Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2020 Jun 5;15(6):e0234215. doi: 10.1371/journal.pone.0234215

Docking studies and molecular dynamics simulations of the binding characteristics of waldiomycin and its methyl ester analog to Staphylococcus aureus histidine kinase

Awwad Radwan 1,2,*, Gamal M Mahrous 1
Editor: Concepción Gonzalez-Bello3
PMCID: PMC7274439  PMID: 32502195

Abstract

Bacterial histidine kinases (HKs) are considered attractive drug targets because of their ability to govern adaptive responses coupled with their ubiquity. There are several classes of HK inhibitors; however, they suffer from drug resistance, poor bioavailability, and a lack of selectivity. The 3D structure of Staphylococcus aureus HK was not isolated in high-resolution coordinates, precluding further disclosure of structure-dependent binding to the specific antibiotics. To elucidate structure-dependent binding, the 3D structure of the catalytic domain WalK of S. aureus HK was constructed using homology modeling to investigate the WalK–ligand binding mechanisms through molecular docking studies and molecular dynamics simulations. The binding free energies of the waldiomycin and its methyl ester analog were calculated using molecular mechanics/generalized born surface area scoring. The key residues for protein–ligand binding were postulated. The structural divergence responsible for the 7.4-fold higher potency of waldiomycin than that of its ester analog was clearly observed. The optimized 3D macromolecule–ligand binding modes shed light on the S. aureus HK/WalK–ligand interactions that afford a means to assess binding affinity to design new HK/WalK inhibitors.

Introduction

Protein kinases (PKs) catalyze the phosphorylation of other proteins and thus play crucial roles not only in cell regulation but also in signal transduction. Cell dysregulation is often associated with a disease. Thus, PKs are considered as important targets in drug discovery programs that seek to alter their function [1,2]. Particularly, histidine kinases (HKs) are responsible for sensing conditions that allow microbes to adopt a pathogenic lifestyle through the expression of virulence factors. Thus, the inhibition of factors that are distinctly unique to the bacterial cell and express virulence of pathogenic bacteria offers a chance for specific interference from the host biochemical processes. Nowadays, it is clear that virulence is an adaptive genetic response involving the genes encoding the induction of virulence factors [3,4]. This response implies that pathogenic microbes can sense when it is in an invasion state. Sensing of environmental stimuli occurs through two-component signaling (TCS) systems that appears almost ubiquitously in bacteria. Bacteria respond to extracellular changes through TCS pathways, which are mediated by HKs and are absent in humans [5]. The adaptive and response ability of bacteria is vital for survival in severe environments that they encounter [6].

The Staphylococcus aureus bacteria TCS system comprises HK protein, containing a membrane-bound sensor histidine kinase (WalK), and a response regulator (RR) protein, containing a conserved DNA-binding domain (WalR) [7]. WalK is a membrane-linked HK that consists of two domains. One domain is a catalytic or ATP-binding domain and the second is a dimerization domain that includes a conserved phosphorylated histidine residue.

HKs are activated as a result of the interaction of external stimuli with its extracellular signaling domain. The activated HKs bind ATP at its catalytic domain WalK, which catalyzes the transfer of the a-phosphate group of ATP to a conserved histidine residue on the HK dimerization and histidine phosphotransfer (DHp) domain. The phosphoryl group transfers from the conserved phosphorylated histidine on a HK to aspartic acid conserved in an RR domain protein, the HK’s associated signaling partner. The RR proteins harbor both the RR domain and the DNA-binding domain. The phosphorylation of RR results in its activation and subsequently triggers it to bind to DNA and stimulate gene transcription, producing a response to the extracellular stimulus (Fig 1) [5,8]. Antibacterial activity can be afforded by inhibiting the microbial HK using ATP competitive inhibitors [9,10].

Fig 1. Two-Component Signaling (TCS) system pathway.

Fig 1

HK (blue), histidine kinase; RR (beige), response regulator; H, conserved histidine residue; D, conserved aspartic acid residue.

The TCS-WalK/WalR system that is unique to gram-positive bacteria, including S. aureus [11], Bacillus subtilis [12], and Enterococcus faecalis [13], is crucial for survival and is attracting growing interest as a novel target of antibacterial agents [14]. Representative inhibitors, RWJ-49815, closantel, and tetrachlorosalicylanilide, have been reported as active inhibitors against several bacterial TCS transduction systems [1517]. Unfortunately, few TCS inhibitors have been reported against S. aureus, a major pathogen with a high virulence factor. The amino acid sequence of the WalK/WalR TCS transduction histidine protein kinase of S. aureus (strain Mu50/ATCC 700699) ARLS_STAAM (Q7A2R7) [18] is well defined; however, the 3D structure is unavailable in the protein databank (PDB) (https://www.rcsb.org/).

For case in which the atomic structures of enzyme macromolecules are absent, homology modeling is a powerful tool to understand the general fold of the enzyme, catalytic site, and conserved residues, in addition to the substrate and inhibitor binding sites. This information is crucial for structure-based drug design approaches for the development of potential enzyme inhibitors for therapeutic application [19]. In addition, waldiomycin and its methyl ester analog, produced by Streptomyces sp., were reported as inhibitors of S. aureus HK with IC50 values of 10.2 and 75.8 μM, respectively, and are effective as antibacterial agents against S. aureus species [20]. This information is also helpful for the ligand-based drug discovery of novel lead compounds with potential activity against S. aureus bacteria.

In this study, we performed in silico modeling to explore the mode of inhibition of the S. aureus HK/WalK enzyme by waldiomycin and its methyl ester analog. The study includes homology modeling, molecular docking, molecular dynamics (MD) simulations, and molecular mechanics/generalized born surface area (MM/GBSA) calculations. The calculations were performed on the S. aureus kinase structure (WalK catalytic domain) to investigate the macromolecule–ligand complex and predict the 3D structure of S. aureus HK/WalK. The S. aureus HK/WalK conformation changes persuaded in the MD simulations and the ligand binding free energies of waldiomycin and its ester analog, as calculated using the MM/GBSA method, confirm the dependencies of site activity on the ligands’ electrostatic and hydrophobic properties. Lys100 was found to be a crucial residue for the 7.4-fold higher potency of waldiomycin than that of its ester analog against S. aureus HK/WalK.

Methods

Overall workflow

The schematic representations of the overall study workflow are illustrated in Fig 2. Firstly, the S. aureus kinase protein sequence was used for searching in the PDB. The BLAST [21] program was utilized to select a template protein possessing the highest similarity. An initial S. aureus kinase 3D structure was built by means of homology modeling followed by structure optimization using an MD simulation. The optimized macromolecule structure was then used for docking analyses. Because of their almost similar structures and the large difference in their potencies as S. aureus HK/WalK inhibitors, waldiomycin and its ester analog [20] were selected and their 3D structures were also built, optimized, and then docked into the homology-built and optimized S. aureus HK/WalK structure. The S. aureus HK/WalK–ligand complexes were further subjected to optimization via MD simulations. Lately, the MD simulation trajectories were used as inputs for the MM/GBSA calculation of binding free energies of the ligands and investigation of their binding mechanism.

Fig 2. Sketch representation of the overall workflow.

Fig 2

Homology modeling

The homology modeling of S. aureus HK/WalK secondary structure was performed to develop the initial structure needed for the MD simulation study. Briefly, the BLAST sequence database search module was utilized to obtain the X-ray structures similar to the target sequence Q7A2R7 [22] and sort them according to similarity percent. Using the MODELLER program, the top-scoring PDB entry was suggested as a template for alignment of the target Q7A2R7 to build the homolog model. During the modeling process, the position of the crystallized ligand was kept in its original position for rebuilding the binding cavity. The obtained homology model was checked for missing atoms and/or residues using the PROCHECK program [23]. Gaps and missing coordinates were repaired using the MODELLER program [24] and the resulting structure was used as the initial structure in the next MD simulation optimization process.

Protein preparation

The AMBER18 program [25] was utilized to perform the structural minimization and MD simulations using the ff14SB force field amino acid parameters. The tleap module of AmberTools18 was used to add the complementary hydrogen atoms that could not be detected by X-ray crystallography. The protein molecule was explicitly solvated with water molecules and sodium counter-ions were also added to obtain a neutral system. A water box with 10 Å thickness was created using the TIP3P water solvent model to completely surround the protein molecule. Simulation was performed using a periodic boundary condition. Long-range electrostatic interactions were processed using the particle–mesh Ewald protocol [26] and a cut-off value of 8 Å was used for non-bonding interactions. Initial minimization of the water molecules and counter-ions was performed using 1000-cycle minimizations; subsequently, the whole system was minimized using 1000-cycle minimizations. Then, a system temperature increase to 298.15 K was performed and MD simulations at 298.15 K were executed with the fixed protein coordinates. The equilibrations were performed under the constant normal temperature and pressure (NPT) ensemble for 170 ps. In the first 20 ps, the waters and counter-ions were equilibrated, while the solutes were restrained. In the next 50 ps, the side chains of the protein amino acids were relaxed, then all the restraints were removed in the last 100 ps. Lastly, MD simulations of 10 ns were processed at 1 atm and 298.15 K under the NPT ensemble with a time step of two femtoseconds (fs). The covalent bonds with hydrogen atoms were constrained by the use of the SHAKE algorithm [27] and Langevin dynamics were used to control the system temperature. Throughout the MD simulations, all of the atom coordinates of the system were saved every 1 ps.

Ligand preparation

Waldiomycin and its methyl ester 1, 2 were reported as inhibitors of S. aureus HK with IC50 values of 10.2 and 75.8 μM, respectively [20]. The chemical structures and compound names are given in Fig 3. The antechamber module [28,29] in the AmberTools18 program was used to generate the force field parameters of the ligands. The general AMBER force field was applied to the ligands, and the Leap program was used to generate the topology and parameter files.

Fig 3. Chemical structures of S. aureus HK/WalK inhibitors.

Fig 3

(1) Waldiomycin (IC50 10.2 μM) and (2) waldiomycin methyl ester (IC50 75.8 μM).

Docking study

The docking procedure was performed using the Dock6.4 program [30] on an ubunto 14.2-supported Dell 5000 workstation (Processor: Intel(R) Core(TM) i7-5500U CPU @ 2.40GHz with a 64-bit operating system, ×64-based processor, memory of 7.7 GiB). The docking method was validated using 5C93 proteins and co-crystalized ligand ACP using the default parameters, except with a sphere of 10 Å diameter around the binding site center. The co-crystallized ligand ACP was docked in the original binding pocket of the protein structure. The docking protocol was validated according to the root-mean-square deviation (RMSD) of the docking pose relative to the ACP X-ray structure.

The refined tertiary structures of S. aureus HK/WalK and HK/WalK inhibitor compounds 1, 2 were prepared in their physiological protonation states. Compounds 1 and 2 were docked using the same settings for the validation process of the docking program. The resulting solutions were clustered based on the heavy atom RMSD values (1 Å). Finally, the top-scoring binding pose was taken into consideration for ligand–receptor interactions and was selected for further refinement using MD simulations.

MD simulations of complexes

MD simulations were performed for the HK–ligand complexes obtained via docking using the same procedure explained in the MD optimization described above, including system minimization, heating, and equilibration. MD simulations were performed for a time period of 35 ns for each macromolecule–ligand complex. The atom coordinates in the complex system were saved at 1-ps time intervals throughout the simulations. Using the initial structures of the MD simulation as the reference structures, the CPPTRAJ module of AmberTools18 was used for determining RMSDs to validate the convergence of the MD simulation processes. Structural flexibilities of the protein were estimated by calculating root-mean-square fluctuations (RMSFs). The trajectories during last 20 ns of equilibration MD simulations were used for calculating the RMSF values, whereas the average structures of the final 20 ns trajectories were used as the reference structures.

Hydrogen bond analysis

The CPPTRAJ module in Amber18 was used to analyze hydrogen bonding between the ligand and the surrounding amino acid residues. Hydrogen bonds with high occupancy were observed during the last 8.0 ns of the simulations for the two ligand complex systems.

Pairwise per-residue free energy decomposition

Molecular mechanics Poisson–Boltzmann surface area (MM/PBSA) pairwise decomposition was used to calculate the interaction energy of the residues involved in high-occupancy hydrogen bonding to the ligand during the last 8 ns of the simulation.

Binding free energy calculation

The MM/GBSA method [31] was used to calculate the binding free energy (ΔGbinding), Eq (1) for receptor–binder complex systems. One thousand snapshots were taken at 20–30 ns time intervals throughout the MD simulation trajectory to compute the MM/GBSA free energy difference. For each snapshot, Eq (1) was used to calculate the binding free energy of the complex system.

ΔGbinding=Gcomplex(Gprotein+Gligand), (1)
ΔG=ΔGgas+ΔGsolvTΔS, (2)
ΔGgas=ΔEelectrostatic+ΔEvdw, (3)
ΔGsolv=ΔGGB+ΔGSA, (4)
ΔGSA=γ×SASA+b, (5)

where Gcomplex, Gprotein, and Gligand are the free energies of the complex, protein, and ligand, respectively; ΔGgas is the gas-phase molecular mechanics free energy, including electrostatic (ΔEelectrostatic) and van der Waals (ΔEvdw) contributions; and ΔGsolv is the solvation free energy, which includes polar (ΔGGB) and nonpolar (ΔGSA) contributions.

ΔGGB is the polar contribution that was estimated using the modified GB model reported by Onufriev et al. [32] using εw = 80 and εp = 1.0, while the solvent-accessible surface area (SASA) is calculated using linear combinations of pairwise overlaps [33]. The surface tension proportionality constant (γ) was set to 0.0072 kcal mol–1Å–2 while the nonpolar solvation free energy for a point solute (b) was set to 0.00 kcal mol–1. The SASA was calculated using a probe sphere radius set to 1.4 Å. The calculated free energies were used for comparisons between the waldiomycin and waldiomycin methyl esters as S. aureus kinase enzyme binders. The final binding free energy for the two binders was calculated as the average value from 1000 snapshots in the last 10 ns of the MD simulations.

Results and discussion

Structure optimization

With an identity of 36%, the sequence of S. aureus UID-Q7A2R7 [18] has the highest-ranked homology in the BLAST search for E. coli histidine kinase, 5C93 [34], on proteins in the PDB. Generally, it is assumed that two proteins topologies are usually similar once their sequences share about 30% identity [35]. Moreover, Dimitrii et al. [36] developed structures of average quality (RMSD < 4.0 Å) based on target-template sequence identity ≥ 20%. The crystal structure of 5C93 was recently obtained at a resolution of 2.52 Å [34]. Therefore, the 3D structure of S. aureus HK, built through a homology model to the 5C93 template structure, should be appropriate for modeling S. aureus HK protein binding data. The initial alignment of Q7A2R7 with the template sequence 5C93 was obtained using ClustalW. The final alignment (36% identity) used in the homology modeling and in the secondary structure prediction is shown in Fig 4. The initial homology models of S. aureus HK were built using MODELLER and used for further optimization.

Fig 4. Staphylococcus aureus Q7A2R7 sequence (target) alignment result with template sequence (5c93).

Fig 4

The obtained secondary structure is shown, where H represents the helix; E represents the strand; and “-” represents the loop. Identical residues are highlighted.

MD simulation is usually practiced to improve homology models [37]. Using MD simulation, the initial 3D structure of S. aureus HK/WalK, obtained from homology modeling, was optimized in a solvent to simulate the real physiological environments. During the MD simulation, the stability of the structure was calculated by its deviance from the starting conformation in RMSD terms. Fig 5A shows the RMSD values of S. aureus HK/WalK protein structure through the whole MD simulation trajectory. After 7 ns, the 3D structures of HK/WalK reached a stable state where the RMSDs of the protein backbone atoms and of all atoms converged to 1.25 Å and 2.25 Å, respectively. Fig 5B shows the calculated RMSFs of the S. aureus HK/WalK structure generated during the MD simulation, which characterizes the mobility of individual residues. The Ramachandran plot of the optimized S. aureus HK/WalK structure showed that 100% of residues were located in the allowable regions (S1 Fig), higher than that of the initial homology modeled 3D S. aureus HK (S2 Fig), which showed 93.4% of residues in the allowed region, indicating the higher stability of the optimized S. aureus HK structure.

Fig 5. Molecular Dynamics (MD) profiles of the S. aureus HK 3D structure optimization.

Fig 5

a) Root-mean-square deviation (RMSD) values (y-axis) along the time frame (x-axis) for atoms of the protein backbone (red lines) and all protein atoms (black line), and b) root-mean-square fluctuation (RMSF) values (y-axis) in the MD simulation for individual residues (x-axis).

Binding modes generated by molecular docking

Redocking of the ACP X-ray structure in its original binding site resulted in a solution pose with 0.704 Å and RMSD value S3 Fig.

To shed light on the possible binding modes of compounds 1 and 2, a large box was defined to include all potential binding sites. Hence, the two ligands adopted the most favorable binding poses that were searched and ranked according to their docking scores. The conformation with the lowermost score, suggesting the most probable binding mode of a ligand, was suggested for additional analysis.

The two binders were efficiently docked inside the binding site of the HK/WalK structure. The binding poses of the two binders showed similar V-shaped orientations inside a binding site delineated with the side chains of residues Lys63, Lys100, Leu102, Hie103, Pro104, Leu131, Ile132, Asp135, Ile138, Lys139, Arg183, Val184, Asp185, and Asn195. The hydrophobic part of the binders fits inside one binding pocket near Val184, while the carboxyl and ester groups of the two compounds fit inside a second binding pocket near Lys100 (Fig 6A and 6B).

Fig 6. Docking results of homolog modeling.

Fig 6

Binding conformations of waldiomycin (a) and waldiomycin methyl ester (b) at the binding site of the homolog model of S. aureus HK/WalK.

The 10 top-scoring conformations of the waldiomycin and waldiomycin ester are shown in S4 Fig and S5 Fig mol2 files, respectively. The docking score and hydrogen bond information of these 10 conformations are also listed in S1 and S2 Files, respectively. The Grid score of these binding poses is in the range of –63.629486 to –62.780089 kcal mol–1 for waldiomycin and –66.089516 to –64.42131 kcal mol–1 for waldiomycin methyl ester, which is incomparable with the difference in their experimental inhibition of S. aureus HK/WalK, with IC50 values of 10.2 and 75.8 μM, respectively. The top-scoring binding poses of the two binders were selected for further investigations using MD simulation.

Although docking studies have been successfully used in calculating the binding poses of ligands for many proteins, they failed in assessing ligand binding affinity [38]. The treatment of proteins as “rigid” molecules does not consider their conformational changes during the docking of some ligands [39]. Conformational changes during protein–ligand interactions could be studied during MD simulations. The conformation changes in protein–ligand interactions have been broadly studied through MD simulation methods [40]. Considering both protein structure flexibility and protein–ligand electrostatic interactions, MD simulations were deemed necessary to justify the 7.4-fold potency difference in anti-S. aureus HK/WalK activity between waldiomycin and its ester analog and to refine the understanding of waldiomycin (ester)-HK/WalK binding modes obtained from molecular docking. Ligand (waldiomycin or its methyl ester) conformation changes were observed through all the MD simulation time frames. MD simulation was initiated from the docked minimized complex and continued for over 35 ns. Using RMSD changes during the MD simulations, the dynamic stabilities of the two complex systems were calculated and are plotted in Fig 7A and 7B.

Fig 7. Root-Mean-Square Deviation (RMSD) in Molecular Dynamics (MD) simulations.

Fig 7

(a) RMSD of protein backbones (black lines) and waldiomycin (red lines). (b) RMSD of protein backbones (in black lines) and of waldiomycin methyl ester (in similar color) with the four orientations of waldiomycin methyl ester through the MD simulation time intervals. 0–11 ns (beige color), 11–14 ns (cyan color), 14–14.5 ns (pink color), and 14.5–35 ns (green color).

For the complexes of S. aureus HK/WalK bound with waldiomycin, Fig 7A, both the waldiomycin and the protein structures were equilibrated with unobvious RMSD fluctuations through the 35 ns time frames. Interestingly, trajectory analysis revealed that the carboxyl group of waldiomycin is engaged in stable hydrogen bond with Lys100. The presence of a hydrogen bond between the Lys100 residue of HK/WalK and waldiomycin (active anti-HK) and its absence with the waldiomycin methyl ester (inactive anti-HK) suggests that Lys100 is a crucial residue for S. aureus HK activity.

The complexes of S. aureus HK/WalK bound with waldiomycin methyl ester (Fig 7B) showed unobvious RMSD fluctuations over the first 11 ns, then a gradual increase from 11–14 ns, followed by a rapid increase from 14 to 14.5 ns, and convergence from 14.5 to 35 ns. The large RMSD fluctuations suggest a large conformational change and repositioning of the waldiomycin ester inside the binding site resulting in four conformational changes (Fig 7B; beige color): holds for 0–11 ns, (7b; cyan color), holds during the 11–14 ns, (Fig 7B; pink color), holds during the 14–14.5 ns (Fig 7B; green color), and continues over the last 14.5–35 ns. Remarkably, trajectory analysis revealed that the ester group of the waldiomycin methyl ester exited the binding pocket after 14.5 ns; this conformational instability during MD simulation is unsurprising since a weak binding affinity was reported for this binder.

The superimposition of binding conformation from the final snapshots of the MD simulations is shown in Fig 8 (waldiomycin: gray; waldiomycin methyl ester: cyan) and reveals different ligand binding conformations, though the MD simulations started from docking poses with similar orientations.

Fig 8. The final snapshots (35 ns) of ligand–protein complexes in the Molecular Dynamics (MD) simulations.

Fig 8

The ligands are represented as sticks (gray: waldiomycin; cyan: waldiomycin methyl ester). The proteins are represented as wires of constant colors.

The CPPTRAJ hydrogen bond analyses are listed in Table 1 and show the hydrogen bonds with high occupancy for the two simulation systems. The table shows two different hydrogen bond sets for the two complex systems. For the waldiomycin-HK/WalK complex, the amino acid residues Lys63, Ser98, Lys100, Gln128, Asp135, Lys139, and Val184 showed a hydrogen bond with the ligand. For the waldiomycin ester-HK/WalK complex, the amino acid residues, Lys63, Leu102, and His103 showed a hydrogen bond with the ligand. The hydrogen bond pattern difference between the two simulation systems is an indication of the two different binding conformations of waldiomycin and its ester analog (Fig 8).

Table 1. Results of pairwise energy decomposition analysis of the high-occupancy hydrogen bonding of residues to the ligands (kcal mol-1).

Hk/WalK residues Lys63 Ser98 Lys100 Leu102 Hie103 Gln128 Asp135 Lys139 Val184
Waldiomycin –1.093 –0.04 –4.371 –0.043 –0.836 –0.111 –2.722 –3.29 –4.136
Waldiomycin ester –1.575 –0.088 –0.096 –0.025 –0.092 –1.064 –1.497 –2.536 –1.345

The MM/PBSA-pairwise decomposition [41] of residue interaction energies developed an insight into the interactions between the binders and their binding sites for HK/WalK. The results demonstrated that the amino acid residues Lys100, Asp135, Lys139, and Val140 play a crucial role in effective binding interactions with waldiomycin and showed absolute decomposed energy in the range of –2.722 to –4.371 kcal mol-1 (Table 1).

Although the MM/GBSA approach does not consider the entropy associated with the binding, it is an efficient and powerful tool for the correct ranking of ligands. Although the inclusion of the conformational entropy is crucial for calculating the absolute free energies of binding, it is not necessary for ranking the binding affinities of similar inhibitors [42].

The binding free energies of the two ligands were calculated using MM/GBSA methods according to the MD simulation trajectories from 25 to 35 ns. The total binding free energy ΔGMM/GBSA and the related energy terms determined using the MM/GBSA method are provided in Table 2. As it was used in direct comparisons with ΔGMM-GBSA, the experimental binding affinity (ΔGexpt) was estimated from binding affinity data using the equation ΔGexpt ≈–RTlnIC50 [43] (Table 2). Interestingly, the binding free energy ΔGMM/GBSA (waldiomycin, –43.73; waldiomycin ester, –27.75 kcal mol-1) was in accordance with the experimental binding affinity, ΔGexpt (waldiomycin, –26.47 kcal mol-1; waldiomycin ester, –21.85 kcal mol-1) estimated from the IC50 values (waldiomycin, 10.2 μM; waldiomycin methyl ester, 75.8 μM). Analyzing the components in the MM/GBSA binding free energies revealed that electrostatic energy was the major contributor to the two binders. For example, ΔGelectrostatic was –17.631 kcal mol–1 for waldiomycin containing carboxyl group, whereas ΔGelectrostatic was –10.093 kcal mol–1 for the waldiomycin containing methyl ester. The strong hydrogen bond formation between waldiomycin and the key residue Lys100 was recognized and postulated to be the reason for the 7.4-fold higher potency of waldiomycin than that of its ester analog.

Table 2. Calculated binding free energies in comparison with experimental data (kcal mol–1 a).

Ligand ΔGvdw ΔGelec ΔGpolarb ΔGSurfc ΔGMM/GBSA ΔGExptd
Wald –58.144 –17.631 40.174 –8.132 –43.733 −26.468
Wald ester –47.868 –10.093 37.403 –7.195 –27.753 −21.849

a Average of 1000 frames

b Whole electrostatic contribution: ΔGelec = ΔGelectrostatic + ΔGpolar

c Whole nonpolar contribution: ΔGnp = ΔGvdw + ΔGsurf

d ΔGexpt = RTlnIC50, T = 277 K, R = 8.314 JK–1mol–1

Conclusions

In a recent study, an inhibitors of S. aureus HK/WalK enzyme, waldiomycin, was reported to have 7.4‐fold higher potency than its other inhibitor, waldiomycin ester. The two inhibitors have the same structures but differ with regard to only one pharmacophore. Knowledge of the compounds’ binding free energies is crucial to evaluate its potential antibacterial activity. The tertiary structures of S. aureus HK/WalK are crucial to understanding HK/WalK–ligand interactions for evaluating potential anti-S. aureus activity. The amino acid residue Lys100 was crucial for hydrogen bonding with waldiomycin at the binding site and possibly played an essential role in the enzyme activity. The results of our work on binding interactions between the ligands and S. aureus HK/WalK would be useful for further studies, including the designing of more efficient antibacterial agents that target S. aureus HK/WalK.

Supporting information

S1 Fig. Ramachandran plot of the optimized S. aureus HK/WalK structure.

(PDF)

S2 Fig. Ramachandran plot of the initial homology modeled S. aureus HK/WalK structure.

(PDF)

S3 Fig

The co-crystallized ACP (from 5c93.pdb, colored magenta) and the redocked ACP structure (colored forest green), superimposed inside the binding site of 5c93.

(TIF)

S4 Fig. The 10 top-scored conformations of the waldiomycin in mol2 format.

(TIF)

S5 Fig. The 10 top-scoring conformations of the waldiomycin methyl ester in mol2 format.

(TIF)

S1 File. The docking score and hydrogen bond information of the top 10 scored conformations of waldiomycin.

(INFO)

S2 File. The docking score and hydrogen bond information of the top 10 scored conformations of waldiomycin methyl ester.

(INFO)

Acknowledgments

The authors extend their appreciation to the Deanship of Scientific Research at King Saud University.

Data Availability

All relevant data are within the paper and its Supporting Information files.

Funding Statement

A A Radwan. RG-1435-080. Deanship of the Scientific Research at King Saud University. dsrs.ksu.edu.sa/en The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Hynes NE. Tyrosine kinase signaling in breast cancer. Breast Cancer Res 2000; 2:154–157. 10.1186/bcr48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hynes NE, Boulay A. The mTOR pathway in breast cancer. J Mammary Gland Biol Neoplasia 2006; 11:53–61. 10.1007/s10911-006-9012-6 [DOI] [PubMed] [Google Scholar]
  • 3.Szurmant H, White RA, Hoch JA. Sensor complexes regulating two-component signal transduction. Curr Opin Struct Biol 2007; 17: 706–715. 10.1016/j.sbi.2007.08.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bronner S, Monteil H, Prevost G. Regulation of virulence determinants in Staphylococcus aureus: complexity and applications. FEMS Microbiol Rev 2004; 28: 183–200. 10.1016/j.femsre.2003.09.003 [DOI] [PubMed] [Google Scholar]
  • 5.Stock AM, Robinson VL, Goudreau PN. Two-component signal transduction. Annu Rev Biochem. 2000; 69:183–215. 10.1146/annurev.biochem.69.1.183 [DOI] [PubMed] [Google Scholar]
  • 6.Gotoh Y, Eguchi Y, Watanabe T, Okamoto S, Doi A, Utsumi R. Two-component signal transduction as potential drug targets in pathogenic bacteria. Curr Opin Microbiol. 2010;13(2):232–239. 10.1016/j.mib.2010.01.008 [DOI] [PubMed] [Google Scholar]
  • 7.Gao R, Stock AM. Biological insights from structures of two-component proteins. Annu Rev Microbiol 2009; 63: 133–154. 10.1146/annurev.micro.091208.073214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gao R, Stock AM. Biological insights from structures of two-component proteins. Annu Rev Microbiol. 2009;63:133–154. 10.1146/annurev.micro.091208.073214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wilke KE, Carlson EE. All signals lost. Sci Transl Med. 2013;5(203):212–212. 10.1126/scitranslmed.3006670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bem AE, Velikova N, Pellicer MT, Baarlen PV, Marina A, Wells JM. Bacterial histidine kinases as novel antibacterial drug targets. ACS Chem Biol. 2015;10:213–224. 10.1021/cb5007135 [DOI] [PubMed] [Google Scholar]
  • 11.Martin PK, Li T, Sun D, Biek DP, Schmid MB. Role in cell permeability of an essential two-component system in Staphylococcus aureus. J Bacteriol 1999; 181: 3666–3673. PMCID: PMC93842 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fabret C, Hoch JA. A two-component signal transduction system essential for growth of Bacillus subtilis: implications for anti-infective therapy. J Bacteriol 1998; 180: 6375–6383. PMCID: PMC107725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hancock L, Perego M. Two-component signal transduction in Enterococcus faecalis. J Bacteriol 2002; 184: 5819–5825. 10.1128/jb.184.21.5819-5825.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gotoh Y, Eguchi Y, Watanabe T, Okamoto S, Doi A, Utsumi R. Two-component signal transduction as potential drug targets in pathogenic bacteria. Curr. Opin. Microbiol 2010; 13: 232–239. 10.1016/j.mib.2010.01.008 [DOI] [PubMed] [Google Scholar]
  • 15.Barrett JF, Goldschmidt RM, Lawrence LE, Foleno B, Chen R, Demers JP, et al. Antibacterial agents that inhibit two-component signal transduction systems. Proc Natl Acad Sci USA 1998; 95: 5317–5322. 10.1073/pnas.95.9.5317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Macielag M, Demers JP, Fraga-Spano SA, Hlasta DJ, Johnson SG, Kanojia RM, et al. Substituted salicylanilides as inhibitors of two-component regulatory systems in bacteria. J Med Chem 1998; 4: 2939–2945. 10.1021/jm9803572 [DOI] [PubMed] [Google Scholar]
  • 17.Sui Z, Guan J, Hlasta DJ, Macielag MJ, Foleno BD, Goldschmidt RM, et al. SAR studies of diaryltriazoles against bacterial two-component regulatory systems and their antibacterial activities. Bioorg Med Chem Lett 1998; 8:1929–1934. 10.1016/s0960-894x(98)00325-4 [DOI] [PubMed] [Google Scholar]
  • 18.Liu Qian, Yeo Won-Sik, and Bae Taeok. The SaeRS Two-Component System of Staphylococcus aureus. Genes (Basel) 2016; 7: E81 10.3390/genes7100081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bharadwaja Vadloori A. K. Sharath N. Prakash Prabhu and Radheshyam Maurya. Homology modelling, molecular docking, and molecular dynamics simulations reveal the inhibition of Leishmania donovani dihydrofolate reductase-thymidylate synthase enzyme by Withaferin-A. Vadloori et al. BMC Res Notes (2018) 11:246–252. 10.1186/s13104-018-3354-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Igarashi M, Watanabe T, Hashida T, Umekita M, Hatano M, Yanagida Y, et al. Waldiomycin, a novel WalK-histidine kinase inhibitor from Streptomyces sp. MK844-mF10. J Antibiot (Tokyo) 2013, 66(8):459–64. 10.1038/ja.2013.33 [DOI] [PubMed] [Google Scholar]
  • 21.Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol 1990, 215(3):403–410. 10.1016/S0022-2836(05)80360-2 [DOI] [PubMed] [Google Scholar]
  • 22.Kuroda M, Ohta T, Uchiyama I, Baba T, Yuzawa H, Kobayashi I, et al. Whole genome sequencing of meticillin-resistant Staphylococcus aureus. Lancet 2001, 357:1225–1240. 10.1016/s0140-6736(00)04403-2 [DOI] [PubMed] [Google Scholar]
  • 23.Laskowski RA, Moss DS, Thornton JM. PROCHECK-a program to check the stereochemical quality of protein structures. J App Cryst 1993, 26:283–291. 10.1107/S0021889892009944 [DOI] [Google Scholar]
  • 24.Webb B, Sali A. Comparative Protein Structure Modeling Using Modeller. Curr Protoc Bioinformatics 2016, 54:5.6.1–5.6.37. 10.1002/cpbi.3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Case DA, Ben-Shalom IY, Brozell SR, Cerutti DS, Cheatham TE, Cruzeiro VWD, et al. AMBER 2018, University of California, San Francisco. [Google Scholar]
  • 26.Yue Y, Sergio EW, Felice CL. Understanding a substrate’s product regioselectivity in a family of enzymes: A case study of acetaminophen binding in cytochrome P450s. PLoS One 2014; 9: e87058 10.1371/journal.pone.0087058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Turner M, Mutter ST, Kennedy-Britten OD, Platts JA. Molecular dynamics simulation of aluminium binding to amyloid-β and its effect on peptide structure. PLoS ONE 2019; 14: e0217992 10.1371/journal.pone.0217992 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wang J, Wang W, Kollman PA, Case DA. Automatic atom type and bond type perception in molecular mechanical calculations. J Mol Graph Model 2006; 25: 247–260. 10.1016/j.jmgm.2005.12.005 [DOI] [PubMed] [Google Scholar]
  • 29.Wang J, Wolf RM, Caldwell JW, Kollman PA. Case DA. Development and testing of a general AMBER force field. J Comput Chem 2004; 25: 1157–1174. 10.1002/jcc.20035 [DOI] [PubMed] [Google Scholar]
  • 30.Allen WJ, Balius TE, Mukherjee S, Brozell SR, Moustakas DT, Lang PT, et al. DOCK 6: Impact of new features and current docking performance. J Comput Chem. 2015; 36: 1132–1156. 10.1002/jcc.23905 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kollman PA, Massova I, Reyes C, Kuhn B, Huo S, Chong L, et al. Calculating Structures and Free Energies of Complex Molecules: Combining Molecular Mechanics and Continuum Models. Acc Chem Res 2000; 33: 889–897. 10.1021/ar000033j [DOI] [PubMed] [Google Scholar]
  • 32.Onufriev A, Bashford D, Case DA. Exploring protein native states and large-scale conformational changes with a modified generalized born model. Proteins 2004; 55: 383–394. 10.1002/prot.20033 [DOI] [PubMed] [Google Scholar]
  • 33.Weiser J, Shenkin PS, Still WC. Approximate atomic surfaces from linear combinations of pairwise overlaps (LCPO). J Comput Chem 1999; 20: 217–230. [DOI] [Google Scholar]
  • 34.Cai Y, Su M, Ahmad A, Hu X, Sang J, Kong L, et al. Conformational dynamics of the essential sensor histidine kinase walk. Acta Crystallogr D Struct 2017; 73: 793–803. 10.1107/S2059798317013043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rost B. Twilight zone of protein sequence alignments. Protein Eng 1999; 12:85–94. 10.1093/protein/12.2.85 [DOI] [PubMed] [Google Scholar]
  • 36.Nikolaev Dmitrii M., Shtyrov Andrey A., Panov Maxim S., Jamal Adeel, Chakchir Oleg B., Kochemirovsky Vladimir A., Olivucci Massimo, and Ryazantsev Mikhail N. A Comparative study of modern homology modeling algorithms for rhodopsin structure prediction. ACS Omega 2018; 3(7): 7555–7566. 10.1021/acsomega.8b00721 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Shen J, Li W, Liu G, Tang Y, Jiang H. Computational Insights into the Mechanism of Ligand Unbinding and Selectivity of Estrogen Receptors. J Phys ChemB 2009; 113: 10436–10444. 10.1021/jp903785h [DOI] [PubMed] [Google Scholar]
  • 38.Cheng T, Li Q, Zhou Z, Wang Y, Bryant S. Structure-based virtual screening for drug discovery: a problem-centric review. AAPS J 2012; 14: 133–141. 10.1208/s12248-012-9322-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Heitz F, Van Mau N. Protein structural changes induced by their uptake at interfaces. Biochim Biophys Acta 2002; 1597: 1–11. 10.1016/s0167-4838(02)00273-x [DOI] [PubMed] [Google Scholar]
  • 40.Li W, Shen J, Liu G, Tang Y, Hoshino T. Exploring coumarin egress channels in human cytochrome p450 2a6 by random acceleration and steered molecular dynamics simulations. Proteins 2011; 79: 271–281. 10.1002/prot.22880 [DOI] [PubMed] [Google Scholar]
  • 41.Lee VS, Tue-ngeun P, Nangola S, Kitidee K, Jitonnom J, Nimmanpipug P, et al. Pairwise decomposition of residue interaction energies of single chain Fv with HIV-1 p17 epitope variants. Mol Immunol 2010; 47: 982–990. 10.1016/j.molimm.2009.11.021 [DOI] [PubMed] [Google Scholar]
  • 42.Hou T, Wang J, Li Y, Wang W. Assessing the performance of the MM/PBSA and MM/GBSA methods. 1. The accuracy of binding free energy calculations based on molecular dynamics simulations. J Chem Inf Model. 2011;51(1):69–82. 10.1021/ci100275a [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chan DC, Chutkowski CT, Kim PS. Evidence that a prominent cavity in the coiled coil of HIV type 1 gp41 is an attractive drug target. Proc Natl Acad Sci 1998; 95: 15613–15617. 10.1073/pnas.95.26.15613 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Concepción Gonzalez-Bello

31 Mar 2020

PONE-D-20-05993

Docking studies and molecular dynamics simulations of the binding characteristics of waldiomycin and its methyl ester analogue to histidine kinase of Staphylococcus aureus

PLOS ONE

Dear Dr Awwad Radwan,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

I fully agree with the comments indicated by the referees. All the points indicated need to be addressed in detail. Additionally, the authors must consider and discuss accordingly the possibility that the ester may be a proform of the carboxylic acid. Therefore, it is important to address the importance of having a negatively charged ligand for recognition, as noted in the comments. This is quite often. 

We would appreciate receiving your revised manuscript by April 30th. When you are ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter.

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). This letter should be uploaded as separate file and labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. This file should be uploaded as separate file and labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. This file should be uploaded as separate file and labeled 'Manuscript'.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We look forward to receiving your revised manuscript.

Kind regards,

Concepción Gonzalez-Bello, Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2.  We suggest you thoroughly copyedit your manuscript for language usage, spelling, and grammar. If you do not know anyone who can help you do this, you may wish to consider employing a professional scientific editing service.  

Whilst you may use any professional scientific editing service of your choice, PLOS has partnered with both American Journal Experts (AJE) and Editage to provide discounted services to PLOS authors. Both organizations have experience helping authors meet PLOS guidelines and can provide language editing, translation, manuscript formatting, and figure formatting to ensure your manuscript meets our submission guidelines. To take advantage of our partnership with AJE, visit the AJE website (http://learn.aje.com/plos/) for a 15% discount off AJE services. To take advantage of our partnership with Editage, visit the Editage website (www.editage.com) and enter referral code PLOSEDIT for a 15% discount off Editage services.  If the PLOS editorial team finds any language issues in text that either AJE or Editage has edited, the service provider will re-edit the text for free.

Upon resubmission, please provide the following:

  • The name of the colleague or the details of the professional service that edited your manuscript

  • A copy of your manuscript showing your changes by either highlighting them or using track changes (uploaded as a *supporting information* file)

  • A clean copy of the edited manuscript (uploaded as the new *manuscript* file)

Additional Editor Comments (if provided):

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Partly

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The authors of this paper study the binding properties of waldiomycin and its methyl ester analog to histidine kinase (HK) of Staphylococcus aureus. To this purpose, the 3D structure of the catalytic domain of S. aureus HK was first generated using a homology model. Then, they study the docking of the two molecules and perform MD simulations on the more stable predicted complexes. Finally, the binding free energies of the two derivatives were calculated using the generalized Born surface area approach (MM-GBSA). According to the calculations, the higher affinity displayed by waldiomycin relative to its methylated analog can be explained because the carboxylate group of the former is engaged in a hydrogen bond with Lys100 of the protein. This stabilizing interaction is not possible in the methyl ester variant.

The work is well conducted, and the reviewer believes that it can deserve publication in PLOS ONE.

However, before it is published, authors should clarify the followings:

1) The homology model is based on a protein that presents only an identity of 36%. This result could lead to erroneous conclusions. This point should be explicitly discussed in the manuscript.

2) Is there any experimental evidence that supports the significance of Lys100 for the recognition of waldiomycin?

3) The MM-GBSA approach does not consider the entropy associated with the binding. This point should be discussed in the paper.

Minor points:

1) Page 6. Lines 142-144. The following paragraph should be rewritten: 'Using the TIP3P water solvent model, the protein molecule was included in a water box with 10 Å thickness and using sodium ions were as counter ions and using explicit solvation'.

2) Page 7. Lines 163-164. The sentence is not correct. The GAFF force field is applied only for the ligands and not for the complexes. In this regard, what is the approach used to calculate the partial charges of the ligands (BCC, HF/6-31G*, etc.)? What is the starting structure used for these ligands? The X-ray structure of waldiomycin has been previously reported (deposition number: CCDC 892997)

3) The structure of the best ten candidates derived from the docking calculations, together with their corresponding score, should be included in the Supporting Information.

4) The MD simulations were done for a short time (35 ns). What is the reason for using this simulation time?

5) Page 12. The interaction between the ligands and the receptor should be explained in detail (hydrogen bonding, CH/p, etc.).

6) Figure 2: the font used is tiny and difficult to read. Why are the authors showing two different binding sites?

7) Figure 3: the double bond in one of the aromatic rings of both molecules is not adequately represented.

8) Figure 5: the number are difficult to read. In Figure 5a, is it true that the red line corresponds to all atoms of the protein? There is a typo in the Figure caption: 'toms' should be 'atoms’.

9) Figure 6: The labels are difficult to read.

10) Figure 7: An interaction diagram should be added for both complexes.

11) Combine Figure 8 and 9 and use the same color code as the one used in Figure 9 to show the RMSD of the methylated derivative.

Reviewer #2: Bacterial histidine kinases (HK) are interesting targets for the design of antibacterial agents. The authors here report the computational studies of the binding to the S. aureus HK/Walk domain of two known inhibitors: waldiomycin and its methyl ester analogue. The interest of the research is based in the difference in the potency of these two ligands: waldiomcyin presents 7.4‐ fold stronger potency for over waldiomycin ester. The authors provide a 3D perspective about their biding modes correlated with this affinity difference. They propose a homology model of the S. aureus HK enzyme and use it for docking and MD simulations. The computational work has been conducted in a professional manner. The here reported studies are relevant in the context of finding novel antimicrobial agents and providing new insights into the SAR of waldiomycin as S. aureus HK inhibitor.

However, the results are not sufficiently well detailed and discussed and some additional studies are missed.

The antimicrobial activity (MIC) of waldiomycin is reported in ref. 20 (Igarashi et al., 2013). At the conclusions, the authors state that “waldiomcyin presents 7.4‐ fold stronger potency for over waldiomycin ester”. This statement should be justified in the main text and also the context where “potency” is used. In any case, the concepts of “affinity” and “potency” are mixed along the manuscript. This should be revised and clarified.

Line 87. “Unfortunately, few inhibitors of TCS have been described against S. aureus, a major pathogen with high virulence factor”. A reference should be cited here. Which inhibitors are these? They should be docked in order to compare with waldiomycin. For example, the compounds cited in ref. 20 (Igarashi et al., 2013) could be good examples to be docked and compared with waldiomycin.

Also the lack or low binding of waldiomycin towards human kinases could be studied (docking) as control, in order to avoid secondary/toxic effects. Lys100 was found to be crucial residue for S aureus HK binding of waldiomcin. Is this Lys present in human kinases?

The discussion about the docking poses and the results about the binding pose after the MD simulation should be enriched: more details about the interactions, pockets and residues that are important for the binding.

It is not clearly mentioned which is the protonation state considered for waldiomycin (protonation state under physiological condition). Is it docked as carboxylate or as carboxylic acid? The authors mention the H-bond with Lys100 (from the figures, it seems that with the side chain ammonium). This interaction drives the carboxylic/carboxylate group towards the Lys100 ammonium, while in the case of the waldiomycin methyl ester is not clear which interactions are not stable, and which new interactions are stablished in the new binding pose after the simulation (why now the new interactions do stabilize the new pose).

Why does waldiomycin methyl ester migrates to other binding pocket? Was this pocket reached by any predicted docked pose? This compound has a reported low affinity but it remains bound during the simulation. What does the energetics analysis say about the interactions? The contributions to the binding free energy from each residue should be analyzed from the MM-GBSA and discussed in the light of the new binding pose for the waldiomycin methyl ester.

Can the authors deepen into the required interactions for a ligand to be a good S. aureus HK inhibitors?

Minor corrections

-Line 279-284. Refs 37 and 38 referring to limitations in ligand-protein docking (2012 and 2002) and ref 39 as publication to illustrate advances in MD simulations (2011) are obsolete. More recent references should be cited.

-FIG5.

(a) Scale of the time should be adapted at the actual time of the simulation (0 to 30 ns?).

(b) The residue numbers should (x-axis) should be cut before 250. A label should be added indicating this corresponds to residue numbering.

-FIG.6 should go to SI. It´s a calculation made as validation, it´s not a main result of the work.

-Typos:

Line 90- “the 3D structure is unavailable in protein databank”.

It probably should say “the 3D structure is unavailable in the protein databank”. The PDB web page should be added as reference.

Additional work is required, which I think is necessary for the sake of sound conclusions, impact of the work and usefulness to the scientific community. In view of the comments noted above, I would recommend its acceptance for publication in PLOS ONE only when the corrections have been addressed.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files to be viewed.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2020 Jun 5;15(6):e0234215. doi: 10.1371/journal.pone.0234215.r002

Author response to Decision Letter 0


24 Apr 2020

Dear sir

I'm so thankful for your valuable comments and suggestions.

Addition work was performed and included in the manuscript based on the reviewer comment. The work includes hydrogen bond analysis cpptraj analysis and the pairwise energy decomposition analysis using mmpbsa calculations in Amber18.

The revised submission includes:

The response for reviewer file includes point by point answer of the reviewer comments.

Mansuscript with track changes.

There is also a clear manuscript file after language edition through editage service.

Certificate of editing from editage service is included.

Supporting information are uploaded as zip compressed archive file.

Wit my grateful appreciation to your help and communications.

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 1

Concepción Gonzalez-Bello

18 May 2020

PONE-D-20-05993R1

Docking studies and molecular dynamics simulations of the binding characteristics of waldiomycin and its methyl ester analog to Staphylococcus aureus histidine kinase

PLOS ONE

Dear Dr. Radwan,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

The manuscript requires to address the points highlighted by the two referees to be suitable for publication.

We would appreciate receiving your revised manuscript by May 25th 2020. When you are ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter.

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). This letter should be uploaded as separate file and labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. This file should be uploaded as separate file and labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. This file should be uploaded as separate file and labeled 'Manuscript'.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We look forward to receiving your revised manuscript.

Kind regards,

Concepción Gonzalez-Bello, Ph.D.

Academic Editor

PLOS ONE

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: All my comments have been addressed, and the manuscript is now acceptable for publication.

Minor points:

1) Units of the grid score should be indicated in both the main text and Supporting Information.

2) Units of pairwise energy decomposition should be shown in Table 1 and the main text. Is this energy given in Kcal or Kcal/mol? (see main text: 'range value -2.722 to -4.371 Kcal (Table 1)'

Reviewer #2: The authors have successfully addressed all the concerns and corrections.

Minor correction:

Line 301: Units should be specified. And probably E values can be rounded to two decimals (this is sufficiently precise in this order of magnitude).

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files to be viewed.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2020 Jun 5;15(6):e0234215. doi: 10.1371/journal.pone.0234215.r004

Author response to Decision Letter 1


18 May 2020

Dear Sir

The corrections were done in view of the reviewer comments.

The point by point answer of the reviewer comments are listed below:

6. Review Comments to the Author

Reviewer #1: All my comments have been addressed, and the manuscript is now acceptable for publication.

Minor points:

**1) Units of the grid score should be indicated in both the main text and Supporting Information.

Answer:

>>Page 14, line 284,285: The unit, “kcal mol–1 “, was included in the main test; and also in S6 and S7 files of the Supporting Information.

**2) Units of pairwise energy decomposition should be shown in Table 1 and the main text. Is this energy given in Kcal or Kcal/mol? (see main text: 'range value -2.722 to -4.371 Kcal (Table 1)'

Answer:

>>Page 17, line 348. The unit was changed into “kcal mol-1).

Reviewer #2: The authors have successfully addressed all the concerns and corrections.

Minor correction:

Line 301: Units should be specified. And probably E values can be rounded to two decimals (this is sufficiently precise in this order of magnitude).

Answer:

>>Page 18, lines 362, 363. E values were rounded to two decimals and units “kcal mol-1” were specified.

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 2

Concepción Gonzalez-Bello

21 May 2020

Docking studies and molecular dynamics simulations of the binding characteristics of waldiomycin and its methyl ester analog to Staphylococcus aureus histidine kinase

PONE-D-20-05993R2

Dear Dr. A. Radwan,

We are pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it complies with all outstanding technical requirements.

Within one week, you will receive an e-mail containing information on the amendments required prior to publication. When all required modifications have been addressed, you will receive a formal acceptance letter and your manuscript will proceed to our production department and be scheduled for publication.

Shortly after the formal acceptance letter is sent, an invoice for payment will follow. To ensure an efficient production and billing process, please log into Editorial Manager at https://www.editorialmanager.com/pone/, click the "Update My Information" link at the top of the page, and update your user information. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to enable them to help maximize its impact. If they will be preparing press materials for this manuscript, you must inform our press team as soon as possible and no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

With kind regards,

Concepción Gonzalez-Bello, Ph.D.

Academic Editor

PLOS ONE

Reviewers' comments:

Acceptance letter

Concepción Gonzalez-Bello

26 May 2020

PONE-D-20-05993R2

Docking studies and molecular dynamics simulations of the binding characteristics of waldiomycin and its methyl ester analog to Staphylococcus aureus histidine kinase

Dear Dr. Radwan:

I am pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximize its impact. If they will be preparing press materials for this manuscript, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

For any other questions or concerns, please email plosone@plos.org.

Thank you for submitting your work to PLOS ONE.

With kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Concepción Gonzalez-Bello

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Ramachandran plot of the optimized S. aureus HK/WalK structure.

    (PDF)

    S2 Fig. Ramachandran plot of the initial homology modeled S. aureus HK/WalK structure.

    (PDF)

    S3 Fig

    The co-crystallized ACP (from 5c93.pdb, colored magenta) and the redocked ACP structure (colored forest green), superimposed inside the binding site of 5c93.

    (TIF)

    S4 Fig. The 10 top-scored conformations of the waldiomycin in mol2 format.

    (TIF)

    S5 Fig. The 10 top-scoring conformations of the waldiomycin methyl ester in mol2 format.

    (TIF)

    S1 File. The docking score and hydrogen bond information of the top 10 scored conformations of waldiomycin.

    (INFO)

    S2 File. The docking score and hydrogen bond information of the top 10 scored conformations of waldiomycin methyl ester.

    (INFO)

    Attachment

    Submitted filename: Response to Reviewers.docx

    Attachment

    Submitted filename: Response to Reviewers.docx

    Data Availability Statement

    All relevant data are within the paper and its Supporting Information files.


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES