A molecular model to study FosA enzyme inhibition

https://doi.org/10.1016/j.jmgm.2021.107978Get rights and content

Highlights

  • FosA reaction leads fosfomycin to lose its pharmacological effect and promotes fosfomycin resistance.

  • We have elucidated the dynamic interactions between the enzyme FosA and the inhibitor ANY1.

  • The molecular model developed here describes the protein-ligand affinity with R2 of 77.51%.

  • Additional residues should be considered in the efforts to design novel attractive FosA inhibitors.

Abstract

Fosfomycin resistance protein (FosA) is a metalloenzyme known for catalyzing a nucleophilic addition reaction of glutathione to the epoxide ring of Fosfomycin, a broad-spectrum antibiotic used to combat Gram-positive pathogens. The reaction leads fosfomycin to lose its pharmacological effect, thus promotes antibiotic resistance. A small-molecule FosA inhibitor has been discovered. ANY1 (3-bromo-6-[3-(3-bromo-2-oxo-1H-pyrazolo[1,5-a]pyrimidin-6-yl)-4-nitro-1H-pyrazol-5-yl]-1H-pyrazolo[1,5-a]pyrimidin-2-one) is competitive with the antibiotic for binding the active site of the enzyme. Through Molecular Mechanics methods, using the AMBER force field, we carry out molecular dynamics simulations and binding free energy calculations to investigate the most important interactions between the enzyme and inhibitor. Our results were able to reproduce the trend of experimental data with R2 of 77.51%. Furthermore, we have shown that electrostatic and van der Waals interactions, as well as cavitation energies, are favorable for maintaining the enzyme-inhibitor complex, while reactive field energies and non-polar interactions act in an unfavorable way for interactions between FosA and ANY1.

Introduction

Fosfomycin (FOF) is a broad-spectrum antibiotic with a safety and effectiveness history administration in humans [1]. Its bactericidal activity is exerted by covalent binding to UDP-N-acetylglucosamine-enolpyruvyl transferase (MurA), an enzyme that catalyzes the first step in cell wall biosynthesis. It is important to note that Fosfomycin is highly active against Escherichia coli, including beta-lactamase producers [2]. In gram-negative bacteria, the clinically relevant resistance mechanism involves the presence of the FosA metalloenzyme, which is capable of catalyzing the degradation of the fosfomycin epoxide ring by adding nucleophilic glutathione (GSH), metabolizing the antibiotic structure and thus breaking its pharmacological effect [3,4].

The FosA enzyme is a homodimer composed of two active sites coordinating Mn2+ ions that bind to fosfomycin and act as Lewis acids during the nucleophilic attack by glutathione. A K+ ion also binds near the active site, balancing negative charges and increasing the rate of reaction [3]. The fosA gene is often found on chromosomes of many Gram-negative bacteria, such as Klebsiella pneumoniae, conferring intrinsic resistance to fosfomycin. Escherichia coli has already been reported presenting the fosA3 gene in its plasmid. This gene is responsible for the synthesis of FosA3 enzymes. FosAKP and FosAPA are enzymes derived from the chromosomes of the bacterias Klebsiella pneumoniae and Pseudomonas aeruginosa. Experimental results report that FosA3 and FosAKP exhibit higher resistances to fosfomycin than FosAPA, since the previous enzymes have larger loops at the interface of their dimers, compared with FosAPA, and that the interface of these loops has an influence on FosA activity [3].

A competitive small-molecule inhibitor was found to increase fosfomycin activity against Gram-negative bacteria [2]. Among several molecules with potential inhibition of FosA, ANY1 (3-bromo-6-[3-(3-bromine-2-oxo-1H-pyrazolo [1,5-a]pyrimidin-6-yl)-4-nitro-1H-pyrazole-5-yl]-1H-pyrazolo [1,5-a]pyrimidin-2-one) (Fig. 1) was the most potent of them with IC50 of 5.1 ± 2.2 μM. This inhibithor can bind to the active site of the metaloenzyme, ceasing the metabolization of FOF in the reaction with GSH. Furthermore, the citotoxicity of ANY1 in association with Fosfomycin was experimentally evaluated and it was found that administration of this compound with the antibiotic is safe and ANY1 had a minor impact in cell viability both in human kidney cells and human peripheral mononuclear cells [2]. Thus, this small-molecule is an advantageous booster of Fosfomycin antibacterial activity (see Fig. 1).

Therefore, we conjecture that the discovery and development of FosA inhibitors can significantly expand the use of fosfomycin against gram-negative pathogens that have the gene responsible for the production of this metalloenzyme. In this work, the interactions between two FosA enzymes and the inhibitor ANY1 were evaluated. Their affinities were estimated by the calculation of binding free energies based on molecular mechanics (MM) methods and molecular dynamics (MD) trajectory. Our results show at a molecular level the dynamics of the interactions between ANY1 and the active site residue that contribute to the inhibitor's affinity with the protein. Additionally, we found a great correlation between the predicted results in silico and the results obtained experimentally.

Section snippets

Molecular model

The starting models for the simulations of FosA enzyme structures in complex with the inhibitor were obtained from the RCSB Protein Data Bank (http://rcsb.org) with the codes 5WEW and 5WEP, corresponding to the FosAKP-ANY1 and FosA3EC-ANY1 systems, respectively [2]. From these systems, the FosAKP enzyme has the divalent manganese metal as a cofactor while FosA3EC presents the divalent zinc metal in its structure. The H++ server (http://biophysics.cs.vt.edu/) was used to add hydrogen atoms to

Evaluating the dynamic effect on the complex FosA-ANY1

Recently, it has been shown that fosfomycin binding modulates cooperative motions in the K+-binding loop, glutathione channel, and dimer interface loop [3]. We used molecular dynamics simulation to analyze FosA flexibility when complexed with ANY1, as measured by the RMSD of the movement of backbone atoms, and residue flexibility, as measured by the RMSF. The RMSD plot of the enzymes backbone atoms shows average values of 1.78 Å for the FosAKP-ANY1 complex and 1.35 Å for FosA3-ANY1, considering

Conclusions

An analysis at the molecular level allowed us to elucidate the dynamic interactions between the enzyme FosA and the inhibitor ANY1. Through MD simulations and free energy calculations, it was possible to quantify these interactions and analyze them from an atomistic point of view. The molecular dynamics simulations of the enzyme-inhibitor systems showed that the FosA enzymes have deviations less than 1.80 Å in the structures of their backbones. The binding free energy calculations have shown

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

The authors would like to thank PROPESP/UFPA and CNPq for the financial support.

References (18)

There are more references available in the full text version of this article.

Cited by (1)

View full text