3301
|
Krammer EM, Vu GT, Homblé F, Prévost M. Dual mechanism of ion permeation through VDAC revealed with inorganic phosphate ions and phosphate metabolites. PLoS One 2015; 10:e0121746. [PMID: 25860993 PMCID: PMC4393092 DOI: 10.1371/journal.pone.0121746] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/03/2015] [Indexed: 11/19/2022] Open
Abstract
In the exchange of metabolites and ions between the mitochondrion and the cytosol, the voltage-dependent anion channel (VDAC) is a key element, as it forms the major transport pathway for these compounds through the mitochondrial outer membrane. Numerous experimental studies have promoted the idea that VDAC acts as a regulator of essential mitochondrial functions. In this study, using a combination of molecular dynamics simulations, free-energy calculations, and electrophysiological measurements, we investigated the transport of ions through VDAC, with a focus on phosphate ions and metabolites. We showed that selectivity of VDAC towards small anions including monovalent phosphates arises from short-lived interactions with positively charged residues scattered throughout the pore. In dramatic contrast, permeation of divalent phosphate ions and phosphate metabolites (AMP and ATP) involves binding sites along a specific translocation pathway. This permeation mechanism offers an explanation for the decrease in VDAC conductance measured in the presence of ATP or AMP at physiological salt concentration. The binding sites occur at similar locations for the divalent phosphate ions, AMP and ATP, and contain identical basic residues. ATP features a marked affinity for a central region of the pore lined by two lysines and one arginine of the N-terminal helix. This cluster of residues together with a few other basic amino acids forms a "charged brush" which facilitates the passage of the anionic metabolites through the pore. All of this reveals that VDAC controls the transport of the inorganic phosphates and phosphate metabolites studied here through two different mechanisms.
Collapse
Affiliation(s)
- Eva-Maria Krammer
- Structure et Fonction des Membranes Biologiques, Centre de Biologie Structurale et de Bioinformatique, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Giang Thi Vu
- Structure et Fonction des Membranes Biologiques, Centre de Biologie Structurale et de Bioinformatique, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fabrice Homblé
- Structure et Fonction des Membranes Biologiques, Centre de Biologie Structurale et de Bioinformatique, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Martine Prévost
- Structure et Fonction des Membranes Biologiques, Centre de Biologie Structurale et de Bioinformatique, Université Libre de Bruxelles (ULB), Brussels, Belgium
- * E-mail: (MP)
| |
Collapse
|
3302
|
Vilseck JZ, Tirado-Rives J, Jorgensen WL. Determination of partial molar volumes from free energy perturbation theory. Phys Chem Chem Phys 2015; 17:8407-15. [PMID: 25589343 PMCID: PMC4872387 DOI: 10.1039/c4cp05304d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Partial molar volume is an important thermodynamic property that gives insights into molecular size and intermolecular interactions in solution. Theoretical frameworks for determining the partial molar volume (V°) of a solvated molecule generally apply Scaled Particle Theory or Kirkwood-Buff theory. With the current abilities to perform long molecular dynamics and Monte Carlo simulations, more direct methods are gaining popularity, such as computing V° directly as the difference in computed volume from two simulations, one with a solute present and another without. Thermodynamically, V° can also be determined as the pressure derivative of the free energy of solvation in the limit of infinite dilution. Both approaches are considered herein with the use of free energy perturbation (FEP) calculations to compute the necessary free energies of solvation at elevated pressures. Absolute and relative partial molar volumes are computed for benzene and benzene derivatives using the OPLS-AA force field. The mean unsigned error for all molecules is 2.8 cm(3) mol(-1). The present methodology should find use in many contexts such as the development and testing of force fields for use in computer simulations of organic and biomolecular systems, as a complement to related experimental studies, and to develop a deeper understanding of solute-solvent interactions.
Collapse
Affiliation(s)
- Jonah Z Vilseck
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, USA.
| | | | | |
Collapse
|
3303
|
Chandramouli B, Di Maio D, Mancini G, Barone V, Brancato G. Breaking the hydrophobicity of the MscL pore: insights into a charge-induced gating mechanism. PLoS One 2015; 10:e0120196. [PMID: 25825909 PMCID: PMC4380313 DOI: 10.1371/journal.pone.0120196] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/06/2015] [Indexed: 01/17/2023] Open
Abstract
The mechanosensitive channel of large conductance (MscL) is a protein that responds to membrane tension by opening a transient pore during osmotic downshock. Due to its large pore size and functional reconstitution into lipid membranes, MscL has been proposed as a promising artificial nanovalve suitable for biotechnological applications. For example, site-specific mutations and tailored chemical modifications have shown how MscL channel gating can be triggered in the absence of tension by introducing charged residues at the hydrophobic pore level. Recently, engineered MscL proteins responsive to stimuli like pH or light have been reported. Inspired by experiments, we present a thorough computational study aiming at describing, with atomistic detail, the artificial gating mechanism and the molecular transport properties of a light-actuated bacterial MscL channel, in which a charge-induced gating mechanism has been enabled through the selective cleavage of photo-sensitive alkylating agents. Properties such as structural transitions, pore dimension, ion flux and selectivity have been carefully analyzed. Besides, the effects of charge on alternative sites of the channel with respect to those already reported have been addressed. Overall, our results provide useful molecular insights into the structural events accompanying the engineered MscL channel gating and the interplay of electrostatic effects, channel opening and permeation properties. In addition, we describe how the experimentally observed ionic current in a single-subunit charged MscL mutant is obtained through a hydrophobicity breaking mechanism involving an asymmetric inter-subunit motion.
Collapse
Affiliation(s)
| | - Danilo Di Maio
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126, Pisa, Italy
- Istituto Nazionale di Fisica Nucleare (INFN), sezione di Pisa, Largo Bruno Pontecorvo 3, 56127, Pisa, Italy
| | - Giordano Mancini
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126, Pisa, Italy
- Istituto Nazionale di Fisica Nucleare (INFN), sezione di Pisa, Largo Bruno Pontecorvo 3, 56127, Pisa, Italy
| | - Vincenzo Barone
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126, Pisa, Italy
- Istituto Nazionale di Fisica Nucleare (INFN), sezione di Pisa, Largo Bruno Pontecorvo 3, 56127, Pisa, Italy
| | - Giuseppe Brancato
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126, Pisa, Italy
- Istituto Nazionale di Fisica Nucleare (INFN), sezione di Pisa, Largo Bruno Pontecorvo 3, 56127, Pisa, Italy
- * E-mail:
| |
Collapse
|
3304
|
Schröder C, Steinhauser O, Sasisanker P, Weingärtner H. Orientational alignment of amyloidogenic proteins in pre-aggregated solutions. PHYSICAL REVIEW LETTERS 2015; 114:128101. [PMID: 25860772 DOI: 10.1103/physrevlett.114.128101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Indexed: 06/04/2023]
Abstract
In the present study we combine dielectric relaxation spectroscopy with generalized Born simulations to explore the role of orientational order for protein aggregation in solutions of bovine pancreatic insulin at various pH conditions. Under aggregation-prone conditions at low pH, insulin monomers prefer antiparallel dipole alignments, which are consistent with the orientation of the monomeric subunits in the dimer structure. This alignment is also true for two dimers, suggesting that already at moderate protein concentrations the species assemble in equilibrium clusters, in which the molecules adopt preferred orientations also found for the protomers of the corresponding oligomers.
Collapse
Affiliation(s)
- C Schröder
- Department of Computational Biological Chemistry, University of Vienna, Währingerstrasse 17, 1090 Vienna, Austria
| | - O Steinhauser
- Department of Computational Biological Chemistry, University of Vienna, Währingerstrasse 17, 1090 Vienna, Austria
| | - P Sasisanker
- Department of Physical Chemistry II, Ruhr-University of Bochum, Germany and Praj Matrix The Innovation Center Urawade, Pune 412108, India
| | - H Weingärtner
- Department of Physical Chemistry II, Ruhr-University of Bochum, Building NC 6-25, 44780 Bochum, Germany
| |
Collapse
|
3305
|
Denisov IG, Grinkova YV, Baylon JL, Tajkhorshid E, Sligar SG. Mechanism of drug-drug interactions mediated by human cytochrome P450 CYP3A4 monomer. Biochemistry 2015; 54:2227-39. [PMID: 25777547 DOI: 10.1021/acs.biochem.5b00079] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Using Nanodiscs, we quantitate the heterotropic interaction between two different drugs mediated by monomeric CYP3A4 incorporated into a nativelike membrane environment. The mechanism of this interaction is deciphered by global analysis of multiple-turnover experiments performed under identical conditions using the pure substrates progesterone (PGS) and carbamazepine (CBZ) and their mixtures. Activation of CBZ epoxidation and simultaneous inhibition of PGS hydroxylation are measured and quantitated through differences in their respective affinities for both a remote allosteric site and the productive catalytic site near the heme iron. Preferred binding of PGS at the allosteric site and a stronger preference for CBZ binding at the productive site give rise to a nontrivial drug-drug interaction. Molecular dynamics simulations indicate functionally important conformational changes caused by PGS binding at the allosteric site and by two CBZ molecules positioned inside the substrate binding pocket. Structural changes involving Phe-213, Phe-219, and Phe-241 are thought to be responsible for the observed synergetic effects and positive allosteric interactions between these two substrates. Such a mechanism is likely of general relevance to the mutual heterotropic effects caused by biologically active compounds that exhibit different patterns of interaction with the distinct allosteric and productive sites of CYP3A4, as well as other xenobiotic metabolizing cytochromes P450 that are also involved in drug-drug interactions. Importantly, this work demonstrates that a monomeric CYP3A4 can display the full spectrum of activation and cooperative effects that are observed in hepatic membranes.
Collapse
Affiliation(s)
- Ilia G Denisov
- †Department of Biochemistry, ‡Center for Biophysics and Computational Biology, and §Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yelena V Grinkova
- †Department of Biochemistry, ‡Center for Biophysics and Computational Biology, and §Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Javier L Baylon
- †Department of Biochemistry, ‡Center for Biophysics and Computational Biology, and §Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Emad Tajkhorshid
- †Department of Biochemistry, ‡Center for Biophysics and Computational Biology, and §Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Stephen G Sligar
- †Department of Biochemistry, ‡Center for Biophysics and Computational Biology, and §Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
3306
|
Vermaas JV, Taguchi AT, Dikanov SA, Wraight CA, Tajkhorshid E. Redox potential tuning through differential quinone binding in the photosynthetic reaction center of Rhodobacter sphaeroides. Biochemistry 2015; 54:2104-16. [PMID: 25734689 DOI: 10.1021/acs.biochem.5b00033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ubiquinone forms an integral part of the electron transport chain in cellular respiration and photosynthesis across a vast number of organisms. Prior experimental results have shown that the photosynthetic reaction center (RC) from Rhodobacter sphaeroides is only fully functional with a limited set of methoxy-bearing quinones, suggesting that specific interactions with this substituent are required to drive electron transport and the formation of quinol. The nature of these interactions has yet to be determined. Through parameterization of a CHARMM-compatible quinone force field and subsequent molecular dynamics simulations of the quinone-bound RC, we have investigated and characterized the interactions of the protein with the quinones in the Q(A) and Q(B) sites using both equilibrium simulation and thermodynamic integration. In particular, we identify a specific interaction between the 2-methoxy group of ubiquinone in the Q(B) site and the amide nitrogen of GlyL225 that we implicate in locking the orientation of the 2-methoxy group, thereby tuning the redox potential difference between the quinones occupying the Q(A) and Q(B) sites. Disruption of this interaction leads to weaker binding in a ubiquinone analogue that lacks a 2-methoxy group, a finding supported by reverse electron transfer electron paramagnetic resonance experiments of the Q(A)⁻Q(B)⁻ biradical and competitive binding assays.
Collapse
Affiliation(s)
- Josh V Vermaas
- †Center for Biophysics and Computational Biology, ‡Department of Biochemistry, §Beckman Institute, and ∥Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Alexander T Taguchi
- †Center for Biophysics and Computational Biology, ‡Department of Biochemistry, §Beckman Institute, and ∥Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sergei A Dikanov
- †Center for Biophysics and Computational Biology, ‡Department of Biochemistry, §Beckman Institute, and ∥Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Colin A Wraight
- †Center for Biophysics and Computational Biology, ‡Department of Biochemistry, §Beckman Institute, and ∥Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Emad Tajkhorshid
- †Center for Biophysics and Computational Biology, ‡Department of Biochemistry, §Beckman Institute, and ∥Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
3307
|
Trujillo K, Paoletta S, Kiselev E, Jacobson KA. Molecular modeling of the human P2Y14 receptor: A template for structure-based design of selective agonist ligands. Bioorg Med Chem 2015; 23:4056-64. [PMID: 25868749 DOI: 10.1016/j.bmc.2015.03.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/11/2015] [Accepted: 03/13/2015] [Indexed: 10/23/2022]
Abstract
The P2Y14 receptor (P2Y14R) is a Gi protein-coupled receptor that is activated by uracil nucleotides UDP and UDP-glucose. The P2Y14R structure has yet to be solved through X-ray crystallography, but the recent agonist-bound crystal structure of the P2Y12R provides a potentially suitable template for its homology modeling for rational structure-based design of selective and high-affinity ligands. In this study, we applied ligand docking and molecular dynamics refinement to a P2Y14R homology model to qualitatively explain structure-activity relationships of previously published synthetic nucleotide analogues and to probe the quality of P2Y14R homology modeling as a template for structure-based design. The P2Y14R model supports the hypothesis of a conserved binding mode of nucleotides in the three P2Y12-like receptors involving functionally conserved residues. We predict phosphate group interactions with R253(6.55), K277(7.35), Y256(6.58) and Q260(6.62), nucleobase (anti-conformation) π-π stacking with Y102(3.33) and the role of F191(5.42) as a means for selectivity among P2Y12-like receptors. The glucose moiety of UDP-glucose docked in a secondary subpocket at the P2Y14R homology model. Thus, P2Y14R homology modeling may allow detailed prediction of interactions to facilitate the design of high affinity, selective agonists as pharmacological tools to study the P2Y14R.
Collapse
Affiliation(s)
- Kevin Trujillo
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, MD 20892-0810, USA
| | - Silvia Paoletta
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, MD 20892-0810, USA
| | - Evgeny Kiselev
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, MD 20892-0810, USA
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, MD 20892-0810, USA.
| |
Collapse
|
3308
|
Wang Q, Edupuganti R, Tavares CDJ, Dalby KN, Ren P. Using docking and alchemical free energy approach to determine the binding mechanism of eEF2K inhibitors and prioritizing the compound synthesis. Front Mol Biosci 2015; 2:9. [PMID: 25988177 PMCID: PMC4429643 DOI: 10.3389/fmolb.2015.00009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/03/2015] [Indexed: 01/09/2023] Open
Abstract
A-484954 is a known eEF2K inhibitor with submicromolar IC50 potency. However, the binding mechanism and the crystal structure of the kinase remains unknown. Here, we employ a homology eEF2K model, docking and alchemical free energy simulations to probe the binding mechanism of eEF2K, and in turn, guide the optimization of potential lead compounds. The inhibitor was docked into the ATP-binding site of a homology model first. Three different binding poses, hypothesis 1, 2, and 3, were obtained and subsequently applied to molecular dynamics (MD) based alchemical free energy simulations. The calculated relative binding free energy of the analogs of A-484954 using the binding pose of hypothesis 1 show a good correlation with the experimental IC50 values, yielding an r2 coefficient of 0.96 after removing an outlier (compound 5). Calculations using another two poses show little correlation with experimental data, (r2 of less than 0.5 with or without removing any outliers). Based on hypothesis 1, the calculated relative free energy suggests that bigger cyclic groups, at R1 e.g., cyclobutyl and cyclopentyl promote more favorable binding than smaller groups, such as cyclopropyl and hydrogen. Moreover, this study also demonstrates the ability of the alchemical free energy approach in combination with docking and homology modeling to prioritize compound synthesis. This can be an effective means of facilitating structure-based drug design when crystal structures are not available.
Collapse
Affiliation(s)
- Qiantao Wang
- Division of Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin Austin, TX, USA ; Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin Austin, TX, USA
| | - Ramakrishna Edupuganti
- Division of Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin Austin, TX, USA
| | - Clint D J Tavares
- Graduate Program in Cell and Molecular Biology, The University of Texas at Austin Austin, TX, USA
| | - Kevin N Dalby
- Division of Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin Austin, TX, USA ; Graduate Program in Cell and Molecular Biology, The University of Texas at Austin Austin, TX, USA
| | - Pengyu Ren
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin Austin, TX, USA
| |
Collapse
|
3309
|
Hill BL, Wong J, May BM, Huerta FB, Manley TE, Sullivan PRF, Olsen KW, Ballicora MA. Conserved residues of the Pro103-Arg115 loop are involved in triggering the allosteric response of the Escherichia coli ADP-glucose pyrophosphorylase. Protein Sci 2015; 24:714-28. [PMID: 25620658 DOI: 10.1002/pro.2644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 11/09/2022]
Abstract
The synthesis of glycogen in bacteria and starch in plants is allosterically controlled by the production of ADP-glucose by ADP-glucose pyrophosphorylase. Using computational studies, site-directed mutagenesis, and kinetic characterization, we found a critical region for transmitting the allosteric signal in the Escherichia coli ADP-glucose pyrophosphorylase. Molecular dynamics simulations and structural comparisons with other ADP-glucose pyrophosphorylases provided information to hypothesize that a Pro103-Arg115 loop is part of an activation path. It had strongly correlated movements with regions of the enzyme associated with regulation and ATP binding, and a network analysis showed that the optimal network pathways linking ATP and the activator binding Lys39 mainly involved residues of this loop. This hypothesis was biochemically tested by mutagenesis. We found that several alanine mutants of the Pro103-Arg115 loop had altered activation profiles for fructose-1,6-bisphosphate. Mutants P103A, Q106A, R107A, W113A, Y114A, and R115A had the most altered kinetic profiles, primarily characterized by a lack of response to fructose-1,6-bisphosphate. This loop is a distinct insertional element present only in allosterically regulated sugar nucleotide pyrophosphorylases that could have been acquired to build a triggering mechanism to link proto-allosteric and catalytic sites.
Collapse
Affiliation(s)
- Benjamin L Hill
- Department of Chemistry and Biochemistry, Loyola University Chicago, 1068 W Sheridan Road, Chicago, Illinois
| | | | | | | | | | | | | | | |
Collapse
|
3310
|
Feig M, Harada R, Mori T, Yu I, Takahashi K, Sugita Y. Complete atomistic model of a bacterial cytoplasm for integrating physics, biochemistry, and systems biology. J Mol Graph Model 2015; 58:1-9. [PMID: 25765281 DOI: 10.1016/j.jmgm.2015.02.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/18/2015] [Accepted: 02/22/2015] [Indexed: 01/10/2023]
Abstract
A model for the cytoplasm of Mycoplasma genitalium is presented that integrates data from a variety of sources into a physically and biochemically consistent model. Based on gene annotations, core genes expected to be present in the cytoplasm were determined and a metabolic reaction network was reconstructed. The set of cytoplasmic genes and metabolites from the predicted reactions were assembled into a comprehensive atomistic model consisting of proteins with predicted structures, RNA, protein/RNA complexes, metabolites, ions, and solvent. The resulting model bridges between atomistic and cellular scales, between physical and biochemical aspects, and between structural and systems views of cellular systems and is meant as a starting point for a variety of simulation studies.
Collapse
Affiliation(s)
- Michael Feig
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Department of Chemistry, Michigan State University, East Lansing, MI 48824, United States; Quantitative Biology Center, RIKEN, International Medical Device Alliance (IMDA) 6F, 1-6-5 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Ryuhei Harada
- Advanced Institute for Computational Science, RIKEN, 7-1-26 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Quantitative Biology Center, RIKEN, International Medical Device Alliance (IMDA) 6F, 1-6-5 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takaharu Mori
- Quantitative Biology Center, RIKEN, International Medical Device Alliance (IMDA) 6F, 1-6-5 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Theoretical Molecular Science Laboratory and iTHES, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Isseki Yu
- Theoretical Molecular Science Laboratory and iTHES, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Koichi Takahashi
- Quantitative Biology Center, RIKEN, Laboratory for Biochemical Simulation, Suita, Osaka 565-0874, Japan; Institute for Advanced Biosciences, Keio University, Fujisawa 252-8520, Japan
| | - Yuji Sugita
- Advanced Institute for Computational Science, RIKEN, 7-1-26 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Quantitative Biology Center, RIKEN, International Medical Device Alliance (IMDA) 6F, 1-6-5 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Theoretical Molecular Science Laboratory and iTHES, RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| |
Collapse
|
3311
|
Lakkaraju SK, Yu W, Raman EP, Hershfeld AV, Fang L, Deshpande DA, MacKerell AD. Mapping functional group free energy patterns at protein occluded sites: nuclear receptors and G-protein coupled receptors. J Chem Inf Model 2015; 55:700-8. [PMID: 25692383 PMCID: PMC4372819 DOI: 10.1021/ci500729k] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Occluded ligand-binding pockets (LBP)
such as those found in nuclear
receptors (NR) and G-protein coupled receptors (GPCR) represent a
significant opportunity and challenge for computer-aided drug design.
To determine free energies maps of functional groups of these LBPs,
a Grand-Canonical Monte Carlo/Molecular Dynamics (GCMC/MD) strategy
is combined with the Site Identification by Ligand Competitive Saturation
(SILCS) methodology. SILCS-GCMC/MD is shown to map functional group
affinity patterns that recapitulate locations of functional groups
across diverse classes of ligands in the LBPs of the androgen (AR)
and peroxisome proliferator-activated-γ (PPARγ) NRs and
the metabotropic glutamate (mGluR) and β2-adreneric
(β2AR) GPCRs. Inclusion of protein flexibility identifies
regions of the binding pockets not accessible in crystal conformations
and allows for better quantitative estimates of relative ligand binding
affinities in all the proteins tested. Differences in functional group
requirements of the active and inactive states of the β2AR LBP were used in virtual screening to identify high efficacy
agonists targeting β2AR in Airway Smooth Muscle (ASM)
cells. Seven of the 15 selected ligands were found to effect ASM relaxation
representing a 46% hit rate. Hence, the method will be of use for
the rational design of ligands in the context of chemical biology
and the development of therapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | | | - Deepak A Deshpande
- §Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, 1020 Locust Street, Philadelphia, Pennsylvania 19107, United States
| | | |
Collapse
|
3312
|
Molecular dynamics, monte carlo simulations, and langevin dynamics: a computational review. BIOMED RESEARCH INTERNATIONAL 2015; 2015:183918. [PMID: 25785262 PMCID: PMC4345249 DOI: 10.1155/2015/183918] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/05/2014] [Indexed: 01/08/2023]
Abstract
Macromolecular structures, such as neuraminidases, hemagglutinins, and monoclonal antibodies, are not rigid entities. Rather, they are characterised by their flexibility, which is the result of the interaction and collective motion of their constituent atoms. This conformational diversity has a significant impact on their physicochemical and biological properties. Among these are their structural stability, the transport of ions through the M2 channel, drug resistance, macromolecular docking, binding energy, and rational epitope design. To assess these properties and to calculate the associated thermodynamical observables, the conformational space must be efficiently sampled and the dynamic of the constituent atoms must be simulated. This paper presents algorithms and techniques that address the abovementioned issues. To this end, a computational review of molecular dynamics, Monte Carlo simulations, Langevin dynamics, and free energy calculation is presented. The exposition is made from first principles to promote a better understanding of the potentialities, limitations, applications, and interrelations of these computational methods.
Collapse
|
3313
|
Raman EP, MacKerell AD. Spatial analysis and quantification of the thermodynamic driving forces in protein-ligand binding: binding site variability. J Am Chem Soc 2015; 137:2608-21. [PMID: 25625202 DOI: 10.1021/ja512054f] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The thermodynamic driving forces behind small molecule-protein binding are still not well-understood, including the variability of those forces associated with different types of ligands in different binding pockets. To better understand these phenomena we calculate spatially resolved thermodynamic contributions of the different molecular degrees of freedom for the binding of propane and methanol to multiple pockets on the proteins Factor Xa and p38 MAP kinase. Binding thermodynamics are computed using a statistical thermodynamics based end-point method applied on a canonical ensemble comprising the protein-ligand complexes and the corresponding free states in an explicit solvent environment. Energetic and entropic contributions of water and ligand degrees of freedom computed from the configurational ensemble provide an unprecedented level of detail into the mechanisms of binding. Direct protein-ligand interaction energies play a significant role in both nonpolar and polar binding, which is comparable to water reorganization energy. Loss of interactions with water upon binding strongly compensates these contributions leading to relatively small binding enthalpies. For both solutes, the entropy of water reorganization is found to favor binding in agreement with the classical view of the "hydrophobic effect". Depending on the specifics of the binding pocket, both energy-entropy compensation and reinforcement mechanisms are observed. It is notable to have the ability to visualize the spatial distribution of the thermodynamic contributions to binding at atomic resolution showing significant differences in the thermodynamic contributions of water to the binding of propane versus methanol.
Collapse
Affiliation(s)
- E Prabhu Raman
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy , 20 Penn Street HSF II, Baltimore, Maryland 21201, United States
| | | |
Collapse
|
3314
|
Villarreal OD, Chen LY, Whetten RL, Yacaman MJ. Ligand-modulated interactions between charged monolayer-protected Au144(SR)60 gold nanoparticles in physiological saline. Phys Chem Chem Phys 2015; 17:3680-8. [PMID: 25556346 PMCID: PMC4303505 DOI: 10.1039/c4cp05137h] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In order to determine how functionalized gold nanoparticles (AuNPs) interact in a near-physiological environment, we performed all-atom molecular dynamics simulations on the icosahedral Au144 nanoparticles each coated with a homogeneous set of 60 thiolates selected from one of these five (5) types: 11-mercapto-1-undecanesulfonate -SC11H22(SO3(-)), 5-mercapto-1-pentanesulfonate -SC5H10(SO3(-)), 5-mercapto-1-pentaneamine -SC5H10(NH3(+)), 4-mercapto-benzoate -SPh(COO(-)), or 4-mercapto-benzamide -SPh(CONH3(+)). These thiolates were selected to elucidate how the aggregation behavior of AuNPs depends on ligand parameters, including the charge of the terminal group (anionic vs. cationic), and its length and conformational flexibility. For this purpose, each functionalized AuNP was paired with a copy of itself, placed in an aqueous cell, neutralized by 120 Na(+)/Cl(-) counter-ions and salinated with a 150 mM concentration of NaCl, to form five (5) systems of like-charged AuNPs pairs in a saline. We computed the potential of mean force (the reversible work of separation) as a function of the intra-pair distance and, based on which, the aggregation affinities. We found that the AuNPs coated with negatively charged, short ligands have very high affinities. Structurally, a significant number of Na(+) counter-ions reside on a plane between the AuNPs, mediating the interaction. Each such ion forms a "salt bridge" (or "ionic bonds") to both of the AuNPs when they are separated by its diameter plus 0.2-0.3 nm. The positively charged AuNPs have much weaker affinities, as Cl(-) counter-ions form fewer and weaker salt bridges between the AuNPs. In the case of Au144(SC11H22(SO3(-)))60 pair, the flexible ligands fluctuate much more than the other four cases. The large fluctuations disfavor the forming of salt bridges between two AuNPs, but enable hydrophobic contact between the exposed hydrocarbon chains of the two AuNPs, which are subject to an effective attraction at a separation much greater than the AuNP diameter and involve a higher concentration of counter ions in the inter-pair space.
Collapse
Affiliation(s)
- Oscar D Villarreal
- Department of Physics and Astronomy, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA.
| | | | | | | |
Collapse
|
3315
|
Mirjalili V, Feig M. Interactions of amino acid side-chain analogs within membrane environments. J Phys Chem B 2015; 119:2877-85. [PMID: 25621811 DOI: 10.1021/jp511712u] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The interactions among four amino acid analog pairs (Asn, Ser, Phe, and Val) within the membrane environment were investigated using umbrella sampling molecular dynamics simulations. The results confirm generally expected qualitative trends of preferential association of polar compounds inside the membrane vs preferential interaction of hydrophobic compounds outside the membrane. Furthermore, correlations between amino acid interactions, membrane insertion, and membrane deformations are discussed and a detailed analysis of pair interaction energies is presented. A comparison of the energetics obtained from explicit lipid simulations with those from implicit membrane models reveals significant deviations and an improved parametrization of the heterogeneous dielectric generalized Born implicit model is provided that partially corrects for deficiencies in the implicit membrane model when compared with the new reference data from this study.
Collapse
Affiliation(s)
- Vahid Mirjalili
- Department of Mechanical Engineering, ‡Department of Biochemistry and Molecular Biology, and §Department of Chemistry, Michigan State University , East Lansing, Michigan 48824, United States
| | | |
Collapse
|
3316
|
Yu W, Lakkaraju SK, Raman EP, Fang L, MacKerell AD. Pharmacophore modeling using site-identification by ligand competitive saturation (SILCS) with multiple probe molecules. J Chem Inf Model 2015; 55:407-20. [PMID: 25622696 DOI: 10.1021/ci500691p] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Receptor-based pharmacophore modeling is an efficient computer-aided drug design technique that uses the structure of the target protein to identify novel leads. However, most methods consider protein flexibility and desolvation effects in a very approximate way, which may limit their use in practice. The Site-Identification by Ligand Competitive Saturation (SILCS) assisted pharmacophore modeling protocol (SILCS-Pharm) was introduced recently to address these issues, as SILCS naturally takes both protein flexibility and desolvation effects into account by using full molecular dynamics simulations to determine 3D maps of the functional group-affinity patterns on a target receptor. In the present work, the SILCS-Pharm protocol is extended to use a wider range of probe molecules including benzene, propane, methanol, formamide, acetaldehyde, methylammonium, acetate and water. This approach removes the previous ambiguity brought by using water as both the hydrogen-bond donor and acceptor probe molecule. The new SILCS-Pharm protocol is shown to yield improved screening results, as compared to the previous approach based on three target proteins. Further validation of the new protocol using five additional protein targets showed improved screening compared to those using common docking methods, further indicating improvements brought by the explicit inclusion of additional feature types associated with the wider collection of probe molecules in the SILCS simulations. The advantage of using complementary features and volume constraints, based on exclusion maps of the protein defined from the SILCS simulations, is presented. In addition, reranking using SILCS-based ligand grid free energies is shown to enhance the diversity of identified ligands for the majority of targets. These results suggest that the SILCS-Pharm protocol will be of utility in rational drug design.
Collapse
Affiliation(s)
- Wenbo Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland , Baltimore, Maryland 21201, United States
| | | | | | | | | |
Collapse
|
3317
|
Sun X, Cheng J, Wang X, Tang Y, Ågren H, Tu Y. Residues remote from the binding pocket control the antagonist selectivity towards the corticotropin-releasing factor receptor-1. Sci Rep 2015; 5:8066. [PMID: 25628267 PMCID: PMC4308710 DOI: 10.1038/srep08066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/02/2015] [Indexed: 01/01/2023] Open
Abstract
The corticotropin releasing factors receptor-1 and receptor-2 (CRF1R and CRF2R) are therapeutic targets for treating neurological diseases. Antagonists targeting CRF1R have been developed for the potential treatment of anxiety disorders and alcohol addiction. It has been found that antagonists targeting CRF1R always show high selectivity, although CRF1R and CRF2R share a very high rate of sequence identity. This has inspired us to study the origin of the selectivity of the antagonists. We have therefore built a homology model for CRF2R and carried out unbiased molecular dynamics and well-tempered metadynamics simulations for systems with the antagonist CP-376395 in CRF1R or CRF2R to address this issue. We found that the side chain of Tyr(6.63) forms a hydrogen bond with the residue remote from the binding pocket, which allows Tyr(6.63) to adopt different conformations in the two receptors and results in the presence or absence of a bottleneck controlling the antagonist binding to or dissociation from the receptors. The rotameric switch of the side chain of Tyr356(6.63) allows the breaking down of the bottleneck and is a perquisite for the dissociation of CP-376395 from CRF1R.
Collapse
Affiliation(s)
- Xianqiang Sun
- Division of Theoretical Chemistry and Biology, School of Biotechnology, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| | - Jianxin Cheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xu Wang
- Division of Theoretical Chemistry and Biology, School of Biotechnology, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hans Ågren
- Division of Theoretical Chemistry and Biology, School of Biotechnology, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| | - Yaoquan Tu
- Division of Theoretical Chemistry and Biology, School of Biotechnology, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| |
Collapse
|
3318
|
Equilibrated atomic models of outward-facing P-glycoprotein and effect of ATP binding on structural dynamics. Sci Rep 2015; 5:7880. [PMID: 25600711 PMCID: PMC4389535 DOI: 10.1038/srep07880] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/16/2014] [Indexed: 12/14/2022] Open
Abstract
P-glycoprotein (Pgp) is an ATP-binding cassette (ABC) transporter that alternates between inward- and outward-facing conformations to capture and force substrates out of cells like a peristaltic pump. The high degree of similarity in outward-facing structures across evolution of ABC transporters allowed construction of a high-confidence outward-facing Pgp atomic model based on crystal structures of outward-facing Sav1866 and inward-facing Pgp. The model adhered to previous experimentally determined secondary- and tertiary- configurations during all-atom molecular dynamics simulations in the presence or absence of MgATP. Three long lasting (>100 ns) meta-stable states were apparent in the presence of MgATP revealing new insights into alternating access. The two ATP-binding pockets are highly asymmetric resulting in differential control of overall structural dynamics and allosteric regulation of the drug-binding pocket. Equilibrated Pgp has a considerably different electrostatic profile compared to Sav1866 that implicates significant kinetic and thermodynamic differences in transport mechanisms.
Collapse
|
3319
|
Müller CS, Knehans T, Davydov DR, Bounds PL, von Mandach U, Halpert JR, Caflisch A, Koppenol WH. Concurrent cooperativity and substrate inhibition in the epoxidation of carbamazepine by cytochrome P450 3A4 active site mutants inspired by molecular dynamics simulations. Biochemistry 2015; 54:711-21. [PMID: 25545162 PMCID: PMC4310618 DOI: 10.1021/bi5011656] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Cytochrome P450 3A4 (CYP3A4) is the
major human P450 responsible
for the metabolism of carbamazepine (CBZ). To explore the mechanisms
of interactions of CYP3A4 with this anticonvulsive drug, we carried
out multiple molecular dynamics (MD) simulations, starting with the
complex of CYP3A4 manually docked with CBZ. On the basis of these
simulations, we engineered CYP3A4 mutants I369F, I369L, A370V, and
A370L, in which the productive binding orientation was expected to
be stabilized, thus leading to increased turnover of CBZ to the 10,11-epoxide
product. In addition, we generated CYP3A4 mutant S119A as a control
construct with putative destabilization of the productive binding
pose. Evaluation of the kinetics profiles of CBZ epoxidation demonstrate
that CYP3A4-containing bacterial membranes (bactosomes) as well as
purified CYP3A4 (wild-type and mutants I369L/F) exhibit substrate
inhibition in reconstituted systems. In contrast, mutants S119A and
A370V/L exhibit S-shaped profiles that are indicative of homotropic
cooperativity. MD simulations with two to four CBZ molecules provide
evidence that the substrate-binding pocket of CYP3A4 can accommodate
more than one molecule of CBZ. Analysis of the kinetics profiles of
CBZ metabolism with a model that combines the formalism of the Hill
equation with an allowance for substrate inhibition demonstrates that
the mechanism of interactions of CBZ with CYP3A4 involves multiple
substrate-binding events (most likely three). Despite the retention
of the multisite binding mechanism in the mutants, functional manifestations
reveal an exquisite sensitivity to even minor structural changes in
the binding pocket that are introduced by conservative substitutions
such as I369F, I369L, and A370V.
Collapse
Affiliation(s)
- Christian S Müller
- Department of Obstetrics, University Hospital Zurich , Zürich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
3320
|
Chen LY. Erythritol predicted to inhibit permeation of water and solutes through the conducting pore of P. falciparum aquaporin. Biophys Chem 2015; 198:14-21. [PMID: 25637890 DOI: 10.1016/j.bpc.2015.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 01/04/2015] [Accepted: 01/07/2015] [Indexed: 12/21/2022]
Abstract
Plasmodium falciparum aquaporin (PfAQP) is a multifunctional channel protein in the plasma membrane of the malarial parasite that causes the most severe form of malaria infecting more than a million people a year. This channel protein facilitates transport of water and several solutes across the cell membrane. In order to better elucidate the fundamental interactions between PfAQP and its permeants and among the permeants, I conducted over three microseconds in silico experiments of atomistic models of the PfAQP-membrane system to obtain the free-energy profiles of five permeants (erythritol, water, glycerol, urea, and ammonia) throughout the amphipathic conducting pore of PfAQP. The profiles are analyzed in light of and shown to be consistent with the existent in vitro data. The binding affinities are computed using the free-energy profiles and the permeant fluctuations inside the channel. On this basis, it is predicted that erythritol, a permeant of PfAQP itself having a deep ditch in its permeation passageway, inhibits PfAQP's functions of transporting water and other solutes with an IC50 in the range of high nanomolars. This leads to the possibility that erythritol, a sweetener generally considered safe, may inhibit or kill the malarial parasite in vivo without causing undesired side effects. Experimental studies are hereby called for to directly test this theoretical prediction of erythritol strongly inhibiting PfAQP in vitro and possibly inhibiting P. falciparum in vivo.
Collapse
Affiliation(s)
- Liao Y Chen
- Department of Physics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA.
| |
Collapse
|
3321
|
Holden JK, Kang S, Hollingsworth SA, Li H, Lim N, Chen S, Huang H, Xue F, Tang W, Silverman RB, Poulos TL. Structure-based design of bacterial nitric oxide synthase inhibitors. J Med Chem 2015; 58:994-1004. [PMID: 25522110 PMCID: PMC4306518 DOI: 10.1021/jm501723p] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
![]()
Inhibition
of bacterial nitric oxide synthase (bNOS) has the potential to improve
the efficacy of antimicrobials used to treat infections by Gram-positive
pathogens Staphylococcus aureus and Bacillus anthracis. However, inhibitor specificity
toward bNOS over the mammalian NOS (mNOS) isoforms remains a challenge
because of the near identical NOS active sites. One key structural
difference between the NOS isoforms is the amino acid composition
of the pterin cofactor binding site that is adjacent to the NOS active
site. Previously, we demonstrated that a NOS inhibitor targeting both
the active and pterin sites was potent and functioned as an antimicrobial
(Holden, , Proc. Natl. Acad.
Sci. U.S.A.2013, 110, 1812724145412). Here we present additional crystal structures, binding
analyses, and bacterial killing studies of inhibitors that target
both the active and pterin sites of a bNOS and function as antimicrobials.
Together, these data provide a framework for continued development
of bNOS inhibitors, as each molecule represents an excellent chemical
scaffold for the design of isoform selective bNOS inhibitors.
Collapse
Affiliation(s)
- Jeffrey K Holden
- Departments of Molecular Biology and Biochemistry, Pharmaceutical Sciences and Chemistry, University of California , 2206 Nat. Sci. 1, Irvine, California 92697-3900, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3322
|
Lopes PEM, Guvench O, MacKerell AD. Current status of protein force fields for molecular dynamics simulations. Methods Mol Biol 2015; 1215:47-71. [PMID: 25330958 PMCID: PMC4554537 DOI: 10.1007/978-1-4939-1465-4_3] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The current status of classical force fields for proteins is reviewed. These include additive force fields as well as the latest developments in the Drude and AMOEBA polarizable force fields. Parametrization strategies developed specifically for the Drude force field are described and compared with the additive CHARMM36 force field. Results from molecular simulations of proteins and small peptides are summarized to illustrate the performance of the Drude and AMOEBA force fields.
Collapse
Affiliation(s)
- Pedro E M Lopes
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, 20 Penn Street HSFII, Baltimore, MD, 21201, USA
| | | | | |
Collapse
|
3323
|
Site Identification by Ligand Competitive Saturation (SILCS) simulations for fragment-based drug design. Methods Mol Biol 2015; 1289:75-87. [PMID: 25709034 DOI: 10.1007/978-1-4939-2486-8_7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Fragment-based drug design (FBDD) involves screening low molecular weight molecules ("fragments") that correspond to functional groups found in larger drug-like molecules to determine their binding to target proteins or nucleic acids. Based on the principle of thermodynamic additivity, two fragments that bind nonoverlapping nearby sites on the target can be combined to yield a new molecule whose binding free energy is the sum of those of the fragments. Experimental FBDD approaches, like NMR and X-ray crystallography, have proven very useful but can be expensive in terms of time, materials, and labor. Accordingly, a variety of computational FBDD approaches have been developed that provide different levels of detail and accuracy.The Site Identification by Ligand Competitive Saturation (SILCS) method of computational FBDD uses all-atom explicit-solvent molecular dynamics (MD) simulations to identify fragment binding. The target is "soaked" in an aqueous solution with multiple fragments having different identities. The resulting computational competition assay reveals what small molecule types are most likely to bind which regions of the target. From SILCS simulations, 3D probability maps of fragment binding called "FragMaps" can be produced. Based on the probabilities relative to bulk, SILCS FragMaps can be used to determine "Grid Free Energies (GFEs)," which provide per-atom contributions to fragment binding affinities. For essentially no additional computational overhead relative to the production of the FragMaps, GFEs can be used to compute Ligand Grid Free Energies (LGFEs) for arbitrarily complex molecules, and these LGFEs can be used to rank-order the molecules in accordance with binding affinities.
Collapse
|
3324
|
Abstract
Molecular dynamics simulations are an effective tool to study the structure, dynamics, and thermodynamics of carbohydrates and proteins. However, the simulations of heterogeneous glycoprotein systems have been limited due to the lack of appropriate molecular force field parameters describing the linkage between the carbohydrate and the protein regions as well as the tools to prepare these systems for modeling studies. In this work we outline the recent developments in the CHARMM carbohydrate force field to treat glycoproteins and describe in detail the step-by-step procedures involved in building glycoprotein geometries using CHARMM-GUI Glycan Reader.
Collapse
Affiliation(s)
- Sairam S. Mallajosyula
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn St., HSF II-629, Baltimore, MD 21201
| | - Sunhwan Jo
- Department of Molecular Biosciences and Center for Bioinformatics, The University of Kansas, 2030 Becker Drive Lawrence, KS 66047, USA
| | - Wonpil Im
- Department of Molecular Biosciences and Center for Bioinformatics, The University of Kansas, 2030 Becker Drive Lawrence, KS 66047, USA
| | - Alexander D. MacKerell
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn St., HSF II-629, Baltimore, MD 21201
| |
Collapse
|
3325
|
Wu Y, Canturk B, Jo H, Ma C, Gianti E, Klein M, Pinto LH, Lamb RA, Fiorin G, Wang J, DeGrado WF. Flipping in the pore: discovery of dual inhibitors that bind in different orientations to the wild-type versus the amantadine-resistant S31N mutant of the influenza A virus M2 proton channel. J Am Chem Soc 2014; 136:17987-95. [PMID: 25470189 PMCID: PMC4286326 DOI: 10.1021/ja508461m] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Indexed: 12/13/2022]
Abstract
Influenza virus infections lead to numerous deaths and millions of hospitalizations each year. One challenge facing anti-influenza drug development is the heterogeneity of the circulating influenza viruses, which comprise several strains with variable susceptibility to antiviral drugs. For example, the wild-type (WT) influenza A viruses, such as the seasonal H1N1, tend to be sensitive to antiviral drugs, amantadine and rimantadine, while the S31N mutant viruses, such as the pandemic 2009 H1N1 (H1N1pdm09) and seasonal H3N2, are resistant to this class of drugs. Thus, drugs targeting both WT and the S31N mutant are highly desired. We report our design of a novel class of dual inhibitors along with their ion channel blockage and antiviral activities. The potency of the most active compound 11 in inhibiting WT and the S31N mutant influenza viruses is comparable with that of amantadine in inhibiting WT influenza virus. Solution NMR studies and molecular dynamics (MD) simulations of drug-M2 interactions supported our design hypothesis: namely, the dual inhibitor binds in the WT M2 channel with an aromatic group facing down toward the C-terminus, while the same drug binds in the S31N M2 channel with its aromatic group facing up toward the N-terminus. The flip-flop mode of drug binding correlates with the structure-activity relationship (SAR) and has paved the way for the next round of rational design of broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- Yibing Wu
- Department
of Pharmaceutical Chemistry, University
of California, Mission Bay Box 3122, San Francisco, California 94158, United States
| | - Belgin Canturk
- Department
of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Hyunil Jo
- Department
of Pharmaceutical Chemistry, University
of California, Mission Bay Box 3122, San Francisco, California 94158, United States
| | - Chunlong Ma
- Department
of Pharmacology and Toxicology and the BIO5 Institute, The University of Arizona, 1501 N. Campbell Avenue, Tucson, Arizona 85721, United States
| | - Eleonora Gianti
- Institute
for Computational and Molecular Science, Science Education and Research
Center (035-07), Temple University, 1925 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | - Michael
L. Klein
- Institute
for Computational and Molecular Science, Science Education and Research
Center (035-07), Temple University, 1925 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | - Lawrence H. Pinto
- Department
of Neurobiology, Northwestern University, 2205 Tech Drive, Evanston, Illinois 60208, United States
| | - Robert A. Lamb
- Department
of Molecular Biosciences, Northwestern University, 2205 Tech Drive, Evanston, Illinois 60208, United States
- Howard
Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Giacomo Fiorin
- Institute
for Computational and Molecular Science, Science Education and Research
Center (035-07), Temple University, 1925 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | - Jun Wang
- Department
of Pharmacology and Toxicology and the BIO5 Institute, The University of Arizona, 1501 N. Campbell Avenue, Tucson, Arizona 85721, United States
| | - William F. DeGrado
- Department
of Pharmaceutical Chemistry, University
of California, Mission Bay Box 3122, San Francisco, California 94158, United States
| |
Collapse
|
3326
|
Cheng B, Ceriotti M. Direct path integral estimators for isotope fractionation ratios. J Chem Phys 2014; 141:244112. [DOI: 10.1063/1.4904293] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
3327
|
Ou SC, Cui D, Patel S. Association of alkanes with the aqueous liquid-vapor interface: a reference system for interpreting hydrophobicity generally through interfacial fluctuations. Phys Chem Chem Phys 2014; 16:26779-85. [PMID: 25372502 DOI: 10.1039/c4cp03170a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report free energy calculations and fluctuation profiles of single alkanes (from methane to pentane) along the direction normal to the air-water interface. The induced fluctuations and the interfacial stabilities of alkanes are found to be correlated and similar to the results of inorganic monovalent ions (Ou et al., J. Phys. Chem. B, 2013, 117, 11732). This suggests that hydrophobic solvation of solutes and ions is important in determining the adsorption behavior.
Collapse
Affiliation(s)
- Shu-Ching Ou
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA.
| | | | | |
Collapse
|
3328
|
Tuszynski JA, Winter P, White D, Tseng CY, Sahu KK, Gentile F, Spasevska I, Omar SI, Nayebi N, Churchill CDM, Klobukowski M, El-Magd RMA. Mathematical and computational modeling in biology at multiple scales. Theor Biol Med Model 2014; 11:52. [PMID: 25542608 PMCID: PMC4396153 DOI: 10.1186/1742-4682-11-52] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/25/2014] [Indexed: 01/08/2023] Open
Abstract
A variety of topics are reviewed in the area of mathematical and computational modeling in biology, covering the range of scales from populations of organisms to electrons in atoms. The use of maximum entropy as an inference tool in the fields of biology and drug discovery is discussed. Mathematical and computational methods and models in the areas of epidemiology, cell physiology and cancer are surveyed. The technique of molecular dynamics is covered, with special attention to force fields for protein simulations and methods for the calculation of solvation free energies. The utility of quantum mechanical methods in biophysical and biochemical modeling is explored. The field of computational enzymology is examined.
Collapse
Affiliation(s)
- Jack A Tuszynski
- />Department of Physics and Department of Oncology, University of Alberta, Edmonton, Canada
| | - Philip Winter
- />Department of Oncology, University of Alberta, Edmonton, Canada
| | - Diana White
- />Department of Oncology, University of Alberta, Edmonton, Canada
| | - Chih-Yuan Tseng
- />Department of Oncology, University of Alberta, Edmonton, Canada
| | - Kamlesh K Sahu
- />Department of Physics, University of Alberta, Edmonton, Canada
| | - Francesco Gentile
- />Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
| | - Ivana Spasevska
- />Department of Biology, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Niloofar Nayebi
- />Department of Physics, University of Alberta, Edmonton, Canada
| | | | | | | |
Collapse
|
3329
|
Systematic study of binding of μ-conotoxins to the sodium channel NaV1.4. Toxins (Basel) 2014; 6:3454-70. [PMID: 25529306 PMCID: PMC4280544 DOI: 10.3390/toxins6123454] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/01/2014] [Accepted: 12/10/2014] [Indexed: 01/25/2023] Open
Abstract
Voltage-gated sodium channels (NaV) are fundamental components of the nervous system. Their dysfunction is implicated in a number of neurological disorders, such as chronic pain, making them potential targets for the treatment of such disorders. The prominence of the NaV channels in the nervous system has been exploited by venomous animals for preying purposes, which have developed toxins that can block the NaV channels, thereby disabling their function. Because of their potency, such toxins could provide drug leads for the treatment of neurological disorders associated with NaV channels. However, most toxins lack selectivity for a given target NaV channel, and improving their selectivity profile among the NaV1 isoforms is essential for their development as drug leads. Computational methods will be very useful in the solution of such design problems, provided accurate models of the protein-ligand complex can be constructed. Using docking and molecular dynamics simulations, we have recently constructed a model for the NaV1.4-μ-conotoxin-GIIIA complex and validated it with the ample mutational data available for this complex. Here, we use the validated NaV1.4 model in a systematic study of binding other μ-conotoxins (PIIIA, KIIIA and BuIIIB) to NaV1.4. The binding mode obtained for each complex is shown to be consistent with the available mutation data and binding constants. We compare the binding modes of PIIIA, KIIIA and BuIIIB to that of GIIIA and point out the similarities and differences among them. The detailed information about NaV1.4-μ-conotoxin interactions provided here will be useful in the design of new NaV channel blocking peptides.
Collapse
|
3330
|
Shanak S, Helms V. Hydration properties of natural and synthetic DNA sequences with methylated adenine or cytosine bases in the R.DpnI target and BDNF promoter studied by molecular dynamics simulations. J Chem Phys 2014; 141:22D512. [PMID: 25494783 DOI: 10.1063/1.4897525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Adenine and cytosine methylation are two important epigenetic modifications of DNA sequences at the levels of the genome and transcriptome. To characterize the differential roles of methylating adenine or cytosine with respect to their hydration properties, we performed conventional MD simulations and free energy perturbation calculations for two particular DNA sequences, namely the brain-derived neurotrophic factor (BDNF) promoter and the R.DpnI-bound DNA that are known to undergo methylation of C5-methyl cytosine and N6-methyl adenine, respectively. We found that a single methylated cytosine has a clearly favorable hydration free energy over cytosine since the attached methyl group has a slightly polar character. In contrast, capping the strongly polar N6 of adenine with a methyl group gives a slightly unfavorable contribution to its free energy of solvation. Performing the same demethylation in the context of a DNA double-strand gave quite similar results for the more solvent-accessible cytosine but much more unfavorable results for the rather buried adenine. Interestingly, the same demethylation reactions are far more unfavorable when performed in the context of the opposite (BDNF or R.DpnI target) sequence. This suggests a natural preference for methylation in a specific sequence context. In addition, free energy calculations for demethylating adenine or cytosine in the context of B-DNA vs. Z-DNA suggest that the conformational B-Z transition of DNA transition is rather a property of cytosine methylated sequences but is not preferable for the adenine-methylated sequences investigated here.
Collapse
Affiliation(s)
- Siba Shanak
- Zentrum für Bioinformatik, Universität des Saarlandes, P.O. Box 15 11 50, 66123 Saarbrücken, Germany
| | - Volkhard Helms
- Zentrum für Bioinformatik, Universität des Saarlandes, P.O. Box 15 11 50, 66123 Saarbrücken, Germany
| |
Collapse
|
3331
|
Wang N, Zhou S, Kekenes-Huskey PM, Li B, McCammon JA. Poisson-Boltzmann versus Size-Modified Poisson-Boltzmann Electrostatics Applied to Lipid Bilayers. J Phys Chem B 2014; 118:14827-32. [PMID: 25426875 PMCID: PMC4280115 DOI: 10.1021/jp511702w] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
![]()
Mean-field
methods, such as the Poisson–Boltzmann equation
(PBE), are often used to calculate the electrostatic properties of
molecular systems. In the past two decades, an enhancement of the
PBE, the size-modified Poisson–Boltzmann equation (SMPBE),
has been reported. Here, the PBE and the SMPBE are reevaluated for
realistic molecular systems, namely, lipid bilayers, under eight different
sets of input parameters. The SMPBE appears to reproduce the molecular
dynamics simulation results better than the PBE only under specific
parameter sets, but in general, it performs no better than the Stern
layer correction of the PBE. These results emphasize the need for
careful discussions of the accuracy of mean-field calculations on
realistic systems with respect to the choice of parameters and call
for reconsideration of the cost-efficiency and the significance of
the current SMPBE formulation.
Collapse
Affiliation(s)
- Nuo Wang
- Department of Chemistry and Biochemistry, ‡Department of Mathematics, §Department of Pharmacology, ⊥Howard Hughes Medical Institute, University of California-San Diego , La Jolla, California 92093, United States
| | - Shenggao Zhou
- Department of Chemistry and Biochemistry, ‡Department of Mathematics, §Department of Pharmacology, ⊥Howard Hughes Medical Institute, University of California-San Diego , La Jolla, California 92093, United States
| | - Peter M Kekenes-Huskey
- Department of Chemistry and Biochemistry, ‡Department of Mathematics, §Department of Pharmacology, ⊥Howard Hughes Medical Institute, University of California-San Diego , La Jolla, California 92093, United States
| | - Bo Li
- Department of Chemistry and Biochemistry, ‡Department of Mathematics, §Department of Pharmacology, ⊥Howard Hughes Medical Institute, University of California-San Diego , La Jolla, California 92093, United States
| | - J Andrew McCammon
- Department of Chemistry and Biochemistry, ‡Department of Mathematics, §Department of Pharmacology, ⊥Howard Hughes Medical Institute, University of California-San Diego , La Jolla, California 92093, United States
| |
Collapse
|
3332
|
Kochanek SE, Clymer TM, Pakkala VS, Hebert SP, Reeping K, Firestine SM, Evanseck JD. Intramolecular charge-assisted hydrogen bond strength in pseudochair carboxyphosphate. J Phys Chem B 2014; 119:1184-91. [PMID: 25405523 PMCID: PMC4306500 DOI: 10.1021/jp506796r] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Carboxyphosphate,
a suspected intermediate in ATP-dependent carboxylases,
has not been isolated nor observed directly by experiment. Consequently,
little is known concerning its structure, stability, and ionization
state. Recently, carboxyphosphate as either a monoanion or dianion
has been shown computationally to adopt a novel pseudochair conformation
featuring an intramolecular charge-assisted hydrogen bond (CAHB).
In this work, additive and subtractive correction schemes to the commonly
employed open–closed method are used to estimate the strength
of the CAHB. Truhlar’s Minnesota M06-2X functional with Dunning’s
aug-cc-pVTZ basis set has been used for geometry optimization, energy
evaluation, and frequency analysis. The CHARMM force field has been
used to approximate the Pauli repulsive terms in the closed and open
forms of carboxyphosphate. From our additive correction scheme, differential
Pauli repulsion contributions between the pseudochair (closed) and
open conformations of carboxyphosphate are found to be significant
in determining the CAHB strength. The additive correction modifies
the CAHB prediction (ΔEclosed–open) of −14 kcal/mol for the monoanion and −12 kcal/mol
for the dianion to −22.9 and −18.4 kcal/mol, respectively.
Results from the subtractive technique reinforce those from our additive
procedure, where the predicted CAHB strength ranges from −17.8
to −25.4 kcal/mol for the monoanion and from −15.7 to
−20.9 kcal/mol for the dianion. Ultimately, we find that the
CAHB in carboxyphosphate meets the criteria for short-strong hydrogen
bonds. However, carboxyphosphate has a unique energy profile that
does not result in the symmetric double-well behavior of low-barrier
hydrogen bonds. These findings provide deeper insight into the pseudochair
conformation of carboxyphosphate, and lead to an improved mechanistic
understanding of this intermediate in ATP-dependent carboxylases.
Collapse
Affiliation(s)
- Sarah E Kochanek
- Center for Computational Sciences and the Department of Chemistry and Biochemistry, Duquesne University , 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282-1530, United States
| | | | | | | | | | | | | |
Collapse
|
3333
|
Babcock JJ, Brancaleon L. The effect of local dynamics of Atto 390-labeled lysozyme on fluorescence anisotropy modeling. Biopolymers 2014; 103:285-95. [PMID: 25431279 DOI: 10.1002/bip.22596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 11/14/2014] [Accepted: 11/18/2014] [Indexed: 11/07/2022]
Abstract
Fluorescence anisotropy decay is a popular optical technique to study the structure, size, shape, and even functions of biomolecules. The method measures the time dependence of the depolarization of a fluorophore and is therefore sensitive to the changes in the rotational motion (e.g., aggregation and binding) or changes in the mobility of segments of biopolymers (such as the ones associated with tertiary structure changes). Fluorescence anisotropy decay often requires the use of fluorescent dyes that need to be covalently attached to the biomolecule. The location of the attachment on the biomolecule (e.g., a protein) and the linker used, affect the mobility of the dye and its anisotropy decay. With this study we have combined the experimental data with molecular dynamic simulations to offer a more correct interpretation of the fluorescence anisotropy decay of a popular fluorescent dye (Atto 390) attached to the N-terminus of Hen Egg White Lysozyme (HEWL). Our model showed how the use of relatively simple molecular dynamics computation to simulate the motion of the dye, provide a model to interpret the experimental fluorescence anisotropy decay that yields a better estimate of the hydrodynamic radius of HEWL. The improvement is provided by a more detailed description of the segmental motion of the dye attached to the protein.
Collapse
Affiliation(s)
- Jeremiah J Babcock
- Department of Physics and Astronomy, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78242
| | | |
Collapse
|
3334
|
Gleed ML, Busath DD. Why bound amantadine fails to inhibit proton conductance according to simulations of the drug-resistant influenza A M2 (S31N). J Phys Chem B 2014; 119:1225-31. [PMID: 25426702 PMCID: PMC4306489 DOI: 10.1021/jp508545d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The mechanisms responsible for drug resistance in the Asn31 variant of the M2 protein of influenza A are not well understood. Molecular dynamics simulations were performed on wild-type (Ser31) and S31N influenza A M2 in the homotetramer configuration. After evaluation of 13 published M2 structures, a solid-state NMR structure with amantadine bound was selected for simulations, an S31N mutant structure was developed and equilibrated, and the native and mutant structures were used to determine the binding behavior of amantadine and the dynamics of water in the two channels. Amantadine is stable in the plugging region of wild-type M2, with the adamantane in contact with the Val27 side chains, while amantadine in S31N M2 has more variable movement and orientation, and spontaneously moves lower into the central cavity of the channel. Free energy profiles from umbrella sampling support this observation. In this configuration, water surrounds the drug and can easily transport protons past it, so the drug binds without blocking proton transport in the S31N M2 channel.
Collapse
Affiliation(s)
- Mitchell L Gleed
- Department of Physiology and Developmental Biology, Brigham Young University , Provo, Utah 84602, United States
| | | |
Collapse
|
3335
|
Poblete H, Oyarzún I, Olivero P, Comer J, Zuñiga M, Sepulveda RV, Báez-Nieto D, González Leon C, González-Nilo F, Latorre R. Molecular determinants of phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) binding to transient receptor potential V1 (TRPV1) channels. J Biol Chem 2014; 290:2086-98. [PMID: 25425643 DOI: 10.1074/jbc.m114.613620] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) has been recognized as an important activator of certain transient receptor potential (TRP) channels. More specifically, TRPV1 is a pain receptor activated by a wide range of stimuli. However, whether or not PI(4,5)P2 is a TRPV1 agonist remains open to debate. Utilizing a combined approach of mutagenesis and molecular modeling, we identified a PI(4,5)P2 binding site located between the TRP box and the S4-S5 linker. At this site, PI(4,5)P2 interacts with the amino acid residues Arg-575 and Arg-579 in the S4-S5 linker and with Lys-694 in the TRP box. We confirmed that PI(4,5)P2 behaves as a channel agonist and found that Arg-575, Arg-579, and Lys-694 mutations to alanine reduce PI(4,5)P2 binding affinity. Additionally, in silico mutations R575A, R579A, and K694A showed that the reduction in binding affinity results from the delocalization of PI(4,5)P2 in the binding pocket. Molecular dynamics simulations indicate that PI(4,5)P2 binding induces conformational rearrangements of the structure formed by S6 and the TRP domain, which cause an opening of the lower TRPV1 channel gate.
Collapse
Affiliation(s)
- Horacio Poblete
- From the Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 2 Norte 685, Talca-Chile
| | - Ingrid Oyarzún
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile
| | - Pablo Olivero
- Escuela de Medicina, Universidad de Valparaíso, Hontaneda 2664, Valparaíso, Chile
| | - Jeffrey Comer
- Institute of Computational Comparative Medicine, Department of Anatomy and Physiology, Kansas State University, P-200 Mosier Hall, Manhattan, Kansas 66506-5802
| | - Matías Zuñiga
- Doctorado Fisicoquímica Molecular, Universidad Andrés Bello, Ave, Republica 275, Santiago, Chile
| | - Romina V Sepulveda
- Doctorado en Biotecnología, Universidad Andrés Bello, Av. Republica 217, Santiago, Chile, and Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. República 239, Santiago, Chile
| | - David Báez-Nieto
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile
| | - Carlos González Leon
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile
| | - Fernando González-Nilo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile, Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. República 239, Santiago, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile,
| |
Collapse
|
3336
|
Gianti E, Carnevale V, DeGrado WF, Klein ML, Fiorin G. Hydrogen-bonded water molecules in the M2 channel of the influenza A virus guide the binding preferences of ammonium-based inhibitors. J Phys Chem B 2014; 119:1173-83. [PMID: 25353315 DOI: 10.1021/jp506807y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tetrameric M2 proton channel of influenza A virus is an integral membrane protein responsible for the acidification of the viral interior. Drugs such as amantadine target the transmembrane region of wild type M2 by acting as pore blockers. However, a number of mutations affecting this domain confer drug resistance, prompting the need for alternative inhibitors. The availability of high-resolution structures of drug-bound M2, paired with computational investigations, revealed that inhibitors can bind at different sites, and provided useful insights in understanding the principles governing proton conduction. Here, we investigated by computation the energetic and geometric factors determining the relative stability of pore blockers at individual sites of different M2 strains. We found that local free energy minima along the translocation pathway of positively charged chemical species correspond to experimentally determined binding sites of inhibitors. Then, by examining the structure of water clusters hydrating each site, as well as of those displaced by binding of hydrophobic scaffolds, we predicted the binding preferences of M2 ligands. This information can be used to guide the identification of novel classes of inhibitors.
Collapse
Affiliation(s)
- Eleonora Gianti
- Institute for Computational Molecular Science, Temple University , SERC Building, 1925 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | | | | | | | | |
Collapse
|
3337
|
Yahya R, Karjiban RA, Basri M, Rahman MBA, Girardi M. Monte Carlo simulation of mixed nonionic Brij surfactants in water. J Mol Model 2014; 20:2512. [PMID: 25381172 DOI: 10.1007/s00894-014-2512-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/21/2014] [Indexed: 11/24/2022]
Abstract
Nonionic surfactants such as the Brij® series are important in the preparation of transdermal drug nanodelivery products using nanoemulsions because of their low toxicity and low irritancy. Here, Monte Carlo (MC) simulation was used to examine the physical behavior of the model deterministic system by using sampling procedures. Metropolis MC simulations were run on three mixtures of two different nonionic surfactants, Brij92 and Brij96, with different compositions in aqueous solution. The system was simulated in the canonical ensemble with constant temperature, volume and number of molecules. Hence, the acceptance ratio for single atom moves of the mixed surfactants increased as the concentration of surfactants increased from 0.494 to 0.591. The lowest total energy for the mixed surfactant systems was -99,039 kcal mol(-1) due to the interaction between all molecules in the system simulated. The physicochemical properties of models such as the radius of gyration and radial distribution function, were also determined. These observations indicate that the behavior and physicochemical of mixed surfactant and PKOEs nanoemulsion systems were described adequately during the simulation.
Collapse
Affiliation(s)
- Ruzanna Yahya
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Selangor, Malaysia
| | | | | | | | | |
Collapse
|
3338
|
Sodt AJ, Mei Y, König G, Tao P, Steele RP, Brooks BR, Shao Y. Multiple environment single system quantum mechanical/molecular mechanical (MESS-QM/MM) calculations. 1. Estimation of polarization energies. J Phys Chem A 2014; 119:1511-23. [PMID: 25321186 PMCID: PMC4353191 DOI: 10.1021/jp5072296] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
In
combined quantum mechanical/molecular mechanical (QM/MM) free
energy calculations, it is often advantageous to have a frozen geometry
for the quantum mechanical (QM) region. For such multiple-environment
single-system (MESS) cases, two schemes are proposed here for estimating
the polarization energy: the first scheme, termed MESS-E, involves
a Roothaan step extrapolation of the self-consistent field (SCF) energy;
whereas the other scheme, termed MESS-H, employs a Newton–Raphson
correction using an approximate inverse electronic Hessian of the
QM region (which is constructed only once). Both schemes are extremely
efficient, because the expensive Fock updates and SCF iterations in
standard QM/MM calculations are completely avoided at each configuration.
They produce reasonably accurate QM/MM polarization energies: MESS-E
can predict the polarization energy within 0.25 kcal/mol in terms
of the mean signed error for two of our test cases, solvated methanol
and solvated β-alanine, using the M06-2X or ωB97X-D functionals;
MESS-H can reproduce the polarization energy within 0.2 kcal/mol for
these two cases and for the oxyluciferin–luciferase complex,
if the approximate inverse electronic Hessians are constructed with
sufficient accuracy.
Collapse
Affiliation(s)
- Alexander J Sodt
- Laboratory of Computational Biology, National Heart, Lung and Blood Institute, National Institutes of Health , 5635 Fishers Lane, T-900 Suite, Rockville, Maryland 20852, United States
| | | | | | | | | | | | | |
Collapse
|
3339
|
Hsu HJ, Sen S, Pearson RM, Uddin S, Král P, Hong S. Poly(ethylene glycol) Corona Chain Length Controls End-Group-Dependent Cell Interactions of Dendron Micelles. Macromolecules 2014; 47:6911-6918. [PMID: 25709141 PMCID: PMC4334293 DOI: 10.1021/ma501258c] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/15/2014] [Indexed: 01/17/2023]
Abstract
To systematically investigate the relationship among surface charge, PEG chain length, and nano-bio interactions of dendron-based micelles (DMs), a series of PEGylated DMs with various end groups (-NH2, -Ac, and -COOH) and PEG chain lengths (600 and 2000 g/mol) are prepared and tested in vitro. The DMs with longer PEG chains (DM2K) do not interact with cells despite their positively charged surfaces. In sharp contrast, the DMs with shorter PEG chains (DM600) exhibit charge-dependent cellular interactions, as observed in both in vitro and molecular dynamics (MD) simulation results. Furthermore, all DMs with different charges display enhanced stability for hydrophobic dye encapsulation compared to conventional linear-block copolymer-based micelles, by allowing only a minimal leakage of the dye in vitro. Our results demonstrate the critical roles of the PEG chain length and polymeric architecture on the terminal charge effect and the stability of micelles, which provides an important design cue for polymeric micelles.
Collapse
Affiliation(s)
- Hao-jui Hsu
- Departments of Biopharmaceutical Sciences, Bioengineering, Chemistry, and Physics, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Soumyo Sen
- Departments of Biopharmaceutical Sciences, Bioengineering, Chemistry, and Physics, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Ryan M. Pearson
- Departments of Biopharmaceutical Sciences, Bioengineering, Chemistry, and Physics, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Sayam Uddin
- Departments of Biopharmaceutical Sciences, Bioengineering, Chemistry, and Physics, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Petr Král
- Departments of Biopharmaceutical Sciences, Bioengineering, Chemistry, and Physics, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Seungpyo Hong
- Departments of Biopharmaceutical Sciences, Bioengineering, Chemistry, and Physics, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| |
Collapse
|
3340
|
Arcario MJ, Mayne CG, Tajkhorshid E. Atomistic models of general anesthetics for use in in silico biological studies. J Phys Chem B 2014; 118:12075-86. [PMID: 25303275 PMCID: PMC4207551 DOI: 10.1021/jp502716m] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
While small molecules have been used
to induce anesthesia in a
clinical setting for well over a century, a detailed understanding
of the molecular mechanism remains elusive. In this study, we utilize
ab initio calculations to develop a novel set of CHARMM-compatible
parameters for the ubiquitous modern anesthetics desflurane, isoflurane,
sevoflurane, and propofol for use in molecular dynamics (MD) simulations.
The parameters generated were rigorously tested against known experimental
physicochemical properties including dipole moment, density, enthalpy
of vaporization, and free energy of solvation. In all cases, the anesthetic
parameters were able to reproduce experimental measurements, signifying
the robustness and accuracy of the atomistic models developed. The
models were then used to study the interaction of anesthetics with
the membrane. Calculation of the potential of mean force for inserting
the molecules into a POPC bilayer revealed a distinct energetic minimum
of 4–5 kcal/mol relative to aqueous solution at the level of
the glycerol backbone in the membrane. The location of this minimum
within the membrane suggests that anesthetics partition to the membrane
prior to binding their ion channel targets, giving context to the
Meyer–Overton correlation. Moreover, MD simulations of these
drugs in the membrane give rise to computed membrane structural parameters,
including atomic distribution, deuterium order parameters, dipole
potential, and lateral stress profile, that indicate partitioning
of anesthetics into the membrane at the concentration range studied
here, which does not appear to perturb the structural integrity of
the lipid bilayer. These results signify that an indirect, membrane-mediated
mechanism of channel modulation is unlikely.
Collapse
Affiliation(s)
- Mark J Arcario
- Center for Biophysics and Computational Biology, Department of Biochemistry, College of Medicine, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | | | | |
Collapse
|
3341
|
Charlier L, Louet M, Chaloin L, Fuchs P, Martinez J, Muriaux D, Favard C, Floquet N. Coarse-grained simulations of the HIV-1 matrix protein anchoring: revisiting its assembly on membrane domains. Biophys J 2014; 106:577-85. [PMID: 24507598 DOI: 10.1016/j.bpj.2013.12.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 11/21/2013] [Accepted: 12/06/2013] [Indexed: 11/25/2022] Open
Abstract
In the accepted model for human immunodeficiency virus preassembly in infected host cells, the anchoring to the intracellular leaflet of the membrane of the matrix domain (MA) that lies at the N-terminus of the viral Gag protein precursor appears to be one of the crucial steps for particle assembly. In this study, we simulated the membrane anchoring of human immunodeficiency virus-1 myristoylated MA protein using a coarse-grained representation of both the protein and the membrane. Our calculations first suggest that the myristoyl group could spontaneously release from its initial hydrophobic pocket before MA protein interacts with the lipid membrane. All-atom simulations confirmed this possibility with a related energy cost estimated to be ~5 kcal.mol(-1). The phosphatidylinositol (4,5) bisphosphate (PI(4,5)P2) head binds preferentially to the MA highly basic region as described in available NMR data, but interestingly without flipping of its 2' acyl chain into the MA protein. Moreover, MA was able to confine PI(4,5)P2 lipids all around its molecular surface after having found a stable orientation at the membrane surface. Our results suggest that this orientation is dependent on Myr anchoring and that this confinement induces a lateral segregation of PI(4,5)P2 in domains. This is consistent with a PI(4,5)P2 enrichment of the virus envelope as compared to the host cell membrane.
Collapse
Affiliation(s)
- Landry Charlier
- Institut des Biomolécules Max Mousseron (IBMM), CNRS UMR5247, Université Montpellier 1, Université Montpellier 2, Faculté de Pharmacie, Montpellier cedex 05, France
| | - Maxime Louet
- Institut des Biomolécules Max Mousseron (IBMM), CNRS UMR5247, Université Montpellier 1, Université Montpellier 2, Faculté de Pharmacie, Montpellier cedex 05, France
| | - Laurent Chaloin
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé (CPBS) CNRS-UMR 5236, Université Montpellier 1 - Université Montpellier 2, Montpellier Cedex 5, France
| | - Patrick Fuchs
- Dynamique des Structures et Interactions des Macromolécules Biologiques INTS, INSERM UMR-S665, Université Paris Diderot, Alexandre Cabanel, Paris
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron (IBMM), CNRS UMR5247, Université Montpellier 1, Université Montpellier 2, Faculté de Pharmacie, Montpellier cedex 05, France
| | - Delphine Muriaux
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé (CPBS) CNRS-UMR 5236, Université Montpellier 1 - Université Montpellier 2, Montpellier Cedex 5, France
| | - Cyril Favard
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé (CPBS) CNRS-UMR 5236, Université Montpellier 1 - Université Montpellier 2, Montpellier Cedex 5, France.
| | - Nicolas Floquet
- Institut des Biomolécules Max Mousseron (IBMM), CNRS UMR5247, Université Montpellier 1, Université Montpellier 2, Faculté de Pharmacie, Montpellier cedex 05, France.
| |
Collapse
|
3342
|
Pevzner Y, Frugier E, Schalk V, Caflisch A, Woodcock HL. Fragment-based docking: development of the CHARMMing Web user interface as a platform for computer-aided drug design. J Chem Inf Model 2014; 54:2612-20. [PMID: 25151852 PMCID: PMC4170817 DOI: 10.1021/ci500322k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Indexed: 01/24/2023]
Abstract
Web-based user interfaces to scientific applications are important tools that allow researchers to utilize a broad range of software packages with just an Internet connection and a browser. One such interface, CHARMMing (CHARMM interface and graphics), facilitates access to the powerful and widely used molecular software package CHARMM. CHARMMing incorporates tasks such as molecular structure analysis, dynamics, multiscale modeling, and other techniques commonly used by computational life scientists. We have extended CHARMMing's capabilities to include a fragment-based docking protocol that allows users to perform molecular docking and virtual screening calculations either directly via the CHARMMing Web server or on computing resources using the self-contained job scripts generated via the Web interface. The docking protocol was evaluated by performing a series of "re-dockings" with direct comparison to top commercial docking software. Results of this evaluation showed that CHARMMing's docking implementation is comparable to many widely used software packages and validates the use of the new CHARMM generalized force field for docking and virtual screening.
Collapse
Affiliation(s)
- Yuri Pevzner
- Department
of Chemistry, University of South Florida, 4202 E. Fowler Ave., CHE205, Tampa, Florida 33620-5250, United States
| | - Emilie Frugier
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Vinushka Schalk
- Department
of Chemistry, University of South Florida, 4202 E. Fowler Ave., CHE205, Tampa, Florida 33620-5250, United States
- Department
of Natural Sciences, New College of Florida, Sarasota, Florida 34243, United States
| | - Amedeo Caflisch
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - H. Lee Woodcock
- Department
of Chemistry, University of South Florida, 4202 E. Fowler Ave., CHE205, Tampa, Florida 33620-5250, United States
| |
Collapse
|
3343
|
Correcting for the free energy costs of bond or angle constraints in molecular dynamics simulations. Biochim Biophys Acta Gen Subj 2014; 1850:932-943. [PMID: 25218695 DOI: 10.1016/j.bbagen.2014.09.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/28/2014] [Accepted: 09/01/2014] [Indexed: 11/22/2022]
Abstract
BACKGROUND Free energy simulations are an important tool in the arsenal of computational biophysics, allowing the calculation of thermodynamic properties of binding or enzymatic reactions. This paper introduces methods to increase the accuracy and precision of free energy calculations by calculating the free energy costs of constraints during post-processing. The primary purpose of employing constraints for these free energy methods is to increase the phase space overlap between ensembles, which is required for accuracy and convergence. METHODS The free energy costs of applying or removing constraints are calculated as additional explicit steps in the free energy cycle. The new techniques focus on hard degrees of freedom and use both gradients and Hessian estimation. Enthalpy, vibrational entropy, and Jacobian free energy terms are considered. RESULTS We demonstrate the utility of this method with simple classical systems involving harmonic and anharmonic oscillators, four-atomic benchmark systems, an alchemical mutation of ethane to methanol, and free energy simulations between alanine and serine. The errors for the analytical test cases are all below 0.0007kcal/mol, and the accuracy of the free energy results of ethane to methanol is improved from 0.15 to 0.04kcal/mol. For the alanine to serine case, the phase space overlaps of the unconstrained simulations range between 0.15 and 0.9%. The introduction of constraints increases the overlap up to 2.05%. On average, the overlap increases by 94% relative to the unconstrained value and precision is doubled. CONCLUSIONS The approach reduces errors arising from constraints by about an order of magnitude. Free energy simulations benefit from the use of constraints through enhanced convergence and higher precision. GENERAL SIGNIFICANCE The primary utility of this approach is to calculate free energies for systems with disparate energy surfaces and bonded terms, especially in multi-scale molecular mechanics/quantum mechanics simulations. This article is part of a Special Issue entitled Recent developments of molecular dynamics.
Collapse
|
3344
|
Pougach K, Voet A, Kondrashov FA, Voordeckers K, Christiaens JF, Baying B, Benes V, Sakai R, Aerts J, Zhu B, Van Dijck P, Verstrepen KJ. Duplication of a promiscuous transcription factor drives the emergence of a new regulatory network. Nat Commun 2014; 5:4868. [PMID: 25204769 PMCID: PMC4172970 DOI: 10.1038/ncomms5868] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/31/2014] [Indexed: 11/08/2022] Open
Abstract
The emergence of new genes throughout evolution requires rewiring and extension of regulatory networks. However, the molecular details of how the transcriptional regulation of new gene copies evolves remain largely unexplored. Here we show how duplication of a transcription factor gene allowed the emergence of two independent regulatory circuits. Interestingly, the ancestral transcription factor was promiscuous and could bind different motifs in its target promoters. After duplication, one paralogue evolved increased binding specificity so that it only binds one type of motif, whereas the other copy evolved a decreased activity so that it only activates promoters that contain multiple binding sites. Interestingly, only a few mutations in both the DNA-binding domains and in the promoter binding sites were required to gradually disentangle the two networks. These results reveal how duplication of a promiscuous transcription factor followed by concerted cis and trans mutations allows expansion of a regulatory network.
Collapse
Affiliation(s)
- Ksenia Pougach
- Laboratory for Genetics and Genomics, Department M2S, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, B-3001 Leuven, Belgium
- Laboratory for Systems biology, Vlaams Instituut voor Biotechnologie (VIB), B-3001 Leuven, Belgium
| | - Arnout Voet
- Structural Bioinformatics, Center for Life Science Technologies (CLST), RIKEN, 230-0045 Yokohama, Japan
| | - Fyodor A. Kondrashov
- Laboratory of Evolutionary Genomics, Centre for genomic regulation (CRG), 08003 Barcelona, Spain
| | - Karin Voordeckers
- Laboratory for Genetics and Genomics, Department M2S, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, B-3001 Leuven, Belgium
- Laboratory for Systems biology, Vlaams Instituut voor Biotechnologie (VIB), B-3001 Leuven, Belgium
| | - Joaquin F. Christiaens
- Laboratory for Genetics and Genomics, Department M2S, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, B-3001 Leuven, Belgium
- Laboratory for Systems biology, Vlaams Instituut voor Biotechnologie (VIB), B-3001 Leuven, Belgium
| | - Bianka Baying
- Genomics Core Facility, European Molecular Biology Laboratory Heidelberg (EMBL), 69117 Heidelberg, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory Heidelberg (EMBL), 69117 Heidelberg, Germany
| | - Ryo Sakai
- Department of Electrical Engineering, STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics, KU Leuven, B-3001 Leuven, Belgium
- iMinds Medical Information Technologies Department, KU Leuven, B-3001 Leuven, Belgium
| | - Jan Aerts
- Department of Electrical Engineering, STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics, KU Leuven, B-3001 Leuven, Belgium
- iMinds Medical Information Technologies Department, KU Leuven, B-3001 Leuven, Belgium
| | - Bo Zhu
- Laboratory for Genetics and Genomics, Department M2S, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, B-3001 Leuven, Belgium
- Laboratory for Systems biology, Vlaams Instituut voor Biotechnologie (VIB), B-3001 Leuven, Belgium
| | - Patrick Van Dijck
- Molecular Microbiology and Biotechnology Section, KU Leuven, B-3001 Leuven, Belgium
- Department of Molecular Microbiology, VIB, B-3001 Leuven, Belgium
| | - Kevin J. Verstrepen
- Laboratory for Genetics and Genomics, Department M2S, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, B-3001 Leuven, Belgium
- Laboratory for Systems biology, Vlaams Instituut voor Biotechnologie (VIB), B-3001 Leuven, Belgium
| |
Collapse
|
3345
|
OptMAVEn--a new framework for the de novo design of antibody variable region models targeting specific antigen epitopes. PLoS One 2014; 9:e105954. [PMID: 25153121 PMCID: PMC4143332 DOI: 10.1371/journal.pone.0105954] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/29/2014] [Indexed: 01/12/2023] Open
Abstract
Antibody-based therapeutics provides novel and efficacious treatments for a number of diseases. Traditional experimental approaches for designing therapeutic antibodies rely on raising antibodies against a target antigen in an immunized animal or directed evolution of antibodies with low affinity for the desired antigen. However, these methods remain time consuming, cannot target a specific epitope and do not lead to broad design principles informing other studies. Computational design methods can overcome some of these limitations by using biophysics models to rationally select antibody parts that maximize affinity for a target antigen epitope. This has been addressed to some extend by OptCDR for the design of complementary determining regions. Here, we extend this earlier contribution by addressing the de novo design of a model of the entire antibody variable region against a given antigen epitope while safeguarding for immunogenicity (Optimal Method for Antibody Variable region Engineering, OptMAVEn). OptMAVEn simulates in silico the in vivo steps of antibody generation and evolution, and is capable of capturing the critical structural features responsible for affinity maturation of antibodies. In addition, a humanization procedure was developed and incorporated into OptMAVEn to minimize the potential immunogenicity of the designed antibody models. As case studies, OptMAVEn was applied to design models of neutralizing antibodies targeting influenza hemagglutinin and HIV gp120. For both HA and gp120, novel computational antibody models with numerous interactions with their target epitopes were generated. The observed rates of mutations and types of amino acid changes during in silico affinity maturation are consistent with what has been observed during in vivo affinity maturation. The results demonstrate that OptMAVEn can efficiently generate diverse computational antibody models with both optimized binding affinity to antigens and reduced immunogenicity.
Collapse
|
3346
|
Jo S, Cheng X, Islam SM, Huang L, Rui H, Zhu A, Lee HS, Qi Y, Han W, Vanommeslaeghe K, MacKerell AD, Roux B, Im W. CHARMM-GUI PDB manipulator for advanced modeling and simulations of proteins containing nonstandard residues. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 96:235-65. [PMID: 25443960 DOI: 10.1016/bs.apcsb.2014.06.002] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
CHARMM-GUI, http://www.charmm-gui.org, is a web-based graphical user interface to prepare molecular simulation systems and input files to facilitate the usage of common and advanced simulation techniques. Since it is originally developed in 2006, CHARMM-GUI has been widely adopted for various purposes and now contains a number of different modules designed to setup a broad range of simulations including free energy calculation and large-scale coarse-grained representation. Here, we describe functionalities that have recently been integrated into CHARMM-GUI PDB Manipulator, such as ligand force field generation, incorporation of methanethiosulfonate spin labels and chemical modifiers, and substitution of amino acids with unnatural amino acids. These new features are expected to be useful in advanced biomolecular modeling and simulation of proteins.
Collapse
Affiliation(s)
- Sunhwan Jo
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Xi Cheng
- Department of Molecular Biosciences and Center for Bioinformatics, The University of Kansas, Lawrence, Kansas, USA
| | - Shahidul M Islam
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Lei Huang
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Huan Rui
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Allen Zhu
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Hui Sun Lee
- Department of Molecular Biosciences and Center for Bioinformatics, The University of Kansas, Lawrence, Kansas, USA
| | - Yifei Qi
- Department of Molecular Biosciences and Center for Bioinformatics, The University of Kansas, Lawrence, Kansas, USA
| | - Wei Han
- Beckman Institute and Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Kenno Vanommeslaeghe
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Benoît Roux
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA.
| | - Wonpil Im
- Department of Molecular Biosciences and Center for Bioinformatics, The University of Kansas, Lawrence, Kansas, USA.
| |
Collapse
|
3347
|
Li J, Ziemba BP, Falke J, Voth GA. Interactions of protein kinase C-α C1A and C1B domains with membranes: a combined computational and experimental study. J Am Chem Soc 2014; 136:11757-66. [PMID: 25075641 PMCID: PMC4140453 DOI: 10.1021/ja505369r] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Indexed: 02/01/2023]
Abstract
Protein kinase C-α (PKCα) has been studied widely as a paradigm for conventional PKCs, with two C1 domains (C1A and C1B) being important for the regulation and function of the kinase. However, it is challenging to explore these domains in membrane-bound environments with either simulations or experiments alone. In this work, we have combined modeling, simulations, and experiments to understand the molecular basis of the PKCα C1A and C1B domain interactions with membranes. Our atomistic simulations of the PKCα C1 domains reveal the dynamic interactions of the proteins with anionic lipids, as well as the conserved hydrogen bonds and the distinct nonpolar contacts formed with lipid activators. Corroborating evidence is obtained from additional simulations and experiments in terms of lipid binding and protein diffusion. Overall, our study, for the first time, explains with atomistic detail how the PKCα C1A and C1B domains interact differently with various lipids. On the molecular level, the information provided by our study helps to shed light on PKCα regulation and activation mechanism. The combined computational/experimental approach demonstrated in this work is anticipated to enable further studies to explore the roles of C1 domains in many signaling proteins and to better understand their molecular mechanisms in normal cellular function and disease development.
Collapse
Affiliation(s)
- Jianing Li
- Department
of Chemistry, Institute for Biophysical Dynamics, James Franck Institute
and Computation Institute, The University
of Chicago, 5735 South
Ellis Avenue, Chicago, Illinois 60637, United States
| | - Brian P. Ziemba
- Department
of Chemistry and Biochemistry and the Molecular Biophysics Program, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Joseph
J. Falke
- Department
of Chemistry and Biochemistry and the Molecular Biophysics Program, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Gregory A. Voth
- Department
of Chemistry, Institute for Biophysical Dynamics, James Franck Institute
and Computation Institute, The University
of Chicago, 5735 South
Ellis Avenue, Chicago, Illinois 60637, United States
| |
Collapse
|
3348
|
Local anesthetic and antiepileptic drug access and binding to a bacterial voltage-gated sodium channel. Proc Natl Acad Sci U S A 2014; 111:13057-62. [PMID: 25136136 DOI: 10.1073/pnas.1408710111] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Voltage-gated sodium (Nav) channels are important targets in the treatment of a range of pathologies. Bacterial channels, for which crystal structures have been solved, exhibit modulation by local anesthetic and anti-epileptic agents, allowing molecular-level investigations into sodium channel-drug interactions. These structures reveal no basis for the "hinged lid"-based fast inactivation, seen in eukaryotic Nav channels. Thus, they enable examination of potential mechanisms of use- or state-dependent drug action based on activation gating, or slower pore-based inactivation processes. Multimicrosecond simulations of NavAb reveal high-affinity binding of benzocaine to F203 that is a surrogate for FS6, conserved in helix S6 of Domain IV of mammalian sodium channels, as well as low-affinity sites suggested to stabilize different states of the channel. Phenytoin exhibits a different binding distribution owing to preferential interactions at the membrane and water-protein interfaces. Two drug-access pathways into the pore are observed: via lateral fenestrations connecting to the membrane lipid phase, as well as via an aqueous pathway through the intracellular activation gate, despite being closed. These observations provide insight into drug modulation that will guide further developments of Nav inhibitors.
Collapse
|
3349
|
Lv X, Liu H, Ke M, Gong H. Exploring the pH-dependent substrate transport mechanism of FocA using molecular dynamics simulation. Biophys J 2014; 105:2714-23. [PMID: 24359743 DOI: 10.1016/j.bpj.2013.11.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/29/2013] [Accepted: 11/01/2013] [Indexed: 12/19/2022] Open
Abstract
FocA belongs to the formate-nitrate transporter family and plays an essential role in the export and uptake of formate in organisms. According to the available crystal structures, the N-terminal residues of FocA are structurally featureless at physiological conditions but at reduced pH form helices to harbor the cytoplasmic entrance of the substrate permeation pathway, which apparently explains the cessation of electrical signal observed in electrophysiological experiments. In this work, we found by structural analysis and molecular dynamics simulations that those N-terminal helices cannot effectively preclude the substrate permeation. Equilibrium simulations and thermodynamic calculations suggest that FocA is permeable to both formate and formic acid, the latter of which is transparent to electrophysiological studies as an electrically neutral species. Hence, the cease of electrical current at acidic pH may be caused by the change of the transported substrate from formate to formic acid. In addition, the mechanism of formate export at physiological pH is discussed.
Collapse
Affiliation(s)
- Xiaoying Lv
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Huihui Liu
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Meng Ke
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haipeng Gong
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
3350
|
Shang Y, LeRouzic V, Schneider S, Bisignano P, Pasternak G, Filizola M. Mechanistic insights into the allosteric modulation of opioid receptors by sodium ions. Biochemistry 2014; 53:5140-9. [PMID: 25073009 PMCID: PMC4131901 DOI: 10.1021/bi5006915] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/21/2014] [Indexed: 01/09/2023]
Abstract
The idea of sodium ions altering G-protein-coupled receptor (GPCR) ligand binding and signaling was first suggested for opioid receptors (ORs) in the 1970s and subsequently extended to other GPCRs. Recently published ultra-high-resolution crystal structures of GPCRs, including that of the δ-OR subtype, have started to shed light on the mechanism underlying sodium control in GPCR signaling by revealing details of the sodium binding site. Whether sodium accesses different receptor subtypes from the extra- or intracellular sides, following similar or different pathways, is still an open question. Earlier experiments in brain homogenates suggested a differential sodium regulation of ligand binding to the three major OR subtypes, in spite of their high degree of sequence similarity. Intrigued by this possibility, we explored the dynamic nature of sodium binding to δ-OR, μ-OR, and κ-OR by means of microsecond-scale, all-atom molecular dynamics (MD) simulations. Rapid sodium permeation was observed exclusively from the extracellular milieu, and following similar binding pathways in all three ligand-free OR systems, notwithstanding extra densities of sodium observed near nonconserved residues of κ-OR and δ-OR, but not in μ-OR. We speculate that these differences may be responsible for the differential increase in antagonist binding affinity of μ-OR by sodium resulting from specific ligand binding experiments in transfected cells. On the other hand, sodium reduced the level of binding of subtype-specific agonists to all OR subtypes. Additional biased and unbiased MD simulations were conducted using the δ-OR ultra-high-resolution crystal structure as a model system to provide a mechanistic explanation for this experimental observation.
Collapse
MESH Headings
- Animals
- Binding Sites
- Crystallography, X-Ray
- Humans
- Ligands
- Mice
- Models, Molecular
- Molecular Dynamics Simulation
- Protein Conformation
- Radioligand Assay
- Receptors, Opioid/chemistry
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/chemistry
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/metabolism
- Sodium/metabolism
Collapse
Affiliation(s)
- Yi Shang
- Department
of Structural and Chemical Biology, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Valerie LeRouzic
- Molecular
Pharmacology and Chemistry Program, Memorial
Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - Sebastian Schneider
- Department
of Structural and Chemical Biology, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Paola Bisignano
- Department
of Structural and Chemical Biology, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Gavril
W. Pasternak
- Molecular
Pharmacology and Chemistry Program, Memorial
Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - Marta Filizola
- Department
of Structural and Chemical Biology, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|