol. biol. and drug discovery has expanded dramatically in recent years. These simulations capture the behavior of proteins and other biomols. in full at. detail and at very fine temporal resoln. Major improvements in simulation speed, accuracy, and accessibility, together with the proliferation of exptl. structural data, have increased the appeal of biomol. simulation to experimentalists-a trend particularly noticeable in, although certainly not limited to, neuroscience. Simulations have proven valuable in deciphering functional mechanisms of proteins and other biomols., in uncovering the structural basis for disease, and in the design and optimization of small mols., peptides, and proteins. Here we describe, in practical terms, the types of information MD simulations can provide and the ways in which they typically motivate further exptl. work.
- 2Groenhof, G.; Modi, V.; Morozov, D. Observe while it happens: catching photoactive proteins in the act with non-adiabatic molecular dynamics simulations. Curr. Opin. Struct. Biol. 2020, 61, 106– 112, DOI: 10.1016/j.sbi.2019.12.013Google Scholar2Observe while it happens: catching photoactive proteins in the act with non-adiabatic molecular dynamics simulationsGroenhof, Gerrit; Modi, Vaibhav; Morozov, DmitryCurrent Opinion in Structural Biology (2020), 61 (), 106-112CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Organisms use photo-receptors to react to light. The first step is usually the absorption of a photon by a prosthetic group embedded inside the photo-receptor, often a conjugated chromophore. The electronic changes in the chromophore induced by photo-absorption can trigger a cascade of structural or chem. transformations that culminate into a response to light. Understanding how these proteins have evolved to mediate their activation process has remained challenging because the required time and spacial resolns. are notoriously difficult to achieve exptl. Therefore, mechanistic insights into photoreceptor activation have been predominantly obtained with computer simulations. Here we briefly outline the challenges assocd. with such computations and review the progress made in this field.
- 3Warshel, A.; Sussman, F.; King, G. Free energy of charges in solvated proteins: microscopic calculations using a reversible charging process. Biochemistry 1986, 25, 8368– 8372, DOI: 10.1021/bi00374a006Google Scholar3Free energy of charges in solvated proteins: microscopic calculations using a reversible charging processWarshel, A.; Sussman, F.; King, G.Biochemistry (1986), 25 (26), 8368-72CODEN: BICHAW; ISSN:0006-2960.Evaluation of the free energy of ionization of acidic groups in proteins may be used as a powerful and general test case for detg. the reliability of calcns. of electrostatic energies in macromols. This test case was addressed by using an adiabatic charging process that evaluates the changes in free energies assocd. with ionizing the acidic groups of aspartate-3 and glutamate-7 in bovine pancreatic trypsin inhibitor and aspartic acid in soln. The results of these free energy calcns. are very encouraging; the error range is ∼1 kcal/mol for these values, which are ∼70 kcal/mol. Thus, the stage of obtaining quant. results in modeling the energetics of solvated proteins is finally being approached.
- 4Alexov, E.; Mehler, E. L.; Baker, N.; Baptista, A. M.; Huang, Y.; Milletti, F.; Erik Nielsen, J.; Farrell, D.; Carstensen, T.; Olsson, M. H. M.; Shen, J. K.; Warwicker, J.; Williams, S.; Word, J. M. Progress in the prediction of pKa values in proteins. Proteins: Struct., Funct., Bioinf. 2011, 79, 3260– 3275, DOI: 10.1002/prot.23189Google Scholar4Progress in the prediction of pKa values in proteinsAlexov, Emil; Mehler, Ernest L.; Baker, Nathan; Baptista, Antonio M.; Huang, Yong; Milletti, Francesca; Erik Nielsen, Jens; Farrell, Damien; Carstensen, Tommy; Olsson, Mats H. M.; Shen, Jana K.; Warwicker, Jim; Williams, Sarah; Word, J. MichaelProteins: Structure, Function, and Bioinformatics (2011), 79 (12), 3260-3275CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)A review. The pKa-cooperative aims to provide a forum for exptl. and theor. researchers interested in protein pKa values and protein electrostatics in general. The first round of the pKa-cooperative, which challenged computational labs to carry out blind predictions against pKas exptl. detd. in the lab. of Bertrand Garcia-Moreno, was completed and results discussed at the Telluride meeting (July 6-10, 2009). This article serves as an introduction to the reports submitted by the blind prediction participants that will be published in a special issue of PROTEINS: Structure, Function and Bioinformatics. Here, the authors briefly outline existing approaches for pKa calcns., emphasizing methods that were used by the participants in calcg. the blind pKa values in the first round of the cooperative. The authors then point out some of the difficulties encountered by the participating groups in making their blind predictions, and finally try to provide some insights for future developments aimed at improving the accuracy of pKa calcns. Proteins 2011; © 2011 Wiley-Liss, Inc.
- 5Chen, W.; Morrow, B. H.; Shi, C.; Shen, J. K. Recent development and application of constant pH molecular dynamics. Mol. Simul. 2014, 40, 830– 838, DOI: 10.1080/08927022.2014.907492Google Scholar5Recent development and application of constant pH molecular dynamicsChen, Wei; Morrow, Brian H.; Shi, Chuanyin; Shen, Jana K.Molecular Simulation (2014), 40 (10-11), 830-838CODEN: MOSIEA; ISSN:0892-7022. (Taylor & Francis Ltd.)A review. Soln. pH is a crit. environmental factor for chem. and biol. processes. Over the last decade, significant efforts have been made in the development of const. pH mol. dynamics (pHMD) techniques for gaining detailed insights into pH-coupled dynamical phenomena. In this article, we review the advancement of this field in the past five years, placing a special emphasis on the development of the all-atom continuous pHMD technique. We discuss various applications, including the prediction of pKa shifts for proteins, nucleic acids and surfactant assemblies, elucidation of pH-dependent population shifts, protein-protein and protein-RNA binding, as well as the mechanisms of pH-dependent self-assembly and phase transitions of surfactants and peptides. We also discuss future directions for the further improvement of the pHMD techniques.
- 6Zeng, X.; Mukhopadhyay, S.; Brooks, C. L., III Residue-level resolution of alphavirus envelope protein interactions in pH-dependent fusion. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 2034– 2039, DOI: 10.1073/pnas.1414190112Google Scholar6Residue-level resolution of alphavirus envelope protein interactions in pH-dependent fusionZeng, Xiancheng; Mukhopadhyay, Suchetana; Brooks, Charles L., IIIProceedings of the National Academy of Sciences of the United States of America (2015), 112 (7), 2034-2039CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Alphavirus envelope proteins, organized as trimers of E2-E1 heterodimers on the surface of the pathogenic alphavirus, mediate the low pH-triggered fusion of viral and endosomal membranes in human cells. The lack of specific treatment for alphaviral infections motivates our exploration of potential antiviral approaches by inhibiting one or more fusion steps in the common endocytic viral entry pathway. In this work, we performed const. pH mol. dynamics based on an at. model of the alphavirus envelope with icosahedral symmetry. We have identified pH-sensitive residues that cause the largest shifts in thermodn. driving forces under neutral and acidic pH conditions for various fusion steps. A series of conserved interdomain His residues is identified to be responsible for the pH-dependent conformational changes in the fusion process, and ligand binding sites in their vicinity are anticipated to be potential drug targets aimed at inhibiting viral infections.
- 7Law, S. M.; Zhang, B. W.; Brooks, C. L., III pH-sensitive residues in the p19 RNA silencing suppressor protein from carnation Italian ringspot virus affect siRNA binding stability. Protein Sci. 2013, 22, 595– 604, DOI: 10.1002/pro.2243Google Scholar7pH-sensitive residues in the p19 RNA silencing suppressor protein from carnation Italian ringspot virus affect siRNA binding stabilityLaw, Sean M.; Zhang, Bin W.; Brooks, Charles L., IIIProtein Science (2013), 22 (5), 595-604CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Tombusviruses, such as Carnation Italian ringspot virus (CIRV), encode a protein homodimer called p19 that is capable of suppressing RNA silencing in their infected hosts by binding to and sequestering short-interfering RNA (siRNA) away from the RNA silencing pathway. P19 binding stability has been shown to be sensitive to changes in pH but the specific amino acid residues involved have remained unclear. Using const. pH mol. dynamics simulations, we have identified key pH-dependent residues that affect CIRV p19-siRNA binding stability at various pH ranges based on calcd. changes in the free energy contribution from each titratable residue. At high pH, the deprotonation of Lys60, Lys67, Lys71, and Cys134 has the largest effect on the binding stability. Similarly, deprotonation of several acidic residues (Asp9, Glu12, Asp20, Glu35, and/or Glu41) at low pH results in a decrease in binding stability. At neutral pH, residues Glu17 and His132 provide a small increase in the binding stability and we find that the optimal pH range for siRNA binding is between 7.0 and 10.0. Overall, our findings further inform recent expts. and are in excellent agreement with data on the pH-dependent binding profile.
- 8Ellis, C. R.; Shen, J. pH-dependent population shift regulates BACE1 activity and inhibition. J. Am. Chem. Soc. 2015, 137, 9543– 9546, DOI: 10.1021/jacs.5b05891Google Scholar8pH-Dependent Population Shift Regulates BACE1 Activity and InhibitionEllis, Christopher R.; Shen, JanaJournal of the American Chemical Society (2015), 137 (30), 9543-9546CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)BACE1, a major therapeutic target for treatment of Alzheimer's disease, functions within a narrow pH range. Despite tremendous effort and progress in the development of BACE1 inhibitors, details of the underlying pH-dependent regulatory mechanism remain unclear. Here the authors elucidate the pH-dependent conformational mechanism that regulates BACE1 activity using continuous const.-pH mol. dynamics (MD). The simulations reveal that BACE1 mainly occupies three conformational states and that the relative populations of the states shift according to pH. At intermediate pH, when the catalytic dyad is monoprotonated, a binding-competent state is highly populated, while at low and high pH a Tyr-inhibited state is dominant. The authors' data provide strong evidence supporting conformational selection as a major mechanism for substrate and peptide-inhibitor binding. These new insights, while consistent with expt., greatly extend the knowledge of BACE1 and have implications for further optimization of inhibitors and understanding potential side effects of targeting BACE1. Finally, the work highlights the importance of properly modeling protonation states in MD simulations.
- 9Kim, M. O.; Blachly, P. G.; McCammon, J. A. Conformational dynamics and binding free energies of inhibitors of BACE-1: from the perspective of protonation equilibria. PLoS computational biology 2015, 11, e1004341, DOI: 10.1371/journal.pcbi.1004341Google Scholar9Conformational dynamics and binding free energies of inhibitors of BACE-1: from the perspective of protonation equilibriaKim, M. Olivia; Blachly, Patrick G.; McCammon, J. AndrewPLoS Computational Biology (2015), 11 (10), e1004341/1-e1004341/28CODEN: PCBLBG; ISSN:1553-7358. (Public Library of Science)BACE-1 is the β-secretase responsible for the initial amyloidogenesis in Alzheimer's disease, catalyzing hydrolytic cleavage of substrate in a pH-sensitive manner. The catalytic mechanism of BACE-1 requires water-mediated proton transfer from aspartyl dyad to the substrate, as well as structural flexibility in the flap region. Thus, the coupling of protonation and conformational equil. is essential to a full in silico characterization of BACE-1. In this work, we perform const. pH replica exchange mol. dynamics simulations on both apo BACE-1 and five BACE-1-inhibitor complexes to examine the effect of pH on dynamics and inhibitor binding properties of BACE-1. In our simulations, we find that soln. pH controls the conformational flexibility of apo BACE-1, whereas bound inhibitors largely limit the motions of the holo enzyme at all levels of pH. The microscopic pKa values of titratable residues in BACE-1 including its aspartyl dyad are computed and compared between apo and inhibitor-bound states. Changes in protonation between the apo and holo forms suggest a thermodn. linkage between binding of inhibitors and protons localized at the dyad. Utilizing our recently developed computational protocol applying the binding polynomial formalism to the const. pH mol. dynamics (CpHMD) framework, we are able to obtain the pH-dependent binding free energy profiles for various BACE-1-inhibitor complexes. Our results highlight the importance of correctly addressing the binding-induced protonation changes in protein-ligand systems where binding accompanies a net proton transfer. This work comprises the first application of our CpHMD-based free energy computational method to protein-ligand complexes and illustrates the value of CpHMD as an all-purpose tool for obtaining pH-dependent dynamics and binding free energies of biol. systems.
- 10Sarkar, A.; Gupta, P. L.; Roitberg, A. E. pH-dependent conformational changes due to ionizable residues in a hydrophobic protein interior: The study of L25K and L125K variants of SNase. J. Phys. Chem. B 2019, 123, 5742– 5754, DOI: 10.1021/acs.jpcb.9b03816Google Scholar10pH-Dependent Conformational Changes Due to Ionizable Residues in a Hydrophobic Protein Interior: The Study of L25K and L125K Variants of SNaseSarkar, Ankita; Gupta, Pancham Lal; Roitberg, Adrian E.Journal of Physical Chemistry B (2019), 123 (27), 5742-5754CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Ionizable residues in the hydrophobic interior of certain proteins are known to play important roles in life processes like energy transduction and enzyme catalysis. These internal ionizable residues show exptl. apparent pKa values having large shifts as compared to their values in soln. In the present work, we study the pH-dependent conformational changes undergone by two variants of staphylococcal nuclease (SNase), L25K and L125K, using pH replica exchange mol. dynamics (pH-REMD) in explicit solvent. Our results show that the obsd. pKa of Lys25 and Lys125 are significantly different than their pKa in soln. We obsd. that the internal lysine residues prefer to be water-exposed when protonated at low pH, but they remain buried within the hydrophobic pocket when deprotonated at high pH. Using thermodn. laws, we est. the microscopic conformation-specific pKa of the water-exposed and buried conformations of the internal lysine residues and explain their relation to the macroscopic obsd. pKa values. We present the differences in the microscopic mechanisms that lead to similar exptl. obsd. apparent pKa of Lys25 and Lys125, and explain the need of thermodn. models of different complexities to account for our calcns. We see that L25K displays pH-dependent fluctuations throughout the entire β barrel and the α1 helix. In contrast, pH-independent fluctuations are obsd. in L125K, primarily limited to the α3 helix. The present computational study offers a detailed atomistic understanding of the determinants of the obsd. anomalous pKa of internal ionizable residues, bolstering the exptl. findings.
- 11Sarkar, A.; Roitberg, A. E. PH-Dependent Conformational Changes Lead to a Highly Shifted p K a for a Buried Glutamic Acid Mutant of SNase. J. Phys. Chem. B 2020, 124, 11072– 11080, DOI: 10.1021/acs.jpcb.0c07136Google Scholar11pH-Dependent Conformational Changes Lead to a Highly Shifted pKa for a Buried Glutamic Acid Mutant of SNaseSarkar, Ankita; Roitberg, Adrian E.Journal of Physical Chemistry B (2020), 124 (49), 11072-11080CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Ionizable residues are rarely present in the hydrophobic interior of proteins, but when they are, they play important roles in biol. processes such as energy transduction and enzyme catalysis. Internal ionizable residues have anomalous exptl. pKa values with respect to their pKa in bulk water. This work studies the atomistic cause of the highly shifted pKa of the internal Glu23 in the artificially mutated variant V23E of Staphylococcal Nuclease (SNase) using pH replica exchange mol. dynamics (pH-REMD) simulations. The pKa of Glu23 obtained from the authors' calcns. is 6.55, which is elevated with respect to the glutamate pKa of 4.40 in bulk water. The calcd. value is close to the exptl. pKa of 7.10. The authors' simulations show that the highly shifted pKa of Glu23 is the product of a pH-dependent conformational change, which was obsd. exptl. and also seen in the authors' simulations. The authors carry out an anal. of this pH-dependent conformational change in response to the protonation state change of Glu23. Using a four-state thermodn. model, the authors est. the two conformation-specific pKa values of Glu23 and describe the coupling between the conformational and ionization equil.
- 12Baptista, A. M.; Martel, P. J.; Petersen, S. B. Simulation of protein conformational freedom as a function of pH: constant-pH molecular dynamics using implicit titration. Proteins: Struct., Funct., Bioinf. 1997, 27, 523– 544, DOI: 10.1002/(SICI)1097-0134(199704)27:4<523::AID-PROT6>3.0.CO;2-BGoogle Scholar12Simulation of protein conformational freedom as a function of pH: constant-pH molecular dynamics using implicit titrationBaptista, Antonio M.; Martel, Paulo J.; Petersen, Steffen B.Proteins: Structure, Function, and Genetics (1997), 27 (4), 523-544CODEN: PSFGEY; ISSN:0887-3585. (Wiley-Liss)Soln. pH is a determinant parameter on protein function and stability, and its inclusion in mol. dynamics simulations is attractive for studies at the mol. level. Current mol. dynamics simulations can consider pH only in a very limited way, through a somewhat arbitrary choice of a set of fixed charges on the titrable sites. Conversely, continuum electrostatic methods that explicitly treat pH effects assume a single protein conformation whose choice is not clearly defined. In this paper we describe a general method that combines both titrn. and conformational freedom. The method is based on a potential of mean force for implicit titrn. and combines both usual mol. dynamics and pH-dependent calcns. based on continuum methods. A simple implementation of the method, using a mean field approxn., is presented and applied to the bovine pancreatic trypsin inhibitor. We believe that this const.-pH mol. dynamics method, by correctly sampling both charges and conformation, can become a valuable help in the understanding of the dependence of protein function and stability on pH.
- 13Baptista, A. M.; Teixeira, V. H.; Soares, C. M. Constant-pH molecular dynamics using stochastic titration. J. Chem. Phys. 2002, 117, 4184– 4200, DOI: 10.1063/1.1497164Google Scholar13Constant-pH molecular dynamics using stochastic titrationBaptista, Antonio M.; Teixeira, Vitor H.; Soares, Claudio M.Journal of Chemical Physics (2002), 117 (9), 4184-4200CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)A new method is proposed for performing const.-pH mol. dynamics (MD) simulations, i.e., MD simulations where pH is one of the external thermodn. parameters, like the temp. or the pressure. The protonation state of each titrable site in the solute is allowed to change during a mol. mechanics (MM) MD simulation, the new states being obtained from a combination of continuum electrostatics (CE) calcns. and Monte Carlo (MC) simulation of protonation equil. The coupling between the MM/MD and CE/MC algorithms is done in a way that ensures a proper Markov chain, sampling from the intended semigrand canonical distribution. This stochastic titrn. method is applied to succinic acid, aimed at illustrating the method and examg. the choice of its adjustable parameters. The complete titrn. of succinic acid, using const.-pH MD simulations at different pH values, gives a clear picture of the coupling between the trans/gauche isomerization and the protonation process, making it possible to reconcile some apparently contradictory results of previous studies. The present const.-pH MD method is shown to require a moderate increase of computational cost when compared to the usual MD method.
- 14Bürgi, R.; Kollman, P. A.; van Gunsteren, W. F. Simulating proteins at constant pH: An approach combining molecular dynamics and Monte Carlo simulation. Proteins: Struct., Funct., Bioinf. 2002, 47, 469– 480, DOI: 10.1002/prot.10046Google Scholar14Simulating proteins at constant pH: an approach combining molecular dynamics and Monte Carlo simulationBurgi, Roland; Kollman, Peter A.; Van Gunsteren, Wilfred F.Proteins: Structure, Function, and Genetics (2002), 47 (4), 469-480CODEN: PSFGEY; ISSN:0887-3585. (Wiley-Liss, Inc.)For the structure and function of proteins, the pH of the soln. is one of the detg. parameters. Current mol. dynamics (MD) simulations account for the soln. pH only in a limited way by keeping each titratable site in a chosen protonation state. We present an algorithm that generates trajectories at a Boltzmann distributed ensemble of protonation states by a combination of MD and Monte Carlo (MC) simulation. The algorithm is useful for pH-dependent structural studies and to investigate in detail the titrn. behavior of proteins. The method is tested on the acidic residues of the protein hen egg white lysozyme. It is shown that small structural changes may have a big effect on the pKA values of titratable residues.
- 15Mongan, J.; Case, D. A.; McCammon, J. A. Constant pH molecular dynamics in generalized Born implicit solvent. J. Comput. Chem. 2004, 25, 2038– 2048, DOI: 10.1002/jcc.20139Google Scholar15Constant pH molecular dynamics in generalized Born implicit solventMongan, John; Case, David A.; McCammon, J. AndrewJournal of Computational Chemistry (2004), 25 (16), 2038-2048CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)A new method is proposed for const. pH mol. dynamics (MD), employing generalized Born (GB) electrostatics. Protonation states are modeled with different charge sets, and titrating residues sample a Boltzmann distribution of protonation states as the simulation progresses, using Monte Carlo sampling based on GB-derived energies. The method is applied to four different crystal structures of hen egg-white lysozyme (HEWL). PKa predictions derived from the simulations have root-mean-square (RMS) error of 0.82 relative to exptl. values. Similarity of results between the four crystal structures shows the method to be independent of starting crystal structure; this is in contrast to most electrostatics-only models. A strong correlation between conformation and protonation state is noted and quant. analyzed, emphasizing the importance of sampling protonation states in conjunction with dynamics.
- 16Meng, Y.; Roitberg, A. E. Constant pH Replica Exchange Molecular Dynamics in Biomolecules Using a Discrete Protonation Model. J. Chem. Theory Comput. 2010, 6, 1401– 1412, DOI: 10.1021/ct900676bGoogle Scholar16Constant pH Replica Exchange Molecular Dynamics in Biomolecules Using a Discrete Protonation ModelMeng, Yilin; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2010), 6 (4), 1401-1412CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A const. pH replica exchange mol. dynamics (REMD) method is proposed and implemented to improve coupled protonation and conformational state sampling. By mixing conformational sampling at const. pH (with discrete protonation states) with a temp. ladder, this method avoids conformational trapping. Our method was tested and applied to seven different biol. systems. The const. pH REMD not only predicted pKa correctly for small, model compds. but also converged faster than const. pH mol. dynamics (MD). We further tested our const. pH REMD on a heptapeptide from the ovomucoid third domain (OMTKY3). Although const. pH REMD and MD produced very close pKa values, the const. pH REMD showed its advantage in the efficiency of conformational and protonation state samplings.
- 17Itoh, S. G.; Damjanovic, A.; Brooks, B. R. pH replica-exchange method based on discrete protonation states. Proteins: Struct., Funct., Bioinf. 2011, 79, 3420– 3436, DOI: 10.1002/prot.23176Google Scholar17pH replica-exchange method based on discrete protonation statesItoh, Satoru G.; Damjanovic, Ana; Brooks, Bernard R.Proteins: Structure, Function, and Bioinformatics (2011), 79 (12), 3420-3436CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The authors propose a new algorithm for obtaining proton titrn. curves of ionizable residues. The algorithm is a pH replica-exchange method (PHREM), which is based on the const. pH algorithm of J. Mongan et al. (2004). In the original replica-exchange method, simulations of different replicas are performed at different temps., and the temps. are exchanged between the replicas. In the authors' PHREM, simulations of different replicas are performed at different pH values, and the pHs are exchanged between the replicas. The PHREM was applied to a blocked amino acid and to two protein systems (snake cardiotoxin and turkey ovomucoid third domain), in conjunction with a generalized Born implicit solvent. The performance and accuracy of this algorithm and the original const. pH method (PHMD) were compared. For a single set of simulations at different pHs, the use of PHREM yields more accurate Hill coeffs. of titratable residues. By performing multiple sets of const. pH simulations started with different initial states, the accuracy of predicted pKa values and Hill coeffs. obtained with PHREM and PHMD methods becomes comparable. However, the PHREM algorithm exhibits better samplings of the protonation states of titratable residues and less scatter of the titrn. points and thus better precision of measured pKa values and Hill coeffs. In addn., PHREM exhibits faster convergence of individual simulations than the original const. pH algorithm. Proteins 2011; © 2011 Wiley-Liss, Inc.
- 18Swails, J. M.; York, D. M.; Roitberg, A. E. Constant pH Replica Exchange Molecular Dynamics in Explicit Solvent Using Discrete Protonation States: Implementation, Testing, and Validation. J. Chem. Theory Comput. 2014, 10, 1341– 1352, DOI: 10.1021/ct401042bGoogle Scholar18Constant pH Replica Exchange Molecular Dynamics in Explicit Solvent Using Discrete Protonation States: Implementation, Testing, and ValidationSwails, Jason M.; York, Darrin M.; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2014), 10 (3), 1341-1352CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)By utilizing Graphics Processing Units, we show that const. pH mol. dynamics simulations (CpHMD) run in Generalized Born (GB) implicit solvent for long time scales can yield poor pKa predictions as a result of sampling unrealistic conformations. To address this shortcoming, we present a method for performing const. pH mol. dynamics simulations (CpHMD) in explicit solvent using a discrete protonation state model. The method involves std. mol. dynamics (MD) being propagated in explicit solvent followed by protonation state changes being attempted in GB implicit solvent at fixed intervals. Replica exchange along the pH-dimension (pH-REMD) helps to obtain acceptable titrn. behavior with the proposed method. We analyzed the effects of various parameters and settings on the titrn. behavior of CpHMD and pH-REMD in explicit solvent, including the size of the simulation unit cell and the length of the relaxation dynamics following protonation state changes. We tested the method with the amino acid model compds., a small pentapeptide with two titratable sites, and hen egg white lysozyme (HEWL). The proposed method yields superior predicted pKa values for HEWL over hundreds of nanoseconds of simulation relative to corresponding predicted values from simulations run in implicit solvent.
- 19Swails, J. M.; Roitberg, A. E. Enhancing conformation and protonation state sampling of hen egg white lysozyme using pH replica exchange molecular dynamics. J. Chem. Theory Comput. 2012, 8, 4393– 4404, DOI: 10.1021/ct300512hGoogle Scholar19Enhancing Conformation and Protonation State Sampling of Hen Egg White Lysozyme Using pH Replica Exchange Molecular DynamicsSwails, Jason M.; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (11), 4393-4404CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We evaluate the efficiency of the pH replica exchange mol. dynamics (pH-REMD) method proposed by Itoh et al. (Proteins 2011, 79, 3420-3436) by using it to predict the pKa values of the titratable residues in hen egg white lysozyme (HEWL). PKa values predicted using pH-REMD converge significantly faster than those calcd. using const. pH mol. dynamics (CpHMD). Furthermore, increasing the frequency between exchange attempts in pH-REMD simulations improves protonation and conformational state sampling. By enabling the simulation to sample both conformational and protonation states more rapidly, pH-REMD simulations provide valuable insight into the pH-dependence of HEWL that the CpHMD simulations failed to capture. We present an efficient and highly scalable implementation of pH-REMD as an attractive enhancement to traditional CpHMD methods.
- 20Börjesson, U.; Hünenberger, P. H. Explicit-solvent molecular dynamics simulation at constant pH: Methodology and application to small amines. J. Chem. Phys. 2001, 114, 9706– 9719, DOI: 10.1063/1.1370959Google Scholar20Explicit-solvent molecular dynamics simulation at constant pH: Methodology and application to small aminesBorjesson, Ulf; Hunenberger, Philippe H.Journal of Chemical Physics (2001), 114 (22), 9706-9719CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)A method is developed for performing classical explicit-solvent mol. dynamics (MD) simulations at const. pH, where the protonation state of each ionizable (titratable) group in a simulated compd. is allowed to fluctuate in time, depending on the instantaneous system configuration and the imposed pH. In this method, each ionizable group is treated as a mixed state, i.e., the interaction-function parameters for the group are a linear combination of those of the protonated state and those of the deprotonated state. Free protons are not handled explicitly. Instead, the extent of deprotonation of each group is relaxed towards its equil. value by weak coupling to a "proton bath. " The method relies on precalibrated empirical functions, one for each type of ionizable group present in the simulated compd., which are obtained through multiple MD simulations of monofunctional model compds. In this study, the method is described in detail and its application illustrated by a series of const.-pH MD simulations of small monofunctional amines. In particular, we investigate the influence of the relaxation time used in the weak-coupling scheme, the choice of appropriate model compds. for the calibration of the required empirical functions, and corrections for finite-size effects linked with the small size of the simulation box.
- 21Lee, M. S.; Salsbury, F. R., Jr.; Brooks, C. L., III Constant-pH molecular dynamics using continuous titration coordinates. Proteins: Struct., Funct., Bioinf. 2004, 56, 738– 752, DOI: 10.1002/prot.20128Google Scholar21Constant-pH molecular dynamics using continuous titration coordinatesLee, Michael S.; Salsbury, Freddie R., Jr.; Brooks, Charles L., IIIProteins: Structure, Function, and Bioinformatics (2004), 56 (4), 738-752CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)In this work, we explore the question of whether pKa calcns. based on a microscopic description of the protein and a macroscopic description of the solvent can be implemented to examine conformationally dependent proton shifts in proteins. To this end, we introduce a new method for performing const.-pH mol. dynamics (PHMD) simulations utilizing the generalized Born implicit solvent model. This approach employs an extended Hamiltonian in which continuous titrn. coordinates propagate simultaneously with the at. motions of the system. The values adopted by these coordinates are modulated by potentials of mean force of isolated titratable model groups and the pH to control the proton occupation at particular sites in the polypeptide. Our results for four different proteins yield an abs. av. error of ∼1.6 pK units, and point to the role that thermally driven relaxation of the protein environment in the vicinity of titrating groups plays in modulating the local pKa, thereby influencing the obsd. pK1/2 values. While the accuracy of our method is not yet equiv. to methods that obtain pK1/2 values through the ad hoc scaling of electrostatics, the present approach and const. pH methods in general provide a useful framework for studying pH-dependent phenomena. Further work to improve our model to approach quant. agreement with expt. is outlined.
- 22Khandogin, J.; Brooks, C. L. Constant pH Molecular Dynamics with proton tautomerism. Biophys. J. 2005, 89, 141– 157, DOI: 10.1529/biophysj.105.061341Google Scholar22Constant pH molecular dynamics with proton tautomerismKhandogin, Jana; Brooks, Charles L., IIIBiophysical Journal (2005), 89 (1), 141-157CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The current article describes a new two-dimensional λ-dynamics method to include proton tautomerism in continuous const. pH mol. dynamics (CPHMD) simulations. The two-dimensional λ-dynamics framework is used to devise a tautomeric state titrn. model for the CPHMD simulations involving carboxyl and histidine residues. Combined with the GBSW implicit solvent model, the new method is tested on titrn. simulations of blocked histidine and aspartic acid as well as two benchmark proteins, turkey ovomucoid third domain (OMTKY3) and RNase A (RNase A). A detailed anal. of the errors inherent to the CPHMD methodol. as well as those due to the underlying solvation model is given. The av. abs. error for the computed pKa values in OMTKY3 is 1.0 pK unit. In RNase A the av. abs. errors for the carboxyl and histidine residues are 1.6 and 0.6 pK units, resp. In contrast to the previous work, the new model predicts the correct sign for all the pKa shifts, but one, in the benchmark proteins. The predictions of the tautomeric states of His12 and His48 and the conformational states of His48 and His119 are in agreement with expt. Based on the simulations of OMTKY3 and RNase A, the current work has demonstrated the capability of the CPHMD technique in revealing pH-coupled conformational dynamics of protein side chains.
- 23Donnini, S.; Tegeler, F.; Groenhof, G.; Grubmüller, H. Constant pH molecular dynamics in explicit solvent with λ-dynamics. J. Chem. Theory Comput. 2011, 7, 1962– 1978, DOI: 10.1021/ct200061rGoogle Scholar23Constant pH Molecular Dynamics in Explicit Solvent with λ-DynamicsDonnini, Serena; Tegeler, Florian; Groenhof, Gerrit; Grubmuller, HelmutJournal of Chemical Theory and Computation (2011), 7 (6), 1962-1978CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)PH is an important parameter in condensed-phase systems, because it dets. the protonation state of titratable groups and thus influences the structure, dynamics, and function of mols. in soln. In most force field simulation protocols, however, the protonation state of a system (rather than its pH) is kept fixed and cannot adapt to changes of the local environment. Here, we present a method, implemented within the MD package GROMACS, for const. pH mol. dynamics simulations in explicit solvent that is based on the λ-dynamics approach. In the latter, the dynamics of the titrn. coordinate λ, which interpolates between the protonated and deprotonated states, is driven by generalized forces between the protonated and deprotonated states. The hydration free energy, as a function of pH, is included to facilitate const. pH simulations. The protonation states of titratable groups are allowed to change dynamically during a simulation, thus reproducing av. protonation probabilities at a certain pH. The accuracy of the method is tested against titrn. curves of single amino acids and a dipeptide in explicit solvent.
- 24Wallace, J. A.; Shen, J. K. Continuous Constant pH Molecular Dynamics in Explicit Solvent with pH-Based Replica Exchange. J. Chem. Theory Comput. 2011, 7, 2617– 2629, DOI: 10.1021/ct200146jGoogle Scholar24Continuous Constant pH Molecular Dynamics in Explicit Solvent with pH-Based Replica ExchangeWallace, Jason A.; Shen, Jana K.Journal of Chemical Theory and Computation (2011), 7 (8), 2617-2629CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A computational tool that offers accurate pKa values and atomically detailed knowledge of protonation-coupled conformational dynamics is valuable for elucidating mechanisms of energy transduction processes in biol., such as enzyme catalysis and electron transfer as well as proton and drug transport. Toward this goal the authors present a new technique of embedding continuous const. pH mol. dynamics within an explicit-solvent representation. In this technique the authors make use of the efficiency of the generalized-Born (GB) implicit-solvent model for estg. the free energy of protein solvation while propagating conformational dynamics using the more accurate explicit-solvent model. Also, the authors employ a pH-based replica exchange scheme to significantly enhance both protonation and conformational state sampling. Benchmark data of five proteins including HP36, NTL9, BBL, HEWL, and SNase yield an av. abs. deviation of 0.53 and a root mean squared deviation of 0.74 from exptl. data. This level of accuracy is obtained with 1 ns simulations per replica. Detailed anal. reveals that explicit-solvent sampling provides increased accuracy relative to the previous GB-based method by preserving the native structure, providing a more realistic description of conformational flexibility of the hydrophobic cluster, and correctly modeling solvent mediated ion-pair interactions. Thus, the authors anticipate that the new technique will emerge as a practical tool to capture ionization equil. while enabling an intimate view of ionization coupled conformational dynamics that is difficult to delineate with exptl. techniques alone.
- 25Wallace, J. A.; Shen, J. K. Charge-leveling and proper treatment of long-range electrostatics in all-atom molecular dynamics at constant pH. J. Chem. Phys. 2012, 137, 184105, DOI: 10.1063/1.4766352Google Scholar25Charge-leveling and proper treatment of long-range electrostatics in all-atom molecular dynamics at constant pHWallace, Jason A.; Shen, Jana K.Journal of Chemical Physics (2012), 137 (18), 184105/1-184105/9CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Recent development of const. pH mol. dynamics (CpHMD) methods has offered promise for adding pH-stat in mol. dynamics simulations. However, until now the working pH mol. dynamics (pHMD) implementations are dependent in part or whole on implicit-solvent models. Here we show that proper treatment of long-range electrostatics and maintaining charge neutrality of the system are crit. for extending the continuous pHMD framework to the all-atom representation. The former is achieved here by adding forces to titrn. coordinates due to long-range electrostatics based on the generalized reaction field method, while the latter is made possible by a charge-leveling technique that couples proton titrn. with simultaneous ionization or neutralization of a co-ion in soln. We test the new method using the pH-replica-exchange CpHMD simulations of a series of aliph. dicarboxylic acids with varying carbon chain length. The av. abs. deviation from the exptl. pKa values is merely 0.18 units. The results show that accounting for the forces due to extended electrostatics removes the large random noise in propagating titrn. coordinates, while maintaining charge neutrality of the system improves the accuracy in the calcd. electrostatic interaction between ionizable sites. Thus, we believe that the way is paved for realizing pH-controlled all-atom mol. dynamics in the near future. (c) 2012 American Institute of Physics.
- 26Goh, G. B.; Knight, J. L.; Brooks, C. L. Constant pH Molecular Dynamics Simulations of Nucleic Acids in Explicit Solvent. J. Chem. Theory Comput. 2012, 8, 36– 46, DOI: 10.1021/ct2006314Google Scholar26Constant pH Molecular Dynamics Simulations of Nucleic Acids in Explicit SolventGoh, Garrett B.; Knight, Jennifer L.; Brooks, Charles L.Journal of Chemical Theory and Computation (2012), 8 (1), 36-46CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The nucleosides of adenine and cytosine have pKa values of 3.50 and 4.08, resp., and are assumed to be unprotonated under physiol. conditions. However, evidence from recent NMR and X-ray crystallog. studies has revealed the prevalence of protonated adenine and cytosine in RNA macromols. Such nucleotides with elevated pKa values may play a role in stabilizing RNA structure and participate in the mechanism of ribozyme catalysis. With the work presented here, we establish the framework and demonstrate the first const. pH MD simulations (CPHMD) for nucleic acids in explicit solvent, in which the protonation state is coupled to the dynamical evolution of the RNA system via λ-dynamics. We adopt the new functional form λNexp for λ that was recently developed for multisite λ-dynamics (MSλD) and demonstrate good sampling characteristics in which rapid and frequent transitions between the protonated and unprotonated states at pH = pKa are achieved. Our calcd. pKa values of simple nucleotides are in a good agreement with exptl. measured values, with a mean abs. error of 0.24 pKa units. This work demonstrates that CPHMD can be used as a powerful tool to investigate pH-dependent biol. properties of RNA macromols.
- 27Chen, W.; Wallace, J. A.; Yue, Z.; Shen, J. K. Introducing Titratable Water to All-Atom Molecular Dynamics at Constant pH. Biophys. J. 2013, 105, L15– L17, DOI: 10.1016/j.bpj.2013.06.036Google Scholar27Introducing Titratable Water to All-Atom Molecular Dynamics at Constant pHChen, Wei; Wallace, Jason A.; Yue, Zhi; Shen, Jana K.Biophysical Journal (2013), 105 (4), L15-L17CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Recent development of titratable coions has paved the way for realizing all-atom mol. dynamics at const. pH. To further improve phys. realism, here we describe a technique in which proton titrn. of the solute is directly coupled to the interconversion between water and hydroxide or hydronium. We test the new method in replica-exchange continuous const. pH mol. dynamics simulations of three proteins, HP36, BBL, and HEWL. The calcd. pKa values based on 10-ns sampling per replica have the av. abs. and root-mean-square errors of 0.7 and 0.9 pH units, resp. Introducing titratable water in mol. dynamics offers a means to model proton exchange between solute and solvent, thus opening a door to gaining new insights into the intricate details of biol. phenomena involving proton translocation.
- 28Lee, J.; Miller, B. T.; Damjanovic, A.; Brooks, B. R. Constant pH Molecular Dynamics in Explicit Solvent with Enveloping Distribution Sampling and Hamiltonian Exchange. J. Chem. Theory Comput. 2014, 10, 2738– 2750, DOI: 10.1021/ct500175mGoogle Scholar28Constant pH Molecular Dynamics in Explicit Solvent with Enveloping Distribution Sampling and Hamiltonian ExchangeLee, Juyong; Miller, Benjamin T.; Damjanovic, Ana; Brooks, Bernard R.Journal of Chemical Theory and Computation (2014), 10 (7), 2738-2750CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We present a new computational approach for const. pH simulations in explicit solvent based on the combination of the enveloping distribution sampling (EDS) and Hamiltonian replica exchange (HREX) methods. Unlike const. pH methods based on variable and continuous charge models, our method is based on discrete protonation states. EDS generates a hybrid Hamiltonian of different protonation states. A smoothness parameter s is used to control the heights of energy barriers of the hybrid-state energy landscape. A small s value facilitates state transitions by lowering energy barriers. Replica exchange between EDS potentials with different s values allows us to readily obtain a thermodynamically accurate ensemble of multiple protonation states with frequent state transitions. The anal. is performed with an ensemble obtained from an EDS Hamiltonian without smoothing, s = ∞, which strictly follows the min. energy surface of the end states. The accuracy and efficiency of this method is tested on aspartic acid, lysine, and glutamic acid, which have two protonation states, a histidine with three states, a four-residue peptide with four states, and snake cardiotoxin with eight states. The pKa values estd. with the EDS-HREX method agree well with the exptl. pKa values. The mean abs. errors of small benchmark systems range from 0.03 to 0.17 pKa units, and those of three titratable groups of snake cardiotoxin range from 0.2 to 1.6 pKa units. This study demonstrates that EDS-HREX is a potent theor. framework, which gives the correct description of multiple protonation states and good calcd. pKa values.
- 29Dobrev, P.; Donnini, S.; Groenhof, G.; Grubmüller, H. Accurate Three States Model for Amino Acids with Two Chemically Coupled Titrating Sites in Explicit Solvent Atomistic Constant pH Simulations and p K a Calculations. J. Chem. Theory Comput. 2017, 13, 147– 160, DOI: 10.1021/acs.jctc.6b00807Google Scholar29Accurate Three States Model for Amino Acids with Two Chemically Coupled Titrating Sites in Explicit Solvent Atomistic Constant pH Simulations and pKa CalculationsDobrev, Plamen; Donnini, Serena; Groenhof, Gerrit; Grubmueller, HelmutJournal of Chemical Theory and Computation (2017), 13 (1), 147-160CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Correct protonation of titratable groups in biomols. is crucial for their accurate description by mol. dynamics simulations. In the context of const. pH simulations, an addnl. protonation degree of freedom is introduced for each titratable site, allowing the protonation state to change dynamically with changing structure or electrostatics. Here, we implement a second reaction coordinate which switches between two tautomeric states of an amino acid with chem. coupled titratable sites, such as aspartate (Asp), glutamate (Glu), and histidine (His). To this aim, we test a scheme involving three protonation states. To facilitate charge neutrality as required for periodic boundary conditions and Particle Mesh Ewald (PME) electrostatics, titrn. of each resp. amino acid is coupled to a "water" mol. that is charged in opposite direction. Addnl., a force field modification for Amber99sb is introduced and tested for the description of carboxyl group protonation. Our three states model is tested by titrn. simulations of Asp, Glu, and His, yielding a good agreement, reproducing the correct geometry of the groups in their different protonation forms. We further show that the ion concn. change due to the neutralizing "water" mols. does not significantly affect the protonation free energies of the titratable groups, suggesting that the three states model provides a good description of biomol. dynamics at const. pH.
- 30Huang, Y.; Chen, W.; Wallace, J. A.; Shen, J. All-atom continuous constant pH molecular dynamics with particle mesh Ewald and titratable water. J. Chem. Theory Comput. 2016, 12, 5411– 5421, DOI: 10.1021/acs.jctc.6b00552Google Scholar30All-Atom Continuous Constant pH Molecular Dynamics With Particle Mesh Ewald and Titratable WaterHuang, Yandong; Chen, Wei; Wallace, Jason A.; Shen, JanaJournal of Chemical Theory and Computation (2016), 12 (11), 5411-5421CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Development of a pH stat to properly control soln. pH in biomol. simulations has been a long-standing goal in the community. Towards this goal recent years have witnessed the emergence of the so-called const. pH mol. dynamics methods. However, the accuracy and generality of these methods have been hampered by the use of implicit-solvent models or electrostatic truncation schemes. Here the authors report the implementation of the particle mesh Ewald (PME) scheme into the all-atom continuous const. pH mol. dynamics (CpHMD) method, enabling CpHMD to be performed with a std. MD engine at a fractional added computational cost. The authors demonstrate the performance using pH replica-exchange CpHMD simulations with titratable water for a stringent test set of proteins, HP38, BBL, HEWL and SNase. With the sampling time of 10 ns per replica, most pKa's are converged, yielding the av. abs. and root-mean-square deviations of 0.61 and 0.77, resp., from expt. Linear regression of the calcd. vs. exptl. pKa shifts gives a correlation coeff. of 0.79, a slope of 1 and an intercept near 0. Anal. reveals inadequate sampling of structure relaxation accompanying a protonation-state switch as a major source of the remaining errors, which are reduced as simulation prolongs. These data suggest PME-based CpHMD can be used as a general tool for pH-controlled simulations of macromol. systems in various environments, enabling at. insights into pH-dependent phenomena involving not only sol. proteins but also transmembrane proteins, nucleic acids, surfactants and polysaccharides.
- 31Huang, Y.; Harris, R. C.; Shen, J. Generalized Born based continuous constant pH molecular dynamics in Amber: Implementation, benchmarking and analysis. J. Chem. Inf. Model. 2018, 58, 1372– 1383, DOI: 10.1021/acs.jcim.8b00227Google Scholar31Generalized Born Based Continuous Constant pH Molecular Dynamics in Amber: Implementation, Benchmarking and AnalysisHuang, Yandong; Harris, Robert C.; Shen, JanaJournal of Chemical Information and Modeling (2018), 58 (7), 1372-1383CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Soln. pH plays an important role in structure and dynamics of biomol. systems; however, pH effects cannot be accurately accounted for in conventional mol. dynamics simulations based on fixed protonation states. Continuous const. pH mol. dynamics (CpHMD) based on the λ-dynamics framework calcs. protonation states on the fly during dynamical simulation at a specified pH condition. Here the authors report the CPU-based implementation of the CpHMD method based on the GBNeck2 generalized Born (GB) implicit-solvent model in the pmemd engine of the Amber mol. dynamics package. The performance of the method was tested using pH replica-exchange titrn. simulations of Asp, Glu and His side chains in 4 miniproteins and 7 enzymes with exptl. known pKa's, some of which are significantly shifted from the model values. The added computational cost due to CpHMD titrn. ranges from 11 to 33% for the data set and scales roughly linearly as the ratio between the titrable sites and no. of solute atoms. Comparison of the exptl. and calcd. pKa's using 2 ns per replica sampling yielded a mean unsigned error of 0.70, a root-mean-squared error of 0.91, and a linear correlation coeff. of 0.79. Though this level of accuracy is similar to the GBSW-based CpHMD in CHARMM, in contrast to the latter, the current implementation was able to reproduce the exptl. orders of the pKa's of the coupled carboxylic dyads. The authors quantified the sampling errors, which revealed that prolonged simulation is needed to converge pKa's of several titratable groups involved in salt-bridge-like interactions or deeply buried in the protein interior. The authors' benchmark data demonstrate that GBNeck2-CpHMD is an attractive tool for protein pKa predictions.
- 32Harris, R. C.; Shen, J. GPU-accelerated implementation of continuous constant pH molecular dynamics in amber: pKa predictions with single-pH simulations. J. Chem. Inf. Model. 2019, 59, 4821– 4832, DOI: 10.1021/acs.jcim.9b00754Google Scholar32GPU-Accelerated Implementation of Continuous Constant pH Molecular Dynamics in Amber: pKa Predictions with Single-pH SimulationsHarris, Robert C.; Shen, JanaJournal of Chemical Information and Modeling (2019), 59 (11), 4821-4832CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The authors present a GPU implementation of the continuous const. pH mol. dynamics (CpHMD) based on the most recent generalized Born implicit-solvent model in the pmemd engine of the Amber mol. dynamics package. To test the accuracy of the tool for rapid pKa predictions, a series of 2-ns single-pH simulations were performed for over 120 titratable residues in 10 benchmark proteins that were previously used to test the various continuous CpHMD methods. The calcd. pKa's showed a root-mean-square deviation of 0.80 and correlation coeff. of 0.83 with respect to expt. 90% of the pKa's were converged with estd. errors <0.1 pH units. Surprisingly, this level of accuracy is similar to the authors' previous replica-exchange simulations with 2 ns per replica and an exchange attempt frequency of 2 ps-1 (Huang, Harris and Shen, J Chem Info Model, 2018). For the linked titrn. sites in two enzymes, although residue-specific protonation state sampling in the single-pH simulations was not converged within 2 ns, the protonation fraction of the linked residues appeared to be largely converged, and the exptl. macroscopic {\pka} values were reproduced to within 1 pH unit. Comparison with replica-exchange simulations with different exchange attempt frequencies showed that the splitting between the two macroscopic pKa's is underestimated with frequent exchange attempts such as 2 ps$̂{-1}$, while single-pH simulations overestimate the splitting. The same trend is seen for the single-pH vs. replica-exchange simulations of a hydrogen-bonded aspartyl dyad in a much larger protein. A 2-ns single-pH simulation of a 400-residue protein takes about one hour on a single NVIDIA GeForce RTX 2080 graphics card, which is over 1000 times faster than a CpHMD run on a single CPU core of a high-performance computing cluster node. Thus, the authors envision that GPU-accelerated continuous CpHMD may be used in routine pKa predictions for a variety of applications, from assisting MD simulations with protonation state assignment to offering pH-dependent corrections of binding free energies and identifying reactive hot spots for covalent drug design.
- 33Harris, R. C.; Liu, R.; Shen, J. Predicting reactive cysteines with implicit-solvent-based continuous constant pH molecular dynamics in amber. J. Chem. Theory Comput. 2020, 16, 3689– 3698, DOI: 10.1021/acs.jctc.0c00258Google Scholar33Predicting Reactive Cysteines with Implicit-Solvent-Based Continuous Constant pH Molecular Dynamics in AmberHarris, Robert C.; Liu, Ruibin; Shen, JanaJournal of Chemical Theory and Computation (2020), 16 (6), 3689-3698CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Cysteines existing in the deprotonated thiolate form or having a tendency to become deprotonated are important players in enzymic and cellular redox functions and frequently exploited in covalent drug design; however, most computational studies assume cysteines as protonated. Thus, developing an efficient tool that can make accurate and reliable predictions of cysteine protonation states is timely needed. The authors recently implemented a generalized Born (GB) based continuous const. pH mol. dynamics (CpHMD) method in Amber for protein pKa calcns. on CPUs and GPUs. Here the authors benchmark the performance of GB-CpHMD for predictions of cysteine pKa's and reactivities using a data set of 24 proteins with both down- and upshifted cysteine pKa's. The authors found that 10 ns single-pH or 4 ns replica-exchange CpHMD titrns. gave root-mean-square errors of 1.2-1.3 and correlation coeffs. of 0.8-0.9 with respect to expt. The accuracy of predicting thiolates or reactive cysteines at physiol. pH with single-pH titrns. is 86 or 81% with a precision of 100 or 90%, resp. This performance well surpasses the traditional structure-based methods, particularly a widely used empirical pKa tool that gives an accuracy less than 50%. The authors discuss simulation convergence, dependence on starting structures, common determinants of the pKa downshifts and upshifts, and the origin of the discrepancies from the structure-based calcns. The work suggests that CpHMD titrns. can be performed on a desktop computer equipped with a single GPU card to predict cysteine protonation states for a variety of applications, from understanding biol. functions to covalent drug design.
- 34Grünewald, F.; Souza, P. C.; Abdizadeh, H.; Barnoud, J.; de Vries, A. H.; Marrink, S. J. Titratable Martini model for constant pH simulations. J. Chem. Phys. 2020, 153, 024118, DOI: 10.1063/5.0014258Google Scholar34Titratable Martini model for constant pH simulationsGruenewald, Fabian; Souza, Paulo C. T.; Abdizadeh, Haleh; Barnoud, Jonathan; de Vries, Alex H.; Marrink, Siewert J.Journal of Chemical Physics (2020), 153 (2), 024118CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The authors deliver a proof of concept for a fast method that introduces pH effects into classical coarse-grained (CG) mol. dynamics simulations. The authors' approach is based upon the latest version of the popular Martini CG model to which explicit proton mimicking particles were added. The authors verify the authors' approach against exptl. data involving several different mols. and different environmental conditions. In particular, the authors compute titrn. curves, pH dependent free energies of transfer, and lipid bilayer membrane affinities as a function of pH. Using oleic acid as an example compd., the authors further illustrate that the authors' method can be used to study passive translocation in lipid bilayers via protonation. Finally, the authors' model reproduces qual. the expansion of the macromol. dendrimer poly(propylene imine) as well as the assocd. pKa shift of its different generations. This example demonstrates that the authors' model is able to pick up collective interactions between titratable sites in large mols. comprising many titratable functional groups. (c) 2020 American Institute of Physics.
- 35Mongan, J.; Case, D. A. Biomolecular simulations at constant pH. Curr. Opin. Struct. Biol. 2005, 15, 157– 163, DOI: 10.1016/j.sbi.2005.02.002Google Scholar35Biomolecular simulations at constant pHMongan, John; Case, David A.Current Opinion in Structural Biology (2005), 15 (2), 157-163CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Like temp. and pressure, the soln. pH is an important thermodn. variable that is commonly varied in expts. and is used by cells to influence biochem. function. It is now becoming feasible to carry out practical mol. dynamics simulations that mimic the thermodn. of such expts., by allowing proton transfer between the system of interest and a hypothetical bath of protons at a given pH. These are demanding calcns., because the energetics of charge changes upon protonation or deprotonation must be accurately modeled, and because such simulations must sample both mol. configurations and the large no. of protonation states that are possible for a mol. with many titrating sites.
- 36Damjanovic, A.; Miller, B. T.; Okur, A.; Brooks, B. R. Reservoir pH replica exchange. J. Chem. Phys. 2018, 149, 072321, DOI: 10.1063/1.5027413Google Scholar36Reservoir pH replica exchangeDamjanovic, Ana; Miller, Benjamin T.; Okur, Asim; Brooks, Bernard R.Journal of Chemical Physics (2018), 149 (7), 072321/1-072321/12CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)We present the reservoir pH replica exchange (R-pH-REM) method for const. pH simulations. The R-pH-REM method consists of a two-step procedure; the first step involves generation of one or more reservoirs of conformations. Each reservoir is obtained from a std. or enhanced mol. dynamics simulation with a constrained (fixed) protonation state. In the second step, fixed charge constraints are relaxed, as the structures from one or more reservoirs are periodically injected into a const. pH or a pH-replica exchange (pH-REM) simulation. The benefit of this two-step process is that the computationally intensive part of conformational search can be decoupled from const. pH simulations, and various techniques for enhanced conformational sampling can be applied without the need to integrate such techniques into the pH-REM framework. Simulations on blocked Lys, KK, and KAAE peptides were used to demonstrate an agreement between pH-REM and R-pH-REM simulations. While the reservoir simulations are not needed for these small test systems, the real need arises in cases when ionizable mols. can sample two or more conformations sepd. by a large energy barrier, such that adequate sampling is not achieved on a time scale of std. const. pH simulations. Such problems might be encountered in protein systems that exploit conformational transitions for function. A hypothetical case is studied, a small mol. with a large torsional barrier; while results of pH-REM simulations depend on the starting structure, R-pH-REM calcns. on this model system are in excellent agreement with a theor. model. (c) 2018 American Institute of Physics.
- 37Chen, Y.; Roux, B. Constant-pH Hybrid Nonequilibrium Molecular Dynamics - Monte Carlo Simulation Method. J. Chem. Theory Comput. 2015, 11, 3919– 3931, DOI: 10.1021/acs.jctc.5b00261Google Scholar37Constant-pH Hybrid Nonequilibrium Molecular Dynamics-Monte Carlo Simulation MethodChen, Yunjie; Roux, BenoitJournal of Chemical Theory and Computation (2015), 11 (8), 3919-3931CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A computational method is developed to carry out explicit solvent simulations of complex mol. systems under conditions of const. pH. In const.-pH simulations, preidentified ionizable sites are allowed to spontaneously protonate and deprotonate as a function of time in response to the environment and the imposed pH. The method consists of carrying out short nonequil. mol. dynamics (neMD) switching trajectories to generate phys. plausible configurations with changed protonation states that are subsequently accepted or rejected according to a Metropolis Monte Carlo (MC) criterion. To ensure microscopic detailed balance arising from such nonequil. switches, the at. momenta are altered according to the sym. two-ends momentum reversal prescription. To achieve higher efficiency, the original neMD-MC scheme is sepd. into two steps, reducing the need for generating a large no. of unproductive and costly nonequil. trajectories. In the first step, the protonation state of a site is randomly attributed via a Metropolis MC process on the basis of an intrinsic pKa; an attempted nonequil. switch is generated only if this change in protonation state is accepted. This hybrid two-step inherent pKa neMD-MC simulation method is tested with single amino acids in soln. (Asp, Glu, and His) and then applied to turkey ovomucoid third domain and hen egg-white lysozyme. Because of the simple linear increase in the computational cost relative to the no. of titratable sites, the present method is naturally able to treat extremely large systems.
- 38Kong, X.; Brooks, C. L., III λ-dynamics: A new approach to free energy calculations. J. Chem. Phys. 1996, 105, 2414– 2423, DOI: 10.1063/1.472109Google ScholarThere is no corresponding record for this reference.
- 39Simonson, T.; Carlsson, J.; Case, D. A. Proton binding to proteins: p K a calculations with explicit and implicit solvent models. J. Am. Chem. Soc. 2004, 126, 4167– 4180, DOI: 10.1021/ja039788mGoogle Scholar39Proton Binding to Proteins: pKa Calculations with Explicit and Implicit Solvent ModelsSimonson, Thomas; Carlsson, Jens; Case, David A.Journal of the American Chemical Society (2004), 126 (13), 4167-4180CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Ionizable residues play important roles in protein structure and activity, and proton binding is a valuable reporter of electrostatic interactions in these systems. The authors use mol. dynamics free energy simulations (MDFE) to compute proton pKa shifts, relative to a model compd. in soln., for three aspartate side chains in two proteins. Simulations with explicit solvent and with an implicit, dielec. continuum solvent are reported. The implicit solvent simulations use the generalized Born (GB) model, which provides an approx., anal. soln. to Poisson's equation. With explicit solvent, the direction of the pKa shifts is correct in all three cases with one force field (AMBER) and in two out of three cases with another (CHARMM). For two aspartates, the dielec. response to ionization is found to be linear, even though the sep. protein and solvent responses can be nonlinear. For thioredoxin Asp-26, nonlinearity arises from the presence of two substates that correspond to the two possible orientations of the protonated carboxylate. For this side chain, which is partly buried and has a large pKa upshift, very long simulations are needed to correctly sample several slow degrees of freedom that reorganize in response to the ionization. Thus, nearby Lys-57 rotates to form a salt bridge and becomes buried, while three waters intercalate along the opposite edge of Asp-26. Such strong and anisotropic reorganization is very difficult to predict with Poisson-Boltzmann methods that only consider electrostatic interactions and employ a single protein structure. In contrast, MDFE with a GB dielec. continuum solvent, used for the first time for pKa calcns., can describe protein reorganization accurately and gives encouraging agreement with expt. and with the explicit solvent simulations.
- 40Chen, W.; Shen, J. K. Effects of System Net Charge and Electrostic Truncation on All-Atom Constant pH Molecular Dynamics. J. Comput. Chem. 2014, 35, 1986– 1996, DOI: 10.1002/jcc.23713Google Scholar40Effects of system net charge and electrostatic truncation on all-atom constant pH molecular dynamicsChen, Wei; Shen, Jana K.Journal of Computational Chemistry (2014), 35 (27), 1986-1996CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Const. pH mol. dynamics offers a means to rigorously study the effects of soln. pH on dynamical processes. Here, we address two crit. questions arising from the most recent developments of the all-atom continuous const. pH mol. dynamics (CpHMD) method: (1) What is the effect of spatial electrostatic truncation on the sampling of protonation states. (2) Is the enforcement of elec. neutrality necessary for const. pH simulations. We first examd. how the generalized reaction field and force-shifting schemes modify the electrostatic forces on the titrn. coordinates. Free energy simulations of model compds. were then carried out to delineate the errors in the deprotonation free energy and salt-bridge stability due to electrostatic truncation and system net charge. Finally, CpHMD titrn. of a mini-protein HP36 was used to understand the manifestation of the two types of errors in the calcd. pKa values. The major finding is that enforcing charge neutrality under all pH conditions and at all time via co-titrating ions significantly improves the accuracy of protonation-state sampling. We suggest that such finding is also relevant for simulations with particle mesh Ewald, considering the known artifacts due to charge-compensating background plasma.
- 41Marrink, S. J.; Risselada, H. J.; Yefimov, S.; Tieleman, D. P.; De Vries, A. H. The MARTINI force field: coarse grained model for biomolecular simulations. J. Phys. Chem. B 2007, 111, 7812– 7824, DOI: 10.1021/jp071097fGoogle Scholar41The MARTINI Force Field: Coarse Grained Model for Biomolecular SimulationsMarrink, Siewert J.; Risselada, H. Jelger; Yefimov, Serge; Tieleman, D. Peter; De Vries, Alex H.Journal of Physical Chemistry B (2007), 111 (27), 7812-7824CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)We present an improved and extended version of our coarse grained lipid model. The new version, coined the MARTINI force field, is parametrized in a systematic way, based on the reprodn. of partitioning free energies between polar and apolar phases of a large no. of chem. compds. To reproduce the free energies of these chem. building blocks, the no. of possible interaction levels of the coarse-grained sites has increased compared to those of the previous model. Application of the new model to lipid bilayers shows an improved behavior in terms of the stress profile across the bilayer and the tendency to form pores. An extension of the force field now also allows the simulation of planar (ring) compds., including sterols. Application to a bilayer/cholesterol system at various concns. shows the typical cholesterol condensation effect similar to that obsd. in all atom representations.
- 42Donnini, S.; Ullmann, R. T.; Groenhof, G.; Grubmüller, H. Charge-neutral constant pH molecular dynamics simulations using a parsimonious proton buffer. J. Chem. Theory Comput. 2016, 12, 1040– 1051, DOI: 10.1021/acs.jctc.5b01160Google Scholar42Charge-Neutral Constant pH Molecular Dynamics Simulations Using a Parsimonious Proton BufferDonnini, Serena; Ullmann, R. Thomas; Groenhof, Gerrit; Grubmuller, HelmutJournal of Chemical Theory and Computation (2016), 12 (3), 1040-1051CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)In const. pH mol. dynamics simulations, the protonation states of titratable sites can respond to changes of the pH and of their electrostatic environment. Consequently, the no. of protons bound to the biomol., and therefore the overall charge of the system, fluctuates during the simulation. To avoid artifacts assocd. with a non-neutral simulation system, we introduce an approach to maintain neutrality of the simulation box in const. pH mol. dynamics simulations, while maintaining an accurate description of all protonation fluctuations. Specifically, we introduce a proton buffer that, like a buffer in expt., can exchange protons with the biomol. enabling its charge to fluctuate. To keep the total charge of the system const., the uptake and release of protons by the buffer are coupled to the titrn. of the biomol. with a constraint. We find that, because the fluctuation of the total charge (no. of protons) of a typical biomol. is much smaller than the no. of titratable sites of the biomol., the no. of buffer sites required to maintain overall charge neutrality without compromising the charge fluctuations of the biomol., is typically much smaller than the no. of titratable sites, implying markedly enhanced simulation and sampling efficiency.
- 43Dobrev, P.; Vemulapalli, S. P. B.; Nath, N.; Griesinger, C.; Grubmüller, H. Probing the accuracy of explicit solvent constant pH molecular dynamics simulations for peptides. J. Chem. Theory Comput. 2020, 16, 2561– 2569, DOI: 10.1021/acs.jctc.9b01232Google Scholar43Probing the Accuracy of Explicit Solvent Constant pH Molecular Dynamics Simulations for PeptidesDobrev, Plamen; Vemulapalli, Sahithya Phani Babu; Nath, Nilamoni; Griesinger, Christian; Grubmueller, HelmutJournal of Chemical Theory and Computation (2020), 16 (4), 2561-2569CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Protonation states of titratable amino acids play a key role in many biomol. processes. Knowledge of protonatable residue charges at a given pH is essential for a correct understanding of protein catalysis, inter- and intramol. interactions, substrate binding, and protein dynamics for instance. However, acquiring exptl. values for individual amino acid protonation states of complex systems is not straightforward; therefore, several in silico approaches have been developed to tackle this issue. In this work, the authors assess the accuracy of the previously developed const. pH MD approach by comparing the theor. obtained pKa values for titratable residues with exptl. values from an equiv. NMR study. The authors selected a set of four pentapeptides, of adequately small size to ensure comprehensive sampling, but concurrently, due to their charge compn., posing a challenge for protonation state calcn. The comparison of the pKa values shows good agreement of the exptl. and the theor. approach with a largest difference of 0.25 pKa units. Further, the corresponding titrn. curves are in fair agreement, although the shift of the Hill coeff. from a value of 1 was not always reproduced in simulations. The phase space overlap in Cartesian space between trajectories generated in const. pH and std. MD simulations is fair and suggests that the const. pH MD approach reasonably well preserves the dynamics of the system, allowing dynamic protonation MD simulations without introducing structural artifacts.
- 44Darden, T.; York, D.; Pedersen, L. Particle mesh Ewald: An Nlog(N) method for Ewald sums in large systems. J. Chem. Phys. 1993, 98, 10089– 10092, DOI: 10.1063/1.464397Google Scholar44Particle mesh Ewald: an N·log(N) method for Ewald sums in large systemsDarden, Tom; York, Darrin; Pedersen, LeeJournal of Chemical Physics (1993), 98 (12), 10089-92CODEN: JCPSA6; ISSN:0021-9606.An N·log(N) method for evaluating electrostatic energies and forces of large periodic systems is presented. The method is based on interpolation of the reciprocal space Ewald sums and evaluation of the resulting convolution using fast Fourier transforms. Timings and accuracies are presented for three large cryst. ionic systems.
- 45Essmann, U.; Perera, L.; Berkowitz, M. L.; Darden, T.; Lee, H.; Pedersen, L. G. A smooth particle mesh Ewald method. J. Chem. Phys. 1995, 103, 8577– 8593, DOI: 10.1063/1.470117Google Scholar45A smooth particle mesh Ewald methodEssmann, Ulrich; Perera, Lalith; Berkowitz, Max L.; Darden, Tom; Lee, Hsing; Pedersen, Lee G.Journal of Chemical Physics (1995), 103 (19), 8577-93CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The previously developed particle mesh Ewald method is reformulated in terms of efficient B-spline interpolation of the structure factors. This reformulation allows a natural extension of the method to potentials of the form 1/rp with p ≥ 1. Furthermore, efficient calcn. of the virial tensor follows. Use of B-splines in the place of Lagrange interpolation leads to analytic gradients as well as a significant improvement in the accuracy. The authors demonstrate that arbitrary accuracy can be achieved, independent of system size N, at a cost that scales as N log(N). For biomol. systems with many thousands of atoms and this method permits the use of Ewald summation at a computational cost comparable to that of a simple truncation method of 10 Å or less.
- 46Knight, J. L.; Brooks, C. L., III Multisite λ dynamics for simulated structure-activity relationship studies. J. Chem. Theory Comput. 2011, 7, 2728– 2739, DOI: 10.1021/ct200444fGoogle Scholar46Multisite λ Dynamics for Simulated Structure-Activity Relationship StudiesKnight, Jennifer L.; Brooks, Charles L., IIIJournal of Chemical Theory and Computation (2011), 7 (9), 2728-2739CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Multisite λ dynamics (MSλD) is a new free energy simulation method that is based on λ dynamics. It has been developed to enable multiple substituents at multiple sites on a common ligand core to be modeled simultaneously and their free energies assessed. The efficacy of MSλD for estg. relative hydration free energies and relative binding affinties is demonstrated using three test systems. Model compds. representing multiple identical benzene, dihydroxybenzene, and dimethoxybenzene mols. show that total combined MSλD trajectory lengths of ∼1.5 ns are sufficient to reliably achieve relative hydration free energy ests. within 0.2 kcal/mol and are less sensitive to the no. of trajectories that are used to generate these ests. for hybrid ligands that contain up to 10 substituents modeled at a single site or five substituents modeled at each of two sites. Relative hydration free energies among six benzene derivs. calcd. from MSλD simulations are in very good agreement with those from alchem. free energy simulations (with av. unsigned differences of 0.23 kcal/mol and R2 = 0.991) and the expt. (with av. unsigned errors of 1.8 kcal/mol and R2 = 0.959). Ests. of the relative binding affinities among 14 inhibitors of HIV-1 reverse transcriptase obtained from MSλD simulations are in reasonable agreement with those from traditional free energy simulations and the expt. (av. unsigned errors of 0.9 kcal/mol and R2 = 0.402). For the same level of accuracy and precision, MSλD simulations are achieved ∼20-50 times faster than traditional free energy simulations and thus with reliable force field parameters can be used effectively to screen tens to hundreds of compds. in structure-based drug design applications.
- 47Goh, G. B.; Hulbert, B. S.; Zhou, H.; Brooks, C. L., III Constant pH molecular dynamics of proteins in explicit solvent with proton tautomerism. Proteins: Struct., Funct., Bioinf. 2014, 82, 1319– 1331, DOI: 10.1002/prot.24499Google Scholar47Constant pH molecular dynamics of proteins in explicit solvent with proton tautomerismGoh, Garrett B.; Hulbert, Benjamin S.; Zhou, Huiqing; Brooks, Charles L., IIIProteins: Structure, Function, and Bioinformatics (2014), 82 (7), 1319-1331CODEN: PSFBAF ISSN:. (Wiley-Blackwell)PH is a ubiquitous regulator of biol. activity, including protein-folding, protein-protein interactions, and enzymic activity. Existing const. pH mol. dynamics (CPHMD) models that were developed to address questions related to the pH-dependent properties of proteins are largely based on implicit solvent models. However, implicit solvent models are known to underestimate the desolvation energy of buried charged residues, increasing the error assocd. with predictions that involve internal ionizable residue that are important in processes like hydrogen transport and electron transfer. Furthermore, discrete water and ions cannot be modeled in implicit solvent, which are important in systems like membrane proteins and ion channels. We report on an explicit solvent const. pH mol. dynamics framework based on multi-site λ-dynamics (CPHMDMSλD). In the CPHMDMSλD framework, we performed seamless alchem. transitions between protonation and tautomeric states using multi-site λ-dynamics, and designed novel biasing potentials to ensure that the phys. end-states are predominantly sampled. We show that explicit solvent CPHMDMSλD simulations model realistic pH-dependent properties of proteins such as the Hen-Egg White Lysozyme (HEWL), binding domain of 2-oxoglutarate dehydrogenase (BBL) and N-terminal domain of ribosomal protein L9 (NTL9), and the pKa predictions are in excellent agreement with exptl. values, with a RMSE ranging from 0.72 to 0.84 pKa units. With the recent development of the explicit solvent CPHMDMSλD framework for nucleic acids, accurate modeling of pH-dependent properties of both major class of biomols.-proteins and nucleic acids is now possible.
- 48Knight, J. L.; Brooks, C. L., III Applying efficient implicit nongeometric constraints in alchemical free energy simulations. Journal of computational chemistry 2011, 32, 3423– 3432, DOI: 10.1002/jcc.21921Google Scholar48Applying efficient implicit nongeometric constraints in alchemical free energy simulationsKnight, Jennifer L.; Brooks, Charles L.Journal of Computational Chemistry (2011), 32 (16), 3423-3432CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Several strategies have been developed for satisfying bond lengths, angle, and other geometric constraints in mol. dynamics simulations. Advanced variations of alchem. free energy perturbation simulations, however, also require nongeometric constraints. In our recently developed multisite λ-dynamics simulation method, the conventional λ parameters that are assocd. with the progress variables in alchem. transformations are treated as dynamic variables and are constrained such that: 0 ≤ λi ≤ 1 and .sum.i λi = 1. Here, we present four functional forms of λ that implicitly satisfy these nongeometric constraints, whose values and forces are facile to compute and that yield stable simulations using a 2 fs integration timestep. Using model systems, we present the sampling characteristics of these functional forms and demonstrate the enhanced sampling profiles and improved convergence rates that are achieved by a functional form of λi that oscillates between λi = 0 and λi = 1 and has relatively steep transitions between these endpoints. Copyright for JCC Journal: © 2011 Wiley Periodicals, Inc.; J. Comput. Chem., 2011.
- 49Abraham, M. J.; Murtola, T.; Schulz, R.; Páll, S.; Smith, J. C.; Hess, B.; Lindahl, E. GROMACS: High performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 2015, 1, 19– 25, DOI: 10.1016/j.softx.2015.06.001Google ScholarThere is no corresponding record for this reference.
- 50Singhal, A. K.; Chien, K. Y.; Wu, W. G.; Rule, G. S. Solution structure of cardiotoxin V from Naja naja atra. Biochemistry 1993, 32, 8036– 8044, DOI: 10.1021/bi00082a026Google Scholar50Solution structure of cardiotoxin V from Naja naja atraSinghal, Arun K.; Chien, Kun Yi; Wu, Wen Guey; Rule, Gordon S.Biochemistry (1993), 32 (31), 8036-44CODEN: BICHAW; ISSN:0006-2960.To det. whether the unique activity of this cardiotoxin (CTX) is attributable to its tertiary structure, the soln. structure of CTX V was detd. by NMR methods. On the basis of these studies, this cardiotoxin has the same general topol. as other members of the family, and thus its unusual properties do not arise from any gross structural differences that are detectable by soln. NMR methods. Mol. dynamics calcns. indicate that residues 36-50 show concerted fluctuations. On the basis of sequence similarity, the authors postulate that residues 30-34 are important in detg. the specificity of cardiotoxins for fusion vs. lysis of vesicles.
- 51Ramanadham, M.; Sieker, L.; Jensen, L. Refinement of triclinic lysozyme: II. The method of stereochemically restrained least squares. Acta Crystallographica Section B: Structural Science 1990, 46, 63– 69, DOI: 10.1107/S0108768189009195Google Scholar51Refinement of triclinic lysozyme: II. The method of stereochemically restrained least squaresRamanadham M; Sieker L C; Jensen L HActa crystallographica. Section B, Structural science (1990), 46 ( Pt 1) (), 63-9 ISSN:0108-7681.Refinement of triclinic lysozyme by restrained least squares against the 2 A resolution X-ray data is described, beginning with the model from cycle 17 of the preceding paper [Hodsdon, Brown, Sieker & Jensen (1990). Acta Cryst. B46, 54-62]. After 20 refinement cycles, R stood at 0.172. Nevertheless, serious errors involving both main-chain and side-chain atoms still remained, requiring numerous model rebuilding sessions interleaved with refinement cycles. After 63 cycles R = 0.124 for the model which includes all protein atoms, 249 water oxygen sites and five nitrate ions. Although the overall B is relatively low, 10.5 A2, B's for atoms in the region of residues 101-103, toward the termini of some of the longer side chains, and in the region of the C terminus of the main chain exceed 20 A2, indicating relatively high atomic mobilities, disorder, or remaining errors in the model.
- 52Sauguet, L.; Poitevin, F.; Murail, S.; Van Renterghem, C.; Moraga-Cid, G.; Malherbe, L.; Thompson, A. W.; Koehl, P.; Corringer, P.-J.; Baaden, M.; Delarue, M. Structural basis for ion permeation mechanism in pentameric ligand-gated ion channels. EMBO journal 2013, 32, 728– 741, DOI: 10.1038/emboj.2013.17Google Scholar52Structural basis for ion permeation mechanism in pentameric ligand-gated ion channelsSauguet, Ludovic; Poitevin, Frederic; Murail, Samuel; Van Renterghem, Catherine; Moraga-Cid, Gustavo; Malherbe, Laurie; Thompson, Andrew W.; Koehl, Patrice; Corringer, Pierre-Jean; Baaden, Marc; Delarue, MarcEMBO Journal (2013), 32 (5), 728-741CODEN: EMJODG; ISSN:0261-4189. (Nature Publishing Group)To understand the mol. mechanism of ion permeation in pentameric ligand-gated ion channels (pLGIC), the structure of an open form of GLIC, a prokaryotic pLGIC, was solved at 2.4 Å. Anomalous diffraction data were used to place bound anions and cations. This reveals ordered water mols. at the level of two rings of hydroxylated residues (named Ser6' and Thr2') that contribute to the ion selectivity filter. Two water pentagons are obsd., a self-stabilized ice-like water pentagon and a second wider water pentagon, with one sodium ion between them. Single-channel electrophysiol. shows that the side-chain hydroxyl of Ser6' is crucial for ion translocation. Simulations and electrostatics calcns. complemented the description of hydration in the pore and suggest that the water pentagons obsd. in the crystal are important for the ion to cross hydrophobic constriction barriers. Simulations that pull a cation through the pore reveal that residue Ser6' actively contributes to ion translocation by reorienting its side chain when the ion is going through the pore. Generalization of these findings to the pLGIC family is proposed.
- 53Lee, T.-W.; Qasim, M., Jr; Laskowski, M., Jr; James, M. N. Structural insights into the non-additivity effects in the sequence-to-reactivity algorithm for serine peptidases and their inhibitors. Journal of molecular biology 2007, 367, 527– 546, DOI: 10.1016/j.jmb.2007.01.008Google Scholar53Structural Insights into the Non-additivity Effects in the Sequence-to-Reactivity Algorithm for Serine Peptidases and their InhibitorsLee, Ting-Wai; Qasim, M. A.; Laskowski, Michael; James, Michael N. G.Journal of Molecular Biology (2007), 367 (2), 527-546CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)Sequence-to-reactivity algorithms (SRAs) for proteins have the potential of being broadly applied in mol. design. Recently, Laskowski et al. have reported an additivity-based SRA that accurately predicts most of the std. free energy changes of assocn. for variants of turkey ovomucoid third domain (OMTKY3) with six serine peptidases, one of which is streptogrisin B (commonly known as Streptomyces griseus peptidase B, SGPB). Non-additivity effects for residues 18I and 32I, and for residues 20I and 32I of OMTKY3 occurred when the assocns. with SGPB were predicted using the SRA. To elucidate precisely the mechanics of these non-additivity effects in structural terms, we have detd. the crystal structures of the unbound OMTKY3 (with Gly32I as in the wild-type amino acid sequence) at a resoln. of 1.16 Å, the unbound Ala32I variant of OMTKY3 at a resoln. of 1.23 Å, and the SGPB:OMTKY3-Ala32I complex (equil. assocn. const. Ka = 7.1×109 M-1 at 21(±2) C°, pH 8.3) at a resoln. of 1.70 Å. Extensive comparisons with the crystal structure of the unbound OMTKY3 confirm our understanding of some previously addressed non-additivity effects. Unexpectedly, SGPB and OMTKY3-Ala32I form a 1:2 complex in the crystal. Comparison with the SGPB:OMTKY3 complex shows a conformational change in the SGPB:OMTKY3-Ala32I complex, resulting from a hinged rigid-body rotation of the inhibitor caused by the steric hindrance between the Me group of Ala32IA of the inhibitor and Pro192BE of the peptidase. This perturbs the interactions among residues 18I, 20I, 32I and 36I of the inhibitor, probably resulting in the above non-additivity effects. This conformational change also introduces residue 10I as an addnl. hyper-variable contact residue to the SRA.
- 54Huang, J.; MacKerell, A. D., Jr CHARMM36 all-atom additive protein force field: Validation based on comparison to NMR data. Journal of computational chemistry 2013, 34, 2135– 2145, DOI: 10.1002/jcc.23354Google Scholar54CHARMM36 all-atom additive protein force field: Validation based on comparison to NMR dataHuang, Jing; MacKerell, Alexander D. JrJournal of Computational Chemistry (2013), 34 (25), 2135-2145CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Protein structure and dynamics can be characterized on the atomistic level with both NMR expts. and mol. dynamics (MD) simulations. Here, the authors quantify the ability of the recently presented CHARMM36 (C36) force field (FF) to reproduce various NMR observables using MD simulations. The studied NMR properties include backbone scalar couplings across hydrogen bonds, residual dipolar couplings (RDCs) and relaxation order parameter, as well as scalar couplings, RDCs, and order parameters for side-chain amino- and methyl-contg. groups. The C36 FF leads to better correlation with exptl. data compared to the CHARMM22/CMAP FF and suggest using C36 in protein simulations. Although both CHARMM FFs contains the same nonbond parameters, the authors' results show how the changes in the internal parameters assocd. with the peptide backbone via CMAP and the χ1 and χ2 dihedral parameters leads to improved treatment of the analyzed nonbond interactions. This highlights the importance of proper treatment of the internal covalent components in modeling nonbond interactions with mol. mechanics FFs.
- 55Buslaev, P.; Aho, N.; Jansen, A.; Bauer, P.; Hess, B.; Groenhof, G. Best practices in constant pH MD simulations: accuracy and precision. J. Chem. Theory Comput. 2022, DOI: 10.1021/acs.jctc.2c00517 .Google ScholarThere is no corresponding record for this reference.
- 56Martini-website, General purpose coarse-grained force field. http://cgmartini.nl/index.php/tools2/proteins-and-bilayers/204-martinize (accessed October 01, 2021).Google ScholarThere is no corresponding record for this reference.
- 57Kaminski, G. A.; Friesner, R. A.; Tirado-Rives, J.; Jorgensen, W. L. Evaluation and reparametrization of the OPLS-AA force field for proteins via comparison with accurate quantum chemical calculations on peptides. J. Phys. Chem. B 2001, 105, 6474– 6487, DOI: 10.1021/jp003919dGoogle Scholar57Evaluation and Reparametrization of the OPLS-AA Force Field for Proteins via Comparison with Accurate Quantum Chemical Calculations on PeptidesKaminski, George A.; Friesner, Richard A.; Tirado-Rives, Julian; Jorgensen, William L.Journal of Physical Chemistry B (2001), 105 (28), 6474-6487CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)We present results of improving the OPLS-AA force field for peptides by means of refitting the key Fourier torsional coeffs. The fitting technique combines using accurate ab initio data as the target, choosing an efficient fitting subspace of the whole potential-energy surface, and detg. wts. for each of the fitting points based on magnitudes of the potential-energy gradient. The av. energy RMS deviation from the LMP2/cc-pVTZ(-f)//HF/6-31G** data is reduced by ∼40% from 0.81 to 0.47 kcal/mol as a result of the fitting for the electrostatically uncharged dipeptides. Transferability of the parameters is demonstrated by using the same alanine dipeptide-fitted backbone torsional parameters for all of the other dipeptides (with the appropriate side-chain refitting) and the alanine tetrapeptide. Parameters of nonbonded interactions have also been refitted for the sulfur-contg. dipeptides (cysteine and methionine), and the validity of the new Coulombic charges and the van der Waals σ's and ε's is proved through reproducing gas-phase energies of complex formation heats of vaporization and densities of pure model liqs. Moreover, a novel approach to fitting torsional parameters for electrostatically charged mol. systems has been presented and successfully tested on five dipeptides with charged side chains.
- 58Buck, M.; Bouguet-Bonnet, S.; Pastor, R. W.; MacKerell, A. D., Jr Importance of the CMAP correction to the CHARMM22 protein force field: dynamics of hen lysozyme. Biophysical journal 2006, 90, L36– L38, DOI: 10.1529/biophysj.105.078154Google Scholar58Importance of the CMAP correction to the CHARMM22 protein force field: dynamics of hen lysozymeBuck, Matthias; Bouguet-Bonnet, Sabine; Pastor, Richard W.; MacKerell, Alexander D., Jr.Biophysical Journal (2006), 90 (4), L36-L38CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The recently developed CMAP correction to the CHARMM22 force field (C22) is evaluated from 25 ns mol. dynamics simulations on hen lysozyme. Substantial deviations from exptl. backbone root mean-square fluctuations and N-H NMR order parameters obtained in the C22 trajectories (esp. in the loops) are eliminated by the CMAP correction. Thus, the C22/CMAP force field yields improved dynamical and structural properties of proteins in mol. dynamics simulations.
- 59Durell, S. R.; Brooks, B. R.; Ben-Naim, A. Solvent-induced forces between two hydrophilic groups. J. Phys. Chem. 1994, 98, 2198– 2202, DOI: 10.1021/j100059a038Google Scholar59Solvent-Induced Forces between Two Hydrophilic GroupsDurell, Stewart R.; Brooks, Bernard R.; Ben-Naim, AriehJournal of Physical Chemistry (1994), 98 (8), 2198-202CODEN: JPCHAX; ISSN:0022-3654.Mol. dynamics simulations were used to calc. the force between two simple hydrophilic solutes in dil. aq. soln. The "solutes" were two water mols. in the same relative orientation as the next-nearest neighbors in hexagonal ice I. Both the direct and solvent-induced contributions to the force were calcd. as a function of sepn. distance. The total force between the solutes was found to be most attractive at 5.0 Å (-1.6 kcal/mol/Å). The potential of mean force had a min. at 4.3 Å, which is 0.2 Å closer than the next-nearest-neighbor distance in ice. A parallel set of simulations were conducted with the partial charges on the "solutes" removed to examine hydrophobic analogs. In this case, the total force was most attractive at 3.5 Å (-0.9 kcal/mol/Å), and the min. of the potential was at the contact distance of 3.2 Å. In agreement with earlier predictions, the max. solvent-induced contribution to the potential was ca. 4 times more neg. for the hydrophilic "solutes" than for the hydrophobic ones. These differences are shown to be due predominantly to a solvent water mol. which simultaneously hydrogen bonds to both hydrophilic "solutes". The results support earlier assertions that solvent-induced interactions between polar amino acid residues are more important in protein folding and stability than generally considered.
- 60Neria, E.; Fischer, S.; Karplus, M. Simulation of activation free energies in molecular systems. J. Chem. Phys. 1996, 105, 1902– 1921, DOI: 10.1063/1.472061Google Scholar60Simulation of activation free energies in molecular systemsNeria, Eyal; Fischer, Stefan; Karplus, MartinJournal of Chemical Physics (1996), 105 (5), 1902-1921CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)A method is presented for detg. activation free energies in complex mol. systems. The method relies on knowledge of the min. energy path and bases the activation free energy calcn. on moving along this path from a min. to a saddle point. Use is made of a local reaction coordinate which describes the advance of the reaction in each segment of the min. energy path. The activation free energy is formulated as a sum of two terms. The first is due to the change in the local reaction coordinate between the endpoints of each segment of the path. The second is due to the change in direction of the min. energy path between consecutive segments. Both contributions can be obtained by mol. dynamics simulations with a constraint on the local reaction coordinate. The method is illustrated by applying it to a model potential and to the C7eq to C7ax transition in the alanine dipeptide. It is found that the term due to the change of direction in the reaction path can make a substantial contribution to the activation free energy.
- 61Yesylevskyy, S. O.; Schäfer, L. V.; Sengupta, D.; Marrink, S. J. Polarizable water model for the coarse-grained MARTINI force field. PLoS computational biology 2010, 6, e1000810, DOI: 10.1371/journal.pcbi.1000810Google Scholar61Polarizable water model for the coarse-grained MARTINI force fieldYesylevskyy Semen O; Schafer Lars V; Sengupta Durba; Marrink Siewert JPLoS computational biology (2010), 6 (6), e1000810 ISSN:.Coarse-grained (CG) simulations have become an essential tool to study a large variety of biomolecular processes, exploring temporal and spatial scales inaccessible to traditional models of atomistic resolution. One of the major simplifications of CG models is the representation of the solvent, which is either implicit or modeled explicitly as a van der Waals particle. The effect of polarization, and thus a proper screening of interactions depending on the local environment, is absent. Given the important role of water as a ubiquitous solvent in biological systems, its treatment is crucial to the properties derived from simulation studies. Here, we parameterize a polarizable coarse-grained water model to be used in combination with the CG MARTINI force field. Using a three-bead model to represent four water molecules, we show that the orientational polarizability of real water can be effectively accounted for. This has the consequence that the dielectric screening of bulk water is reproduced. At the same time, we parameterized our new water model such that bulk water density and oil/water partitioning data remain at the same level of accuracy as for the standard MARTINI force field. We apply the new model to two cases for which current CG force fields are inadequate. First, we address the transport of ions across a lipid membrane. The computed potential of mean force shows that the ions now naturally feel the change in dielectric medium when moving from the high dielectric aqueous phase toward the low dielectric membrane interior. In the second application we consider the electroporation process of both an oil slab and a lipid bilayer. The electrostatic field drives the formation of water filled pores in both cases, following a similar mechanism as seen with atomistically detailed models.
- 62Berendsen, H. J.; Postma, J. P.; van Gunsteren, W. F.; Hermans, J. Intermolecular forces; Springer, 1981; pp 331– 342.Google ScholarThere is no corresponding record for this reference.
- 63De Jong, D. H.; Baoukina, S.; Ingólfsson, H. I.; Marrink, S. J. Martini straight: Boosting performance using a shorter cutoff and GPUs. Comput. Phys. Commun. 2016, 199, 1– 7, DOI: 10.1016/j.cpc.2015.09.014Google Scholar63Martini straight: Boosting performance using a shorter cutoff and GPUsde Jong, Djurre H.; Baoukina, Svetlana; Ingolfsson, Helgi I.; Marrink, Siewert J.Computer Physics Communications (2016), 199 (), 1-7CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)In mol. dynamics simulations, sufficient sampling is of key importance and a continuous challenge in the field. The coarse grain Martini force field has been widely used to enhance sampling. In its original implementation, this force field applied a shifted Lennard-Jones potential for the non-bonded van der Waals interactions, to avoid problems related to a relatively short cutoff. Here we investigate the use of a straight cutoff Lennard-Jones potential with potential modifiers. Together with a Verlet neighbor search algorithm, the modified potential allows the use of GPUs to accelerate the computations in Gromacs. We find that this alternative potential has little influence on most of the properties studied, including partitioning free energies, bulk liq. properties and bilayer properties. At the same time, energy conservation is kept within reasonable bounds. We conclude that the newly proposed straight cutoff approach is a viable alternative to the std. shifted potentials used in Martini, offering significant speedup even in the absence of GPUs.
- 64Bussi, G.; Donadio, D.; Parrinello, M. Canonical sampling through velocity rescaling. J. Chem. Phys. 2007, 126, 014101, DOI: 10.1063/1.2408420Google Scholar64Canonical sampling through velocity rescalingBussi, Giovanni; Donadio, Davide; Parrinello, MicheleJournal of Chemical Physics (2007), 126 (1), 014101/1-014101/7CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The authors present a new mol. dynamics algorithm for sampling the canonical distribution. In this approach the velocities of all the particles are rescaled by a properly chosen random factor. The algorithm is formally justified and it is shown that, in spite of its stochastic nature, a quantity can still be defined that remains const. during the evolution. In numerical applications this quantity can be used to measure the accuracy of the sampling. The authors illustrate the properties of this new method on Lennard-Jones and TIP4P water models in the solid and liq. phases. Its performance is excellent and largely independent of the thermostat parameter also with regard to the dynamic properties.
- 65Parrinello, M.; Rahman, A. Polymorphic transitions in single crystals: A new molecular dynamics method. J. Appl. Phys. 1981, 52, 7182– 7190, DOI: 10.1063/1.328693Google Scholar65Polymorphic transitions in single crystals: A new molecular dynamics methodParrinello, M.; Rahman, A.Journal of Applied Physics (1981), 52 (12), 7182-90CODEN: JAPIAU; ISSN:0021-8979.A Lagrangian formulation is introduced; it can be used to make mol. dynamics (MD) calcns. on systems under the most general, externally applied, conditions of stress. In this formulation the MD cell shape and size can change according to dynamic equations given by this Lagrangian. This MD technique was used to the study of structural transitions of a Ni single crystal under uniform uniaxial compressive and tensile loads. Some results regarding the stress-strain relation obtained by static calcns. are invalid at finite temp. Under compressive loading, the model of Ni shows a bifurcation in its stress-strain relation; this bifurcation provides a link in configuration space between cubic and hexagonal close packing. Such a transition could perhaps be obsd. exptl. under extreme conditions of shock.
- 66Hess, B.; Bekker, H.; Berendsen, H. J.; Fraaije, J. G. LINCS: a linear constraint solver for molecular simulations. Journal of computational chemistry 1997, 18, 1463– 1472, DOI: 10.1002/(SICI)1096-987X(199709)18:12<1463::AID-JCC4>3.0.CO;2-HGoogle Scholar66LINCS: a linear constraint solver for molecular simulationsHess, Berk; Bekker, Henk; Berendsen, Herman J. C.; Fraaije, Johannes G. E. M.Journal of Computational Chemistry (1997), 18 (12), 1463-1472CODEN: JCCHDD; ISSN:0192-8651. (Wiley)We present a new LINear Constraint Solver (LINCS) for mol. simulations with bond constraints using the enzyme lysozyme and a 32-residue peptide as test systems. The algorithm is inherently stable, as the constraints themselves are reset instead of derivs. of the constraints, thereby eliminating drift. Although the derivation of the algorithm is presented in terms of matrixes, no matrix matrix multiplications are needed and only the nonzero matrix elements have to be stored, making the method useful for very large mols. At the same accuracy, the LINCS algorithm is 3-4 times faster than the SHAKE algorithm. Parallelization of the algorithm is straightforward.
- 67Miyamoto, S.; Kollman, P. A. Settle: An analytical version of the SHAKE and RATTLE algorithm for rigid water models. Journal of computational chemistry 1992, 13, 952– 962, DOI: 10.1002/jcc.540130805Google Scholar67SETTLE: an analytical version of the SHAKE and RATTLE algorithm for rigid water modelsMiyamoto, Shuichi; Kollman, Peter A.Journal of Computational Chemistry (1992), 13 (8), 952-62CODEN: JCCHDD; ISSN:0192-8651.An anal. algorithm, called SETTLE, for resetting the positions and velocities to satisfy the holonomic constraints on the rigid water model is presented. This method is based on the Cartesian coordinate system and can be used in place of SHAKE and RATTLE. The authors implemented this algorithm in the SPASMS package of mol. mechanics and dynamics. Several series of mol. dynamics simulations were carried out to examine the performance of the new algorithm in comparison with the original RATTLE method. SETTLE is of higher accuracy and is faster than RATTLE with reasonable tolerances by three to nine times on a scalar machine. The performance improvement ranged from factors of 26 to 98 on a vector machine since the method presented is not iterative.
- 68Hub, J. S.; de Groot, B. L.; Grubmüller, H.; Groenhof, G. Quantifying artifacts in Ewald simulations of inhomogeneous systems with a net charge. J. Chem. Theory Comput. 2014, 10, 381– 390, DOI: 10.1021/ct400626bGoogle Scholar68Quantifying Artifacts in Ewald Simulations of Inhomogeneous Systems with a Net ChargeHub, Jochen S.; de Groot, Bert L.; Grubmueller, Helmut; Groenhof, GerritJournal of Chemical Theory and Computation (2014), 10 (1), 381-390CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Ewald summation, which has become the de facto std. for computing electrostatic interactions in biomol. simulations, formally requires that the simulation box is neutral. For non-neutral systems, the Ewald algorithm implicitly introduces a uniform background charge distribution that effectively neutralizes the simulation box. Because a uniform distribution of counter charges typically deviates from the spatial distribution of counterions in real systems, artifacts may arise, in particular in systems with an inhomogeneous dielec. const. Here, we derive an anal. expression for the effect of using an implicit background charge instead of explicit counterions, on the chem. potential of ions in heterogeneous systems, which (i) provides a quant. criterion for deciding if the background charge offers an acceptable trade-off between artifacts arising from sampling problems and artifacts arising from the homogeneous background charge distribution, and (ii) can be used to correct this artifact in certain cases. Our model quantifies the artifact in terms of the difference in charge d. between the non-neutral system with a uniform neutralizing background charge and the real neutral system with a phys. correct distribution of explicit counterions. We show that for inhomogeneous systems, such as proteins and membranes in water, the artifact manifests itself by an overstabilization of ions inside the lower dielec. by tens to even hundreds kilojoules per mol. We have tested the accuracy of our model in mol. dynamics simulations and found that the error in the calcd. free energy for moving a test charge from water into hexadecane, at different net charges of the system and different simulation box sizes, is correctly predicted by the model. The calcns. further confirm that the incorrect distribution of counter charges in the simulation box is solely responsible for the errors in the PMFs.
- 69Thurlkill, R. L.; Grimsley, G. R.; Scholtz, J. M.; Pace, C. N. pK values of the ionizable groups of proteins. Protein science 2006, 15, 1214– 1218, DOI: 10.1110/ps.051840806Google Scholar69pK values of the ionizable groups of proteinsThurlkill, Richard L.; Grimsley, Gerald R.; Scholtz, J. Martin; Pace, C. NickProtein Science (2006), 15 (5), 1214-1218CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)We have used potentiometric titrns. to measure the pK values of the ionizable groups of proteins in alanine pentapeptides with appropriately blocked termini. These pentapeptides provide an improved model for the pK values of the ionizable groups in proteins. Our pK values detd. in 0.1 M KCl at 25°C are: 3.67 ± 0.03 (α-carboxyl), 3.67 ± 0.04 (Asp), 4.25 ± 0.05 (Glu), 6.54 ± 0.04 (His), 8.00 ± 0.03 (α-amino), 8.55 ± 0.03 (Cys), 9.84 ± 0.11 (Tyr), and 10.40 ± 0.08 (Lys). The pK values of some groups differ from the Nozaki and Tanford (N&T) pK values often used in the literature: Asp (3.67 this work vs. 4.0 N&T); His (6.54 this work vs. 6.3 N&T); α-amino (8.00 this work vs. 7.5 N&T); Cys (8.55 this work vs. 9.5 N&T); and Tyr (9.84 this work vs. 9.6 N&T). Our pK values will be useful to those who study pK perturbations in folded and unfolded proteins, and to those who use theory to gain a better understanding of the factors that det. the pK values of the ionizable groups of proteins.
- 70Tanokura, M. 1H-NMR study on the tautomerism of the imidazole ring of histidine residues: I. Microscopic pK values and molar ratios of tautomers in histidine-containing peptides. Biochimica et Biophysica Acta (BBA)-Protein Structure and Molecular Enzymology 1983, 742, 576– 585, DOI: 10.1016/0167-4838(83)90276-5Google ScholarThere is no corresponding record for this reference.
- 71Chiang, C.-M.; Chien, K.-Y.; Lin, H.-j.; Lin, J.-F.; Yeh, H.-C.; Ho, P.-l.; Wu, W.-g. Conformational change and inactivation of membrane phospholipid-related activity of cardiotoxin V from Taiwan cobra venom at acidic pH. Biochemistry 1996, 35, 9167– 9176, DOI: 10.1021/bi952823kGoogle Scholar71Conformational Change and Inactivation of Membrane Phospholipid-Related Activity of Cardiotoxin V from Taiwan Cobra Venom at Acidic pHChiang, Chien-Min; Chien, Kun-Yi; Lin, Hai-jui; Lin, Ji-Fu; Yeh, Hsien-Chi; Ho, Pei-li; Wu, Wen-gueyBiochemistry (1996), 35 (28), 9167-9176CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)The phospholipid binding activity of cardiotoxin V from Naja naja atra (CTX A5) was studied by use of Langmuir monolayers and found to exhibit pH-dependence in binding to phosphatidylcholine membrane with an apparent pKa around 6.0. Proton NMR investigation of the CTX A5 mol. in the presence of phosphatidylcholine micelles reveals a decrease in assocn. of CTX A5 with membranes at low pH as a result of the protonation of His-4 near the membrane binding site of loop I region of CTX. The pH-dependent binding can be attributed mainly, but not solely, to the change in charge content of the CTX mol. upon His-4 protonation at the membrane/water interface. This is shown by analyzing the pH- and ionic strength dependence of binding of CTXs to phospholipid monolayers according to Gouy-Chapman theory. The protonation of the His-4 residue also results in a local conformational change in the loop I region since the chem. shifts of amide protons for the amino acid residues from Cys-3 to Thr-14 are all found to vary as a function of pH with an apparent pKa similar to that of His-4. Interestingly, the effect is relayed to other amino acid residues in the structural core of the protein such as those in C-terminal (Lys-60, Cys-61, and Asn-62) and triple-stranded antiparallel β-sheet (Cys-22, Lys-24, Ala-25, Arg-38, and Ala-41) regions. An addnl. local conformational change in the mol. results around pH 5 as evidenced by CD spectroscopic studies, although this change does not affect the characteristic β-sheet and three-finger loop structure of CTX mol. as revealed by two-dimensional NOESY 1H NMR study. The latter conformational change at acidic pH, however, completely inactivates CTX-induced aggregation/fusion activity of sphingomyelin vesicles. The results suggest that deciphering the functional sites of CTXs on the basis of structure and dynamics detd. at low pH should be done with caution. Since 19 out of 44 CTX homologs with known amino acid sequence contain His-4, the effect of His-4 on the structure and function of CTX mols. is important and is discussed in terms of the diverse membrane targets of CTX subtypes. Also discussed is the pH-induced activation of snake venom proteins in the victim.
- 72Chiang, C.-M.; Chang, S.-L.; Lin, H.-j.; Wu, W.-g. The role of acidic amino acid residues in the structural stability of snake cardiotoxins. Biochemistry 1996, 35, 9177– 9186, DOI: 10.1021/bi960077tGoogle Scholar72The Role of Acidic Amino Acid Residues in the Structural Stability of Snake CardiotoxinsChiang, Chien-Min; Chang, Shou-Lin; Lin, Hai-jui; Wu, Wen-gueyBiochemistry (1996), 35 (28), 9177-9186CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)PH-induced structural transitions occurred in cardiotoxin V from Naja naja atra (CTX A5) at two pH values as judged by the CD ellipticity around 195 nm: an increase in the β-sheet content occurred around pH 4 and followed by a decrease, therein, around pH 2. The pKa of three acidic amino acid residues in CTX A5, i.e., Glu-17, Asp-42, and Asp-59, were detd. to be 4.0, 3.2, and below 2.3, resp., by NMR spectroscopy. The low pKa value of Asp-59 implies salt bridge formation between Lys-2 and Asp-59. Thus, electrostatic interaction may stabilize the three loop structure in addn. to the hydrogen bonds between N- and C-termini of CTX mol. Second, 2,2,2-trifluoroethanol (TFE) and guanidinium chloride (GdmHCl) were found to induce α-helical and random coil formation, resp., in CTX A5 and eight other β-sheet CTXs. Comparison of the relative potencies of TFE and GdmHCl to induce structural changes suggests that the amino acid residue located at position 17 plays a role in the structural stability. Specifically, CTXs contg. neg. charged Glu-17 are least stable. It is suggested that Glu-17 may perturb the interaction between Lys-2 and Asp-59, and thus the overall stability of β-sheet, in the presence of denaturing reagent. In conclusion, the perturbed structural stability of CTXs may partially explain the lower activity CTX exhibits at acidic pH. A structural model to account for the unfolding and refolding of CTX mols. without the breaking of disulfide bonds is also proposed.
- 73Webb, H.; Tynan-Connolly, B. M.; Lee, G. M.; Farrell, D.; O’Meara, F.; Søndergaard, C. R.; Teilum, K.; Hewage, C.; McIntosh, L. P.; Nielsen, J. E. Remeasuring HEWL pKa values by NMR spectroscopy: Methods, analysis, accuracy, and implications for theoretical pKa calculations. Proteins: Struct., Funct., Bioinf. 2011, 79, 685– 702, DOI: 10.1002/prot.22886Google Scholar73Remeasuring HEWL pKa values by NMR spectroscopy: methods, analysis, accuracy, and implications for theoretical pKa calculationsWebb, Helen; Tynan-Connolly, Barbara Mary; Lee, Gregory M.; Farrell, Damien; O'Meara, Fergal; Sondergaard, Chresten R.; Teilum, Kaare; Hewage, Chandralal; McIntosh, Lawrence P.; Nielsen, Jens ErikProteins: Structure, Function, and Bioinformatics (2011), 79 (3), 685-702CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)Site-specific pKa values measured by NMR spectroscopy provide essential information on protein electrostatics, the pH-dependence of protein structure, dynamics and function, and constitute an important benchmark for protein pKa calcn. algorithms. Titrn. curves can be measured by tracking the NMR chem. shifts of several reporter nuclei vs. sample pH. However, careful anal. of these curves is needed to ext. residue-specific pKa values since pH-dependent chem. shift changes can arise from many sources, including through-bond inductive effects, through-space elec. field effects, and conformational changes. We have re-measured titrn. curves for all carboxylates and His 15 in Hen Egg White Lysozyme (HEWL) by recording the pH-dependent chem. shifts of all backbone amide nitrogens and protons, Asp/Glu side chain protons and carboxyl carbons, and imidazole protonated carbons and protons in this protein. We extd. pKa values from the resulting titrn. curves using std. fitting methods, and compared these values to each other, and with those measured previously by 1H NMR. This anal. gives insights into the true accuracy assocd. with exptl. measured pKa values. We find that apparent pKa values frequently differ by 0.5-1.0 units depending upon the nuclei monitored, and that larger differences occasionally can be obsd. The variation in measured pKa values, which reflects the difficulty in fitting and assigning pH-dependent chem. shifts to specific ionization equil., has significant implications for the exptl. procedures used for measuring protein pKa values, for the benchmarking of protein pKa calcn. algorithms, and for the understanding of protein electro-statics in general.
- 74Lee, J.; Miller, B. T.; Damjanovic, A.; Brooks, B. R. Enhancing constant-pH simulation in explicit solvent with a two-dimensional replica exchange method. J. Chem. Theory Comput. 2015, 11, 2560– 2574, DOI: 10.1021/ct501101fGoogle Scholar74Enhancing Constant-pH Simulation in Explicit Solvent with a Two-Dimensional Replica Exchange MethodLee, Juyong; Miller, Benjamin T.; Damjanovic, Ana; Brooks, Bernard R.Journal of Chemical Theory and Computation (2015), 11 (6), 2560-2574CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We present a new method for enhanced sampling for const.-pH simulations in explicit water based on a two-dimensional (2D) replica exchange scheme. The new method is a significant extension of our previously developed const.-pH simulation method, which is based on enveloping distribution sampling (EDS) coupled with a one-dimensional (1D) Hamiltonian exchange method (HREM). EDS constructs a hybrid Hamiltonian from multiple discrete end state Hamiltonians that, in this case, represent different protonation states of the system. The ruggedness and heights of the hybrid Hamiltonian's energy barriers can be tuned by the smoothness parameter. Within the context of the 1D EDS-HREM method, exchanges are performed between replicas with different smoothness parameters, allowing frequent protonation-state transitions and sampling of conformations that are favored by the end-state Hamiltonians. In this work, the 1D method is extended to 2D with an addnl. dimension, external pH. Within the context of the 2D method (2D EDS-HREM), exchanges are performed on a lattice of Hamiltonians with different pH conditions and smoothness parameters. We demonstrate that both the 1D and 2D methods exactly reproduce the thermodn. properties of the semigrand canonical (SGC) ensemble of a system at a given pH. We have tested our new 2D method on aspartic acid, glutamic acid, lysine, a four residue peptide (sequence KAAE), and snake cardiotoxin. In all cases, the 2D method converges faster and without loss of precision; the only limitation is a loss of flexibility in how CPU time is employed. The results for snake cardiotoxin demonstrate that the 2D method enhances protonation-state transitions, samples a wider conformational space with the same amt. of computational resources, and converges significantly faster overall than the original 1D method.
- 75Pezeshkian, W.; König, M.; Wassenaar, T. A.; Marrink, S. J. Backmapping triangulated surfaces to coarse-grained membrane models. Nat. Commun. 2020, 11, 2296, DOI: 10.1038/s41467-020-16094-yGoogle Scholar75Backmapping triangulated surfaces to coarse-grained membrane modelsPezeshkian, Weria; Koenig, Melanie; Wassenaar, Tsjerk A.; Marrink, Siewert J.Nature Communications (2020), 11 (1), 2296CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Many biol. processes involve large-scale changes in membrane shape. Computer simulations of these processes are challenging since they occur across a wide range of spatiotemporal scales that cannot be investigated in full by any single current simulation technique. A potential soln. is to combine different levels of resoln. through a multiscale scheme. Here, we present a multiscale algorithm that backmaps a continuum membrane model represented as a dynamically triangulated surface (DTS) to its corresponding mol. model based on the coarse-grained (CG) Martini force field. Thus, we can use DTS simulations to equilibrate slow large-scale membrane conformational changes and then explore the local properties at CG resoln. We demonstrate the power of our method by backmapping a vesicular bud induced by binding of Shiga toxin and by transforming the membranes of an entire mitochondrion to near-at. resoln. Our approach opens the way to whole cell simulations at mol. detail.
- 76Wolfram Research, Inc. Mathematica, Version 11.3. Champaign: IL, 2018.Google ScholarThere is no corresponding record for this reference.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 75 publications.
- Tomás F. D. Silva, Giovanni Bussi. Characterizing RNA Oligomers Using Stochastic Titration Constant-pH Metadynamics Simulations. Journal of Chemical Information and Modeling 2025, 65
(7)
, 3568-3580. https://doi.org/10.1021/acs.jcim.4c02185
- Shijie Xu, Akira Onoda. Accurate and Rapid Prediction of Protein pKa: Protein Language Models Reveal the Sequence–pKa Relationship. Journal of Chemical Theory and Computation 2025, 21
(7)
, 3752-3764. https://doi.org/10.1021/acs.jctc.4c01288
- Mohannad J. Yousef, Nuno F. B. Oliveira, João N. M. Vitorino, Pedro B. P. S. Reis, Piotr Draczkowski, Maciej Maj, Krzysztof Jozwiak, Miguel Machuqueiro. Toward Accurate pH-Dependent Binding Constant Predictions Using Molecular Docking and Constant-pH MD Calculations. Journal of Chemical Theory and Computation 2025, 21
(5)
, 2655-2667. https://doi.org/10.1021/acs.jctc.4c01291
- Markéta Paloncýová, Mariana Valério, Ricardo Nascimento Dos Santos, Petra Kührová, Martin Šrejber, Petra Čechová, Dimitar A. Dobchev, Akshay Balsubramani, Pavel Banáš, Vikram Agarwal, Paulo C. T. Souza, Michal Otyepka. Computational Methods for Modeling Lipid-Mediated Active Pharmaceutical Ingredient Delivery. Molecular Pharmaceutics 2025, 22
(3)
, 1110-1141. https://doi.org/10.1021/acs.molpharmaceut.4c00744
- Eliane Briand, Bartosz Kohnke, Carsten Kutzner, Helmut Grubmüller. Constant pH Simulation with FMM Electrostatics in GROMACS. (A) Design and Applications. Journal of Chemical Theory and Computation 2025, 21
(4)
, 1762-1786. https://doi.org/10.1021/acs.jctc.4c01318
- Bartosz Kohnke, Eliane Briand, Carsten Kutzner, Helmut Grubmüller. Constant pH Simulation with FMM Electrostatics in GROMACS. (B) GPU Accelerated Hamiltonian Interpolation. Journal of Chemical Theory and Computation 2025, 21
(4)
, 1787-1804. https://doi.org/10.1021/acs.jctc.4c01319
- David Beyer, Pablo M. Blanco, Jonas Landsgesell, Peter Košovan, Christian Holm. How To Correct Erroneous Symmetry-Breaking in Coarse-Grained Constant-pH Simulations. Journal of Chemical Theory and Computation 2025, 21
(3)
, 1396-1404. https://doi.org/10.1021/acs.jctc.4c01010
- Marc Domingo, Horacio V. Guzman, Matej Kanduč, Jordi Faraudo. Electrostatic Interaction between SARS-CoV-2 and Charged Surfaces: Spike Protein Evolution Changed the Game. Journal of Chemical Information and Modeling 2025, 65
(1)
, 240-251. https://doi.org/10.1021/acs.jcim.4c01724
- Craig A. Peeples, Ruibin Liu, Jana Shen. Force Field Limitations of All-Atom Continuous Constant pH Molecular Dynamics. The Journal of Physical Chemistry B 2024, 128
(47)
, 11616-11624. https://doi.org/10.1021/acs.jpcb.4c05971
- Richard S. Hong, Busayo D. Alagbe, Alessandra Mattei, Ahmad Y. Sheikh, Mark E. Tuckerman. Enhanced and Efficient Predictions of Dynamic Ionization through Constant-pH Adiabatic Free Energy Dynamics. Journal of Chemical Theory and Computation 2024, 20
(22)
, 10010-10021. https://doi.org/10.1021/acs.jctc.4c00704
- Noora Aho, Gerrit Groenhof, Pavel Buslaev. What Is the Protonation State of Proteins in Crystals? Insights from Constant pH Molecular Dynamics Simulations. The Journal of Physical Chemistry B 2024, 128
(45)
, 11124-11133. https://doi.org/10.1021/acs.jpcb.4c05947
- Zhongxin Cui, Yuefeng Wang, Lei Zhang, Haishan Qi. Zwitterionic Peptides: From Mechanism, Design Strategies to Applications. ACS Applied Materials & Interfaces 2024, 16
(42)
, 56497-56518. https://doi.org/10.1021/acsami.4c08891
- Wonmuk Hwang, Steven L. Austin, Arnaud Blondel, Eric D. Boittier, Stefan Boresch, Matthias Buck, Joshua Buckner, Amedeo Caflisch, Hao-Ting Chang, Xi Cheng, Yeol Kyo Choi, Jhih-Wei Chu, Michael F. Crowley, Qiang Cui, Ana Damjanovic, Yuqing Deng, Mike Devereux, Xinqiang Ding, Michael F. Feig, Jiali Gao, David R. Glowacki, James E. Gonzales, II, Mehdi Bagerhi Hamaneh, Edward D. Harder, Ryan L. Hayes, Jing Huang, Yandong Huang, Phillip S. Hudson, Wonpil Im, Shahidul M. Islam, Wei Jiang, Michael R. Jones, Silvan Käser, Fiona L. Kearns, Nathan R. Kern, Jeffery B. Klauda, Themis Lazaridis, Jinhyuk Lee, Justin A. Lemkul, Xiaorong Liu, Yun Luo, Alexander D. MacKerell, Jr., Dan T. Major, Markus Meuwly, Kwangho Nam, Lennart Nilsson, Victor Ovchinnikov, Emanuele Paci, Soohyung Park, Richard W. Pastor, Amanda R. Pittman, Carol Beth Post, Samarjeet Prasad, Jingzhi Pu, Yifei Qi, Thenmalarchelvi Rathinavelan, Daniel R. Roe, Benoit Roux, Christopher N. Rowley, Jana Shen, Andrew C. Simmonett, Alexander J. Sodt, Kai Töpfer, Meenu Upadhyay, Arjan van der Vaart, Luis Itza Vazquez-Salazar, Richard M. Venable, Luke C. Warrensford, H. Lee Woodcock, Yujin Wu, Charles L. Brooks, III, Bernard R. Brooks, Martin Karplus. CHARMM at 45: Enhancements in Accessibility, Functionality, and Speed. The Journal of Physical Chemistry B 2024, 128
(41)
, 9976-10042. https://doi.org/10.1021/acs.jpcb.4c04100
- Junjie Shen, Yi Sun, Xuanzhe Liu, Yimin Chai, Chunyang Wang, Jia Xu. Nerve Regeneration Potential of Antioxidant-Modified Black Phosphorus Quantum Dots in Peripheral Nerve Injury. ACS Nano 2024, 18
(34)
, 23518-23536. https://doi.org/10.1021/acsnano.4c07285
- Tomasz K. Piskorz, Laura Perez-Chirinos, Baofu Qiao, Ivan R. Sasselli. Tips and Tricks in the Modeling of Supramolecular Peptide Assemblies. ACS Omega 2024, 9
(29)
, 31254-31273. https://doi.org/10.1021/acsomega.4c02628
- Magnus Lundborg, Christian Wennberg, Erik Lindahl, Lars Norlén. Simulating the Skin Permeation Process of Ionizable Molecules. Journal of Chemical Information and Modeling 2024, 64
(13)
, 5295-5302. https://doi.org/10.1021/acs.jcim.4c00722
- Jonathan Lasham, Amina Djurabekova, Volker Zickermann, Janet Vonck, Vivek Sharma. Role of Protonation States in the Stability of Molecular Dynamics Simulations of High-Resolution Membrane Protein Structures. The Journal of Physical Chemistry B 2024, 128
(10)
, 2304-2316. https://doi.org/10.1021/acs.jpcb.3c07421
- Anton Jansen, Noora Aho, Gerrit Groenhof, Pavel Buslaev, Berk Hess. phbuilder: A Tool for Efficiently Setting up Constant pH Molecular Dynamics Simulations in GROMACS. Journal of Chemical Information and Modeling 2024, 64
(3)
, 567-574. https://doi.org/10.1021/acs.jcim.3c01313
- Tao Luo, Sébastien Le Crom, N. Tan Luong, Khalil Hanna, Jean-François Boily. Goethite-Bound Copper Controls the Fate of Antibiotics in Aquatic Environments. ACS ES&T Water 2024, 4
(2)
, 638-647. https://doi.org/10.1021/acsestwater.3c00666
- Luise Jacobsen, Laura Lydersen, Himanshu Khandelia. ATP-Bound State of the Uncoupling Protein 1 (UCP1) from Molecular Simulations. The Journal of Physical Chemistry B 2023, 127
(45)
, 9685-9696. https://doi.org/10.1021/acs.jpcb.3c03473
- Luís Borges-Araújo, Ilias Patmanidis, Akhil P. Singh, Lucianna H. S. Santos, Adam K. Sieradzan, Stefano Vanni, Cezary Czaplewski, Sergio Pantano, Wataru Shinoda, Luca Monticelli, Adam Liwo, Siewert J. Marrink, Paulo C. T. Souza. Pragmatic Coarse-Graining of Proteins: Models and Applications. Journal of Chemical Theory and Computation 2023, 19
(20)
, 7112-7135. https://doi.org/10.1021/acs.jctc.3c00733
- Varun Mandalaparthy, Madhusmita Tripathy, Nico F. A. van der Vegt. Anions and Cations Affect Amino Acid Dissociation Equilibria via Distinct Mechanisms. The Journal of Physical Chemistry Letters 2023, 14
(41)
, 9250-9256. https://doi.org/10.1021/acs.jpclett.3c02062
- Mykyta V. Prud, Alexander Kyrychenko, Oleg N. Kalugin. pH-Controllable Coating of Silver Nanoparticles with PMMA-b-PDMAEMA Oligomers: A Molecular Dynamics Simulation Study. The Journal of Physical Chemistry C 2023, 127
(24)
, 11748-11759. https://doi.org/10.1021/acs.jpcc.3c02779
- Zhitao Cai, Tengzi Liu, Qiaoling Lin, Jiahao He, Xiaowei Lei, Fangfang Luo, Yandong Huang. Basis for Accurate Protein pKa Prediction with Machine Learning. Journal of Chemical Information and Modeling 2023, 63
(10)
, 2936-2947. https://doi.org/10.1021/acs.jcim.3c00254
- Pavel Buslaev, Noora Aho, Anton Jansen, Paul Bauer, Berk Hess, Gerrit Groenhof. Best Practices in Constant pH MD Simulations: Accuracy and Sampling. Journal of Chemical Theory and Computation 2022, 18
(10)
, 6134-6147. https://doi.org/10.1021/acs.jctc.2c00517
- Jacilene Silva, Matheus Nunes da Rocha, Victor Moreira de Oliveira, Caio Henrique Alexandre Roberto, Francisco Nithael Melo Lúcio, Márcia Machado Marinho, Hélcio Silva dos Santos, Emmanuel Silva Marinho. Allosteric modulation of laeviganoid-based clerodane diterpenes derivatives in muscarinic acetylcholine M1 receptor against tinnitus: a structure-based virtual screening approach. Future Journal of Pharmaceutical Sciences 2025, 11
(1)
https://doi.org/10.1186/s43094-025-00783-w
- Yingqi Qiu, Jiahao Lu, Chenhao Zhao, Yuqiang Xiang, Aiqun Wu, Liqun Shen, Haiou Jiang. Synthesis of N-acylsulfonamide chromone derivatives as efficient anti-Candida albicans agents. Journal of Molecular Structure 2025, 1334 , 141887. https://doi.org/10.1016/j.molstruc.2025.141887
- Aydin J. Hodala, Ian G. Wood, Paola Carbone. Understanding the influence of electrostatic interactions on observed pK shifts in surfactant aggregates using classical simulations. Journal of Molecular Liquids 2025, 427 , 127410. https://doi.org/10.1016/j.molliq.2025.127410
- C. Risueño, I. Carbajo, D. Charro, N. G. A. Abrescia, I. Coluzza. pH-antenna residues trigger a large-scale conformational change in the large extracellular loop domain of the CD81 human receptor. The Journal of Chemical Physics 2025, 162
(15)
https://doi.org/10.1063/5.0251361
- Haiyan Xiong, Qiyuan Chang, Jiayi Ding, Shuyuan Wang, Wenhao Zhang, Yu Li, Yaochen Wu, Pengyan Lin, Chengyu Yang, Miaoxing Liu, Guicun Fang, Yiwei Yang, Jiongfang Xie, Dong Qi, Tao Jiang, Wenfeng Fu, Fen Hu, Yiming Chen, Rongcai Yue, Yanbin Li, Yong Cui, Min Li, Shilong Fan, Yufeng Yang, Yunlu Xu, Dong Li, Fenghua Zhang, Hu Zhao, Congxian Wu, Qingbing Zheng, Kiryl D. Piatkevich, Zhifei Fu. A highly stable monomeric red fluorescent protein for advanced microscopy. Nature Methods 2025, 8 https://doi.org/10.1038/s41592-025-02676-5
- Helge Vatheuer, Oscar Palomino‐Hernández, Janis Müller, Phillip Galonska, Serghei Glinca, Paul Czodrowski. Protonation Effects in Protein‐Ligand Complexes – A Case Study of Endothiapepsin and Pepstatin A with Computational and Experimental Methods. ChemMedChem 2025, 20
(8)
https://doi.org/10.1002/cmdc.202400953
- Varun Mandalaparthy, Nico F. A. van der Vegt. A generic model for pH-sensitive collapse of hydrophobic polymers. Physical Chemistry Chemical Physics 2025, 27
(14)
, 6984-6993. https://doi.org/10.1039/D4CP04756G
- Li-Nan Chen, Hui Zhou, Kun Xi, Shizhuo Cheng, Yongfeng Liu, Yifan Fu, Xiangyu Ma, Ping Xu, Su-Yu Ji, Wei-Wei Wang, Dan-Dan Shen, Huibing Zhang, Qingya Shen, Renjie Chai, Min Zhang, Lin Yang, Feng Han, Chunyou Mao, Xiujun Cai, Yan Zhang. Proton perception and activation of a proton-sensing GPCR. Molecular Cell 2025, 85
(8)
, 1640-1657.e8. https://doi.org/10.1016/j.molcel.2025.02.030
- Ming Zhang, Minmin Chen, Yizhe Yan, Juan Lu, Jun Sheng, Mingying Gui, Xiao Ma. Comprehensive characterisation of bioactive compounds in Boletus edulis as functional foods to improve muscle atrophy; through whole plant targeted metabolomics, network pharmacology, in vivo and in vitro experiments, molecular docking and molecular dynamics analysis. Journal of Ethnopharmacology 2025, 346 , 119685. https://doi.org/10.1016/j.jep.2025.119685
- Luca Manciocchi, Alexandre Bianchi, Valérie Mazan, Mark Potapov, Katharina M. Fromm, Martin Spichty. Modeling the Interaction Between Silver(I) Ion and Proteins with 12-6 Lennard-Jones Potential: A Bottom-Up Parameterization Approach. Biophysica 2025, 5
(1)
, 7. https://doi.org/10.3390/biophysica5010007
- Vaibhav Modi, Dmitry Morzov. FluProCAD: a computational screening workflow for fluorescent protein variants. Molecular Physics 2025, 2 https://doi.org/10.1080/00268976.2025.2458641
- Matthew K. Howard, Nicholas Hoppe, Xi-Ping Huang, Darko Mitrovic, Christian B. Billesbølle, Christian B. Macdonald, Eshan Mehrotra, Patrick Rockefeller Grimes, Donovan D. Trinidad, Lucie Delemotte, Justin G. English, Willow Coyote-Maestas, Aashish Manglik. Molecular basis of proton sensing by G protein-coupled receptors. Cell 2025, 188
(3)
, 671-687.e20. https://doi.org/10.1016/j.cell.2024.11.036
- Qiuyu Han, Yuxin Chen, Ziyang Zhang, Liping Fan, Jiaoyue Qiu, Wenqing Zhang, Qi Chen, Jinhui Xu, Qianlian Wu, Yue Zhang, Hongbo Liu, Zhishu Tang, Bo Li, Huaxu Zhu. Unveiling the impact of self-assembly on ultrafiltration: Insights from salvianolic acid B. Separation and Purification Technology 2025, 354 , 128857. https://doi.org/10.1016/j.seppur.2024.128857
- Ya-Kun Zhang, Zhan Xue, Jian-Bo Tong, Jing Tan, Min Yang, Yan-Rong Zeng, Cheng-Jian Tan. Exploration and computational assessment of ochrocephalamine G from
Oxytropis ochrocephala
as an anti-HBV candidate. Journal of Asian Natural Products Research 2025, 57 , 1-14. https://doi.org/10.1080/10286020.2024.2441773
- Rontu Das, Debashis Kundu. Enlightenment the dynamic behavior of norbornene–modified ’click’ 4–arm polyethylene glycol hydrogel: Delving into framework properties and transport properties through molecular dynamics simulations. Computational Materials Science 2025, 247 , 113516. https://doi.org/10.1016/j.commatsci.2024.113516
- Yasmin Momin, Vilas Beloshe. Pharmacophore modeling in drug design. 2025, 313-324. https://doi.org/10.1016/bs.apha.2025.01.010
- A. Scacchi, M. Vuorte, M. Sammalkorpi. Multiscale modelling of biopolymers. Advances in Physics: X 2024, 9
(1)
https://doi.org/10.1080/23746149.2024.2358196
- Ziyuan Niu, Georgios Kementzidis, Peng Zhang, Ziji Zhang, Bernard Essuman, Karin Hasegawa, Miriam Rafailovich, Marcia Simon, Bertal Huseyin Aktas, Evangelos Papadopoulos, Yuefan Deng. Modelling of SARS-CoV-2 spike protein structures at varying pH values. Molecular Simulation 2024, 50
(17-18)
, 1540-1552. https://doi.org/10.1080/08927022.2024.2415524
- Peter Vogel, David Beyer, Christian Holm, Thomas Palberg. CO
2
-induced drastic decharging of dielectric surfaces in aqueous suspensions. Soft Matter 2024, 20
(46)
, 9261-9272. https://doi.org/10.1039/D4SM00957F
- Zilan Feng, Chuan Li, Xiangzhou Yi, Changfeng Xue, Xia Gao, Lin Liao, Qiongyao Xiang, Xuanri Shen, Zhisheng Pei. Raman spectroscopy and molecular dynamics simulations of protein microgels at the oil-water interface. International Journal of Biological Macromolecules 2024, 279 , 135398. https://doi.org/10.1016/j.ijbiomac.2024.135398
- Hongpeng Yu, Xiaotong Wei, Huan Ding, Shaodan Hu, Feng Sun, Zhenghua Cao, Li Shi. Exploring the potential mechanisms of Mahuang Fuzi Xixin decoction in treating elderly bronchial asthma through network pharmacology, molecular docking, and molecular dynamics simulations. Medicine 2024, 103
(41)
, e39921. https://doi.org/10.1097/MD.0000000000039921
- Xiaocheng Guo, Xinyuan Feng, Yue Yang, He Zhang, Lunhao Bai. Spermidine attenuates chondrocyte inflammation and cellular pyroptosis through the AhR/NF-κB axis and the NLRP3/caspase-1/GSDMD pathway. Frontiers in Immunology 2024, 15 https://doi.org/10.3389/fimmu.2024.1462777
- Dilpreet Singh, Suprit Dilip Saoji. Nanoarchitectonics in Macromolecular Science: Integrating Molecular Dynamics with Smart Materials. Journal of Macromolecular Science, Part B 2024, , 1-10. https://doi.org/10.1080/00222348.2024.2403861
- Ya-Kun Zhang, Jian-Bo Tong, Mu-Xuan Luo, Xiao-Yu Xing, Yu-Lu Yang, Zhi-Peng Qing, Ze-Lei Chang, Yan-Rong Zeng. Design and evaluation of piperidine carboxamide derivatives as potent ALK inhibitors through 3D-QSAR modeling, artificial neural network and computational analysis. Arabian Journal of Chemistry 2024, 17
(9)
, 105863. https://doi.org/10.1016/j.arabjc.2024.105863
- Kelly M. Lee, Vance W. Jaeger. Adsorption of
Staphylococcus aureus
biofilm associated compounds on silica probed with molecular dynamics simulations. Biointerphases 2024, 19
(5)
https://doi.org/10.1116/6.0003870
- Alexander Zlobin, Ivan Smirnov, Andrey Golovin. Dynamic interchange between two protonation states is characteristic of active sites of cholinesterases. Protein Science 2024, 33
(8)
https://doi.org/10.1002/pro.5100
- Simon M Lichtinger, Joanne L Parker, Simon Newstead, Philip C Biggin. The mechanism of mammalian proton-coupled peptide transporters. eLife 2024, 13 https://doi.org/10.7554/eLife.96507
- Simon M Lichtinger, Joanne L Parker, Simon Newstead, Philip C Biggin. The mechanism of mammalian proton-coupled peptide transporters. eLife 2024, 13 https://doi.org/10.7554/eLife.96507.3
- Carter J. Wilson, Bert L. de Groot, Vytautas Gapsys. Resolving coupled pH titrations using alchemical free energy calculations. Journal of Computational Chemistry 2024, 45
(17)
, 1444-1455. https://doi.org/10.1002/jcc.27318
- Azadeh Eskandari, Thean Chor Leow, Mohd Basyaruddin Abdul Rahman, Siti Nurbaya Oslan. Molecular dynamics-guided insight into the adsorption–inhibition mechanism for controlling ice growth/melt of antifreeze protein type IV mutant from longhorn sculpin fish. Chemical Papers 2024, 78
(7)
, 4437-4454. https://doi.org/10.1007/s11696-024-03407-4
- Subramani Selvi, Arun Kannan, John M. Jayaraj, Thangavel Selvi, Muthusamy Karthikeyan, Chidambaram Prahalathan, Natarajan Sampath, Kannupal Srinivasan. Synthesis and biological evaluation of
3‐hydroxypyrazoles
as aquaporin 9 inhibitors. Journal of Heterocyclic Chemistry 2024, 61
(4)
, 669-679. https://doi.org/10.1002/jhet.4793
- Hamed Zahraee, Seyed Shahriar Arab, Zahra Khoshbin, Seyed Mohammad Taghdisi, Khalil Abnous. Molecular dynamics simulation as a promising approach for computational study of liquid crystal-based aptasensors. Journal of Biomolecular Structure and Dynamics 2024, 40 , 1-13. https://doi.org/10.1080/07391102.2024.2315326
- Sunita Kumari, Rudolf Podgornik. On the nature of screening in charge-regulated macroion solutions. The Journal of Chemical Physics 2024, 160
(1)
https://doi.org/10.1063/5.0187324
- Patrick Duchstein, Felix Löffler, Dirk Zahn. Efficient Assessment of ‘Instantaneous
pK’
Values from Molecular Dynamics Simulations. ChemPhysChem 2024, 25
(1)
https://doi.org/10.1002/cphc.202300489
- Oleksandr Khoshaba, Viktor Grechaninov, Tetiana Molodetska, Kostiantyn Zavertailo, Illia Malinich. Structural and Functional Features of the Synthetic Benchmark. 2024, 103-115. https://doi.org/10.1007/978-981-97-7571-2_9
- Fernando Luís Barroso da Silva. Constant-pH Simulation Methods for Biomolecular Systems. 2024, 942-963. https://doi.org/10.1016/B978-0-12-821978-2.00090-8
- Giovanni Settanni. Computational approaches to lipid-based nucleic acid delivery systems. The European Physical Journal E 2023, 46
(12)
https://doi.org/10.1140/epje/s10189-023-00385-5
- Hu Ruixuan, Arghya Majee, Jure Dobnikar, Rudolf Podgornik. Electrostatic interactions between charge regulated spherical macroions. The European Physical Journal E 2023, 46
(11)
https://doi.org/10.1140/epje/s10189-023-00373-9
- Jiaxin Luo, Aoqi Zhang, Yuan Yao, Jun Yuan. Finding small molecular compounds to decrease trimethylamine oxide levels in atherosclerosis by virtual screening. Open Chemistry 2023, 21
(1)
https://doi.org/10.1515/chem-2023-0128
- Shuai Zhang, Chao Zhang, Min Zhang, Xin Liu, Sheng Xue. Model construction and optimization of coal molecular structure. Journal of Molecular Structure 2023, 1290 , 135960. https://doi.org/10.1016/j.molstruc.2023.135960
- Anton O. Chugunov, Elena A. Dvoryakova, Maria A. Dyuzheva, Tatyana R. Simonyan, Valeria F. Tereshchenkova, Irina Yu. Filippova, Roman G. Efremov, Elena N. Elpidina. Fighting Celiac Disease: Improvement of pH Stability of Cathepsin L In Vitro by Computational Design. International Journal of Molecular Sciences 2023, 24
(15)
, 12369. https://doi.org/10.3390/ijms241512369
- Tuuva Kastinen, Dawid Lupa, Piotr Bonarek, Dmitrii Fedorov, Maria Morga, Markus B. Linder, Jodie L. Lutkenhaus, Piotr Batys, Maria Sammalkorpi. pH dependence of the assembly mechanism and properties of poly(
l
-lysine) and poly(
l
-glutamic acid) complexes. Physical Chemistry Chemical Physics 2023, 25
(27)
, 18182-18196. https://doi.org/10.1039/D3CP01421E
- Lorena Zuzic, Jan K Marzinek, Ganesh S Anand, Jim Warwicker, Peter J Bond. A pH-dependent cluster of charges in a conserved cryptic pocket on flaviviral envelopes. eLife 2023, 12 https://doi.org/10.7554/eLife.82447
- Adam L. Harmat, Maria Morga, Jodie L. Lutkenhaus, Piotr Batys, Maria Sammalkorpi. Molecular mechanisms of pH-tunable stability and surface coverage of polypeptide films. Applied Surface Science 2023, 615 , 156331. https://doi.org/10.1016/j.apsusc.2023.156331
- Yu Wang, Hao Yang, Wei He, Peixuan Sun, Wenjin Zhao, Miao Liu. Exploring the Potential Hormonal Effects of Tire Polymers (TPs) on Different Species Based on a Theoretical Computational Approach. Polymers 2023, 15
(7)
, 1719. https://doi.org/10.3390/polym15071719
- Nicolas Pierre Friedrich Mueller, Paolo Carloni, Mercedes Alfonso-Prieto. Molecular determinants of acrylamide neurotoxicity through covalent docking. Frontiers in Pharmacology 2023, 14 https://doi.org/10.3389/fphar.2023.1125871
- Pierre Tufféry, Philippe Derreumaux. A refined pH-dependent coarse-grained model for peptide structure prediction in aqueous solution. Frontiers in Bioinformatics 2023, 3 https://doi.org/10.3389/fbinf.2023.1113928
- Fang-Fang Luo, Zhi-Tao Cai, Yan-Dong Huang, . Progress in protein p<i>K</i><sub>a</sub> prediction. Acta Physica Sinica 2023, 72
(24)
, 248704. https://doi.org/10.7498/aps.72.20231356
- Luca Donati, Marcus Weber. Assessing transition rates as functions of environmental variables. The Journal of Chemical Physics 2022, 157
(22)
https://doi.org/10.1063/5.0109555
- Lisbeth R. Kjølbye, Gilberto P. Pereira, Alessio Bartocci, Martina Pannuzzo, Simone Albani, Alessandro Marchetto, Brian Jiménez-García, Juliette Martin, Giulia Rossetti, Marco Cecchini, Sangwook Wu, Luca Monticelli, Paulo C. T. Souza. Towards design of drugs and delivery systems with the Martini coarse-grained model. QRB Discovery 2022, 3 https://doi.org/10.1017/qrd.2022.16
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Illustration of the additional λ-dependent potential energy terms (B–D). Panel A shows the protonation free energy of a titratable residue in its reference state obtained at the force field level, ΔGiMM(λi) . To compensate for the shortcomings of the force field and obtain a zero free energy difference between the two protonation states (A + B), we add the correction potential, ViMM(λ), shown in panel B. A biasing potential, Vibias(λ), (42) is introduced to avoid sampling of nonphysical states (panel C). To model the proton chemical potential (pH), we add a pH-dependent term, VpH(λi) (panel D). For a titratable residue at pH ≠ pKa, the total λ-dependent potential, including the interpolated force field functions and the three additional terms, is illustrated in panel (A + B + C + D).
Figure 2
Figure 2. Multisite representation illustrated for a histidine side chain. Each possible protonation state is represented by its own λ-coordinate. HSP refers to doubly protonated histidine, HSD, and HSE to histidine singly protonated at the Nδ or Nϵ, respectively. HS0 is a common, nonphysical state of the residue. To restrict the sampling to the plane connecting the physical states, a constraint λ1 + λ2 + λ3 = 1 is applied (gray plane). A biasing potential is also applied to enhance sampling at the end states, where one of the λ-coordinates is close to one, while the other coordinates are close to zero.
Figure 3
Figure 3. Titrations of tripeptide amino acids (Glu, Asp, and His) in water. The top and bottom rows show titrations performed with the modified AA CHARMM36 (55) and CG Martini (41) force fields, respectively. In all simulations, neutrality was maintained by including ten buffer particles in combination with the charge constraint. Dots show the fraction of frames in which the residue is deprotonated, and the dashed lines represent the fits to the Henderson–Hasselbalch equation. For His, the blue color represents the macroscopic pKa, while yellow and red represent the microscopic pKa values for HSD (proton on Nδ) and HSE (proton on Nϵ), respectively. In the Martini 2.0 model, HSD and HSE are indistinguishable and hence only the macroscopic titration curve is shown. Errors of Sdeprot were estimated from the standard error of the mean for the different replicas. From the fits, the pKa values were estimated and listed in Table 1.
Figure 4
Figure 4. Titration curves of the cardiotoxin V protein obtained from constant pH MD simulations with the CHARMM36 (top) and Martini 2.0 force fields (bottom). For each of the four titratable residues in this protein, the dots show the fraction of frames in which the residue is deprotonated. Errors of Sdeprot were estimated from the standard error of the mean for the different replicas. The lines show the best fits to the Henderson–Hasselbalch equation. The pKa values for each titratable residue were estimated from these fits and listed in Table 1. The right panel shows the protein structure with the four titratable residues highlighted in stick representation.
Figure 5
Figure 5. Titration curves of the HEWL protein obtained from constant pH MD simulations with the CHARMM36 (top) and Martini 2.0 force fields (bottom). For each of the ten titratable residues, the dots show the fraction of frames in which that residue is deprotonated. Errors of Sdeprot were estimated from the standard error of the mean for the different replicas. The lines show the best fit to the Henderson-Hasselbach equation. The pKa values for each titratable residue were estimated from these fits and listed in Table 1. The right panel shows the protein structure with the ten titratable residues highlighted in stick representation.
Figure 6
Figure 6. (A) Relative performance of interpolating potentials in a previous implementation of CpHMD into a fork of GROMACS 3.3 release (blue) and of charge interpolation in our new implementation (red) as a function of the number of titratable sites. The simulations were performed for the turkey ovomucoid inhibitor protein (PDB ID: 2GKR (53)), shown in the inset, where the titratable sites are highlighted in stick representation. (B) Comparison of the performance between CPU-only and CPU+GPU implementations for the ligand-gated ion channel GLIC (PDB ID: 4HFI (52)) with 185 titratable sites. In total, the GLIC system contained 292135 atoms.
References
This article references 76 other publications.
- 1Hollingsworth, S. A.; Dror, R. O. Molecular Dynamics Simulation for All. Neuron 2018, 99, 1129– 1143, DOI: 10.1016/j.neuron.2018.08.0111Molecular Dynamics Simulation for AllHollingsworth, Scott A.; Dror, Ron O.Neuron (2018), 99 (6), 1129-1143CODEN: NERNET; ISSN:0896-6273. (Cell Press)A review. The impact of mol. dynamics (MD) simulations in mol. biol. and drug discovery has expanded dramatically in recent years. These simulations capture the behavior of proteins and other biomols. in full at. detail and at very fine temporal resoln. Major improvements in simulation speed, accuracy, and accessibility, together with the proliferation of exptl. structural data, have increased the appeal of biomol. simulation to experimentalists-a trend particularly noticeable in, although certainly not limited to, neuroscience. Simulations have proven valuable in deciphering functional mechanisms of proteins and other biomols., in uncovering the structural basis for disease, and in the design and optimization of small mols., peptides, and proteins. Here we describe, in practical terms, the types of information MD simulations can provide and the ways in which they typically motivate further exptl. work.
- 2Groenhof, G.; Modi, V.; Morozov, D. Observe while it happens: catching photoactive proteins in the act with non-adiabatic molecular dynamics simulations. Curr. Opin. Struct. Biol. 2020, 61, 106– 112, DOI: 10.1016/j.sbi.2019.12.0132Observe while it happens: catching photoactive proteins in the act with non-adiabatic molecular dynamics simulationsGroenhof, Gerrit; Modi, Vaibhav; Morozov, DmitryCurrent Opinion in Structural Biology (2020), 61 (), 106-112CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Organisms use photo-receptors to react to light. The first step is usually the absorption of a photon by a prosthetic group embedded inside the photo-receptor, often a conjugated chromophore. The electronic changes in the chromophore induced by photo-absorption can trigger a cascade of structural or chem. transformations that culminate into a response to light. Understanding how these proteins have evolved to mediate their activation process has remained challenging because the required time and spacial resolns. are notoriously difficult to achieve exptl. Therefore, mechanistic insights into photoreceptor activation have been predominantly obtained with computer simulations. Here we briefly outline the challenges assocd. with such computations and review the progress made in this field.
- 3Warshel, A.; Sussman, F.; King, G. Free energy of charges in solvated proteins: microscopic calculations using a reversible charging process. Biochemistry 1986, 25, 8368– 8372, DOI: 10.1021/bi00374a0063Free energy of charges in solvated proteins: microscopic calculations using a reversible charging processWarshel, A.; Sussman, F.; King, G.Biochemistry (1986), 25 (26), 8368-72CODEN: BICHAW; ISSN:0006-2960.Evaluation of the free energy of ionization of acidic groups in proteins may be used as a powerful and general test case for detg. the reliability of calcns. of electrostatic energies in macromols. This test case was addressed by using an adiabatic charging process that evaluates the changes in free energies assocd. with ionizing the acidic groups of aspartate-3 and glutamate-7 in bovine pancreatic trypsin inhibitor and aspartic acid in soln. The results of these free energy calcns. are very encouraging; the error range is ∼1 kcal/mol for these values, which are ∼70 kcal/mol. Thus, the stage of obtaining quant. results in modeling the energetics of solvated proteins is finally being approached.
- 4Alexov, E.; Mehler, E. L.; Baker, N.; Baptista, A. M.; Huang, Y.; Milletti, F.; Erik Nielsen, J.; Farrell, D.; Carstensen, T.; Olsson, M. H. M.; Shen, J. K.; Warwicker, J.; Williams, S.; Word, J. M. Progress in the prediction of pKa values in proteins. Proteins: Struct., Funct., Bioinf. 2011, 79, 3260– 3275, DOI: 10.1002/prot.231894Progress in the prediction of pKa values in proteinsAlexov, Emil; Mehler, Ernest L.; Baker, Nathan; Baptista, Antonio M.; Huang, Yong; Milletti, Francesca; Erik Nielsen, Jens; Farrell, Damien; Carstensen, Tommy; Olsson, Mats H. M.; Shen, Jana K.; Warwicker, Jim; Williams, Sarah; Word, J. MichaelProteins: Structure, Function, and Bioinformatics (2011), 79 (12), 3260-3275CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)A review. The pKa-cooperative aims to provide a forum for exptl. and theor. researchers interested in protein pKa values and protein electrostatics in general. The first round of the pKa-cooperative, which challenged computational labs to carry out blind predictions against pKas exptl. detd. in the lab. of Bertrand Garcia-Moreno, was completed and results discussed at the Telluride meeting (July 6-10, 2009). This article serves as an introduction to the reports submitted by the blind prediction participants that will be published in a special issue of PROTEINS: Structure, Function and Bioinformatics. Here, the authors briefly outline existing approaches for pKa calcns., emphasizing methods that were used by the participants in calcg. the blind pKa values in the first round of the cooperative. The authors then point out some of the difficulties encountered by the participating groups in making their blind predictions, and finally try to provide some insights for future developments aimed at improving the accuracy of pKa calcns. Proteins 2011; © 2011 Wiley-Liss, Inc.
- 5Chen, W.; Morrow, B. H.; Shi, C.; Shen, J. K. Recent development and application of constant pH molecular dynamics. Mol. Simul. 2014, 40, 830– 838, DOI: 10.1080/08927022.2014.9074925Recent development and application of constant pH molecular dynamicsChen, Wei; Morrow, Brian H.; Shi, Chuanyin; Shen, Jana K.Molecular Simulation (2014), 40 (10-11), 830-838CODEN: MOSIEA; ISSN:0892-7022. (Taylor & Francis Ltd.)A review. Soln. pH is a crit. environmental factor for chem. and biol. processes. Over the last decade, significant efforts have been made in the development of const. pH mol. dynamics (pHMD) techniques for gaining detailed insights into pH-coupled dynamical phenomena. In this article, we review the advancement of this field in the past five years, placing a special emphasis on the development of the all-atom continuous pHMD technique. We discuss various applications, including the prediction of pKa shifts for proteins, nucleic acids and surfactant assemblies, elucidation of pH-dependent population shifts, protein-protein and protein-RNA binding, as well as the mechanisms of pH-dependent self-assembly and phase transitions of surfactants and peptides. We also discuss future directions for the further improvement of the pHMD techniques.
- 6Zeng, X.; Mukhopadhyay, S.; Brooks, C. L., III Residue-level resolution of alphavirus envelope protein interactions in pH-dependent fusion. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 2034– 2039, DOI: 10.1073/pnas.14141901126Residue-level resolution of alphavirus envelope protein interactions in pH-dependent fusionZeng, Xiancheng; Mukhopadhyay, Suchetana; Brooks, Charles L., IIIProceedings of the National Academy of Sciences of the United States of America (2015), 112 (7), 2034-2039CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Alphavirus envelope proteins, organized as trimers of E2-E1 heterodimers on the surface of the pathogenic alphavirus, mediate the low pH-triggered fusion of viral and endosomal membranes in human cells. The lack of specific treatment for alphaviral infections motivates our exploration of potential antiviral approaches by inhibiting one or more fusion steps in the common endocytic viral entry pathway. In this work, we performed const. pH mol. dynamics based on an at. model of the alphavirus envelope with icosahedral symmetry. We have identified pH-sensitive residues that cause the largest shifts in thermodn. driving forces under neutral and acidic pH conditions for various fusion steps. A series of conserved interdomain His residues is identified to be responsible for the pH-dependent conformational changes in the fusion process, and ligand binding sites in their vicinity are anticipated to be potential drug targets aimed at inhibiting viral infections.
- 7Law, S. M.; Zhang, B. W.; Brooks, C. L., III pH-sensitive residues in the p19 RNA silencing suppressor protein from carnation Italian ringspot virus affect siRNA binding stability. Protein Sci. 2013, 22, 595– 604, DOI: 10.1002/pro.22437pH-sensitive residues in the p19 RNA silencing suppressor protein from carnation Italian ringspot virus affect siRNA binding stabilityLaw, Sean M.; Zhang, Bin W.; Brooks, Charles L., IIIProtein Science (2013), 22 (5), 595-604CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Tombusviruses, such as Carnation Italian ringspot virus (CIRV), encode a protein homodimer called p19 that is capable of suppressing RNA silencing in their infected hosts by binding to and sequestering short-interfering RNA (siRNA) away from the RNA silencing pathway. P19 binding stability has been shown to be sensitive to changes in pH but the specific amino acid residues involved have remained unclear. Using const. pH mol. dynamics simulations, we have identified key pH-dependent residues that affect CIRV p19-siRNA binding stability at various pH ranges based on calcd. changes in the free energy contribution from each titratable residue. At high pH, the deprotonation of Lys60, Lys67, Lys71, and Cys134 has the largest effect on the binding stability. Similarly, deprotonation of several acidic residues (Asp9, Glu12, Asp20, Glu35, and/or Glu41) at low pH results in a decrease in binding stability. At neutral pH, residues Glu17 and His132 provide a small increase in the binding stability and we find that the optimal pH range for siRNA binding is between 7.0 and 10.0. Overall, our findings further inform recent expts. and are in excellent agreement with data on the pH-dependent binding profile.
- 8Ellis, C. R.; Shen, J. pH-dependent population shift regulates BACE1 activity and inhibition. J. Am. Chem. Soc. 2015, 137, 9543– 9546, DOI: 10.1021/jacs.5b058918pH-Dependent Population Shift Regulates BACE1 Activity and InhibitionEllis, Christopher R.; Shen, JanaJournal of the American Chemical Society (2015), 137 (30), 9543-9546CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)BACE1, a major therapeutic target for treatment of Alzheimer's disease, functions within a narrow pH range. Despite tremendous effort and progress in the development of BACE1 inhibitors, details of the underlying pH-dependent regulatory mechanism remain unclear. Here the authors elucidate the pH-dependent conformational mechanism that regulates BACE1 activity using continuous const.-pH mol. dynamics (MD). The simulations reveal that BACE1 mainly occupies three conformational states and that the relative populations of the states shift according to pH. At intermediate pH, when the catalytic dyad is monoprotonated, a binding-competent state is highly populated, while at low and high pH a Tyr-inhibited state is dominant. The authors' data provide strong evidence supporting conformational selection as a major mechanism for substrate and peptide-inhibitor binding. These new insights, while consistent with expt., greatly extend the knowledge of BACE1 and have implications for further optimization of inhibitors and understanding potential side effects of targeting BACE1. Finally, the work highlights the importance of properly modeling protonation states in MD simulations.
- 9Kim, M. O.; Blachly, P. G.; McCammon, J. A. Conformational dynamics and binding free energies of inhibitors of BACE-1: from the perspective of protonation equilibria. PLoS computational biology 2015, 11, e1004341, DOI: 10.1371/journal.pcbi.10043419Conformational dynamics and binding free energies of inhibitors of BACE-1: from the perspective of protonation equilibriaKim, M. Olivia; Blachly, Patrick G.; McCammon, J. AndrewPLoS Computational Biology (2015), 11 (10), e1004341/1-e1004341/28CODEN: PCBLBG; ISSN:1553-7358. (Public Library of Science)BACE-1 is the β-secretase responsible for the initial amyloidogenesis in Alzheimer's disease, catalyzing hydrolytic cleavage of substrate in a pH-sensitive manner. The catalytic mechanism of BACE-1 requires water-mediated proton transfer from aspartyl dyad to the substrate, as well as structural flexibility in the flap region. Thus, the coupling of protonation and conformational equil. is essential to a full in silico characterization of BACE-1. In this work, we perform const. pH replica exchange mol. dynamics simulations on both apo BACE-1 and five BACE-1-inhibitor complexes to examine the effect of pH on dynamics and inhibitor binding properties of BACE-1. In our simulations, we find that soln. pH controls the conformational flexibility of apo BACE-1, whereas bound inhibitors largely limit the motions of the holo enzyme at all levels of pH. The microscopic pKa values of titratable residues in BACE-1 including its aspartyl dyad are computed and compared between apo and inhibitor-bound states. Changes in protonation between the apo and holo forms suggest a thermodn. linkage between binding of inhibitors and protons localized at the dyad. Utilizing our recently developed computational protocol applying the binding polynomial formalism to the const. pH mol. dynamics (CpHMD) framework, we are able to obtain the pH-dependent binding free energy profiles for various BACE-1-inhibitor complexes. Our results highlight the importance of correctly addressing the binding-induced protonation changes in protein-ligand systems where binding accompanies a net proton transfer. This work comprises the first application of our CpHMD-based free energy computational method to protein-ligand complexes and illustrates the value of CpHMD as an all-purpose tool for obtaining pH-dependent dynamics and binding free energies of biol. systems.
- 10Sarkar, A.; Gupta, P. L.; Roitberg, A. E. pH-dependent conformational changes due to ionizable residues in a hydrophobic protein interior: The study of L25K and L125K variants of SNase. J. Phys. Chem. B 2019, 123, 5742– 5754, DOI: 10.1021/acs.jpcb.9b0381610pH-Dependent Conformational Changes Due to Ionizable Residues in a Hydrophobic Protein Interior: The Study of L25K and L125K Variants of SNaseSarkar, Ankita; Gupta, Pancham Lal; Roitberg, Adrian E.Journal of Physical Chemistry B (2019), 123 (27), 5742-5754CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Ionizable residues in the hydrophobic interior of certain proteins are known to play important roles in life processes like energy transduction and enzyme catalysis. These internal ionizable residues show exptl. apparent pKa values having large shifts as compared to their values in soln. In the present work, we study the pH-dependent conformational changes undergone by two variants of staphylococcal nuclease (SNase), L25K and L125K, using pH replica exchange mol. dynamics (pH-REMD) in explicit solvent. Our results show that the obsd. pKa of Lys25 and Lys125 are significantly different than their pKa in soln. We obsd. that the internal lysine residues prefer to be water-exposed when protonated at low pH, but they remain buried within the hydrophobic pocket when deprotonated at high pH. Using thermodn. laws, we est. the microscopic conformation-specific pKa of the water-exposed and buried conformations of the internal lysine residues and explain their relation to the macroscopic obsd. pKa values. We present the differences in the microscopic mechanisms that lead to similar exptl. obsd. apparent pKa of Lys25 and Lys125, and explain the need of thermodn. models of different complexities to account for our calcns. We see that L25K displays pH-dependent fluctuations throughout the entire β barrel and the α1 helix. In contrast, pH-independent fluctuations are obsd. in L125K, primarily limited to the α3 helix. The present computational study offers a detailed atomistic understanding of the determinants of the obsd. anomalous pKa of internal ionizable residues, bolstering the exptl. findings.
- 11Sarkar, A.; Roitberg, A. E. PH-Dependent Conformational Changes Lead to a Highly Shifted p K a for a Buried Glutamic Acid Mutant of SNase. J. Phys. Chem. B 2020, 124, 11072– 11080, DOI: 10.1021/acs.jpcb.0c0713611pH-Dependent Conformational Changes Lead to a Highly Shifted pKa for a Buried Glutamic Acid Mutant of SNaseSarkar, Ankita; Roitberg, Adrian E.Journal of Physical Chemistry B (2020), 124 (49), 11072-11080CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Ionizable residues are rarely present in the hydrophobic interior of proteins, but when they are, they play important roles in biol. processes such as energy transduction and enzyme catalysis. Internal ionizable residues have anomalous exptl. pKa values with respect to their pKa in bulk water. This work studies the atomistic cause of the highly shifted pKa of the internal Glu23 in the artificially mutated variant V23E of Staphylococcal Nuclease (SNase) using pH replica exchange mol. dynamics (pH-REMD) simulations. The pKa of Glu23 obtained from the authors' calcns. is 6.55, which is elevated with respect to the glutamate pKa of 4.40 in bulk water. The calcd. value is close to the exptl. pKa of 7.10. The authors' simulations show that the highly shifted pKa of Glu23 is the product of a pH-dependent conformational change, which was obsd. exptl. and also seen in the authors' simulations. The authors carry out an anal. of this pH-dependent conformational change in response to the protonation state change of Glu23. Using a four-state thermodn. model, the authors est. the two conformation-specific pKa values of Glu23 and describe the coupling between the conformational and ionization equil.
- 12Baptista, A. M.; Martel, P. J.; Petersen, S. B. Simulation of protein conformational freedom as a function of pH: constant-pH molecular dynamics using implicit titration. Proteins: Struct., Funct., Bioinf. 1997, 27, 523– 544, DOI: 10.1002/(SICI)1097-0134(199704)27:4<523::AID-PROT6>3.0.CO;2-B12Simulation of protein conformational freedom as a function of pH: constant-pH molecular dynamics using implicit titrationBaptista, Antonio M.; Martel, Paulo J.; Petersen, Steffen B.Proteins: Structure, Function, and Genetics (1997), 27 (4), 523-544CODEN: PSFGEY; ISSN:0887-3585. (Wiley-Liss)Soln. pH is a determinant parameter on protein function and stability, and its inclusion in mol. dynamics simulations is attractive for studies at the mol. level. Current mol. dynamics simulations can consider pH only in a very limited way, through a somewhat arbitrary choice of a set of fixed charges on the titrable sites. Conversely, continuum electrostatic methods that explicitly treat pH effects assume a single protein conformation whose choice is not clearly defined. In this paper we describe a general method that combines both titrn. and conformational freedom. The method is based on a potential of mean force for implicit titrn. and combines both usual mol. dynamics and pH-dependent calcns. based on continuum methods. A simple implementation of the method, using a mean field approxn., is presented and applied to the bovine pancreatic trypsin inhibitor. We believe that this const.-pH mol. dynamics method, by correctly sampling both charges and conformation, can become a valuable help in the understanding of the dependence of protein function and stability on pH.
- 13Baptista, A. M.; Teixeira, V. H.; Soares, C. M. Constant-pH molecular dynamics using stochastic titration. J. Chem. Phys. 2002, 117, 4184– 4200, DOI: 10.1063/1.149716413Constant-pH molecular dynamics using stochastic titrationBaptista, Antonio M.; Teixeira, Vitor H.; Soares, Claudio M.Journal of Chemical Physics (2002), 117 (9), 4184-4200CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)A new method is proposed for performing const.-pH mol. dynamics (MD) simulations, i.e., MD simulations where pH is one of the external thermodn. parameters, like the temp. or the pressure. The protonation state of each titrable site in the solute is allowed to change during a mol. mechanics (MM) MD simulation, the new states being obtained from a combination of continuum electrostatics (CE) calcns. and Monte Carlo (MC) simulation of protonation equil. The coupling between the MM/MD and CE/MC algorithms is done in a way that ensures a proper Markov chain, sampling from the intended semigrand canonical distribution. This stochastic titrn. method is applied to succinic acid, aimed at illustrating the method and examg. the choice of its adjustable parameters. The complete titrn. of succinic acid, using const.-pH MD simulations at different pH values, gives a clear picture of the coupling between the trans/gauche isomerization and the protonation process, making it possible to reconcile some apparently contradictory results of previous studies. The present const.-pH MD method is shown to require a moderate increase of computational cost when compared to the usual MD method.
- 14Bürgi, R.; Kollman, P. A.; van Gunsteren, W. F. Simulating proteins at constant pH: An approach combining molecular dynamics and Monte Carlo simulation. Proteins: Struct., Funct., Bioinf. 2002, 47, 469– 480, DOI: 10.1002/prot.1004614Simulating proteins at constant pH: an approach combining molecular dynamics and Monte Carlo simulationBurgi, Roland; Kollman, Peter A.; Van Gunsteren, Wilfred F.Proteins: Structure, Function, and Genetics (2002), 47 (4), 469-480CODEN: PSFGEY; ISSN:0887-3585. (Wiley-Liss, Inc.)For the structure and function of proteins, the pH of the soln. is one of the detg. parameters. Current mol. dynamics (MD) simulations account for the soln. pH only in a limited way by keeping each titratable site in a chosen protonation state. We present an algorithm that generates trajectories at a Boltzmann distributed ensemble of protonation states by a combination of MD and Monte Carlo (MC) simulation. The algorithm is useful for pH-dependent structural studies and to investigate in detail the titrn. behavior of proteins. The method is tested on the acidic residues of the protein hen egg white lysozyme. It is shown that small structural changes may have a big effect on the pKA values of titratable residues.
- 15Mongan, J.; Case, D. A.; McCammon, J. A. Constant pH molecular dynamics in generalized Born implicit solvent. J. Comput. Chem. 2004, 25, 2038– 2048, DOI: 10.1002/jcc.2013915Constant pH molecular dynamics in generalized Born implicit solventMongan, John; Case, David A.; McCammon, J. AndrewJournal of Computational Chemistry (2004), 25 (16), 2038-2048CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)A new method is proposed for const. pH mol. dynamics (MD), employing generalized Born (GB) electrostatics. Protonation states are modeled with different charge sets, and titrating residues sample a Boltzmann distribution of protonation states as the simulation progresses, using Monte Carlo sampling based on GB-derived energies. The method is applied to four different crystal structures of hen egg-white lysozyme (HEWL). PKa predictions derived from the simulations have root-mean-square (RMS) error of 0.82 relative to exptl. values. Similarity of results between the four crystal structures shows the method to be independent of starting crystal structure; this is in contrast to most electrostatics-only models. A strong correlation between conformation and protonation state is noted and quant. analyzed, emphasizing the importance of sampling protonation states in conjunction with dynamics.
- 16Meng, Y.; Roitberg, A. E. Constant pH Replica Exchange Molecular Dynamics in Biomolecules Using a Discrete Protonation Model. J. Chem. Theory Comput. 2010, 6, 1401– 1412, DOI: 10.1021/ct900676b16Constant pH Replica Exchange Molecular Dynamics in Biomolecules Using a Discrete Protonation ModelMeng, Yilin; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2010), 6 (4), 1401-1412CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A const. pH replica exchange mol. dynamics (REMD) method is proposed and implemented to improve coupled protonation and conformational state sampling. By mixing conformational sampling at const. pH (with discrete protonation states) with a temp. ladder, this method avoids conformational trapping. Our method was tested and applied to seven different biol. systems. The const. pH REMD not only predicted pKa correctly for small, model compds. but also converged faster than const. pH mol. dynamics (MD). We further tested our const. pH REMD on a heptapeptide from the ovomucoid third domain (OMTKY3). Although const. pH REMD and MD produced very close pKa values, the const. pH REMD showed its advantage in the efficiency of conformational and protonation state samplings.
- 17Itoh, S. G.; Damjanovic, A.; Brooks, B. R. pH replica-exchange method based on discrete protonation states. Proteins: Struct., Funct., Bioinf. 2011, 79, 3420– 3436, DOI: 10.1002/prot.2317617pH replica-exchange method based on discrete protonation statesItoh, Satoru G.; Damjanovic, Ana; Brooks, Bernard R.Proteins: Structure, Function, and Bioinformatics (2011), 79 (12), 3420-3436CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The authors propose a new algorithm for obtaining proton titrn. curves of ionizable residues. The algorithm is a pH replica-exchange method (PHREM), which is based on the const. pH algorithm of J. Mongan et al. (2004). In the original replica-exchange method, simulations of different replicas are performed at different temps., and the temps. are exchanged between the replicas. In the authors' PHREM, simulations of different replicas are performed at different pH values, and the pHs are exchanged between the replicas. The PHREM was applied to a blocked amino acid and to two protein systems (snake cardiotoxin and turkey ovomucoid third domain), in conjunction with a generalized Born implicit solvent. The performance and accuracy of this algorithm and the original const. pH method (PHMD) were compared. For a single set of simulations at different pHs, the use of PHREM yields more accurate Hill coeffs. of titratable residues. By performing multiple sets of const. pH simulations started with different initial states, the accuracy of predicted pKa values and Hill coeffs. obtained with PHREM and PHMD methods becomes comparable. However, the PHREM algorithm exhibits better samplings of the protonation states of titratable residues and less scatter of the titrn. points and thus better precision of measured pKa values and Hill coeffs. In addn., PHREM exhibits faster convergence of individual simulations than the original const. pH algorithm. Proteins 2011; © 2011 Wiley-Liss, Inc.
- 18Swails, J. M.; York, D. M.; Roitberg, A. E. Constant pH Replica Exchange Molecular Dynamics in Explicit Solvent Using Discrete Protonation States: Implementation, Testing, and Validation. J. Chem. Theory Comput. 2014, 10, 1341– 1352, DOI: 10.1021/ct401042b18Constant pH Replica Exchange Molecular Dynamics in Explicit Solvent Using Discrete Protonation States: Implementation, Testing, and ValidationSwails, Jason M.; York, Darrin M.; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2014), 10 (3), 1341-1352CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)By utilizing Graphics Processing Units, we show that const. pH mol. dynamics simulations (CpHMD) run in Generalized Born (GB) implicit solvent for long time scales can yield poor pKa predictions as a result of sampling unrealistic conformations. To address this shortcoming, we present a method for performing const. pH mol. dynamics simulations (CpHMD) in explicit solvent using a discrete protonation state model. The method involves std. mol. dynamics (MD) being propagated in explicit solvent followed by protonation state changes being attempted in GB implicit solvent at fixed intervals. Replica exchange along the pH-dimension (pH-REMD) helps to obtain acceptable titrn. behavior with the proposed method. We analyzed the effects of various parameters and settings on the titrn. behavior of CpHMD and pH-REMD in explicit solvent, including the size of the simulation unit cell and the length of the relaxation dynamics following protonation state changes. We tested the method with the amino acid model compds., a small pentapeptide with two titratable sites, and hen egg white lysozyme (HEWL). The proposed method yields superior predicted pKa values for HEWL over hundreds of nanoseconds of simulation relative to corresponding predicted values from simulations run in implicit solvent.
- 19Swails, J. M.; Roitberg, A. E. Enhancing conformation and protonation state sampling of hen egg white lysozyme using pH replica exchange molecular dynamics. J. Chem. Theory Comput. 2012, 8, 4393– 4404, DOI: 10.1021/ct300512h19Enhancing Conformation and Protonation State Sampling of Hen Egg White Lysozyme Using pH Replica Exchange Molecular DynamicsSwails, Jason M.; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (11), 4393-4404CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We evaluate the efficiency of the pH replica exchange mol. dynamics (pH-REMD) method proposed by Itoh et al. (Proteins 2011, 79, 3420-3436) by using it to predict the pKa values of the titratable residues in hen egg white lysozyme (HEWL). PKa values predicted using pH-REMD converge significantly faster than those calcd. using const. pH mol. dynamics (CpHMD). Furthermore, increasing the frequency between exchange attempts in pH-REMD simulations improves protonation and conformational state sampling. By enabling the simulation to sample both conformational and protonation states more rapidly, pH-REMD simulations provide valuable insight into the pH-dependence of HEWL that the CpHMD simulations failed to capture. We present an efficient and highly scalable implementation of pH-REMD as an attractive enhancement to traditional CpHMD methods.
- 20Börjesson, U.; Hünenberger, P. H. Explicit-solvent molecular dynamics simulation at constant pH: Methodology and application to small amines. J. Chem. Phys. 2001, 114, 9706– 9719, DOI: 10.1063/1.137095920Explicit-solvent molecular dynamics simulation at constant pH: Methodology and application to small aminesBorjesson, Ulf; Hunenberger, Philippe H.Journal of Chemical Physics (2001), 114 (22), 9706-9719CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)A method is developed for performing classical explicit-solvent mol. dynamics (MD) simulations at const. pH, where the protonation state of each ionizable (titratable) group in a simulated compd. is allowed to fluctuate in time, depending on the instantaneous system configuration and the imposed pH. In this method, each ionizable group is treated as a mixed state, i.e., the interaction-function parameters for the group are a linear combination of those of the protonated state and those of the deprotonated state. Free protons are not handled explicitly. Instead, the extent of deprotonation of each group is relaxed towards its equil. value by weak coupling to a "proton bath. " The method relies on precalibrated empirical functions, one for each type of ionizable group present in the simulated compd., which are obtained through multiple MD simulations of monofunctional model compds. In this study, the method is described in detail and its application illustrated by a series of const.-pH MD simulations of small monofunctional amines. In particular, we investigate the influence of the relaxation time used in the weak-coupling scheme, the choice of appropriate model compds. for the calibration of the required empirical functions, and corrections for finite-size effects linked with the small size of the simulation box.
- 21Lee, M. S.; Salsbury, F. R., Jr.; Brooks, C. L., III Constant-pH molecular dynamics using continuous titration coordinates. Proteins: Struct., Funct., Bioinf. 2004, 56, 738– 752, DOI: 10.1002/prot.2012821Constant-pH molecular dynamics using continuous titration coordinatesLee, Michael S.; Salsbury, Freddie R., Jr.; Brooks, Charles L., IIIProteins: Structure, Function, and Bioinformatics (2004), 56 (4), 738-752CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)In this work, we explore the question of whether pKa calcns. based on a microscopic description of the protein and a macroscopic description of the solvent can be implemented to examine conformationally dependent proton shifts in proteins. To this end, we introduce a new method for performing const.-pH mol. dynamics (PHMD) simulations utilizing the generalized Born implicit solvent model. This approach employs an extended Hamiltonian in which continuous titrn. coordinates propagate simultaneously with the at. motions of the system. The values adopted by these coordinates are modulated by potentials of mean force of isolated titratable model groups and the pH to control the proton occupation at particular sites in the polypeptide. Our results for four different proteins yield an abs. av. error of ∼1.6 pK units, and point to the role that thermally driven relaxation of the protein environment in the vicinity of titrating groups plays in modulating the local pKa, thereby influencing the obsd. pK1/2 values. While the accuracy of our method is not yet equiv. to methods that obtain pK1/2 values through the ad hoc scaling of electrostatics, the present approach and const. pH methods in general provide a useful framework for studying pH-dependent phenomena. Further work to improve our model to approach quant. agreement with expt. is outlined.
- 22Khandogin, J.; Brooks, C. L. Constant pH Molecular Dynamics with proton tautomerism. Biophys. J. 2005, 89, 141– 157, DOI: 10.1529/biophysj.105.06134122Constant pH molecular dynamics with proton tautomerismKhandogin, Jana; Brooks, Charles L., IIIBiophysical Journal (2005), 89 (1), 141-157CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The current article describes a new two-dimensional λ-dynamics method to include proton tautomerism in continuous const. pH mol. dynamics (CPHMD) simulations. The two-dimensional λ-dynamics framework is used to devise a tautomeric state titrn. model for the CPHMD simulations involving carboxyl and histidine residues. Combined with the GBSW implicit solvent model, the new method is tested on titrn. simulations of blocked histidine and aspartic acid as well as two benchmark proteins, turkey ovomucoid third domain (OMTKY3) and RNase A (RNase A). A detailed anal. of the errors inherent to the CPHMD methodol. as well as those due to the underlying solvation model is given. The av. abs. error for the computed pKa values in OMTKY3 is 1.0 pK unit. In RNase A the av. abs. errors for the carboxyl and histidine residues are 1.6 and 0.6 pK units, resp. In contrast to the previous work, the new model predicts the correct sign for all the pKa shifts, but one, in the benchmark proteins. The predictions of the tautomeric states of His12 and His48 and the conformational states of His48 and His119 are in agreement with expt. Based on the simulations of OMTKY3 and RNase A, the current work has demonstrated the capability of the CPHMD technique in revealing pH-coupled conformational dynamics of protein side chains.
- 23Donnini, S.; Tegeler, F.; Groenhof, G.; Grubmüller, H. Constant pH molecular dynamics in explicit solvent with λ-dynamics. J. Chem. Theory Comput. 2011, 7, 1962– 1978, DOI: 10.1021/ct200061r23Constant pH Molecular Dynamics in Explicit Solvent with λ-DynamicsDonnini, Serena; Tegeler, Florian; Groenhof, Gerrit; Grubmuller, HelmutJournal of Chemical Theory and Computation (2011), 7 (6), 1962-1978CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)PH is an important parameter in condensed-phase systems, because it dets. the protonation state of titratable groups and thus influences the structure, dynamics, and function of mols. in soln. In most force field simulation protocols, however, the protonation state of a system (rather than its pH) is kept fixed and cannot adapt to changes of the local environment. Here, we present a method, implemented within the MD package GROMACS, for const. pH mol. dynamics simulations in explicit solvent that is based on the λ-dynamics approach. In the latter, the dynamics of the titrn. coordinate λ, which interpolates between the protonated and deprotonated states, is driven by generalized forces between the protonated and deprotonated states. The hydration free energy, as a function of pH, is included to facilitate const. pH simulations. The protonation states of titratable groups are allowed to change dynamically during a simulation, thus reproducing av. protonation probabilities at a certain pH. The accuracy of the method is tested against titrn. curves of single amino acids and a dipeptide in explicit solvent.
- 24Wallace, J. A.; Shen, J. K. Continuous Constant pH Molecular Dynamics in Explicit Solvent with pH-Based Replica Exchange. J. Chem. Theory Comput. 2011, 7, 2617– 2629, DOI: 10.1021/ct200146j24Continuous Constant pH Molecular Dynamics in Explicit Solvent with pH-Based Replica ExchangeWallace, Jason A.; Shen, Jana K.Journal of Chemical Theory and Computation (2011), 7 (8), 2617-2629CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A computational tool that offers accurate pKa values and atomically detailed knowledge of protonation-coupled conformational dynamics is valuable for elucidating mechanisms of energy transduction processes in biol., such as enzyme catalysis and electron transfer as well as proton and drug transport. Toward this goal the authors present a new technique of embedding continuous const. pH mol. dynamics within an explicit-solvent representation. In this technique the authors make use of the efficiency of the generalized-Born (GB) implicit-solvent model for estg. the free energy of protein solvation while propagating conformational dynamics using the more accurate explicit-solvent model. Also, the authors employ a pH-based replica exchange scheme to significantly enhance both protonation and conformational state sampling. Benchmark data of five proteins including HP36, NTL9, BBL, HEWL, and SNase yield an av. abs. deviation of 0.53 and a root mean squared deviation of 0.74 from exptl. data. This level of accuracy is obtained with 1 ns simulations per replica. Detailed anal. reveals that explicit-solvent sampling provides increased accuracy relative to the previous GB-based method by preserving the native structure, providing a more realistic description of conformational flexibility of the hydrophobic cluster, and correctly modeling solvent mediated ion-pair interactions. Thus, the authors anticipate that the new technique will emerge as a practical tool to capture ionization equil. while enabling an intimate view of ionization coupled conformational dynamics that is difficult to delineate with exptl. techniques alone.
- 25Wallace, J. A.; Shen, J. K. Charge-leveling and proper treatment of long-range electrostatics in all-atom molecular dynamics at constant pH. J. Chem. Phys. 2012, 137, 184105, DOI: 10.1063/1.476635225Charge-leveling and proper treatment of long-range electrostatics in all-atom molecular dynamics at constant pHWallace, Jason A.; Shen, Jana K.Journal of Chemical Physics (2012), 137 (18), 184105/1-184105/9CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Recent development of const. pH mol. dynamics (CpHMD) methods has offered promise for adding pH-stat in mol. dynamics simulations. However, until now the working pH mol. dynamics (pHMD) implementations are dependent in part or whole on implicit-solvent models. Here we show that proper treatment of long-range electrostatics and maintaining charge neutrality of the system are crit. for extending the continuous pHMD framework to the all-atom representation. The former is achieved here by adding forces to titrn. coordinates due to long-range electrostatics based on the generalized reaction field method, while the latter is made possible by a charge-leveling technique that couples proton titrn. with simultaneous ionization or neutralization of a co-ion in soln. We test the new method using the pH-replica-exchange CpHMD simulations of a series of aliph. dicarboxylic acids with varying carbon chain length. The av. abs. deviation from the exptl. pKa values is merely 0.18 units. The results show that accounting for the forces due to extended electrostatics removes the large random noise in propagating titrn. coordinates, while maintaining charge neutrality of the system improves the accuracy in the calcd. electrostatic interaction between ionizable sites. Thus, we believe that the way is paved for realizing pH-controlled all-atom mol. dynamics in the near future. (c) 2012 American Institute of Physics.
- 26Goh, G. B.; Knight, J. L.; Brooks, C. L. Constant pH Molecular Dynamics Simulations of Nucleic Acids in Explicit Solvent. J. Chem. Theory Comput. 2012, 8, 36– 46, DOI: 10.1021/ct200631426Constant pH Molecular Dynamics Simulations of Nucleic Acids in Explicit SolventGoh, Garrett B.; Knight, Jennifer L.; Brooks, Charles L.Journal of Chemical Theory and Computation (2012), 8 (1), 36-46CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)The nucleosides of adenine and cytosine have pKa values of 3.50 and 4.08, resp., and are assumed to be unprotonated under physiol. conditions. However, evidence from recent NMR and X-ray crystallog. studies has revealed the prevalence of protonated adenine and cytosine in RNA macromols. Such nucleotides with elevated pKa values may play a role in stabilizing RNA structure and participate in the mechanism of ribozyme catalysis. With the work presented here, we establish the framework and demonstrate the first const. pH MD simulations (CPHMD) for nucleic acids in explicit solvent, in which the protonation state is coupled to the dynamical evolution of the RNA system via λ-dynamics. We adopt the new functional form λNexp for λ that was recently developed for multisite λ-dynamics (MSλD) and demonstrate good sampling characteristics in which rapid and frequent transitions between the protonated and unprotonated states at pH = pKa are achieved. Our calcd. pKa values of simple nucleotides are in a good agreement with exptl. measured values, with a mean abs. error of 0.24 pKa units. This work demonstrates that CPHMD can be used as a powerful tool to investigate pH-dependent biol. properties of RNA macromols.
- 27Chen, W.; Wallace, J. A.; Yue, Z.; Shen, J. K. Introducing Titratable Water to All-Atom Molecular Dynamics at Constant pH. Biophys. J. 2013, 105, L15– L17, DOI: 10.1016/j.bpj.2013.06.03627Introducing Titratable Water to All-Atom Molecular Dynamics at Constant pHChen, Wei; Wallace, Jason A.; Yue, Zhi; Shen, Jana K.Biophysical Journal (2013), 105 (4), L15-L17CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Recent development of titratable coions has paved the way for realizing all-atom mol. dynamics at const. pH. To further improve phys. realism, here we describe a technique in which proton titrn. of the solute is directly coupled to the interconversion between water and hydroxide or hydronium. We test the new method in replica-exchange continuous const. pH mol. dynamics simulations of three proteins, HP36, BBL, and HEWL. The calcd. pKa values based on 10-ns sampling per replica have the av. abs. and root-mean-square errors of 0.7 and 0.9 pH units, resp. Introducing titratable water in mol. dynamics offers a means to model proton exchange between solute and solvent, thus opening a door to gaining new insights into the intricate details of biol. phenomena involving proton translocation.
- 28Lee, J.; Miller, B. T.; Damjanovic, A.; Brooks, B. R. Constant pH Molecular Dynamics in Explicit Solvent with Enveloping Distribution Sampling and Hamiltonian Exchange. J. Chem. Theory Comput. 2014, 10, 2738– 2750, DOI: 10.1021/ct500175m28Constant pH Molecular Dynamics in Explicit Solvent with Enveloping Distribution Sampling and Hamiltonian ExchangeLee, Juyong; Miller, Benjamin T.; Damjanovic, Ana; Brooks, Bernard R.Journal of Chemical Theory and Computation (2014), 10 (7), 2738-2750CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We present a new computational approach for const. pH simulations in explicit solvent based on the combination of the enveloping distribution sampling (EDS) and Hamiltonian replica exchange (HREX) methods. Unlike const. pH methods based on variable and continuous charge models, our method is based on discrete protonation states. EDS generates a hybrid Hamiltonian of different protonation states. A smoothness parameter s is used to control the heights of energy barriers of the hybrid-state energy landscape. A small s value facilitates state transitions by lowering energy barriers. Replica exchange between EDS potentials with different s values allows us to readily obtain a thermodynamically accurate ensemble of multiple protonation states with frequent state transitions. The anal. is performed with an ensemble obtained from an EDS Hamiltonian without smoothing, s = ∞, which strictly follows the min. energy surface of the end states. The accuracy and efficiency of this method is tested on aspartic acid, lysine, and glutamic acid, which have two protonation states, a histidine with three states, a four-residue peptide with four states, and snake cardiotoxin with eight states. The pKa values estd. with the EDS-HREX method agree well with the exptl. pKa values. The mean abs. errors of small benchmark systems range from 0.03 to 0.17 pKa units, and those of three titratable groups of snake cardiotoxin range from 0.2 to 1.6 pKa units. This study demonstrates that EDS-HREX is a potent theor. framework, which gives the correct description of multiple protonation states and good calcd. pKa values.
- 29Dobrev, P.; Donnini, S.; Groenhof, G.; Grubmüller, H. Accurate Three States Model for Amino Acids with Two Chemically Coupled Titrating Sites in Explicit Solvent Atomistic Constant pH Simulations and p K a Calculations. J. Chem. Theory Comput. 2017, 13, 147– 160, DOI: 10.1021/acs.jctc.6b0080729Accurate Three States Model for Amino Acids with Two Chemically Coupled Titrating Sites in Explicit Solvent Atomistic Constant pH Simulations and pKa CalculationsDobrev, Plamen; Donnini, Serena; Groenhof, Gerrit; Grubmueller, HelmutJournal of Chemical Theory and Computation (2017), 13 (1), 147-160CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Correct protonation of titratable groups in biomols. is crucial for their accurate description by mol. dynamics simulations. In the context of const. pH simulations, an addnl. protonation degree of freedom is introduced for each titratable site, allowing the protonation state to change dynamically with changing structure or electrostatics. Here, we implement a second reaction coordinate which switches between two tautomeric states of an amino acid with chem. coupled titratable sites, such as aspartate (Asp), glutamate (Glu), and histidine (His). To this aim, we test a scheme involving three protonation states. To facilitate charge neutrality as required for periodic boundary conditions and Particle Mesh Ewald (PME) electrostatics, titrn. of each resp. amino acid is coupled to a "water" mol. that is charged in opposite direction. Addnl., a force field modification for Amber99sb is introduced and tested for the description of carboxyl group protonation. Our three states model is tested by titrn. simulations of Asp, Glu, and His, yielding a good agreement, reproducing the correct geometry of the groups in their different protonation forms. We further show that the ion concn. change due to the neutralizing "water" mols. does not significantly affect the protonation free energies of the titratable groups, suggesting that the three states model provides a good description of biomol. dynamics at const. pH.
- 30Huang, Y.; Chen, W.; Wallace, J. A.; Shen, J. All-atom continuous constant pH molecular dynamics with particle mesh Ewald and titratable water. J. Chem. Theory Comput. 2016, 12, 5411– 5421, DOI: 10.1021/acs.jctc.6b0055230All-Atom Continuous Constant pH Molecular Dynamics With Particle Mesh Ewald and Titratable WaterHuang, Yandong; Chen, Wei; Wallace, Jason A.; Shen, JanaJournal of Chemical Theory and Computation (2016), 12 (11), 5411-5421CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Development of a pH stat to properly control soln. pH in biomol. simulations has been a long-standing goal in the community. Towards this goal recent years have witnessed the emergence of the so-called const. pH mol. dynamics methods. However, the accuracy and generality of these methods have been hampered by the use of implicit-solvent models or electrostatic truncation schemes. Here the authors report the implementation of the particle mesh Ewald (PME) scheme into the all-atom continuous const. pH mol. dynamics (CpHMD) method, enabling CpHMD to be performed with a std. MD engine at a fractional added computational cost. The authors demonstrate the performance using pH replica-exchange CpHMD simulations with titratable water for a stringent test set of proteins, HP38, BBL, HEWL and SNase. With the sampling time of 10 ns per replica, most pKa's are converged, yielding the av. abs. and root-mean-square deviations of 0.61 and 0.77, resp., from expt. Linear regression of the calcd. vs. exptl. pKa shifts gives a correlation coeff. of 0.79, a slope of 1 and an intercept near 0. Anal. reveals inadequate sampling of structure relaxation accompanying a protonation-state switch as a major source of the remaining errors, which are reduced as simulation prolongs. These data suggest PME-based CpHMD can be used as a general tool for pH-controlled simulations of macromol. systems in various environments, enabling at. insights into pH-dependent phenomena involving not only sol. proteins but also transmembrane proteins, nucleic acids, surfactants and polysaccharides.
- 31Huang, Y.; Harris, R. C.; Shen, J. Generalized Born based continuous constant pH molecular dynamics in Amber: Implementation, benchmarking and analysis. J. Chem. Inf. Model. 2018, 58, 1372– 1383, DOI: 10.1021/acs.jcim.8b0022731Generalized Born Based Continuous Constant pH Molecular Dynamics in Amber: Implementation, Benchmarking and AnalysisHuang, Yandong; Harris, Robert C.; Shen, JanaJournal of Chemical Information and Modeling (2018), 58 (7), 1372-1383CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Soln. pH plays an important role in structure and dynamics of biomol. systems; however, pH effects cannot be accurately accounted for in conventional mol. dynamics simulations based on fixed protonation states. Continuous const. pH mol. dynamics (CpHMD) based on the λ-dynamics framework calcs. protonation states on the fly during dynamical simulation at a specified pH condition. Here the authors report the CPU-based implementation of the CpHMD method based on the GBNeck2 generalized Born (GB) implicit-solvent model in the pmemd engine of the Amber mol. dynamics package. The performance of the method was tested using pH replica-exchange titrn. simulations of Asp, Glu and His side chains in 4 miniproteins and 7 enzymes with exptl. known pKa's, some of which are significantly shifted from the model values. The added computational cost due to CpHMD titrn. ranges from 11 to 33% for the data set and scales roughly linearly as the ratio between the titrable sites and no. of solute atoms. Comparison of the exptl. and calcd. pKa's using 2 ns per replica sampling yielded a mean unsigned error of 0.70, a root-mean-squared error of 0.91, and a linear correlation coeff. of 0.79. Though this level of accuracy is similar to the GBSW-based CpHMD in CHARMM, in contrast to the latter, the current implementation was able to reproduce the exptl. orders of the pKa's of the coupled carboxylic dyads. The authors quantified the sampling errors, which revealed that prolonged simulation is needed to converge pKa's of several titratable groups involved in salt-bridge-like interactions or deeply buried in the protein interior. The authors' benchmark data demonstrate that GBNeck2-CpHMD is an attractive tool for protein pKa predictions.
- 32Harris, R. C.; Shen, J. GPU-accelerated implementation of continuous constant pH molecular dynamics in amber: pKa predictions with single-pH simulations. J. Chem. Inf. Model. 2019, 59, 4821– 4832, DOI: 10.1021/acs.jcim.9b0075432GPU-Accelerated Implementation of Continuous Constant pH Molecular Dynamics in Amber: pKa Predictions with Single-pH SimulationsHarris, Robert C.; Shen, JanaJournal of Chemical Information and Modeling (2019), 59 (11), 4821-4832CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The authors present a GPU implementation of the continuous const. pH mol. dynamics (CpHMD) based on the most recent generalized Born implicit-solvent model in the pmemd engine of the Amber mol. dynamics package. To test the accuracy of the tool for rapid pKa predictions, a series of 2-ns single-pH simulations were performed for over 120 titratable residues in 10 benchmark proteins that were previously used to test the various continuous CpHMD methods. The calcd. pKa's showed a root-mean-square deviation of 0.80 and correlation coeff. of 0.83 with respect to expt. 90% of the pKa's were converged with estd. errors <0.1 pH units. Surprisingly, this level of accuracy is similar to the authors' previous replica-exchange simulations with 2 ns per replica and an exchange attempt frequency of 2 ps-1 (Huang, Harris and Shen, J Chem Info Model, 2018). For the linked titrn. sites in two enzymes, although residue-specific protonation state sampling in the single-pH simulations was not converged within 2 ns, the protonation fraction of the linked residues appeared to be largely converged, and the exptl. macroscopic {\pka} values were reproduced to within 1 pH unit. Comparison with replica-exchange simulations with different exchange attempt frequencies showed that the splitting between the two macroscopic pKa's is underestimated with frequent exchange attempts such as 2 ps$̂{-1}$, while single-pH simulations overestimate the splitting. The same trend is seen for the single-pH vs. replica-exchange simulations of a hydrogen-bonded aspartyl dyad in a much larger protein. A 2-ns single-pH simulation of a 400-residue protein takes about one hour on a single NVIDIA GeForce RTX 2080 graphics card, which is over 1000 times faster than a CpHMD run on a single CPU core of a high-performance computing cluster node. Thus, the authors envision that GPU-accelerated continuous CpHMD may be used in routine pKa predictions for a variety of applications, from assisting MD simulations with protonation state assignment to offering pH-dependent corrections of binding free energies and identifying reactive hot spots for covalent drug design.
- 33Harris, R. C.; Liu, R.; Shen, J. Predicting reactive cysteines with implicit-solvent-based continuous constant pH molecular dynamics in amber. J. Chem. Theory Comput. 2020, 16, 3689– 3698, DOI: 10.1021/acs.jctc.0c0025833Predicting Reactive Cysteines with Implicit-Solvent-Based Continuous Constant pH Molecular Dynamics in AmberHarris, Robert C.; Liu, Ruibin; Shen, JanaJournal of Chemical Theory and Computation (2020), 16 (6), 3689-3698CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Cysteines existing in the deprotonated thiolate form or having a tendency to become deprotonated are important players in enzymic and cellular redox functions and frequently exploited in covalent drug design; however, most computational studies assume cysteines as protonated. Thus, developing an efficient tool that can make accurate and reliable predictions of cysteine protonation states is timely needed. The authors recently implemented a generalized Born (GB) based continuous const. pH mol. dynamics (CpHMD) method in Amber for protein pKa calcns. on CPUs and GPUs. Here the authors benchmark the performance of GB-CpHMD for predictions of cysteine pKa's and reactivities using a data set of 24 proteins with both down- and upshifted cysteine pKa's. The authors found that 10 ns single-pH or 4 ns replica-exchange CpHMD titrns. gave root-mean-square errors of 1.2-1.3 and correlation coeffs. of 0.8-0.9 with respect to expt. The accuracy of predicting thiolates or reactive cysteines at physiol. pH with single-pH titrns. is 86 or 81% with a precision of 100 or 90%, resp. This performance well surpasses the traditional structure-based methods, particularly a widely used empirical pKa tool that gives an accuracy less than 50%. The authors discuss simulation convergence, dependence on starting structures, common determinants of the pKa downshifts and upshifts, and the origin of the discrepancies from the structure-based calcns. The work suggests that CpHMD titrns. can be performed on a desktop computer equipped with a single GPU card to predict cysteine protonation states for a variety of applications, from understanding biol. functions to covalent drug design.
- 34Grünewald, F.; Souza, P. C.; Abdizadeh, H.; Barnoud, J.; de Vries, A. H.; Marrink, S. J. Titratable Martini model for constant pH simulations. J. Chem. Phys. 2020, 153, 024118, DOI: 10.1063/5.001425834Titratable Martini model for constant pH simulationsGruenewald, Fabian; Souza, Paulo C. T.; Abdizadeh, Haleh; Barnoud, Jonathan; de Vries, Alex H.; Marrink, Siewert J.Journal of Chemical Physics (2020), 153 (2), 024118CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The authors deliver a proof of concept for a fast method that introduces pH effects into classical coarse-grained (CG) mol. dynamics simulations. The authors' approach is based upon the latest version of the popular Martini CG model to which explicit proton mimicking particles were added. The authors verify the authors' approach against exptl. data involving several different mols. and different environmental conditions. In particular, the authors compute titrn. curves, pH dependent free energies of transfer, and lipid bilayer membrane affinities as a function of pH. Using oleic acid as an example compd., the authors further illustrate that the authors' method can be used to study passive translocation in lipid bilayers via protonation. Finally, the authors' model reproduces qual. the expansion of the macromol. dendrimer poly(propylene imine) as well as the assocd. pKa shift of its different generations. This example demonstrates that the authors' model is able to pick up collective interactions between titratable sites in large mols. comprising many titratable functional groups. (c) 2020 American Institute of Physics.
- 35Mongan, J.; Case, D. A. Biomolecular simulations at constant pH. Curr. Opin. Struct. Biol. 2005, 15, 157– 163, DOI: 10.1016/j.sbi.2005.02.00235Biomolecular simulations at constant pHMongan, John; Case, David A.Current Opinion in Structural Biology (2005), 15 (2), 157-163CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)A review. Like temp. and pressure, the soln. pH is an important thermodn. variable that is commonly varied in expts. and is used by cells to influence biochem. function. It is now becoming feasible to carry out practical mol. dynamics simulations that mimic the thermodn. of such expts., by allowing proton transfer between the system of interest and a hypothetical bath of protons at a given pH. These are demanding calcns., because the energetics of charge changes upon protonation or deprotonation must be accurately modeled, and because such simulations must sample both mol. configurations and the large no. of protonation states that are possible for a mol. with many titrating sites.
- 36Damjanovic, A.; Miller, B. T.; Okur, A.; Brooks, B. R. Reservoir pH replica exchange. J. Chem. Phys. 2018, 149, 072321, DOI: 10.1063/1.502741336Reservoir pH replica exchangeDamjanovic, Ana; Miller, Benjamin T.; Okur, Asim; Brooks, Bernard R.Journal of Chemical Physics (2018), 149 (7), 072321/1-072321/12CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)We present the reservoir pH replica exchange (R-pH-REM) method for const. pH simulations. The R-pH-REM method consists of a two-step procedure; the first step involves generation of one or more reservoirs of conformations. Each reservoir is obtained from a std. or enhanced mol. dynamics simulation with a constrained (fixed) protonation state. In the second step, fixed charge constraints are relaxed, as the structures from one or more reservoirs are periodically injected into a const. pH or a pH-replica exchange (pH-REM) simulation. The benefit of this two-step process is that the computationally intensive part of conformational search can be decoupled from const. pH simulations, and various techniques for enhanced conformational sampling can be applied without the need to integrate such techniques into the pH-REM framework. Simulations on blocked Lys, KK, and KAAE peptides were used to demonstrate an agreement between pH-REM and R-pH-REM simulations. While the reservoir simulations are not needed for these small test systems, the real need arises in cases when ionizable mols. can sample two or more conformations sepd. by a large energy barrier, such that adequate sampling is not achieved on a time scale of std. const. pH simulations. Such problems might be encountered in protein systems that exploit conformational transitions for function. A hypothetical case is studied, a small mol. with a large torsional barrier; while results of pH-REM simulations depend on the starting structure, R-pH-REM calcns. on this model system are in excellent agreement with a theor. model. (c) 2018 American Institute of Physics.
- 37Chen, Y.; Roux, B. Constant-pH Hybrid Nonequilibrium Molecular Dynamics - Monte Carlo Simulation Method. J. Chem. Theory Comput. 2015, 11, 3919– 3931, DOI: 10.1021/acs.jctc.5b0026137Constant-pH Hybrid Nonequilibrium Molecular Dynamics-Monte Carlo Simulation MethodChen, Yunjie; Roux, BenoitJournal of Chemical Theory and Computation (2015), 11 (8), 3919-3931CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)A computational method is developed to carry out explicit solvent simulations of complex mol. systems under conditions of const. pH. In const.-pH simulations, preidentified ionizable sites are allowed to spontaneously protonate and deprotonate as a function of time in response to the environment and the imposed pH. The method consists of carrying out short nonequil. mol. dynamics (neMD) switching trajectories to generate phys. plausible configurations with changed protonation states that are subsequently accepted or rejected according to a Metropolis Monte Carlo (MC) criterion. To ensure microscopic detailed balance arising from such nonequil. switches, the at. momenta are altered according to the sym. two-ends momentum reversal prescription. To achieve higher efficiency, the original neMD-MC scheme is sepd. into two steps, reducing the need for generating a large no. of unproductive and costly nonequil. trajectories. In the first step, the protonation state of a site is randomly attributed via a Metropolis MC process on the basis of an intrinsic pKa; an attempted nonequil. switch is generated only if this change in protonation state is accepted. This hybrid two-step inherent pKa neMD-MC simulation method is tested with single amino acids in soln. (Asp, Glu, and His) and then applied to turkey ovomucoid third domain and hen egg-white lysozyme. Because of the simple linear increase in the computational cost relative to the no. of titratable sites, the present method is naturally able to treat extremely large systems.
- 38Kong, X.; Brooks, C. L., III λ-dynamics: A new approach to free energy calculations. J. Chem. Phys. 1996, 105, 2414– 2423, DOI: 10.1063/1.472109There is no corresponding record for this reference.
- 39Simonson, T.; Carlsson, J.; Case, D. A. Proton binding to proteins: p K a calculations with explicit and implicit solvent models. J. Am. Chem. Soc. 2004, 126, 4167– 4180, DOI: 10.1021/ja039788m39Proton Binding to Proteins: pKa Calculations with Explicit and Implicit Solvent ModelsSimonson, Thomas; Carlsson, Jens; Case, David A.Journal of the American Chemical Society (2004), 126 (13), 4167-4180CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Ionizable residues play important roles in protein structure and activity, and proton binding is a valuable reporter of electrostatic interactions in these systems. The authors use mol. dynamics free energy simulations (MDFE) to compute proton pKa shifts, relative to a model compd. in soln., for three aspartate side chains in two proteins. Simulations with explicit solvent and with an implicit, dielec. continuum solvent are reported. The implicit solvent simulations use the generalized Born (GB) model, which provides an approx., anal. soln. to Poisson's equation. With explicit solvent, the direction of the pKa shifts is correct in all three cases with one force field (AMBER) and in two out of three cases with another (CHARMM). For two aspartates, the dielec. response to ionization is found to be linear, even though the sep. protein and solvent responses can be nonlinear. For thioredoxin Asp-26, nonlinearity arises from the presence of two substates that correspond to the two possible orientations of the protonated carboxylate. For this side chain, which is partly buried and has a large pKa upshift, very long simulations are needed to correctly sample several slow degrees of freedom that reorganize in response to the ionization. Thus, nearby Lys-57 rotates to form a salt bridge and becomes buried, while three waters intercalate along the opposite edge of Asp-26. Such strong and anisotropic reorganization is very difficult to predict with Poisson-Boltzmann methods that only consider electrostatic interactions and employ a single protein structure. In contrast, MDFE with a GB dielec. continuum solvent, used for the first time for pKa calcns., can describe protein reorganization accurately and gives encouraging agreement with expt. and with the explicit solvent simulations.
- 40Chen, W.; Shen, J. K. Effects of System Net Charge and Electrostic Truncation on All-Atom Constant pH Molecular Dynamics. J. Comput. Chem. 2014, 35, 1986– 1996, DOI: 10.1002/jcc.2371340Effects of system net charge and electrostatic truncation on all-atom constant pH molecular dynamicsChen, Wei; Shen, Jana K.Journal of Computational Chemistry (2014), 35 (27), 1986-1996CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Const. pH mol. dynamics offers a means to rigorously study the effects of soln. pH on dynamical processes. Here, we address two crit. questions arising from the most recent developments of the all-atom continuous const. pH mol. dynamics (CpHMD) method: (1) What is the effect of spatial electrostatic truncation on the sampling of protonation states. (2) Is the enforcement of elec. neutrality necessary for const. pH simulations. We first examd. how the generalized reaction field and force-shifting schemes modify the electrostatic forces on the titrn. coordinates. Free energy simulations of model compds. were then carried out to delineate the errors in the deprotonation free energy and salt-bridge stability due to electrostatic truncation and system net charge. Finally, CpHMD titrn. of a mini-protein HP36 was used to understand the manifestation of the two types of errors in the calcd. pKa values. The major finding is that enforcing charge neutrality under all pH conditions and at all time via co-titrating ions significantly improves the accuracy of protonation-state sampling. We suggest that such finding is also relevant for simulations with particle mesh Ewald, considering the known artifacts due to charge-compensating background plasma.
- 41Marrink, S. J.; Risselada, H. J.; Yefimov, S.; Tieleman, D. P.; De Vries, A. H. The MARTINI force field: coarse grained model for biomolecular simulations. J. Phys. Chem. B 2007, 111, 7812– 7824, DOI: 10.1021/jp071097f41The MARTINI Force Field: Coarse Grained Model for Biomolecular SimulationsMarrink, Siewert J.; Risselada, H. Jelger; Yefimov, Serge; Tieleman, D. Peter; De Vries, Alex H.Journal of Physical Chemistry B (2007), 111 (27), 7812-7824CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)We present an improved and extended version of our coarse grained lipid model. The new version, coined the MARTINI force field, is parametrized in a systematic way, based on the reprodn. of partitioning free energies between polar and apolar phases of a large no. of chem. compds. To reproduce the free energies of these chem. building blocks, the no. of possible interaction levels of the coarse-grained sites has increased compared to those of the previous model. Application of the new model to lipid bilayers shows an improved behavior in terms of the stress profile across the bilayer and the tendency to form pores. An extension of the force field now also allows the simulation of planar (ring) compds., including sterols. Application to a bilayer/cholesterol system at various concns. shows the typical cholesterol condensation effect similar to that obsd. in all atom representations.
- 42Donnini, S.; Ullmann, R. T.; Groenhof, G.; Grubmüller, H. Charge-neutral constant pH molecular dynamics simulations using a parsimonious proton buffer. J. Chem. Theory Comput. 2016, 12, 1040– 1051, DOI: 10.1021/acs.jctc.5b0116042Charge-Neutral Constant pH Molecular Dynamics Simulations Using a Parsimonious Proton BufferDonnini, Serena; Ullmann, R. Thomas; Groenhof, Gerrit; Grubmuller, HelmutJournal of Chemical Theory and Computation (2016), 12 (3), 1040-1051CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)In const. pH mol. dynamics simulations, the protonation states of titratable sites can respond to changes of the pH and of their electrostatic environment. Consequently, the no. of protons bound to the biomol., and therefore the overall charge of the system, fluctuates during the simulation. To avoid artifacts assocd. with a non-neutral simulation system, we introduce an approach to maintain neutrality of the simulation box in const. pH mol. dynamics simulations, while maintaining an accurate description of all protonation fluctuations. Specifically, we introduce a proton buffer that, like a buffer in expt., can exchange protons with the biomol. enabling its charge to fluctuate. To keep the total charge of the system const., the uptake and release of protons by the buffer are coupled to the titrn. of the biomol. with a constraint. We find that, because the fluctuation of the total charge (no. of protons) of a typical biomol. is much smaller than the no. of titratable sites of the biomol., the no. of buffer sites required to maintain overall charge neutrality without compromising the charge fluctuations of the biomol., is typically much smaller than the no. of titratable sites, implying markedly enhanced simulation and sampling efficiency.
- 43Dobrev, P.; Vemulapalli, S. P. B.; Nath, N.; Griesinger, C.; Grubmüller, H. Probing the accuracy of explicit solvent constant pH molecular dynamics simulations for peptides. J. Chem. Theory Comput. 2020, 16, 2561– 2569, DOI: 10.1021/acs.jctc.9b0123243Probing the Accuracy of Explicit Solvent Constant pH Molecular Dynamics Simulations for PeptidesDobrev, Plamen; Vemulapalli, Sahithya Phani Babu; Nath, Nilamoni; Griesinger, Christian; Grubmueller, HelmutJournal of Chemical Theory and Computation (2020), 16 (4), 2561-2569CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Protonation states of titratable amino acids play a key role in many biomol. processes. Knowledge of protonatable residue charges at a given pH is essential for a correct understanding of protein catalysis, inter- and intramol. interactions, substrate binding, and protein dynamics for instance. However, acquiring exptl. values for individual amino acid protonation states of complex systems is not straightforward; therefore, several in silico approaches have been developed to tackle this issue. In this work, the authors assess the accuracy of the previously developed const. pH MD approach by comparing the theor. obtained pKa values for titratable residues with exptl. values from an equiv. NMR study. The authors selected a set of four pentapeptides, of adequately small size to ensure comprehensive sampling, but concurrently, due to their charge compn., posing a challenge for protonation state calcn. The comparison of the pKa values shows good agreement of the exptl. and the theor. approach with a largest difference of 0.25 pKa units. Further, the corresponding titrn. curves are in fair agreement, although the shift of the Hill coeff. from a value of 1 was not always reproduced in simulations. The phase space overlap in Cartesian space between trajectories generated in const. pH and std. MD simulations is fair and suggests that the const. pH MD approach reasonably well preserves the dynamics of the system, allowing dynamic protonation MD simulations without introducing structural artifacts.
- 44Darden, T.; York, D.; Pedersen, L. Particle mesh Ewald: An Nlog(N) method for Ewald sums in large systems. J. Chem. Phys. 1993, 98, 10089– 10092, DOI: 10.1063/1.46439744Particle mesh Ewald: an N·log(N) method for Ewald sums in large systemsDarden, Tom; York, Darrin; Pedersen, LeeJournal of Chemical Physics (1993), 98 (12), 10089-92CODEN: JCPSA6; ISSN:0021-9606.An N·log(N) method for evaluating electrostatic energies and forces of large periodic systems is presented. The method is based on interpolation of the reciprocal space Ewald sums and evaluation of the resulting convolution using fast Fourier transforms. Timings and accuracies are presented for three large cryst. ionic systems.
- 45Essmann, U.; Perera, L.; Berkowitz, M. L.; Darden, T.; Lee, H.; Pedersen, L. G. A smooth particle mesh Ewald method. J. Chem. Phys. 1995, 103, 8577– 8593, DOI: 10.1063/1.47011745A smooth particle mesh Ewald methodEssmann, Ulrich; Perera, Lalith; Berkowitz, Max L.; Darden, Tom; Lee, Hsing; Pedersen, Lee G.Journal of Chemical Physics (1995), 103 (19), 8577-93CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The previously developed particle mesh Ewald method is reformulated in terms of efficient B-spline interpolation of the structure factors. This reformulation allows a natural extension of the method to potentials of the form 1/rp with p ≥ 1. Furthermore, efficient calcn. of the virial tensor follows. Use of B-splines in the place of Lagrange interpolation leads to analytic gradients as well as a significant improvement in the accuracy. The authors demonstrate that arbitrary accuracy can be achieved, independent of system size N, at a cost that scales as N log(N). For biomol. systems with many thousands of atoms and this method permits the use of Ewald summation at a computational cost comparable to that of a simple truncation method of 10 Å or less.
- 46Knight, J. L.; Brooks, C. L., III Multisite λ dynamics for simulated structure-activity relationship studies. J. Chem. Theory Comput. 2011, 7, 2728– 2739, DOI: 10.1021/ct200444f46Multisite λ Dynamics for Simulated Structure-Activity Relationship StudiesKnight, Jennifer L.; Brooks, Charles L., IIIJournal of Chemical Theory and Computation (2011), 7 (9), 2728-2739CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Multisite λ dynamics (MSλD) is a new free energy simulation method that is based on λ dynamics. It has been developed to enable multiple substituents at multiple sites on a common ligand core to be modeled simultaneously and their free energies assessed. The efficacy of MSλD for estg. relative hydration free energies and relative binding affinties is demonstrated using three test systems. Model compds. representing multiple identical benzene, dihydroxybenzene, and dimethoxybenzene mols. show that total combined MSλD trajectory lengths of ∼1.5 ns are sufficient to reliably achieve relative hydration free energy ests. within 0.2 kcal/mol and are less sensitive to the no. of trajectories that are used to generate these ests. for hybrid ligands that contain up to 10 substituents modeled at a single site or five substituents modeled at each of two sites. Relative hydration free energies among six benzene derivs. calcd. from MSλD simulations are in very good agreement with those from alchem. free energy simulations (with av. unsigned differences of 0.23 kcal/mol and R2 = 0.991) and the expt. (with av. unsigned errors of 1.8 kcal/mol and R2 = 0.959). Ests. of the relative binding affinities among 14 inhibitors of HIV-1 reverse transcriptase obtained from MSλD simulations are in reasonable agreement with those from traditional free energy simulations and the expt. (av. unsigned errors of 0.9 kcal/mol and R2 = 0.402). For the same level of accuracy and precision, MSλD simulations are achieved ∼20-50 times faster than traditional free energy simulations and thus with reliable force field parameters can be used effectively to screen tens to hundreds of compds. in structure-based drug design applications.
- 47Goh, G. B.; Hulbert, B. S.; Zhou, H.; Brooks, C. L., III Constant pH molecular dynamics of proteins in explicit solvent with proton tautomerism. Proteins: Struct., Funct., Bioinf. 2014, 82, 1319– 1331, DOI: 10.1002/prot.2449947Constant pH molecular dynamics of proteins in explicit solvent with proton tautomerismGoh, Garrett B.; Hulbert, Benjamin S.; Zhou, Huiqing; Brooks, Charles L., IIIProteins: Structure, Function, and Bioinformatics (2014), 82 (7), 1319-1331CODEN: PSFBAF ISSN:. (Wiley-Blackwell)PH is a ubiquitous regulator of biol. activity, including protein-folding, protein-protein interactions, and enzymic activity. Existing const. pH mol. dynamics (CPHMD) models that were developed to address questions related to the pH-dependent properties of proteins are largely based on implicit solvent models. However, implicit solvent models are known to underestimate the desolvation energy of buried charged residues, increasing the error assocd. with predictions that involve internal ionizable residue that are important in processes like hydrogen transport and electron transfer. Furthermore, discrete water and ions cannot be modeled in implicit solvent, which are important in systems like membrane proteins and ion channels. We report on an explicit solvent const. pH mol. dynamics framework based on multi-site λ-dynamics (CPHMDMSλD). In the CPHMDMSλD framework, we performed seamless alchem. transitions between protonation and tautomeric states using multi-site λ-dynamics, and designed novel biasing potentials to ensure that the phys. end-states are predominantly sampled. We show that explicit solvent CPHMDMSλD simulations model realistic pH-dependent properties of proteins such as the Hen-Egg White Lysozyme (HEWL), binding domain of 2-oxoglutarate dehydrogenase (BBL) and N-terminal domain of ribosomal protein L9 (NTL9), and the pKa predictions are in excellent agreement with exptl. values, with a RMSE ranging from 0.72 to 0.84 pKa units. With the recent development of the explicit solvent CPHMDMSλD framework for nucleic acids, accurate modeling of pH-dependent properties of both major class of biomols.-proteins and nucleic acids is now possible.
- 48Knight, J. L.; Brooks, C. L., III Applying efficient implicit nongeometric constraints in alchemical free energy simulations. Journal of computational chemistry 2011, 32, 3423– 3432, DOI: 10.1002/jcc.2192148Applying efficient implicit nongeometric constraints in alchemical free energy simulationsKnight, Jennifer L.; Brooks, Charles L.Journal of Computational Chemistry (2011), 32 (16), 3423-3432CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Several strategies have been developed for satisfying bond lengths, angle, and other geometric constraints in mol. dynamics simulations. Advanced variations of alchem. free energy perturbation simulations, however, also require nongeometric constraints. In our recently developed multisite λ-dynamics simulation method, the conventional λ parameters that are assocd. with the progress variables in alchem. transformations are treated as dynamic variables and are constrained such that: 0 ≤ λi ≤ 1 and .sum.i λi = 1. Here, we present four functional forms of λ that implicitly satisfy these nongeometric constraints, whose values and forces are facile to compute and that yield stable simulations using a 2 fs integration timestep. Using model systems, we present the sampling characteristics of these functional forms and demonstrate the enhanced sampling profiles and improved convergence rates that are achieved by a functional form of λi that oscillates between λi = 0 and λi = 1 and has relatively steep transitions between these endpoints. Copyright for JCC Journal: © 2011 Wiley Periodicals, Inc.; J. Comput. Chem., 2011.
- 49Abraham, M. J.; Murtola, T.; Schulz, R.; Páll, S.; Smith, J. C.; Hess, B.; Lindahl, E. GROMACS: High performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 2015, 1, 19– 25, DOI: 10.1016/j.softx.2015.06.001There is no corresponding record for this reference.
- 50Singhal, A. K.; Chien, K. Y.; Wu, W. G.; Rule, G. S. Solution structure of cardiotoxin V from Naja naja atra. Biochemistry 1993, 32, 8036– 8044, DOI: 10.1021/bi00082a02650Solution structure of cardiotoxin V from Naja naja atraSinghal, Arun K.; Chien, Kun Yi; Wu, Wen Guey; Rule, Gordon S.Biochemistry (1993), 32 (31), 8036-44CODEN: BICHAW; ISSN:0006-2960.To det. whether the unique activity of this cardiotoxin (CTX) is attributable to its tertiary structure, the soln. structure of CTX V was detd. by NMR methods. On the basis of these studies, this cardiotoxin has the same general topol. as other members of the family, and thus its unusual properties do not arise from any gross structural differences that are detectable by soln. NMR methods. Mol. dynamics calcns. indicate that residues 36-50 show concerted fluctuations. On the basis of sequence similarity, the authors postulate that residues 30-34 are important in detg. the specificity of cardiotoxins for fusion vs. lysis of vesicles.
- 51Ramanadham, M.; Sieker, L.; Jensen, L. Refinement of triclinic lysozyme: II. The method of stereochemically restrained least squares. Acta Crystallographica Section B: Structural Science 1990, 46, 63– 69, DOI: 10.1107/S010876818900919551Refinement of triclinic lysozyme: II. The method of stereochemically restrained least squaresRamanadham M; Sieker L C; Jensen L HActa crystallographica. Section B, Structural science (1990), 46 ( Pt 1) (), 63-9 ISSN:0108-7681.Refinement of triclinic lysozyme by restrained least squares against the 2 A resolution X-ray data is described, beginning with the model from cycle 17 of the preceding paper [Hodsdon, Brown, Sieker & Jensen (1990). Acta Cryst. B46, 54-62]. After 20 refinement cycles, R stood at 0.172. Nevertheless, serious errors involving both main-chain and side-chain atoms still remained, requiring numerous model rebuilding sessions interleaved with refinement cycles. After 63 cycles R = 0.124 for the model which includes all protein atoms, 249 water oxygen sites and five nitrate ions. Although the overall B is relatively low, 10.5 A2, B's for atoms in the region of residues 101-103, toward the termini of some of the longer side chains, and in the region of the C terminus of the main chain exceed 20 A2, indicating relatively high atomic mobilities, disorder, or remaining errors in the model.
- 52Sauguet, L.; Poitevin, F.; Murail, S.; Van Renterghem, C.; Moraga-Cid, G.; Malherbe, L.; Thompson, A. W.; Koehl, P.; Corringer, P.-J.; Baaden, M.; Delarue, M. Structural basis for ion permeation mechanism in pentameric ligand-gated ion channels. EMBO journal 2013, 32, 728– 741, DOI: 10.1038/emboj.2013.1752Structural basis for ion permeation mechanism in pentameric ligand-gated ion channelsSauguet, Ludovic; Poitevin, Frederic; Murail, Samuel; Van Renterghem, Catherine; Moraga-Cid, Gustavo; Malherbe, Laurie; Thompson, Andrew W.; Koehl, Patrice; Corringer, Pierre-Jean; Baaden, Marc; Delarue, MarcEMBO Journal (2013), 32 (5), 728-741CODEN: EMJODG; ISSN:0261-4189. (Nature Publishing Group)To understand the mol. mechanism of ion permeation in pentameric ligand-gated ion channels (pLGIC), the structure of an open form of GLIC, a prokaryotic pLGIC, was solved at 2.4 Å. Anomalous diffraction data were used to place bound anions and cations. This reveals ordered water mols. at the level of two rings of hydroxylated residues (named Ser6' and Thr2') that contribute to the ion selectivity filter. Two water pentagons are obsd., a self-stabilized ice-like water pentagon and a second wider water pentagon, with one sodium ion between them. Single-channel electrophysiol. shows that the side-chain hydroxyl of Ser6' is crucial for ion translocation. Simulations and electrostatics calcns. complemented the description of hydration in the pore and suggest that the water pentagons obsd. in the crystal are important for the ion to cross hydrophobic constriction barriers. Simulations that pull a cation through the pore reveal that residue Ser6' actively contributes to ion translocation by reorienting its side chain when the ion is going through the pore. Generalization of these findings to the pLGIC family is proposed.
- 53Lee, T.-W.; Qasim, M., Jr; Laskowski, M., Jr; James, M. N. Structural insights into the non-additivity effects in the sequence-to-reactivity algorithm for serine peptidases and their inhibitors. Journal of molecular biology 2007, 367, 527– 546, DOI: 10.1016/j.jmb.2007.01.00853Structural Insights into the Non-additivity Effects in the Sequence-to-Reactivity Algorithm for Serine Peptidases and their InhibitorsLee, Ting-Wai; Qasim, M. A.; Laskowski, Michael; James, Michael N. G.Journal of Molecular Biology (2007), 367 (2), 527-546CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)Sequence-to-reactivity algorithms (SRAs) for proteins have the potential of being broadly applied in mol. design. Recently, Laskowski et al. have reported an additivity-based SRA that accurately predicts most of the std. free energy changes of assocn. for variants of turkey ovomucoid third domain (OMTKY3) with six serine peptidases, one of which is streptogrisin B (commonly known as Streptomyces griseus peptidase B, SGPB). Non-additivity effects for residues 18I and 32I, and for residues 20I and 32I of OMTKY3 occurred when the assocns. with SGPB were predicted using the SRA. To elucidate precisely the mechanics of these non-additivity effects in structural terms, we have detd. the crystal structures of the unbound OMTKY3 (with Gly32I as in the wild-type amino acid sequence) at a resoln. of 1.16 Å, the unbound Ala32I variant of OMTKY3 at a resoln. of 1.23 Å, and the SGPB:OMTKY3-Ala32I complex (equil. assocn. const. Ka = 7.1×109 M-1 at 21(±2) C°, pH 8.3) at a resoln. of 1.70 Å. Extensive comparisons with the crystal structure of the unbound OMTKY3 confirm our understanding of some previously addressed non-additivity effects. Unexpectedly, SGPB and OMTKY3-Ala32I form a 1:2 complex in the crystal. Comparison with the SGPB:OMTKY3 complex shows a conformational change in the SGPB:OMTKY3-Ala32I complex, resulting from a hinged rigid-body rotation of the inhibitor caused by the steric hindrance between the Me group of Ala32IA of the inhibitor and Pro192BE of the peptidase. This perturbs the interactions among residues 18I, 20I, 32I and 36I of the inhibitor, probably resulting in the above non-additivity effects. This conformational change also introduces residue 10I as an addnl. hyper-variable contact residue to the SRA.
- 54Huang, J.; MacKerell, A. D., Jr CHARMM36 all-atom additive protein force field: Validation based on comparison to NMR data. Journal of computational chemistry 2013, 34, 2135– 2145, DOI: 10.1002/jcc.2335454CHARMM36 all-atom additive protein force field: Validation based on comparison to NMR dataHuang, Jing; MacKerell, Alexander D. JrJournal of Computational Chemistry (2013), 34 (25), 2135-2145CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)Protein structure and dynamics can be characterized on the atomistic level with both NMR expts. and mol. dynamics (MD) simulations. Here, the authors quantify the ability of the recently presented CHARMM36 (C36) force field (FF) to reproduce various NMR observables using MD simulations. The studied NMR properties include backbone scalar couplings across hydrogen bonds, residual dipolar couplings (RDCs) and relaxation order parameter, as well as scalar couplings, RDCs, and order parameters for side-chain amino- and methyl-contg. groups. The C36 FF leads to better correlation with exptl. data compared to the CHARMM22/CMAP FF and suggest using C36 in protein simulations. Although both CHARMM FFs contains the same nonbond parameters, the authors' results show how the changes in the internal parameters assocd. with the peptide backbone via CMAP and the χ1 and χ2 dihedral parameters leads to improved treatment of the analyzed nonbond interactions. This highlights the importance of proper treatment of the internal covalent components in modeling nonbond interactions with mol. mechanics FFs.
- 55Buslaev, P.; Aho, N.; Jansen, A.; Bauer, P.; Hess, B.; Groenhof, G. Best practices in constant pH MD simulations: accuracy and precision. J. Chem. Theory Comput. 2022, DOI: 10.1021/acs.jctc.2c00517 .There is no corresponding record for this reference.
- 56Martini-website, General purpose coarse-grained force field. http://cgmartini.nl/index.php/tools2/proteins-and-bilayers/204-martinize (accessed October 01, 2021).There is no corresponding record for this reference.
- 57Kaminski, G. A.; Friesner, R. A.; Tirado-Rives, J.; Jorgensen, W. L. Evaluation and reparametrization of the OPLS-AA force field for proteins via comparison with accurate quantum chemical calculations on peptides. J. Phys. Chem. B 2001, 105, 6474– 6487, DOI: 10.1021/jp003919d57Evaluation and Reparametrization of the OPLS-AA Force Field for Proteins via Comparison with Accurate Quantum Chemical Calculations on PeptidesKaminski, George A.; Friesner, Richard A.; Tirado-Rives, Julian; Jorgensen, William L.Journal of Physical Chemistry B (2001), 105 (28), 6474-6487CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)We present results of improving the OPLS-AA force field for peptides by means of refitting the key Fourier torsional coeffs. The fitting technique combines using accurate ab initio data as the target, choosing an efficient fitting subspace of the whole potential-energy surface, and detg. wts. for each of the fitting points based on magnitudes of the potential-energy gradient. The av. energy RMS deviation from the LMP2/cc-pVTZ(-f)//HF/6-31G** data is reduced by ∼40% from 0.81 to 0.47 kcal/mol as a result of the fitting for the electrostatically uncharged dipeptides. Transferability of the parameters is demonstrated by using the same alanine dipeptide-fitted backbone torsional parameters for all of the other dipeptides (with the appropriate side-chain refitting) and the alanine tetrapeptide. Parameters of nonbonded interactions have also been refitted for the sulfur-contg. dipeptides (cysteine and methionine), and the validity of the new Coulombic charges and the van der Waals σ's and ε's is proved through reproducing gas-phase energies of complex formation heats of vaporization and densities of pure model liqs. Moreover, a novel approach to fitting torsional parameters for electrostatically charged mol. systems has been presented and successfully tested on five dipeptides with charged side chains.
- 58Buck, M.; Bouguet-Bonnet, S.; Pastor, R. W.; MacKerell, A. D., Jr Importance of the CMAP correction to the CHARMM22 protein force field: dynamics of hen lysozyme. Biophysical journal 2006, 90, L36– L38, DOI: 10.1529/biophysj.105.07815458Importance of the CMAP correction to the CHARMM22 protein force field: dynamics of hen lysozymeBuck, Matthias; Bouguet-Bonnet, Sabine; Pastor, Richard W.; MacKerell, Alexander D., Jr.Biophysical Journal (2006), 90 (4), L36-L38CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The recently developed CMAP correction to the CHARMM22 force field (C22) is evaluated from 25 ns mol. dynamics simulations on hen lysozyme. Substantial deviations from exptl. backbone root mean-square fluctuations and N-H NMR order parameters obtained in the C22 trajectories (esp. in the loops) are eliminated by the CMAP correction. Thus, the C22/CMAP force field yields improved dynamical and structural properties of proteins in mol. dynamics simulations.
- 59Durell, S. R.; Brooks, B. R.; Ben-Naim, A. Solvent-induced forces between two hydrophilic groups. J. Phys. Chem. 1994, 98, 2198– 2202, DOI: 10.1021/j100059a03859Solvent-Induced Forces between Two Hydrophilic GroupsDurell, Stewart R.; Brooks, Bernard R.; Ben-Naim, AriehJournal of Physical Chemistry (1994), 98 (8), 2198-202CODEN: JPCHAX; ISSN:0022-3654.Mol. dynamics simulations were used to calc. the force between two simple hydrophilic solutes in dil. aq. soln. The "solutes" were two water mols. in the same relative orientation as the next-nearest neighbors in hexagonal ice I. Both the direct and solvent-induced contributions to the force were calcd. as a function of sepn. distance. The total force between the solutes was found to be most attractive at 5.0 Å (-1.6 kcal/mol/Å). The potential of mean force had a min. at 4.3 Å, which is 0.2 Å closer than the next-nearest-neighbor distance in ice. A parallel set of simulations were conducted with the partial charges on the "solutes" removed to examine hydrophobic analogs. In this case, the total force was most attractive at 3.5 Å (-0.9 kcal/mol/Å), and the min. of the potential was at the contact distance of 3.2 Å. In agreement with earlier predictions, the max. solvent-induced contribution to the potential was ca. 4 times more neg. for the hydrophilic "solutes" than for the hydrophobic ones. These differences are shown to be due predominantly to a solvent water mol. which simultaneously hydrogen bonds to both hydrophilic "solutes". The results support earlier assertions that solvent-induced interactions between polar amino acid residues are more important in protein folding and stability than generally considered.
- 60Neria, E.; Fischer, S.; Karplus, M. Simulation of activation free energies in molecular systems. J. Chem. Phys. 1996, 105, 1902– 1921, DOI: 10.1063/1.47206160Simulation of activation free energies in molecular systemsNeria, Eyal; Fischer, Stefan; Karplus, MartinJournal of Chemical Physics (1996), 105 (5), 1902-1921CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)A method is presented for detg. activation free energies in complex mol. systems. The method relies on knowledge of the min. energy path and bases the activation free energy calcn. on moving along this path from a min. to a saddle point. Use is made of a local reaction coordinate which describes the advance of the reaction in each segment of the min. energy path. The activation free energy is formulated as a sum of two terms. The first is due to the change in the local reaction coordinate between the endpoints of each segment of the path. The second is due to the change in direction of the min. energy path between consecutive segments. Both contributions can be obtained by mol. dynamics simulations with a constraint on the local reaction coordinate. The method is illustrated by applying it to a model potential and to the C7eq to C7ax transition in the alanine dipeptide. It is found that the term due to the change of direction in the reaction path can make a substantial contribution to the activation free energy.
- 61Yesylevskyy, S. O.; Schäfer, L. V.; Sengupta, D.; Marrink, S. J. Polarizable water model for the coarse-grained MARTINI force field. PLoS computational biology 2010, 6, e1000810, DOI: 10.1371/journal.pcbi.100081061Polarizable water model for the coarse-grained MARTINI force fieldYesylevskyy Semen O; Schafer Lars V; Sengupta Durba; Marrink Siewert JPLoS computational biology (2010), 6 (6), e1000810 ISSN:.Coarse-grained (CG) simulations have become an essential tool to study a large variety of biomolecular processes, exploring temporal and spatial scales inaccessible to traditional models of atomistic resolution. One of the major simplifications of CG models is the representation of the solvent, which is either implicit or modeled explicitly as a van der Waals particle. The effect of polarization, and thus a proper screening of interactions depending on the local environment, is absent. Given the important role of water as a ubiquitous solvent in biological systems, its treatment is crucial to the properties derived from simulation studies. Here, we parameterize a polarizable coarse-grained water model to be used in combination with the CG MARTINI force field. Using a three-bead model to represent four water molecules, we show that the orientational polarizability of real water can be effectively accounted for. This has the consequence that the dielectric screening of bulk water is reproduced. At the same time, we parameterized our new water model such that bulk water density and oil/water partitioning data remain at the same level of accuracy as for the standard MARTINI force field. We apply the new model to two cases for which current CG force fields are inadequate. First, we address the transport of ions across a lipid membrane. The computed potential of mean force shows that the ions now naturally feel the change in dielectric medium when moving from the high dielectric aqueous phase toward the low dielectric membrane interior. In the second application we consider the electroporation process of both an oil slab and a lipid bilayer. The electrostatic field drives the formation of water filled pores in both cases, following a similar mechanism as seen with atomistically detailed models.
- 62Berendsen, H. J.; Postma, J. P.; van Gunsteren, W. F.; Hermans, J. Intermolecular forces; Springer, 1981; pp 331– 342.There is no corresponding record for this reference.
- 63De Jong, D. H.; Baoukina, S.; Ingólfsson, H. I.; Marrink, S. J. Martini straight: Boosting performance using a shorter cutoff and GPUs. Comput. Phys. Commun. 2016, 199, 1– 7, DOI: 10.1016/j.cpc.2015.09.01463Martini straight: Boosting performance using a shorter cutoff and GPUsde Jong, Djurre H.; Baoukina, Svetlana; Ingolfsson, Helgi I.; Marrink, Siewert J.Computer Physics Communications (2016), 199 (), 1-7CODEN: CPHCBZ; ISSN:0010-4655. (Elsevier B.V.)In mol. dynamics simulations, sufficient sampling is of key importance and a continuous challenge in the field. The coarse grain Martini force field has been widely used to enhance sampling. In its original implementation, this force field applied a shifted Lennard-Jones potential for the non-bonded van der Waals interactions, to avoid problems related to a relatively short cutoff. Here we investigate the use of a straight cutoff Lennard-Jones potential with potential modifiers. Together with a Verlet neighbor search algorithm, the modified potential allows the use of GPUs to accelerate the computations in Gromacs. We find that this alternative potential has little influence on most of the properties studied, including partitioning free energies, bulk liq. properties and bilayer properties. At the same time, energy conservation is kept within reasonable bounds. We conclude that the newly proposed straight cutoff approach is a viable alternative to the std. shifted potentials used in Martini, offering significant speedup even in the absence of GPUs.
- 64Bussi, G.; Donadio, D.; Parrinello, M. Canonical sampling through velocity rescaling. J. Chem. Phys. 2007, 126, 014101, DOI: 10.1063/1.240842064Canonical sampling through velocity rescalingBussi, Giovanni; Donadio, Davide; Parrinello, MicheleJournal of Chemical Physics (2007), 126 (1), 014101/1-014101/7CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The authors present a new mol. dynamics algorithm for sampling the canonical distribution. In this approach the velocities of all the particles are rescaled by a properly chosen random factor. The algorithm is formally justified and it is shown that, in spite of its stochastic nature, a quantity can still be defined that remains const. during the evolution. In numerical applications this quantity can be used to measure the accuracy of the sampling. The authors illustrate the properties of this new method on Lennard-Jones and TIP4P water models in the solid and liq. phases. Its performance is excellent and largely independent of the thermostat parameter also with regard to the dynamic properties.
- 65Parrinello, M.; Rahman, A. Polymorphic transitions in single crystals: A new molecular dynamics method. J. Appl. Phys. 1981, 52, 7182– 7190, DOI: 10.1063/1.32869365Polymorphic transitions in single crystals: A new molecular dynamics methodParrinello, M.; Rahman, A.Journal of Applied Physics (1981), 52 (12), 7182-90CODEN: JAPIAU; ISSN:0021-8979.A Lagrangian formulation is introduced; it can be used to make mol. dynamics (MD) calcns. on systems under the most general, externally applied, conditions of stress. In this formulation the MD cell shape and size can change according to dynamic equations given by this Lagrangian. This MD technique was used to the study of structural transitions of a Ni single crystal under uniform uniaxial compressive and tensile loads. Some results regarding the stress-strain relation obtained by static calcns. are invalid at finite temp. Under compressive loading, the model of Ni shows a bifurcation in its stress-strain relation; this bifurcation provides a link in configuration space between cubic and hexagonal close packing. Such a transition could perhaps be obsd. exptl. under extreme conditions of shock.
- 66Hess, B.; Bekker, H.; Berendsen, H. J.; Fraaije, J. G. LINCS: a linear constraint solver for molecular simulations. Journal of computational chemistry 1997, 18, 1463– 1472, DOI: 10.1002/(SICI)1096-987X(199709)18:12<1463::AID-JCC4>3.0.CO;2-H66LINCS: a linear constraint solver for molecular simulationsHess, Berk; Bekker, Henk; Berendsen, Herman J. C.; Fraaije, Johannes G. E. M.Journal of Computational Chemistry (1997), 18 (12), 1463-1472CODEN: JCCHDD; ISSN:0192-8651. (Wiley)We present a new LINear Constraint Solver (LINCS) for mol. simulations with bond constraints using the enzyme lysozyme and a 32-residue peptide as test systems. The algorithm is inherently stable, as the constraints themselves are reset instead of derivs. of the constraints, thereby eliminating drift. Although the derivation of the algorithm is presented in terms of matrixes, no matrix matrix multiplications are needed and only the nonzero matrix elements have to be stored, making the method useful for very large mols. At the same accuracy, the LINCS algorithm is 3-4 times faster than the SHAKE algorithm. Parallelization of the algorithm is straightforward.
- 67Miyamoto, S.; Kollman, P. A. Settle: An analytical version of the SHAKE and RATTLE algorithm for rigid water models. Journal of computational chemistry 1992, 13, 952– 962, DOI: 10.1002/jcc.54013080567SETTLE: an analytical version of the SHAKE and RATTLE algorithm for rigid water modelsMiyamoto, Shuichi; Kollman, Peter A.Journal of Computational Chemistry (1992), 13 (8), 952-62CODEN: JCCHDD; ISSN:0192-8651.An anal. algorithm, called SETTLE, for resetting the positions and velocities to satisfy the holonomic constraints on the rigid water model is presented. This method is based on the Cartesian coordinate system and can be used in place of SHAKE and RATTLE. The authors implemented this algorithm in the SPASMS package of mol. mechanics and dynamics. Several series of mol. dynamics simulations were carried out to examine the performance of the new algorithm in comparison with the original RATTLE method. SETTLE is of higher accuracy and is faster than RATTLE with reasonable tolerances by three to nine times on a scalar machine. The performance improvement ranged from factors of 26 to 98 on a vector machine since the method presented is not iterative.
- 68Hub, J. S.; de Groot, B. L.; Grubmüller, H.; Groenhof, G. Quantifying artifacts in Ewald simulations of inhomogeneous systems with a net charge. J. Chem. Theory Comput. 2014, 10, 381– 390, DOI: 10.1021/ct400626b68Quantifying Artifacts in Ewald Simulations of Inhomogeneous Systems with a Net ChargeHub, Jochen S.; de Groot, Bert L.; Grubmueller, Helmut; Groenhof, GerritJournal of Chemical Theory and Computation (2014), 10 (1), 381-390CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Ewald summation, which has become the de facto std. for computing electrostatic interactions in biomol. simulations, formally requires that the simulation box is neutral. For non-neutral systems, the Ewald algorithm implicitly introduces a uniform background charge distribution that effectively neutralizes the simulation box. Because a uniform distribution of counter charges typically deviates from the spatial distribution of counterions in real systems, artifacts may arise, in particular in systems with an inhomogeneous dielec. const. Here, we derive an anal. expression for the effect of using an implicit background charge instead of explicit counterions, on the chem. potential of ions in heterogeneous systems, which (i) provides a quant. criterion for deciding if the background charge offers an acceptable trade-off between artifacts arising from sampling problems and artifacts arising from the homogeneous background charge distribution, and (ii) can be used to correct this artifact in certain cases. Our model quantifies the artifact in terms of the difference in charge d. between the non-neutral system with a uniform neutralizing background charge and the real neutral system with a phys. correct distribution of explicit counterions. We show that for inhomogeneous systems, such as proteins and membranes in water, the artifact manifests itself by an overstabilization of ions inside the lower dielec. by tens to even hundreds kilojoules per mol. We have tested the accuracy of our model in mol. dynamics simulations and found that the error in the calcd. free energy for moving a test charge from water into hexadecane, at different net charges of the system and different simulation box sizes, is correctly predicted by the model. The calcns. further confirm that the incorrect distribution of counter charges in the simulation box is solely responsible for the errors in the PMFs.
- 69Thurlkill, R. L.; Grimsley, G. R.; Scholtz, J. M.; Pace, C. N. pK values of the ionizable groups of proteins. Protein science 2006, 15, 1214– 1218, DOI: 10.1110/ps.05184080669pK values of the ionizable groups of proteinsThurlkill, Richard L.; Grimsley, Gerald R.; Scholtz, J. Martin; Pace, C. NickProtein Science (2006), 15 (5), 1214-1218CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)We have used potentiometric titrns. to measure the pK values of the ionizable groups of proteins in alanine pentapeptides with appropriately blocked termini. These pentapeptides provide an improved model for the pK values of the ionizable groups in proteins. Our pK values detd. in 0.1 M KCl at 25°C are: 3.67 ± 0.03 (α-carboxyl), 3.67 ± 0.04 (Asp), 4.25 ± 0.05 (Glu), 6.54 ± 0.04 (His), 8.00 ± 0.03 (α-amino), 8.55 ± 0.03 (Cys), 9.84 ± 0.11 (Tyr), and 10.40 ± 0.08 (Lys). The pK values of some groups differ from the Nozaki and Tanford (N&T) pK values often used in the literature: Asp (3.67 this work vs. 4.0 N&T); His (6.54 this work vs. 6.3 N&T); α-amino (8.00 this work vs. 7.5 N&T); Cys (8.55 this work vs. 9.5 N&T); and Tyr (9.84 this work vs. 9.6 N&T). Our pK values will be useful to those who study pK perturbations in folded and unfolded proteins, and to those who use theory to gain a better understanding of the factors that det. the pK values of the ionizable groups of proteins.
- 70Tanokura, M. 1H-NMR study on the tautomerism of the imidazole ring of histidine residues: I. Microscopic pK values and molar ratios of tautomers in histidine-containing peptides. Biochimica et Biophysica Acta (BBA)-Protein Structure and Molecular Enzymology 1983, 742, 576– 585, DOI: 10.1016/0167-4838(83)90276-5There is no corresponding record for this reference.
- 71Chiang, C.-M.; Chien, K.-Y.; Lin, H.-j.; Lin, J.-F.; Yeh, H.-C.; Ho, P.-l.; Wu, W.-g. Conformational change and inactivation of membrane phospholipid-related activity of cardiotoxin V from Taiwan cobra venom at acidic pH. Biochemistry 1996, 35, 9167– 9176, DOI: 10.1021/bi952823k71Conformational Change and Inactivation of Membrane Phospholipid-Related Activity of Cardiotoxin V from Taiwan Cobra Venom at Acidic pHChiang, Chien-Min; Chien, Kun-Yi; Lin, Hai-jui; Lin, Ji-Fu; Yeh, Hsien-Chi; Ho, Pei-li; Wu, Wen-gueyBiochemistry (1996), 35 (28), 9167-9176CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)The phospholipid binding activity of cardiotoxin V from Naja naja atra (CTX A5) was studied by use of Langmuir monolayers and found to exhibit pH-dependence in binding to phosphatidylcholine membrane with an apparent pKa around 6.0. Proton NMR investigation of the CTX A5 mol. in the presence of phosphatidylcholine micelles reveals a decrease in assocn. of CTX A5 with membranes at low pH as a result of the protonation of His-4 near the membrane binding site of loop I region of CTX. The pH-dependent binding can be attributed mainly, but not solely, to the change in charge content of the CTX mol. upon His-4 protonation at the membrane/water interface. This is shown by analyzing the pH- and ionic strength dependence of binding of CTXs to phospholipid monolayers according to Gouy-Chapman theory. The protonation of the His-4 residue also results in a local conformational change in the loop I region since the chem. shifts of amide protons for the amino acid residues from Cys-3 to Thr-14 are all found to vary as a function of pH with an apparent pKa similar to that of His-4. Interestingly, the effect is relayed to other amino acid residues in the structural core of the protein such as those in C-terminal (Lys-60, Cys-61, and Asn-62) and triple-stranded antiparallel β-sheet (Cys-22, Lys-24, Ala-25, Arg-38, and Ala-41) regions. An addnl. local conformational change in the mol. results around pH 5 as evidenced by CD spectroscopic studies, although this change does not affect the characteristic β-sheet and three-finger loop structure of CTX mol. as revealed by two-dimensional NOESY 1H NMR study. The latter conformational change at acidic pH, however, completely inactivates CTX-induced aggregation/fusion activity of sphingomyelin vesicles. The results suggest that deciphering the functional sites of CTXs on the basis of structure and dynamics detd. at low pH should be done with caution. Since 19 out of 44 CTX homologs with known amino acid sequence contain His-4, the effect of His-4 on the structure and function of CTX mols. is important and is discussed in terms of the diverse membrane targets of CTX subtypes. Also discussed is the pH-induced activation of snake venom proteins in the victim.
- 72Chiang, C.-M.; Chang, S.-L.; Lin, H.-j.; Wu, W.-g. The role of acidic amino acid residues in the structural stability of snake cardiotoxins. Biochemistry 1996, 35, 9177– 9186, DOI: 10.1021/bi960077t72The Role of Acidic Amino Acid Residues in the Structural Stability of Snake CardiotoxinsChiang, Chien-Min; Chang, Shou-Lin; Lin, Hai-jui; Wu, Wen-gueyBiochemistry (1996), 35 (28), 9177-9186CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)PH-induced structural transitions occurred in cardiotoxin V from Naja naja atra (CTX A5) at two pH values as judged by the CD ellipticity around 195 nm: an increase in the β-sheet content occurred around pH 4 and followed by a decrease, therein, around pH 2. The pKa of three acidic amino acid residues in CTX A5, i.e., Glu-17, Asp-42, and Asp-59, were detd. to be 4.0, 3.2, and below 2.3, resp., by NMR spectroscopy. The low pKa value of Asp-59 implies salt bridge formation between Lys-2 and Asp-59. Thus, electrostatic interaction may stabilize the three loop structure in addn. to the hydrogen bonds between N- and C-termini of CTX mol. Second, 2,2,2-trifluoroethanol (TFE) and guanidinium chloride (GdmHCl) were found to induce α-helical and random coil formation, resp., in CTX A5 and eight other β-sheet CTXs. Comparison of the relative potencies of TFE and GdmHCl to induce structural changes suggests that the amino acid residue located at position 17 plays a role in the structural stability. Specifically, CTXs contg. neg. charged Glu-17 are least stable. It is suggested that Glu-17 may perturb the interaction between Lys-2 and Asp-59, and thus the overall stability of β-sheet, in the presence of denaturing reagent. In conclusion, the perturbed structural stability of CTXs may partially explain the lower activity CTX exhibits at acidic pH. A structural model to account for the unfolding and refolding of CTX mols. without the breaking of disulfide bonds is also proposed.
- 73Webb, H.; Tynan-Connolly, B. M.; Lee, G. M.; Farrell, D.; O’Meara, F.; Søndergaard, C. R.; Teilum, K.; Hewage, C.; McIntosh, L. P.; Nielsen, J. E. Remeasuring HEWL pKa values by NMR spectroscopy: Methods, analysis, accuracy, and implications for theoretical pKa calculations. Proteins: Struct., Funct., Bioinf. 2011, 79, 685– 702, DOI: 10.1002/prot.2288673Remeasuring HEWL pKa values by NMR spectroscopy: methods, analysis, accuracy, and implications for theoretical pKa calculationsWebb, Helen; Tynan-Connolly, Barbara Mary; Lee, Gregory M.; Farrell, Damien; O'Meara, Fergal; Sondergaard, Chresten R.; Teilum, Kaare; Hewage, Chandralal; McIntosh, Lawrence P.; Nielsen, Jens ErikProteins: Structure, Function, and Bioinformatics (2011), 79 (3), 685-702CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)Site-specific pKa values measured by NMR spectroscopy provide essential information on protein electrostatics, the pH-dependence of protein structure, dynamics and function, and constitute an important benchmark for protein pKa calcn. algorithms. Titrn. curves can be measured by tracking the NMR chem. shifts of several reporter nuclei vs. sample pH. However, careful anal. of these curves is needed to ext. residue-specific pKa values since pH-dependent chem. shift changes can arise from many sources, including through-bond inductive effects, through-space elec. field effects, and conformational changes. We have re-measured titrn. curves for all carboxylates and His 15 in Hen Egg White Lysozyme (HEWL) by recording the pH-dependent chem. shifts of all backbone amide nitrogens and protons, Asp/Glu side chain protons and carboxyl carbons, and imidazole protonated carbons and protons in this protein. We extd. pKa values from the resulting titrn. curves using std. fitting methods, and compared these values to each other, and with those measured previously by 1H NMR. This anal. gives insights into the true accuracy assocd. with exptl. measured pKa values. We find that apparent pKa values frequently differ by 0.5-1.0 units depending upon the nuclei monitored, and that larger differences occasionally can be obsd. The variation in measured pKa values, which reflects the difficulty in fitting and assigning pH-dependent chem. shifts to specific ionization equil., has significant implications for the exptl. procedures used for measuring protein pKa values, for the benchmarking of protein pKa calcn. algorithms, and for the understanding of protein electro-statics in general.
- 74Lee, J.; Miller, B. T.; Damjanovic, A.; Brooks, B. R. Enhancing constant-pH simulation in explicit solvent with a two-dimensional replica exchange method. J. Chem. Theory Comput. 2015, 11, 2560– 2574, DOI: 10.1021/ct501101f74Enhancing Constant-pH Simulation in Explicit Solvent with a Two-Dimensional Replica Exchange MethodLee, Juyong; Miller, Benjamin T.; Damjanovic, Ana; Brooks, Bernard R.Journal of Chemical Theory and Computation (2015), 11 (6), 2560-2574CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We present a new method for enhanced sampling for const.-pH simulations in explicit water based on a two-dimensional (2D) replica exchange scheme. The new method is a significant extension of our previously developed const.-pH simulation method, which is based on enveloping distribution sampling (EDS) coupled with a one-dimensional (1D) Hamiltonian exchange method (HREM). EDS constructs a hybrid Hamiltonian from multiple discrete end state Hamiltonians that, in this case, represent different protonation states of the system. The ruggedness and heights of the hybrid Hamiltonian's energy barriers can be tuned by the smoothness parameter. Within the context of the 1D EDS-HREM method, exchanges are performed between replicas with different smoothness parameters, allowing frequent protonation-state transitions and sampling of conformations that are favored by the end-state Hamiltonians. In this work, the 1D method is extended to 2D with an addnl. dimension, external pH. Within the context of the 2D method (2D EDS-HREM), exchanges are performed on a lattice of Hamiltonians with different pH conditions and smoothness parameters. We demonstrate that both the 1D and 2D methods exactly reproduce the thermodn. properties of the semigrand canonical (SGC) ensemble of a system at a given pH. We have tested our new 2D method on aspartic acid, glutamic acid, lysine, a four residue peptide (sequence KAAE), and snake cardiotoxin. In all cases, the 2D method converges faster and without loss of precision; the only limitation is a loss of flexibility in how CPU time is employed. The results for snake cardiotoxin demonstrate that the 2D method enhances protonation-state transitions, samples a wider conformational space with the same amt. of computational resources, and converges significantly faster overall than the original 1D method.
- 75Pezeshkian, W.; König, M.; Wassenaar, T. A.; Marrink, S. J. Backmapping triangulated surfaces to coarse-grained membrane models. Nat. Commun. 2020, 11, 2296, DOI: 10.1038/s41467-020-16094-y75Backmapping triangulated surfaces to coarse-grained membrane modelsPezeshkian, Weria; Koenig, Melanie; Wassenaar, Tsjerk A.; Marrink, Siewert J.Nature Communications (2020), 11 (1), 2296CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Many biol. processes involve large-scale changes in membrane shape. Computer simulations of these processes are challenging since they occur across a wide range of spatiotemporal scales that cannot be investigated in full by any single current simulation technique. A potential soln. is to combine different levels of resoln. through a multiscale scheme. Here, we present a multiscale algorithm that backmaps a continuum membrane model represented as a dynamically triangulated surface (DTS) to its corresponding mol. model based on the coarse-grained (CG) Martini force field. Thus, we can use DTS simulations to equilibrate slow large-scale membrane conformational changes and then explore the local properties at CG resoln. We demonstrate the power of our method by backmapping a vesicular bud induced by binding of Shiga toxin and by transforming the membranes of an entire mitochondrion to near-at. resoln. Our approach opens the way to whole cell simulations at mol. detail.
- 76Wolfram Research, Inc. Mathematica, Version 11.3. Champaign: IL, 2018.There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jctc.2c00516.
The input files and parameters used for MD simulations in the presented work (ZIP)
A Mathematica notebook with instructions and routines to fit VMM (ZIP)
Description of λ-potentials, the effect of neglecting the interpolation of Lennard-Jones interactions, titration results for Asp and Glu within the single-site representation, a comparison of pKa values for HEWL obtained with various λ-dynamics-based constant pH methods, demonstration that charge interpolation requires a single evaluation of the electrostatic potential for both single- and multisite representations (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.