Next Article in Journal
Enhancing Endocannabinoid Signaling via β-Catenin in the Nucleus Accumbens Attenuates PTSD- and Depression-like Behavior of Male Rats
Previous Article in Journal
Long-Term Persistence of Mitochondrial DNA Instability in HIV-Exposed Uninfected Children during and after Exposure to Antiretroviral Drugs and HIV
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Inhibitory Potential of Ferulic Acid Derivatives against the SARS-CoV-2 Main Protease: Molecular Docking, Molecular Dynamics, and ADMET Evaluation

by
Io Antonopoulou
*,†,
Eleftheria Sapountzaki
,
Ulrika Rova
and
Paul Christakopoulos
Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental and Natural Resources Engineering, Luleå University of Technology, SE-97187 Luleå, Sweden
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2022, 10(8), 1787; https://doi.org/10.3390/biomedicines10081787
Submission received: 15 June 2022 / Revised: 19 July 2022 / Accepted: 21 July 2022 / Published: 25 July 2022
(This article belongs to the Section Drug Discovery, Development and Delivery)

Abstract

:
The main protease (Mpro) of SARS-CoV-2 is an appealing target for the development of antiviral compounds, due to its critical role in the viral life cycle and its high conservation among different coronaviruses and the continuously emerging mutants of SARS-CoV-2. Ferulic acid (FA) is a phytochemical with several health benefits that is abundant in plant biomass and has been used as a basis for the enzymatic or chemical synthesis of derivatives with improved properties, including antiviral activity against a range of viruses. This study tested 54 reported FA derivatives for their inhibitory potential against Mpro by in silico simulations. Molecular docking was performed using Autodock Vina, resulting in comparable or better binding affinities for 14 compounds compared to the known inhibitors N3 and GC376. ADMET analysis showed limited bioavailability but significantly improved the solubility for the enzymatically synthesized hits while better bioavailability and druglikeness properties but higher toxicity were observed for the chemically synthesized ones. MD simulations confirmed the stability of the complexes of the most promising compounds with Mpro, highlighting FA rutinoside and compound e27 as the best candidates from each derivative category.

1. Introduction

During the past two years, public health and socioeconomic life have been going through a severe crisis due to the Coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2), which has led to more than 450 million cases and more than 6 million deaths worldwide [1]. SARS-CoV-2 belongs to the family of coronaviruses, which have posed a threat to public health in the past, with the severe acute respiratory syndrome-coronavirus (SARS-CoV) and Middle East respiratory syndrome (MERS-CoV) outbreaks in 2002 and 2012, respectively [2].
The development of vaccines was remarkably quick, allowing many countries to initiate the vaccination process in the beginning of 2021 and, therefore, provide a valuable weapon to boost immunity against the virus [3]. As far as drugs and other non-vaccine therapeutic options are concerned, remdesivir, an RNA-dependent RNA polymerase inhibitor, is the only one to receive FDA approval for use in COVID-19 patients [4]. Currently, 15 other products have received emergency use authorizations by the FDA, including the protease inhibitor Paxlovid [5].
However, new variants continue to emerge, affecting the transmissibility of the virus, the impact of the disease, and the immunity against it. Currently, variants beta, gamma, delta, and omicron are labeled as variants of concern (VOCs) by WHO [6]. Among them, the omicron variant is the most capable of antigenic escape, causing concern over the efficacy of current therapeutic routes, such as vaccines and monoclonal antibody treatments [7,8]. Thus, it is of great importance to develop tools that can remain effective against potential viral mutations.
Therapeutic targets to combat COVID-19 include structural and functional proteins of the virus, and virulence factors and host proteins that are useful for viral proliferation. Among them, the focus of this work is the SARS-CoV-2 main protease (Mpro, 3CLpro, or Nsp5). Mpro is a very promising antiviral target, as it plays a major role in the viral life cycle, while there is also adequate information available on its structure and mechanism to allow further investigation, both in silico and in vitro [9]. Inhibition of Mpro is expected to exhibit high specificity and limited side effects, as there are no other human enzymes that recognize the same sequence and the peptide bond as a cleavage site [10]. In addition, Mpro seems to have fewer mutation hotspots compared to other targets, such as the spike protein [11]. More specifically, the main protease of the SARS-CoV-2 omicron and delta variants are almost identical to that of the wild type, with only one prevalent mutation observed in the case of omicron (P132H) and none in the case of delta [12,13]. Moreover, the protease exhibits high conservation among coronaviruses (e.g., 96% sequence identity between SARS-CoV-2 and SARS-CoV main protease), meaning that its inhibitors are very likely to be effective against variants or other viruses of the same family [14,15].
Taking into consideration all the above, and the fact that there are numerous natural compounds that are being screened for their antiviral properties and have exhibited efficacy in fighting a wide range of viruses, including SARS-CoV-2, plants emerge as a potential valuable source of bioactive compounds, which could be utilized as nutraceuticals to contribute to the protection against a viral infection and potentially aid immunity [16,17,18].
Ferulic acid (FA, 4-hydroxy-3-methoxycinnamic acid) is a phenolic bioactive compound belonging to a group of hydroxycinnamic acids that have drawn attention as nutraceuticals due to their numerous beneficial properties, such as high antioxidant activity, anti-inflammatory, antibacterial, neuro- and photoprotective, anticancer, antidiabetic, and skin-whitening effects [19,20,21]. It has also been identified as an antiviral agent against several viruses [22], including SARS-CoV-2 [23]. The profile of FA as a promising nutraceutical is reinforced by its low toxicity; however, its bioavailability is limited and needs to be improved [24]. Although further investigation of its pharmacokinetic properties is required, its absorption and metabolism appear to be dose and form dependent, indicating that further investigation of FA derivatives could lead to compounds with improved pharmacokinetic properties [25,26,27].
FA is abundantly present in vegetables, fruits, cereals, flowers, leaves, beans, coffee seeds, and nuts [28,29]. It can be found in monocots (rice, wheat, etc.) and dicots, (e.g., sugar beet pulp, spinach, glasswort, carrot) [30] and is the most commonly found hydroxycinnamic acid in plant cell walls, where it is esterified to polysaccharides. In monocots, it is bound to xylan at the O-5 of its-l-arabinose moieties while in dicots, it is often bound to the neutral side chains of pectin, esterified to the O-2 of a-l-arabinose or to the O-6 of b-d-galactose units. It also forms dimers and trimers, which create crosslinks between polysaccharide chains and lignin, contributing to the rigidity of the lignocellulosic matrix of plant cell walls. Overall, it is observed mainly in its trans- isomeric form and esterified with mono- and disaccharides, glycoproteins, polyamines, hydroxylated fatty acids, alcohols, and flavonoids, apart from plant cell wall polysaccharides [28,29,31]. Various studies have been carried out on possible routes for the derivatization of FA towards compounds with preferable bioactive and pharmacokinetic properties [32,33,34,35,36]. Such derivatives can be synthesized chemically or enzymatically, and many of them have exhibited promising antiviral properties [22].
This work focuses on the evaluation of a broad range of FA derivatives for their ability to bind to and, therefore, inhibit Mpro. The goal is to determine which compounds are more likely to form a stable complex with Mpro and to provide further information on the effect of derivatization on the inhibitory efficacy of the compounds, and their pharmacokinetic properties. By examining enzymatically and chemically synthesized derivatives with varying structural characteristics, additional insight is gained into the potential of the different routes for the development of antiviral nutraceuticals.

2. Materials and Methods

2.1. Ligand and Receptor Preparation for Docking Simulations

The phytochemicals selected were FA derivatives reported in the literature. Their structures were adopted from the original publications and then constructed in Chemsketch (ACD/Labs, Toronto, ON, Canada). The docking simulation essentially calculates the lowest possible energy and the conformation that leads to it of the complex between a larger macromolecule (receptor), in this case SARS-CoV-2 Mpro, and a smaller molecule (ligand), which in this case is a respective inhibitor or FA derivative. For the docking to be executed, the receptor and the ligand need to be defined and prepared. The 3D structure of each ligand, drawn in Chemsketch, was optimized through the same program, and saved in a .mol file, before being imported and energy minimized in YASARA Version 20.12.2 [37]. The protease (Mpro) structure used for the simulation is available in the Protein Data Bank (PDB) (https://www.rcsb.org/ accessed on 1 May 2022) under the PDB ID 6LU7, with a resolution of 2.16 Å. This structure was selected among the numerous structures deposited in the PDB because it was the first one to be made available and the one that is more often used in studies featuring molecular docking simulations. The ligand that is bound to the protease in this particular co-crystallization structure was deleted, the receptor was cleaned, and its hydrogen network was optimized. A simulation cell was built as a cube centered in the atoms of the catalytic dyad His41 and Cys145 and extended as many Å as needed for its side to be 2–3 Å longer than the length of each ligand. This margin allowed for flexibility as the ligand may acquire different conformations onto the active site; however, it was not so large that it increased the uncertainty of the docking simulation.

2.2. Molecular Docking Simulation and Data Output

Molecular docking was performed using the embedded macro in YASARA, AutoDock Vina, using the default parameters. The program calculates the binding energy of the possible receptor–ligand complexes taking into consideration steric, hydrophobic, and hydrogen-bonding interactions [38]. During the simulation, the program performs 25 docking runs, which produce 25 possible ligand–receptor-binding conformations. The different conformations that are arranged around the same hotspot and have an RMSD smaller than 5 Å from each other form a cluster. The number of clusters differs depending on the simulation, and the binding energy of the most favorable conformation within the cluster is reported.
After each simulation, the program generated a report documenting the output data for each run and cluster. This includes the binding energy (in kcal/mol), the dissociation constant (in pM), and the contacting residues for each case (including hydrogen bond, hydrophobic, pi-pi, cation–pi, and ionic interactions). The binding energy is calculated by subtracting the energy of the ligand–receptor complex in the bound state from the energy when the ligand is at an infinite distance from the receptor and is given as output from the software as a positive number. Based on the used simulation, a higher binding energy indicates a higher binding affinity. Reference to other works attributes the binding energies as negative values; therefore, in such cases, the lowest binding energies are regarded as the ones resulting in a higher binding affinity. The software manual states that the only difference between the positive binding energy values reported and the negative energies of binding reported in other works is the flipped sign. Therefore, we report the binding energies as negative values to be in accordance with the relevant literature.

2.3. Validation of the Docking Method

Apart from FA and its derivatives, two known Mpro inhibitors with available co-crystallization structures in complex with the protease deposited at the PDB were docked to Mpro. The docked complex was compared with the PDB co-crystallization complex in order to confirm the reliability of molecular docking and the selected software in particular as a tool to accurately simulate the targeted protein–ligand interactions. The selected inhibitors were N3 (PDB ID: 6LU7) and GC376 (PDB ID: 7D1M). The first cluster given as a simulation output was superposed to the co-crystallization structure and the reproducibility of the binding mode of the ligand in the docking simulation was evaluated based on the root-mean-square deviation (RMSD) and the intramolecular interactions.

2.4. Visualization of Binding Modes and Mpro–Ligand Interactions

The results were visualized using PyMOL Version 2.4.1 [39]. The hydrogen bonds reported are the ones calculated by PyMOL while hydrophobic and pi-pi interactions were calculated in YASARA. Intramolecular interactions of the most promising compounds were depicted using LIGPLOT v.4.5.3 [40].

2.5. ADMET Prediction

The canonical SMILE notations of the selected phytochemicals were obtained through Chemsketch and used as an input to available online software tools that provide data on the druglikeness, pharmacokinetic properties, and toxicity of the compounds. More specifically, SwissADME (http://www.swissadme.ch/index.php accessed on 1 May 2022) was used to obtain data on the physicochemical properties, lipophilicity, water solubility, pharmacokinetics, druglikeness, and medicinal chemistry of the compounds. A free online tool provided by Molsoft (http://molsoft.com/mprop/ accessed on 1 May 2022) was also utilized to calculate the molecular properties and overall druglikeness of the compounds. The toxicity of the compounds under evaluation was predicted through the freely available webserver ProTox II (https://tox-new.charite.de/protox_II/index.php?site=compound_input accessed on 1 May 2022), which calculated the median lethal dose (LD50) and provides an estimation of the acute toxicity, hepatotoxicity, carcinogenicity, immunotoxicity, mutagenicity, and cytotoxicity.

2.6. Molecular Dynamics Simulation

Molecular dynamics (MD) simulation was performed to further investigate the stability of the complexes of selected docked compounds with Mpro. The co-crystallization complex of the known inhibitor N3 with Mpro (PDB ID: 6LU7) was used as a reference. The simulation was run using YASARA Structure Version 20.12.24 for 10–30 ns, as this was considered a time within which the complexes reached equilibrium. The conditions set for the simulation were a pH of 7.4 [41], NaCl ions at a concentration of 0.9%, temperature of 298 K, and pressure of 1 atm. The setup included optimization of the hydrogen bonding network [42], energy minimization of the system, and definition of a simulation cell 20 Å larger than the protein in every direction. The AMBER14 force field [43] was used for the solute and the Van der Waals forces were calculated using a cutoff of 8 Å (the default used by AMBER [44]) while no cutoff was applied to electrostatic forces, which were calculated using the Particle Mesh Ewald algorithm [45]. The simulation trajectories were saved every 250 ps with a timestep of 2.5 fs for bonded interactions and 5.0 fs for non-bonded interactions [46]. The trajectories were analyzed to show the RMSD of the C-alpha atoms of the complexes, the root-mean-square fluctuation (RMSF) of the protein residues and its radius of gyration (RoG), and the hydrogen bonds between the protease and the ligands.

3. Results and Discussion

3.1. Mpro as a Receptor for Docking Simulations

The role of Mpro is the proteolytic cleavage of two viral polyproteins pp1a and pp1ab towards the formation of nonstructural proteins required for further viral reproduction, including its self-release from the aforementioned polyproteins [47,48]. There are more than 11 proteolytic sites, characterized by the sequence (Leu-Gln)-(Ser/Ala/Gly), where the peptide bond being hydrolyzed is the one after Gln [49]. The enzyme functions in a homodimeric form, through a mechanism of nucleophilic addition facilitated by a cysteine-histidine catalytic dyad (Cys145-His41). Each monomer is composed of 306 residues arranged in a polypeptide chain with 3 distinct domains (domain I: residues 8–101; domain II: residues 102–184; and domain III: residues 201–303) (Figure 1). The active site of the enzyme is a cavity formed between domains I and II, which are made of antiparallel β-barrels. Domain III is composed of five α-helices and contributes to the formation of the dimer [50,51,52,53].
Apart from the presence of catalytic residues His41 and Cys145, it is important to mention the existence of four main subsites forming the active site of Mpro, labeled as S1, S1′, S2, and S4. According to Stoddard et al. [2], the S1 subsite consists of the side chains of Phe140, Asn142, Ser144, Cys145, His163, Glu166, His172, and the backbone of Leu141, Gly143, His164, and Met165; the S1′ subsite is formed by the side chains of Thr25, His41, Val42, Asn119, Gly143, Cys145, and the backbone of Thr26; S2 is created by the side chains of His41, Met49, Tyr54, Asp187, and the backbone of Arg 188; and S4 is made up of the side chains of Met165, Leu167, Pro 168, Ala191, Gln192, and the backbones of Glu166, Arg188, and Thr190. Among them, His41, Gly143, Ser144, Cys145, and Glu166 have been pointed out as residues playing a major role in protein–ligand interactions in molecular dynamics studies [14]. Moreover, residues Gly143, Ser144, and Cys145 form an oxyanion hole capable of stabilizing the negative charge of ligands, such as that of the carbonyl oxygen of the scissile bond in the natural substrate [9,10,53].
Regarding the catalytic mechanism of the enzyme, it is suggested that it is established on the formation of a nucleophilic ion pair, through a proton transfer from the thiol group of Cys145 to the imidazole of His41. The catalytic cysteine attacks the carbonyl of the scissile bond, leading to a thiohemiketal intermediate, while the protonated histidine attacks the N-atom of the peptide bond, creating the acyl–enzyme complex intermediate. The participation of a water molecule in the reaction is of great importance, as it attacks the carbonyl carbon of the substrate’s Gln while the catalytic His is being reprotonated, and also stabilizes the polar contacts between residues His41, His164, and Asp187 by interacting with them. The last step of the mechanism is described by the release of Cys145 through the cleavage of its covalent bond with the peptide [53].

3.2. Docking Validation

Both known inhibitors examined, N3 and GC376, led to a docking output comparable to the co-crystallization structure of their complex with Mpro. N3 is the most widely accepted and analyzed inhibitor of Mpro in the literature [51,54,55]. It is often used as a positive control to provide some reference values with which the binding energy and interactions of a purported inhibitor with Mpro can be compared. The binding energy for N3 calculated in this work by Vina is −8.26 kcal/mol, whereas Das et al. [56] reported it to be −7.7 kcal/mol and Ahmed et al. [57] −7.5 kcal/mol, a difference that can be attributed to the different pretreatment of the receptor and the ligand structures before the simulation. Superimposing of the co-crystallized protein–ligand complex and the one calculated by the simulation reveals high similarity between the conformation of N3 in the binding site in the two cases, with an RMSD of 3.16 Å (Figure 2a). In the docking output, the interaction of N3 with Cys145 is a 2 Å hydrogen bond, formed between the pentacyclic ring of N3 and the hydrogen attached to the sulfur atom of the protein residue. Whereas, as determined by the co-crystallization data, it is a 1.8 Å covalent bond between the sulfur atoms of Cys145 and the Cβ atom of the vinyl group of the inhibitor. Although a different part of the inhibitor is bound to the protease, it results in a very similar geometry of the molecule. Moreover, among the seven protein–ligand hydrogen bonds calculated for the co-crystallization structure, involving residues Gly143, His163, His164, Glu166, Gln189, and Thr190, three were also calculated from the simulation (including Glu166 and Gln189). GC376 is another broad-spectrum inhibitor that has demonstrated activity against the main protease of various coronaviruses [58]. In the present simulation, the binding energy calculated for GC376 was −7.80 kcal/mol. The docking output is remarkably similar to the co-crystallized structure, with the ligands in the two conformations having an RMSD as low as 1.0 Å (Figure 2b). In addition, another study featuring the docking of GC376 to PDB 6LU7 with AutoDock Vina reported a binding energy of −8.1 kcal/mol [59], which is very close to the result of our study. The hydrogen bonds that formed appear to be matching to a great extent, as GC376 interacts with His41, Phe140, His164, Glu166, and Gln189 in the co-crystallized complex and with His41, Phe140, His163, Glu166, and Gln189 in the docked complex.
Overall, the docking results for the known inhibitors are highly comparable to the binding modes observed in reality, reinforcing the validity of molecular docking as a tool to provide indications for the inhibitory potential of the compounds under evaluation.

3.3. FA as a ‘Reference’ Ligand

Although FA derivatives have not been extensively studied in silico for their inhibitory potential against SARS-CoV-2 Mpro, FA itself has been part of related studies featuring natural compounds. The binding energy calculated by Autodock Vina for FA in this work was −5.88 kcal/mol for the first among six emerging clusters. The next two clusters did not deviate much from this value, with binding energies of −5.60 and −5.47 kcal/mol, respectively (Figure 3). In the first and third cluster, the phenolic group is situated in the S2 subsite, but the tail of the molecule extends towards the S4 subsite in the first case and towards the S1 subsite in the second. In the case of the second cluster, the phenolic ring is located between the S1 and S1′ subsites, in front of the catalytic dyad, while the other end of the molecule is inserted into the S2 subsite. His41 stands out as an interacting residue in all cases, involved in both hydrophobic and pi-pi interactions, while hydrogen bond interactions involve residues closer to the catalytic dyad in the case of clusters 2 and 3, and S4 site residues for cluster 1. Using the protease PDB structure 6LU7 and the Molecular Operating Environment 2019.0102 (MOE) software for molecular docking, two different studies reported almost identical binding energies of −5.33 [60] and −5.35 kcal/mol [61], and the formation of two and one hydrogen bonds, respectively, with residues Thr190 and Glu199, and Glu166. The latter also provided the orientation of the molecule when bound to the active site, which resembles the first cluster of this work (Figure 3), without being identical, however.
These results are quite similar with the results produced in the present work for the first cluster of FA, as the binding energies do not differ significantly and there is one common residue involved in hydrogen bonding (Thr190). Another work reported a binding energy of −10.63 kcal/mol and hydrogen bonds with residues Leu141, Gly143, Ser144, Cys145, His163, and Glu166 [62] while the orientation of FA in the active is identical to the one observed in the second cluster for FA in this work. In accordance with the present results, a docking score of −6.0 kcal/mol was reported for FA when the Autodock Vina and PDB structure 6W63 were used [63]. Overall, the docking scores for FA are quite low compared to established inhibitors such as the previously mentioned N3 (−8.26 kcal/mol) and GC376 (−7.80 kcal/mol). Therefore, the screening of FA derivatives aims to explore their potentially improved properties both in Mpro inhibition and in terms of pharmacokinetics.

3.4. Docking of Enzymatically Synthesized FA Derivatives

Enzymatic synthesis of FA derivatives has been prevalently dominated by esterification of FA or transesterification of a respective activated donor (such as methyl ferulate, MFA or vinyl ferulate, VFA), under a low water content towards the production of stabilized esters with tailored lipophilicity [36,64,65]. Esterases that are widely used for this purpose are the triaglycerol lipases (EC 3.1.1.3) due to their broad specificity towards glycerides and related substrates. Nevertheless, feruloyl esterases (EC 3.1.1.73), which have specificity towards hydroxynnamic acids, have also been employed for this purpose as they can offer the unique advantage of catalyzing the synthesis of esters with a wide range of substitutions on the phenolic ring, resulting in either more lipophilic (e.g., alkyl esters) or hydrophilic esters (e.g., sugar esters) [66,67,68,69,70]. Transglycosylation of FA with the glucoside rutin has been described [35]. An overview of the major FA derivatives that have been synthesized enzymatically along with the representative synthesis routes is described in Table 1. These derivatives were selected for further in silico simulations. Table 2 presents the results of the docking simulations for the major enzymatically synthesized derivatives.

3.4.1. Alkyl and Alkenyl Esters of FA

The FA derivatives belonging to this category that were investigated include methyl, ethyl, propyl, butyl, isobutyl, pentyl, isopentyl, prenyl, hexyl, octyl, dodecyl, and octadecyl ferulates. Overall, the calculated binding energies ranged from −5.20 to −6.75 kcal/mol, with prenyl ferulate exhibiting the highest binding affinity to the active site of Mpro. Although there is not a definite correlation between the binding energy and the potential of in vitro inhibitory effect, it is not encouraging that these values are significantly lower than the inhibitors N3 and GC376. The total results in terms of the binding energies and interactions calculated are shown in Table 2. It is interesting that prenyl ferulate is the only alkenyl ester among this category, and has better binding compared to the respective alkyl ester (isopentyl ferulate, −6.58 kcal/mol). It is observed that the binding energies become progressively lower as the carbon chain of the substitutions becomes larger, until the point of five carbons. For the larger substitutions, the binding affinity starts to decrease again.
The prevalent conformation observed is the one where the phenolic ring of the FA moiety is stabilized at the S1 subsite, in front of the catalytic residue Cys145, while the carbon chain extends towards the S2/S4 subsites. This particular orientation is seen in the first cluster for all ligands in this category, except for pentyl ferulate, where the orientation corresponds to the second cluster (Figure 4). The binding energy is almost identical between the two clusters. Moreover, no such conformation is observed in the cases of dodecyl and octadecyl ferulate, which possess a long carbon chain that folds differently into the active site.
Another commonly occurring geometry is characterized by the phenolic moiety of the ligands being located at the S2 subsite while the rest of the molecule is orientated towards the S1 or S1′ subsites, as seen, for example, in the case of the third cluster for methyl ferulate or the second cluster for butyl ferulate, respectively (Figure S1). However, taking into consideration the facts that this conformation does not appear as consistently as the previously described one and, when it does, it represents the second or third clusters emerging from the simulation, which often have a considerably lower binding affinity to the active site of the protease, it can be suggested that the most likely position for the phenolic ring is the one in front of the catalytic cysteine. In this spatial arrangement of the molecule, the hydroxyl substitution, which readily contributes to the formation of hydrogen bonds, appears to play an important role in the stabilization of the molecule in a position where access to one of the two catalytic residues is restricted, therefore potentially resulting in more effective inhibition of the catalytic activity of the enzyme.
Regarding comparable results in the literature, the binding of methyl ferulate to Mpro (PDB ID:6W63) was simulated with AutoDock 4.2, which calculated a binding energy of −5.19 kcal/mol, and the formation of hydrogen bonds with the residues Thr190 and Gln192 [100]. The binding energy is comparable to the one calculated in this work (−5.73 kcal/mol), but the hydrogen bond interactions are located in different parts of the active site, as the present study indicated interactions around the catalytic dyad with residues Gly143, Ser144, and Cys145 for the first cluster.

3.4.2. Fatty Esters of FA and Other Related Esters

This category of compounds includes oleyl, glyceryl, diglyceryl, tocopheryl, and β-sitosteryl ferulates. The binding energies calculated via the docking simulation range from −5.17 to −7.81 kcal/mol. The lowest binding affinity was predicted for oleyl ferulate, which has an 18-carbon aliphatic chain attached to the FA moiety and exhibits similar results to these of octadecyl ferulate, which possesses an equally long carbon chain, both in terms of the binding energy (−5.17 and −5.20 kcal/mol, respectively) and the conformation of the first cluster. Glyceryl, diglyceryl, and tocopheryl ferulates do not fall below −7.0 kcal/mol, thus exhibiting a lower binding affinity compared to the inhibitors N3 (−8.26 kcal/mol) and GC376 (−7.80 kcal/mol), which is, however, not high enough to exclude the possibility of effective inhibition. The most promising results were yielded for β-sitosteryl ferulate in the first cluster, where the triterpene moiety of the molecule occupies the S4 and S1 subsites while the FA moiety extends to the S1′ subsite and out of the active site cavity. This ligand emerges as particularly promising, as its binding energy (−7.81 kcal/mol) is very close to that of GC376 (−7.80 kcal/mol). β-Sitosteryl ferulate also forms three hydrogen bonds, with residues Thr25, Gly143, and the catalytic His41. Another molecular docking study also using Autodock Vina and PDB structure 6LU7 reported an identical binding energy of −7.8 kcal/mol for β-sitosteryl ferulate; however, in this case, the compound appears to bind to Mpro at a different site [101].
Regarding other patterns observed in the binding mode to the active site, it is observed that the smaller ligands in this category (glyceryl and diglyceryl ferulate) bind to the active in a similar manner as the FA derivatives described in the previous paragraph, where the FA phenolic moiety is situated in front of Cys145 at the S1 subsite, and its hydroxyl group participates in hydrogen bonding with the neighboring residues while the tail of the molecule is orientated towards the S4/S2 subsites. This orientation is seen in the first cluster of both glyceryl and diglyceryl ferulate. As far as the molecules with the larger carbon chains are concerned (oley and tocopheryl ferulates), no binding patterns were observed while the orientation of tocopheryl ferulate in all three clusters appeared to be more efficiently blocking access to the catalytic dyad compared to oleyl ferulate, where the FA moiety mostly occupied the active site. The structures of the ligands and the conformations described above are shown in Figure 5 for cluster 1 and Figure S2 for clusters 2 and 3.

3.4.3. Sugar Esters of FA

Several FA sugar derivatives were included in this study, as they are an interesting case of enzymatically synthesized derivatives with improved water solubility. The derivatives studied in this work include d-glucose, d-fructose, d-galactose, d-mannose, l-arabinose, d-xylose, d-lactose, d-sucrose, d-maltose, d-cellobiose, Galactobiose, Xylobiose, Raffinose, Arabinobiose, fructooligosaccharide (FOS 1, 2, and 3), d-mannitol, d-sorbitol, and d-xylitol ferulates.
The range of binding energies calculated for the monosaccharide-based esters is quite narrow, ranging from −7.06 kcal/mol for d-fructose ferulate, which is the only FA derivative with a keto-hexose tested, to −7.37 kcal/mol for d-xylose ferulate. In comparison to the inhibitors N3 and GC376, these results are not exceptional, but they are comparable. Therefore, also taking into consideration the improved solubility of these compounds, these sugar esters of FA are worthy of further investigation. As far as the first clusters are concerned, two orientations are more often seen. One is characterized by the positioning of the penta- or hexacyclic ring in the S1 subsite, close to the catalytic Cys145, and the extension of the rest of the molecule horizontally, towards the S4 (or sometimes S2) subsite, as in the case of the first cluster for d-galactose ferulate (Figure 6). With the exception of d-glucose and d-mannose ferulates, where the FA moiety occupies the S2 subsite, and l-arabinose ferulate, for which this conformation is not observed at all in the first three clusters, the interactions observed are almost identical and include hydrogen bonds with residues Leu141, Asn142, Gly143, Ser144, Cys145, His163, Arg188, Thr190, and Gln192. The second most prevalent orientation resembles the dominant orientation described in the previous paragraphs, and is opposite to the aforementioned one, with the phenolic ring of FA being stabilized in front of Cys145 at the S1 subsite and the sugar substitution blocking the S4 subsite. The residues more often involved in hydrogen bonding in this case are quite similar, including Leu141, Gly143, Ser144, Glu166, Thr190, and Gln192. Overall, based on the frequency of occurrence of the presented conformations in the first three clusters resulting from the simulation (Figure 6 and Figure S3), the first binding mode described is the prevailing one. This supposition is supported by the fact that in the cases where this geometry appears in the second cluster instead of the first, the resulting binding energies of the two clusters are almost identical. An example is d-glucose ferulate, with a binding energy of −7.09 kcal/mol for the first cluster and −7.08 kcal/mol for the second cluster.
The sugar esters of FA also include coupling with disaccharides d-lactose, d-sucrose, d-maltose, d-cellobiose, and xylobiose. Their binding energies provide a positive indication of the inhibitory potential, as they fluctuate within a small range comparable to that of the reference inhibitors, from −7.47 kcal/mol for d-maltose ferulate to −7.97 kcal/mol for xylobiose ferulate. Three binding patterns stand out from the evaluation of the first three clusters for each compound, with only one of them being present in all the cases. This involves the FA moiety blocking the S4 subsite, the monosaccharide closer to FA taking up the S1 subsite, and the second monosaccharide extending upwards at the S1′ subsite, as seen in the case of the third cluster for d-lactose ferulate or the first cluster for d-sucrose ferulate (Figure 7).
The occupation of the same regions of the active site is, however, not translated into identical hydrogen bond interactions. The most commonly occurring ones involve residues Gly143 and Thr190 in four out of five ligands and Thr26 in three out of five. Another conformation observed in four out of the five compounds (e.g., the first cluster for d-maltose ferulate) is a vertical one, with the cyclic ring of the sugar closer to FA blocking the catalytic cysteine between the S1 and S1′ subsites and the FA extending towards S1′. An evident motif is observed in the interactions between the compounds in this orientation and the protease active site, with Ser46, Gly143, Ser 144, Cys145, and Glu166 emerging as hydrogen bond hotspots. Lastly, a conformation involving the occupation of all four subsites, S4 by the phenolic ring of FA, S2 by its carbonyl group, S1′ by the monosaccharide ring closer to FA, and S1 by the second monosaccharide, respectively, is observed in two of the five cases (in the second cluster of D-lactose ferulate and the third clusters of D-sucrose ferulate (Figure S4). This binding pattern is stabilized by hydrogen bonds of the ligands with residues Phe140, Leu141, Asn142, Gly143, His163, and Glu166.
Two other disaccharide derivatives include two FA groups, galactobiose ferulate and arabinobiose ferulate, with very promising binding energies of −8.36 and −7.88 kcal/mol, respectively, the former of which is the second highest among the enzymatic derivatives tested. When observing the first three clusters for the two ligands, only the first cluster for galactobiose ferulate had an orientation comparable to any of the binding patterns detected for the other disaccharide derivatives. This conformation involves the two monosaccharides occupying the S1 and S1′ subsites and one of the FA groups extending to the S4 subsite while the other is located above the S1′ subsite, emerging out of the active site cavity (Figure 7).
The only trisaccharide esterified with one FA moiety tested is raffinose ferulate, which exhibited the third best binding affinity (−8.34 kcal/mol) to Mpro among the enzymatic derivatives studied, which also surpasses that of the native inhibitor N3. These results were only exceeded by galactobiose ferulate and one of the structures examined for FOS ferulate (FOS ferulate 3), which will be further described below. Raffinose ferulate forms eight hydrogen bonds with residues Thr26, Ser144, Cys145, His163, Glu66, Thr190, and Gln192, most of which are between the hydroxyl groups of the sugar moieties and residues located at the S1 and S4 subsites. The binding mode of the compound as predicted by the docking simulation appears to be limiting access to Cys145. (Figure 8). Another category of compounds tested is sugar alcohol esters, and, more specifically, the FA esters with D-mannitol, D-sorbitol, and D-xylitol. These compounds had the lowest binding affinity to the active site among the sugar derivatives of FA: −6.38, −6.43, and −6.68 kcal/mol, respectively. In this category, the conformation where the FA ring is in front of Cys145, between the S1 and S1′ subsites, while the polyol is orientated towards the S4 subsite is the one that prevails in the first clusters and is stabilized in all three cases with hydrogen bonds with residues Leu141, Gly143, Ser144, Cys145, Glu166, Arg188, Thr190, and Gln192 (Figure 8).
Fructooligosaccharides (FOSs) are oligomers of fructose units linked by beta (2-1) glycosidic bonds, with the terminal unit often being a glucose. These oligosaccharides can comprise 2 to 60 monomers [102]; therefore, their exact structure is unknown. In this work, the structures of feruloylated FOS fragments proposed by Couto et al. [99] were adopted for the purposes of the simulation. It is stated in the original work that the position of the FA moieties on the oligosaccharide backbone is not confirmed, and the structures proposed are indicative. They include three forms of FOS ferulate, here referred to as FOS ferulate 1, 2, and 3. FOS ferulate 1 is a fragment that is composed of two FA moieties esterified to the O4 and O5 positions of a fructose and exhibits a binding energy of 8.175 kcal/mol, which is significantly high if compared with the rest of the monosaccharide FA esters. This value is very encouraging, being also highly comparable to the respective values for the reference inhibitors. Although FOS is not a monosaccharide, the orientation of the studied monosaccharide fragment, FOS ferulate 1, in its first cluster resembles the prevalent conformation described above for the rest of the monosaccharide-based derivatives, where the monosaccharide is located below Cys145 and the one FA group occupies the S4 subsite while the other extends upwards to block the S1′ subsite (Figure 9). Hydrogen bonds with residues Leu141, Gly143, Ser144, His163, and Thr190 are also common interactions in both cases. FOS ferulate 2 is a fragment depicting two fructose moieties linked with a (1-2) glycosidic bond, where FA is esterified to the O5 position of the first monomer. It exhibited an encouraging binding energy of −7.34 kcal/mol and 8 hydrogen bonds with residues commonly involved in ligand binding, namely Thr26, Gly143, His164, Glu166, and Thr190. As in the case of FOS ferulate 1, although FOS ferulate 2 is an arbitrarily defined fragment rather than a disaccharide, the binding mode of its first cluster can be compared to that of the disaccharides described above, as it follows the predominant pattern observed, where FA blocks the S4 subsite, the fructose linked to it occupies the S1 subsite, and the other fructose monomer extends to the S1′ subsite. FOS ferulate 3 is the largest of the molecules tested, including a fructose trimer with four FA groups esterified to the O5 position of the first monomer, the O6 of the second, and the O4 and O6 of the third, respectively. It showed the highest binding affinity (−8.52 kcal/mol), utilizing its volume to take up the entire active site of Mpro and being stabilized through 14 hydrogen bonds, with residues Ser46, Leu141, Asn142, Gly143, Ser144, Cys145, His163, His164, and Gln192.
Conformations for the second and third cluster results for the simulations regarding trisaccharide, polyol, and polysaccharide esters are presented in Figures S5 and S6.

3.4.4. FA Glycosides

FA rutinoside is a rutinase-synthesized glycoside, which exhibited an exceptionally high binding affinity and a binding energy of 8.404 kcal/mol. It forms several hydrogen bonds with residues Thr24, Thr25, Thr45, Leu141, Gly143, Ser144, Cys145, His163, Glu166, Arg188, and Gln189. The docking simulation output suggests that it binds to the active site of Mpro blocking the S1, S1′, and S4 subsites, with its glucosyl group being stabilized through multiple hydrogen bonds at the S1 subsite, the rhamnosyl group occupying the S4 subsite, and the FA moiety extending upwards into the S1′ subsite (Figure 10). The conformations of the second and third cluster are shown in Figure S7.

3.5. Docking of Chemically Synthesized FA Derivatives

The chemically synthesized FA derivatives included in this study were selected based on their promising results in published works regarding antiviral effects (Table 3). The studies from which the derivatives were selected involve the synthesis of several derivatives and their in vitro evaluation against a range of viruses. The ones selected in this work for investigation are a selection of the compounds that showed the best antiviral potential against the respective tested virus. Such derivatives fall into the categories of FA amides, FA oligomers, and FA derivatives with dithioacetal, acylhydrazone, quinazoline, and chalcone moieties.
The considerable variety in the structure of the ligands in this category is reflected in a broad range of binding energies (from −6.27 to −8.40 kcal/mol) and a wide diversity of binding modes. The binding energy and detailed interactions for each compound are presented in Table 4. A general observation about the binding of the chemically synthesized derivatives, as seen in Figure 11, is that in a considerable number of them, for cluster 1, the phenolic moiety in FA appears to be located between the S2 and S1′ subsites, more towards S2. In this position, it is in the close vicinity of the catalytic dyad, as seen, for example, in the cases of compounds 7a, 2, and g18. Particularly in the cases of compound D4, FA dimer, and trimers, the phenolic ring is stabilized closer to the S1´ subsite, blocking access to the area around the catalytic residues even more effectively. The binding modes of the second and third clusters were also taken into consideration, but no evident binding pattern was observed (Figure S8).
The compounds that stood out are two FA derivatives with a quinazoline moiety, namely e27 and e28, with binding energies of −8.11 and −7.76 kcal/mol respectively; 4n, an FA amide and myricetin derivative with a binding energy of −7.82 kcal/mol; F3, an FA-chalcone ester with a binding energy of −7.80 kcal/mol; and triferulic acid, which exhibited a binding energy of −8.32 kcal/mol. The two quinazoline derivatives form hydrogen bonds with His41 and Gln192; the myricetin derivative with residues Leu141 and Gly143; the chalcone derivative with Thr24, Thr 25, Thr45, and Ser46; and the FA trimer with Thr26, Tyr54, and Asp187. Moreover, Gln189 and Met165 often participate in hydrophobic interactions while His41 appears to be involved in pi-pi stacking. Although the interactions do not exhibit any evident pattern, the ligands in all the cases appear to be occupying the area in front of the catalytic dyad, providing a positive indication of potential inhibitory activity. Compounds e27 and e28 have a very similar structure, which results in an almost identical binding mode. It is also interesting that in the case of compound 4n, where the respective substitution is linked to FA at the same position of FA as in the case of e27, and e28 (at C4 of the phenolic ring) binds to the active site in a way that the FA moiety has the same position as in the quinazoline derivatives. Its bulkier substitution, however, allows for higher coverage of the active site and occupation of the S1 subsite as well, as opposed to only S4, S2, and S1´ in the case of e27 and e28. The FA trimer also appears to be quite flexible and bends in a way that allows blocking of all four subsites while F3 mostly occupies the S4 and S1´ subsites and is stabilized through hydrogen bonds in the upper part of the latter.

3.6. Summary of Docking Results

Taking into consideration the binding scores of the confirmed Mpro inhibitors GC376 and N3 (−7.80 and −8.26 kcal/mol, respectively), the compounds with a binding energy of −7.8 kcal/mol or lower can be highlighted as the most promising ones. These are namely FOS ferulate 3 (−8.52 kcal/mol), FA rutinoside (−8.40 kcal/mol), galactobiose ferulate (−8.36 kcal/mol), raffinose ferulate (−8.34 kcal/mol), FA trimer (−8.32 kcal/mol), FOS ferulate 1 (−8.18 kcal/mol), compound e27 (−8.11 kcal/mol), xylobiose ferulate (−7.97 kcal/mol), arabinobiose ferulate (−7.88 kcal/mol), compound 4n (−7.82 kcal/mol), β-sitosteryl ferulate (−7.81 kcal/mol), compounds F3 (−7.80 kcal/mol) and e28 (−7.76 kcal/mol), and d-sucrose ferulate (−7.77 kcal/mol), in a series of decreasing binding affinity. It is observed that among these hits, the sugar derivatives form significantly more hydrogen bonds with the active site residues, a fact that can be attributed to the presence of several hydroxyl groups. Leu141, Gly143, Ser144, Cys145, His163, and Glu166 very often appear to be involved in these interactions, which is in consonance with the literature that highlights Gly143, Ser144, Cys145, and Glu166 among the key interacting residues, as indicated by computational tools such as molecular dynamics simulations [14,112].

3.7. ADMET Properties of the Selected FA Derivatives

3.7.1. Bioavailability and Druglikeness

The estimation of the druglikeness of the compounds under investigation, performed using the SwissADME and MOLSOFT online servers, is presented in Table 5. The size, flexibility, polarity, and solubility/lipophilicity of a compound, and its ability to form hydrogen bonds, affect how easily it can permeate membranes, be absorbed, distributed, and reach and bind to biological targets. More specifically, low lipophilicity hinders the transfer of a compound through cell membranes, but high lipophilicity is usually related to low absorption and high metabolic turnover, and potential toxicity [113]. The majority of the sugar derivatives are less lipophilic than FA while the aliphatic and chemically synthesized derivatives are more lipophilic.
Water solubility is principally a desired property. Almost all the FA derivatives have acceptable solubility, but the derivatization itself causes an increase in the solubility compared to FA mostly in the case of the sugar derivatives. As expected, the alkyl and alkenyl derivatives become progressively less soluble in water as the carbon chain size of the substitution increases. Oleic acid, tocopherol, and sitosterol esters are poorly soluble in aqueous media. As far as the chemically synthesized derivatives are concerned, they display moderate to low solubility. Hydrogen bond donors and acceptors can contribute to the solubility and binding to the desired targets, but the presence of too many of such groups negatively impacts the permeability through cell membranes [114]. The derivatives tested possess an adequate amount of hydrogen bond donors and acceptors, aside from FA sugar derivatives, in which a high number of such groups is observed.
According to Daina et al. [115], the optimum lipophilicity, size, polarity, insolubility, insaturation, and flexibility for a compound to be considered as having good bioavailability correspond to logPo/w values ranging between −0.7 and +5.0, molecular weight between 150 and 500 g/mol, topological polar surface area (TPSA) between 20 and 130 Å2, logS between 0 and 6, fraction of carbons in the sp3 hybridization between 0.25 and 1, and no more than 9 rotatable bonds. Based on these rules, more than half of the compounds have favorable properties in most of the categories. However, the most positive oral bioavailability indications are for the compounds that have less good binding scores.
Another rule for easily evaluating oral bioavailability is the Lipinski’s rule of 5, according to which a compound is likely to be orally bioavailable if it has less than 5 hydrogen bond donors and less than 10 acceptors, molecular weight lower than 500 g/mol, and an octanol-water partition coefficient lower than 4.15. It is hopeful that 41 out of the 57 compounds satisfy the Lipinski´s rule of 5 regarding oral bioavailability, even though the best hits from the molecular docking simulation are not included in those. However, it is important to note that this is not a strict criterion that can certainly exclude a compound from further investigation. For example, 13.6% of the oral and 50.0% of the non-oral drugs with low solubility and permeability violate the rule [116]. The bioavailability score is another index that demonstrates a similar tendency for the compounds tested as the two previously described criteria.
Overall, the druglikeness scores calculated for the investigated compounds, based on the respective index calculated by MOLSOFT, do not exclude them from being used as drugs. The index is calculated by comparing the molecular properties of the respective compound with a library of selected drug and non-drug compounds [117] and provides an indication of how likely a compound is to function as an oral drug. Scores between 0 and 1 are stronger indicators of druglikeness, but also negative values, particularly over −1, correspond to resemblance to drug molecules. The alkyl and alkenyl derivatives had the most negative scores, the lowest being −0.76, while the FA sugar esters and chemically synthesized derivatives showed more encouraging results, with tocopheryl and sitosteryl ferulate and compound 5 yielding the best scores, with druglikeness scores of 1.14, 1.21, and 1.40, respectively. It is worth mentioning that isobutyl and isopentyl ferulates stood out among the alkyl derivatives in terms of druglikeness, the sugar polyol esters exhibited lower scores compared to the other sugar derivatives, and the chemically designed molecules had an overall higher druglikeness, something that could be expected given the fact that many of them emerged from drug design procedures.

3.7.2. Pharmacokinetics

Computational evaluation of the pharmacokinetic properties of the screened compounds using SwissADME showed high gastrointestinal absorption for the majority of the alkyl and alkenyl FA esters and chemically synthesized FA derivatives but low gastrointestinal absorption for the majority of the FA sugar esters (Table 6).

3.7.3. Toxicity Profile

Altogether, the compounds were predicted to have low toxicity, with LD50 values being greater than 5000 mg/kg for the majority of them, and only a few alkyl, alkenyl, and fatty acid derivatives having a lethal dose lower than 1000 mg/kg. The prediction accuracy estimated by ProTox-II was around 70% for the vast majority of the compounds. Almost all appeared likely to be immunotoxic, but hepatotoxicity, carcinogenicity, mutagenicity, and cytotoxicity were predicted to be of no concern (Table 6).

3.8. Summary of the ADMET Properties of the Most Promising Compounds

Regarding the compounds with the best binding scores, namely FOS ferulate, FA rutinoside, galactobiose ferulate, raffinose ferulate, FA trimer, FOS ferulate 1, compound e27, xylobiose ferulate, arabinobiose ferulate, compound 4n, β-sitosteryl ferulate, compounds F3 and e28, and d-sucrose ferulate, the pharmacokinetic properties were not optimal. Among the best hits, only e27 has molecular weight, rotatable bond, polar surface area, logP, and logS values that indicate good oral bioavailability. The chemically synthesized derivatives, excluding 4n, which showed poor ADMET properties, are the only hits for which an adequate bioavailability score, good balance between water solubility and lipophilicity, and compliance with the Lipinski´s rule were predicted. On the other hand, the enzymatically synthesized derivatives, comprising the sugar esters of FA, exhibit higher solubility and significantly lower acute toxicity, portrayed in the lethal dose values. Only compounds e27, e28, and F3 were predicted to have high gastrointestinal absorption while all of them appear to be unable to cross the BBB. Regarding selectivity, very few are assessed as cytochrome inhibitors, whereas none are a P-glycoprotein substrate.
Based on all the above, FOS ferulate 1 would appear to have the highest balance between lipophilicity and solubility among the enzymatically synthesized derivatives, and an adequate druglikeness score, slight deviation from the oral bioavailability criteria (compared to the rest of the compounds), and a relatively high lethal dose. However, due to their higher binding scores, exceptional solubility, and the fact that they can be more easily synthesized, FA rutinoside and raffinose ferulate are also highlighted as promising hits. Regarding the chemically synthesized derivatives, compound e27 appears to have an edge over the rest of the compounds in all the categories, with the only concern being its relatively low lethal dose value. In any case, despite being a useful indication, these predictions are not definite and cannot exclude the compounds from further evaluation for their potential utilization as drugs or nutraceuticals.

3.9. MD Simulation of Selected Derivatives

MD simulation was performed to more thoroughly evaluate the stability of the complexes of three ligands that emerged as particularly promising from the molecular docking simulation (raffinose ferulate, FA rutinoside, and compound e27) with SARS-CoV-2 Mpro while the complex of the protease with the native inhibitor N3 was also analyzed to provide a reference for the evaluation of the results. The complexes appeared to be particularly stable (Figure 12a), with overall low values and subtle fluctuation of the RMSD (1–2 Å) for FA rutinoside and compound e27, with the results for the latter indicating a complex that is more stable than that of N3 for the majority of the simulation time. Raffinose ferulate exhibited slightly higher RMSD values, which exceeded 2 Å after 6 ns. Although its RMSD values appear to have an ascending tendency, the simulation was run for 30 ns to confirm that equilibrium indeed occurred after 10 ns (results not shown). In any case, the RMSD values of the complex of raffinose ferulate are still below 3 Å, which is considered low enough to characterize the complex as stable [119].
The radius of gyration is a useful measure of the rigidity of the protein–ligand complexes. All the complexes appeared to have comparable results (Figure 12b), which can be regarded as indicators of compactness and stability [120,121]. Compound e27 showed the tightest complex with Mpro. FA rutinoside had very similar RoG compared to N3, and raffinose ferulate exhibited a greater variation in its RoG values, which, however, still compose a low RoG profile. Analysis of the RMSF values allows the acquisition of an image of which residues are more flexible. As seen in Figure 12c, the majority of residues have low RMSF values below 1 Å [120,122] while the terminal residues of the protease exhibit a much larger deviation and are the only ones to exceed 2.5 Å , with the exception of Arg222, Tyr154, Gln273, and Asp155 for compound e27. Therefore, the structure of the protein appears to be quite stable.
As far as protein–ligand interactions are concerned, the total hydrogen bonds formed between the two as a function of time are presented in Figure 12d and the number of total interactions between the ligands and each Mpro residue (when occurring) are shown in Figure 13. The reference inhibitor N3 appears to be more stable in terms of its hydrogen bonds with the protease, which exhibit only slight deviations from the value of 3. Forming fewer hydrogen bonds but also showing good stability, compound e27 appears to have one hydrogen bond contact with the protease for most of the simulation time. The enzymatic FA derivatives FA rutinoside and raffinose ferulate form significantly more hydrogen bonds. In the case of FA rutinoside, the bonds appear to be more stable and fluctuate around the value of 6 while raffinose ferulate exhibits a more sudden drop after 2 ns of simulation to then be quite stable between 2 and 4 hydrogen bonds. Based on the number and stability of the hydrogen bonds, FA rutinoside shows indications of stronger binding to the protease.
Regarding the overall interactions, as seen in Figure 13a, FA rutinoside interacts with numerous residues around the active site, including multiple contacts with oxyanion hole residues Gly143, Ser144, and Cys145. Moreover, Val42 appears to interact with the ligand. Although it is not calculated as a contacting residue in the molecular docking simulation, it is a neighboring residue to the catalytic histidine (His41). Considering that the contacts are calculated based on the distance of the residue atoms from the atoms of the ligands, it can be difficult to identify and separate individual contacts of the ligand with neighboring residues. However, results show the vicinity of the ligand to this area of the active site. FA rutinoside also interacts with residues located at the S1′ (Thr25, Thr26, and Asn28, which neighbor Leu 27, which is given as a contacting residue), S2 (Ser46, Glu47, Leu50), and S4 subsites (His164, Met165, Leu167). Raffinose ferulate also interacts with oxyanion hole residues, including the catalytic cysteine, and the residues Glu47 and Asp48, which are located near the contacting residues Thr45 and Met49 at the S2 subsite (Figure 13b). Moreover, it interacts with several residues of the S4 subsite, with the ones appearing to form the most interactions being Thr190 and Ala193 (neighbor of the contacting residue Gln192), followed by Glu166. Lastly, compound e27 interacts with similar residues covering all four subsites (Figure 13c). Although it does not appear to interact with any of the catalytic residues, it forms contacts with their neighboring residues Val42, Ser144, and Gly146. Another observation is that the majority of the contacts are with residues located at the S4 subsite (Glu166, Leu167, Thr169, Arg188, Thr190, Gln192, Ala193).
Overall, compared to inhibitor N3 (Figure 13d), it is evident that the ligands examined form more contacts with the protease residues. The fact that the confirmed protease inhibitor N3 exhibits fluctuations in the number of contacts with the various contacting residues over time indicates that such fluctuations are normal. Although the contacts established between N3 and the protease appear to be more stable compared to the rest of the ligands, compound e27 also exhibits a consistent interaction profile. The contacts of FA rutinoside are also quite stable and higher in number overall. Raffinose ferulate, confirming the tendency shown from the RMSD and H-bond diagrams, appears to have the least balance in terms of interactions among the three candidates. In general, the MD simulation leads to the conclusion that the complex of compound e27 is the most stable among the three ligands investigated. However, the results yielded for FA rutinoside are also very encouraging and do not deviate much. This fact, combined with the greater number of interactions with the protease, establishes FA rutinoside as a very interesting candidate for the inhibition of Mpro.

4. Conclusions

The difficulties that have arisen in combating the ongoing pandemic highlight the importance of utilizing any available tool for immunity boosting. In this context, phytochemicals emerge as valuable allies. FA is an abundant bioactive compound, whose derivatization has been investigated both through chemical and enzymatic routes, in search for improved properties. This work studied 54 FA derivatives, among which 14 exhibited similar or better in silico binding affinity to the SARS-CoV-2 Mpro compared to confirmed inhibitors. Further computational evaluation of their ADMET properties indicated FA rutinoside, raffinose ferulate, and compound e27 as the most promising hits to be further examined through molecular dynamics simulation. Analysis of the MD trajectories identified FA rutinoside and compound e27 as promising candidates, representing enzymatically and chemically synthesized derivatives, respectively. Several derivatives exhibited good binding affinity to the main protease though, indicating that it would be worth investigating different administration routes in order to overcome the problem of low oral bioavailability, and potential structural modifications that could lead to even higher affinity and more favorable pharmacokinetic properties. Moreover, the favorable results for many enzymatically synthesized derivatives encourage the development and optimization of more sustainable enzymatic processes, which can also include the valorization of biomass in which FA can be found. Even though further in vitro and in vivo research is necessary to provide more reliable data regarding the efficacy of the compounds, this work suggests that FA derivatives are interesting candidates to be considered for their antiviral potential against the current public health concern SARS-CoV-2 and future viral threats.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines10081787/s1, Figure S1: Binding modes of the second and third clusters for alkyl and alkenyl FA esters at the active site of Mpro; Figure S2: Binding modes of the second and third clusters for fatty acid, polyol, tocopherol, and sterol FA derivatives at the active site of Mpro; Figure S3: Binding modes of the second and third clusters for monosaccharide esters of FA at the active site of Mpro; Figure S4: Binding modes of the second and third clusters for disaccharide esters of FA at the active site of Mpro; Figure S5: Binding modes of the second and third clusters for trisaccharide and sugar alcohol esters of FA at the active site of Mpro; Figure S6: Binding modes of the second and third clusters for FOS esters of FA at the active site of Mpro; Figure S7: Binding modes of the second and third clusters for FA rutinoside at the active site of Mpro; Figure S8: Binding modes of the second and third clusters for chemically synthesized FA derivatives at the active site of Mpro.

Author Contributions

Conceptualization, I.A. and E.S.; Methodology, I.A., E.S., U.R. and P.C.; Validation, I.A. and E.S.; Formal Analysis, E.S.; Investigation, I.A. and E.S.; Resources, I.A., U.R. and P.C.; Data Curation, E.S.; Writing—Original Draft Preparation, I.A. and E.S.; Writing—and & Editing, I.A., E.S., U.R. and P.C.; Supervision, I.A., U.R. and P.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article and Supplemental Materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int/ (accessed on 10 March 2022).
  2. Stoddard, S.V.; Stoddard, S.D.; Oelkers, B.K.; Fitts, K.; Whalum, K.; Whalum, K.; Hemphill, A.D.; Manikonda, J.; Martinez, L.M.; Riley, E.G.; et al. Optimization Rules for SARS-CoV-2 Mpro Antivirals: Ensemble Docking and Exploration of the Coronavirus Protease Active Site. Viruses 2020, 12, 942. [Google Scholar] [CrossRef]
  3. Tregoning, J.S.; Flight, K.E.; Higham, S.L.; Wang, Z.; Pierce, B.F. Progress of the COVID-19 Vaccine Effort: Viruses, Vaccines and Variants versus Efficacy, Effectiveness and Escape. Nat. Rev. Immunol. 2021, 21, 626–636. [Google Scholar] [CrossRef]
  4. Rubin, D.; Chan-Tack, K.; Farley, J.; Sherwat, A. FDA Approval of Remdesivir—A Step in the Right Direction. N. Engl. J. Med. 2020, 383, 2598–2600. [Google Scholar] [CrossRef]
  5. U.S. Food and Drug Administration. Emergency Use Authorization. Available online: https://www.fda.gov/emergency-preparedness-and-response/mcm-legal-regulatory-and-policy-framework/emergency-use-authorization#coviddrugs (accessed on 12 March 2022).
  6. European Centre for Disease Prevention and Control. SARS-CoV-2 Variants of Concern as of 11 March 2022. Available online: https://www.ecdc.europa.eu/en/COVID-19/variants-concern (accessed on 14 March 2022).
  7. Mohapatra, R.K.; Tiwari, R.; Sarangi, A.K.; Islam, R.; Chakraborty, C.; Dhama, K. Omicron (B.1.1.529) Variant of SARS-CoV-2: Concerns, Challenges, and Recent Updates. J. Med. Virol. 2022, 94, 2336–2342. [Google Scholar] [CrossRef]
  8. Sun, C.; Kang, Y.-F.; Liu, Y.-T.; Kong, X.-W.; Xu, H.-Q.; Xiong, D.; Xie, C.; Liu, Y.-H.; Peng, S.; Feng, G.-K.; et al. Parallel Profiling of Antigenicity Alteration and Immune Escape of SARS-CoV-2 Omicron and Other Variants. Signal Transduct. Target. Ther. 2022, 7, 42. [Google Scholar] [CrossRef]
  9. Świderek, K.; Moliner, V. Revealing the Molecular Mechanisms of Proteolysis of SARS-CoV-2 Mpro by QM/MM Computational Methods. Chem. Sci. 2020, 11, 10626–10630. [Google Scholar] [CrossRef]
  10. Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal Structure of SARS-CoV-2 Main Protease Provides a Basis for Design of Improved a-Ketoamide Inhibitors. Science 2020, 368, 409–412. [Google Scholar] [CrossRef] [Green Version]
  11. Parvez, S.A.; Saha, M.K.; Araf, Y.; Islam, T.; Ohtsuki, G.; Hosen, M.J. Insights from a Computational Analysis of the SARS-CoV-2 Omicron Variant: Host-Pathogen Interaction, Pathogenicity, and Possible Therapeutics. Immun. Inflamm. Dis. 2022, 10, e639. [Google Scholar] [CrossRef]
  12. Nikolaidis, M.; Papakyriakou, A.; Chlichlia, K.; Markoulatos, P.; Oliver, S.G.; Amoutzias, G.D. Comparative Analysis of SARS-CoV-2 Variants of Concern, Including Omicron, Highlights Their Common and Distinctive Amino Acid Substitution Patterns, Especially at the Spike ORF. Viruses 2022, 14, 707. [Google Scholar] [CrossRef]
  13. Ullrich, S.; Ekanayake, K.B.; Otting, G.; Nitsche, C. Main Protease Mutants of SARS-CoV-2 Variants Remain Susceptible to Nirmatrelvir. Bioorganic Med. Chem. Lett. 2022, 62, 128629. [Google Scholar] [CrossRef]
  14. Yoshino, R.; Yasuo, N.; Sekijima, M. Identification of Key Interactions between SARS-CoV-2 Main Protease and Inhibitor Drug Candidates. Sci. Rep. 2020, 10, 12493. [Google Scholar] [CrossRef]
  15. Macdonald, E.A.; Frey, G.; Namchuk, M.N.; Harrison, S.C.; Hinshaw, S.M.; Windsor, I.W. Recognition of Divergent Viral Substrates by the SARS-CoV-2 Main Protease. Infect. Dis. 2021, 7, 2591–2595. [Google Scholar] [CrossRef]
  16. Lin, L.-T.; Hsu, W.-C.; Lin, C.-C. Antiviral Natural Products and Herbal Medicines. J. Tradit. Complement. Med. 2014, 4, 24–35. [Google Scholar] [CrossRef] [Green Version]
  17. Omrani, M.; Keshavarz, M.; Nejad Ebrahimi, S.; Mehrabi, M.; McGaw, L.J.; Ali Abdalla, M.; Mehrbod, P. Potential Natural Products against Respiratory Viruses: A Perspective to Develop Anti-COVID-19 Medicines. Front. Pharmacol. 2021, 11, 586993. [Google Scholar] [CrossRef]
  18. Antonopoulou, I.; Sapountzaki, E.; Rova, U.; Christakopoulos, P. Inhibition of the Main Protease of SARS-CoV-2 (Mpro) by Repurposing/Designing Drug-like Substances and Utilizing Nature’s Toolbox of Bioactive Compounds. Comput. Struct. Biotechnol. J. 2022, 20, 1306–1344. [Google Scholar] [CrossRef]
  19. Katsimpouras, C.; Antonopoulou, I.; Christakopoulos, P.; Topakas, E. Role and Applications of Feruloyl Esterases in Biomass Bioconversion. In Microbial Enzymes in Bioconversions of Biomass; Gupta, V.K., Ed.; Springer International Publishing: Cham, Switzerland, 2016; pp. 79–123. ISBN 978-3-319-43679-1. [Google Scholar]
  20. Stompor-Gorący, M.; Machaczka, M. Recent Advances in Biological Activity, New Formulations and Prodrugs of Ferulic Acid. Int. J. Mol. Sci. 2021, 22, 12889. [Google Scholar] [CrossRef]
  21. Srinivasan, M.; Sudheer, A.R.; Menon, V.P. Recent Advances in Indian Herbal Drug Research Guest Editor: Thomas Paul Asir Devasagayam Ferulic Acid: Therapeutic Potential Through Its Antioxidant Property. J. Clin. Biochem. Nutr. 2007, 40, 92–100. [Google Scholar] [CrossRef] [Green Version]
  22. Antonopoulou, I.; Sapountzaki, E.; Rova, U.; Christakopoulos, P. Ferulic Acid from Plant Biomass: A Phytochemical with Promising Antiviral Properties. Front. Nutr. 2022, 8, 777576. [Google Scholar] [CrossRef]
  23. Salman, S.; Shah, F.H.; Idrees, J.; Idrees, F.; Velagala, S.; Ali, J.; Khan, A.A. Virtual Screening of Immunomodulatory Medicinal Compounds as Promising Anti-SARS-CoV-2 Inhibitors. Future Virol. 2020, 15, 267–275. [Google Scholar] [CrossRef]
  24. Mancuso, C.; Santangelo, R. Ferulic Acid: Pharmacological and Toxicological Aspects. Food Chem. Toxicol. 2014, 65, 185–195. [Google Scholar] [CrossRef]
  25. Bourne, L.C.; Rice-Evans, C. Bioavailability of Ferulic Acid. Biochem. Biophys. Res. Commun. 1998, 253, 222–227. [Google Scholar] [CrossRef]
  26. Zhao, Z.; Moghadasian, M.H. Chemistry, Natural Sources, Dietary Intake and Pharmacokinetic Properties of Ferulic Acid: A Review. Food Chem. 2008, 109, 691–702. [Google Scholar] [CrossRef]
  27. Li, D.; Rui, Y.; Guo, S.; Luan, F.; Liu, R.; Zeng, N. Ferulic Acid: A Review of Its Pharmacology, Pharmacokinetics and Derivatives. Life Sci. 2021, 284, 119921. [Google Scholar] [CrossRef]
  28. Kumar, N.; Pruthi, V. Potential Applications of Ferulic Acid from Natural Sources. Biotechnol. Rep. 2014, 4, 86–93. [Google Scholar] [CrossRef] [Green Version]
  29. de Oliveira Silva, E.; Batista, R. Ferulic Acid and Naturally Occurring Compounds Bearing a Feruloyl Moiety: A Review on Their Structures, Occurrence, and Potential Health Benefits. Compr. Rev. Food Sci. Food Saf. 2017, 16, 580–616. [Google Scholar] [CrossRef] [Green Version]
  30. Mathew, S.; Abraham, T. Ferulic Acid: An Antioxidant Found Naturally in Plant Cell Walls and Feruloyl Esterases Involved in Its Release and Their Applications. Crit. Rev. Biotechnol. 2004, 24, 59–83. [Google Scholar] [CrossRef]
  31. Meryl Smith, M.; Hartley, R. Occurence and Nature of Ferulic Acid Substitution of Cell-Wall Polysacchariddes in Graminaceous Plants. Carbohydr. Res. 1983, 118, 65–80. [Google Scholar] [CrossRef]
  32. Huang, G.Y.; Cui, C.; Wang, Z.P.; Li, Y.Q.; Xiong, L.X.; Wang, L.Z.; Yu, S.J.; Li, Z.M.; Zhao, W.G. Synthesis and Characteristics of (Hydrogenated) Ferulic Acid Derivatives as Potential Antiviral Agents with Insecticidal Activity. Chem. Cent. J. 2013, 7, 33. [Google Scholar] [CrossRef] [Green Version]
  33. Ren, X.; Li, X.; Yin, L.; Jiang, D.; Hu, D. Design, Synthesis, Antiviral Bioactivity, and Mechanism of the Ferulic Acid Ester-Containing Sulfonamide Moiety. ACS Omega 2020, 5, 19721–19726. [Google Scholar] [CrossRef]
  34. Wang, Y.; He, F.; Wu, S.; Luo, Y.; Wu, R.; Hu, D.; Song, B. Design, Synthesis, Anti-TMV Activity, and Preliminary Mechanism of Cinnamic Acid Derivatives Containing Dithioacetal Moiety. Pestic. Biochem. Physiol. 2020, 164, 115–121. [Google Scholar] [CrossRef]
  35. Katayama, S.; Ohno, F.; Yamauchi, Y.; Kato, M.; Makabe, H.; Nakamura, S. Enzymatic Synthesis of Novel Phenol Acid Rutinosides Using Rutinase and Their Antiviral Activity in Vitro. J. Agric. Food Chem. 2013, 61, 9617–9622. [Google Scholar] [CrossRef]
  36. Antonopoulou, I.; Varriale, S.; Topakas, E.; Rova, U.; Christakopoulos, P.; Faraco, V. Enzymatic Synthesis of Bioactive Compounds with High Potential for Cosmeceutical Application. Appl. Microbiol. Biotechnol. 2016, 100, 6519–6543. [Google Scholar] [CrossRef] [Green Version]
  37. Krieger, E.; Vriend, G. YASARA View—Molecular Graphics for All Devices—From Smartphones to Workstations. Bioinformatics 2014, 30, 2981–2982. [Google Scholar] [CrossRef] [Green Version]
  38. Trott, O.; Olson, A.J. AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [Green Version]
  39. Schrödinger, L. The PyMOL Molecular Graphics System. Version 2.4.1. Schrödinger: New York, NY, USA, 2020. [Google Scholar]
  40. Wallace, A.C.; Laskowski, R.A.; Thornton, J.M. LIGPLOT: A Program to Generate Schematic Diagrams of Protein-Ligand Interactions. Protein Eng. Des. Sel. 1995, 8, 127–134. [Google Scholar] [CrossRef]
  41. Krieger, E.; Nielsen, J.E.; Spronk, C.A.E.M.; Vriend, G. Fast Empirical PKa Prediction by Ewald Summation. J. Mol. Graph. Model. 2006, 25, 481–486. [Google Scholar] [CrossRef]
  42. Krieger, E.; Dunbrack, R.L.J.; Hooft, R.W.W.; Krieger, B. Assignment of Protonation States in Proteins and Ligands: Combining PKa Prediction with Hydrogen Bonding Network Optimization. Methods Mol Biol 2012, 819, 405–421. [Google Scholar] [CrossRef]
  43. Maier, J.A.; Martinez, C.; Kasavajhala, K.; Wickstrom, L.; Hauser, K.E.; Simmerling, C. Ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from Ff99SB. J. Chem. Theory Comput. 2015, 11, 3696–3713. [Google Scholar] [CrossRef] [Green Version]
  44. Hornak, V.; Abel, R.; Okur, A.; Strockbine, B.; Roitberg, A.; Simmerling, C. Comparison of Multiple Amber Force Fields and Development of Improved Protein Backbone Parameters. Proteins 2006, 65, 712–725. [Google Scholar] [CrossRef] [Green Version]
  45. Essmann, U.; Perera, L.; Berkowitz, M.L.; Darden, T.; Lee, H.; Pedersen, L.G. A Smooth Particle Mesh Ewald Method. J. Chem. Phys. 1995, 103, 8577–8593. [Google Scholar] [CrossRef] [Green Version]
  46. Krieger, E.; Vriend, G. New Ways to Boost Molecular Dynamics Simulations. J. Comput. Chem. 2015, 36, 996–1007. [Google Scholar] [CrossRef] [PubMed]
  47. Koudelka, T.; Boger, J.; Henkel, A.; Schönherr, R.; Krantz, S.; Fuchs, S.; Rodríguez, E.; Redecke, L.; Tholey, A. N-Terminomics for the Identification of In Vitro Substrates and Cleavage Site Specificity of the SARS-CoV-2 Main Protease. Proteomics 2021, 21, 2000246. [Google Scholar] [CrossRef] [PubMed]
  48. Hegyi, A.; Ziebuhr, J. Conservation of Substrate Specificities among Coronavirus Main Proteases. J. Gen. Virol. 2002, 83, 595–599. [Google Scholar] [CrossRef] [PubMed]
  49. Lee, J.; Worrall, L.J.; Vuckovic, M.; Rosell, F.I.; Gentile, F.; Ton, A.-T.; Caveney, N.A.; Ban, F.; Cherkasov, A.; Paetzel, M.; et al. Crystallographic Structure of Wild-Type SARS-CoV-2 Main Protease Acyl-Enzyme Intermediate with Physiological C-Terminal Autoprocessing Site. Nat. Commun. 2020, 11, 5877. [Google Scholar] [CrossRef]
  50. Kneller, D.W.; Phillips, G.; O’Neill, H.M.; Jedrzejczak, R.; Stols, L.; Langan, P.; Joachimiak, A.; Coates, L.; Kovalevsky, A. Structural Plasticity of SARS-CoV-2 3CL Mpro Active Site Cavity Revealed by Room Temperature X-Ray Crystallography. Nat. Commun. 2020, 11, 7–12. [Google Scholar] [CrossRef] [PubMed]
  51. Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; et al. Structure of M pro from SARS-CoV-2 and Discovery of Its Inhibitors. Nature 2020, 582, 289–293. [Google Scholar] [CrossRef] [Green Version]
  52. Sacco, M.D.; Ma, C.; Lagarias, P.; Gao, A.; Townsend, J.A.; Meng, X.; Dube, P.; Zhang, X.; Hu, Y.; Kitamura, N.; et al. Structure and Inhibition of the SARS-CoV-2 Main Protease Reveals Strategy for Developing Dual Inhibitors against Mpro and Cathepsin L. bioRxiv 2020. [Google Scholar] [CrossRef]
  53. Kneller, D.W.; Galanie, S.; Phillips, G.; Neill, H.M.O.; Coates, L.; Kovalevsky, A.; Kneller, D.W.; Galanie, S.; Phillips, G.; Neill, H.M.O.; et al. Malleability of the SARS-CoV-2 3CL Mpro Active-Site Cavity Facilitates Binding of Clinical Antivirals. Struct./Fold. Des. 2020, 28, 1313–1320.e3. [Google Scholar] [CrossRef]
  54. Bharadwaj, S.; Azhar, E.I.; Kamal, M.A.; Bajrai, H.; Dubey, A.; Jha, K.; Yadava, U.; Kang, S.G. SARS-CoV-2 M Inhibitors : Identification of Anti- SARS-CoV-2 M Compounds from FDA Approved Drugs. J. Biomol. Struct. Dyn. 2022, 40, 2769–2784. [Google Scholar] [CrossRef]
  55. Arafet, K.; Serrano-Aparicio, N.; Lodola, A.; Mulholland, A.J.; González, F.V.; Świderek, K.; Moliner, V. Mechanism of Inhibition of SARS-CoV-2 Mpro by N3 peptidyl Michael Acceptor Explained by QM/MM Simulations and Design of New Derivatives with Tunable Chemical Reactivity. Chem. Sci. 2021, 12, 1433–1444. [Google Scholar] [CrossRef]
  56. Das, P.; Majumder, R.; Mandal, M.; Basak, P. In-Silico Approach for Identification of Effective and Stable Inhibitors for COVID-19 Main Protease (Mpro) from Flavonoid Based Phytochemical Constituents of Calendula Officinalis. J. Biomol. Struct. Dyn. 2021, 39, 6265–6280. [Google Scholar] [CrossRef] [PubMed]
  57. Ahmed, S.; Id, C.; Merzouk, H.; Mokhtari-Soulimane, N. Potential Bioactive Glycosylated Flavonoids as SARS-CoV-2 Main Protease Inhibitors: A Molecular Docking and Simulation Studies. PLoS ONE 2020, 15, e0240653. [Google Scholar] [CrossRef]
  58. Hu, Y.; Ma, C.; Szeto, T.; Hurst, B.; Tarbet, B.; Wang, J. Boceprevir, Calpain Inhibitors II and XII, and GC-376 Have Broad-Spectrum Antiviral Activity against Coronaviruses. ACS Infect. Dis. 2021, 7, 586–597. [Google Scholar] [CrossRef] [PubMed]
  59. Rakib, A.; Nain, Z.; Sami, S.A.; Mahmud, S.; Islam, A.; Ahmed, S.; Siddiqui, A.B.F.; Babu, S.M.O.F.; Hossain, P.; Shahriar, A.; et al. A Molecular Modelling Approach for Identifying Antiviral Selenium-Containing Heterocyclic Compounds That Inhibit the Main Protease of SARS-CoV-2: An in Silico Investigation. Brief. Bioinform. 2021, 22, 1476–1498. [Google Scholar] [CrossRef]
  60. Allam, A.E.; Amen, Y.; Ashour, A.; Assaf, H.K.; Hassan, H.A.; Abdel-Rahman, I.M.; Sayed, A.M.; Shimizu, K. In Silico Study of Natural Compounds from Sesame against COVID-19 by Targeting Mpro, PLpro and RdRp. RSC Adv. 2021, 11, 22398–22408. [Google Scholar] [CrossRef]
  61. El Gizawy, H.A.; Boshra, S.A.; Mostafa, A.; Mahmoud, S.H.; Ismail, M.I.; Alsfouk, A.A.; Taher, A.T.; Al-Karmalawy, A.A. Pimenta dioica (L.) Merr. Bioactive Constituents Exert Anti-SARS-CoV-2 and Anti-Inflammatory Activities: Molecular Docking and Dynamics, In Vitro, and In Vivo Studies. Molecules 2021, 26, 5844. [Google Scholar] [CrossRef]
  62. Saeed, A.; Ahmad, B.; Majaz, S.; Nouroz, F.; Ahmad, A.; Xie, Y. Targeting Omicron and Other Reported SARS-CoV-2 Lineages by Potent Inhibitors of Main Protease 3CL Mpro: Molecular Simulation Analysis. J. Infect. 2022, 84, e133–e136. [Google Scholar] [CrossRef]
  63. Joshi, T.; Bhat, S.; Pundir, H.; Chandra, S. Identification of Berbamine, Oxyacanthine and Rutin from Berberis Asiatica as Anti-SARS-CoV-2 Compounds: An in Silico Study. J. Mol. Graph. Model. 2021, 109, 108028. [Google Scholar] [CrossRef]
  64. Oliveira, D.M.; Mota, T.R.; Oliva, B.; Segato, F.; Marchiosi, R.; Ferrarese-Filho, O.; Faulds, C.B.; dos Santos, W.D. Feruloyl Esterases: Biocatalysts to Overcome Biomass Recalcitrance and for the Production of Bioactive Compounds. Bioresour. Technol. 2019, 278, 408–423. [Google Scholar] [CrossRef] [Green Version]
  65. Grajales-Hernández, D.A.; Armendáriz-Ruiz, M.A.; Gallego, F.L.; Mateos-Díaz, J.C. Approaches for the Enzymatic Synthesis of Alkyl Hydroxycinnamates and Applications Thereof. Appl. Microbiol. Biotechnol. 2021, 105, 3901–3917. [Google Scholar] [CrossRef]
  66. Cerullo, G.; Varriale, S.; Bozonnet, S.; Antonopoulou, I.; Christakopoulos, P.; Rova, U.; Gherbovet, O.; Fauré, R.; Piechot, A.; Jütten, P.; et al. Directed Evolution of the Type C Feruloyl Esterase from Fusarium Oxysporum FoFaeC and Molecular Docking Analysis of Its Improved Variants. New Biotechnol. 2019, 51, 14–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Karnaouri, A.; Antonopoulou, I.; Zerva, A.; Dimarogona, M.; Topakas, E.; Rova, U.; Christakopoulos, P. Thermophilic Enzyme Systems for Efficient Conversion of Lignocellulose to Valuable Products: Structural Insights and Future Perspectives for Esterases and Oxidative Catalysts. Bioresour. Technol. 2019, 279, 362–372. [Google Scholar] [CrossRef] [PubMed]
  68. Hunt, C.J.; Antonopoulou, I.; Tanksale, A.; Rova, U.; Christakopoulos, P.; Haritos, V.S. Insights into Substrate Binding of Ferulic Acid Esterases by Arabinose and Methyl Hydroxycinnamate Esters and Molecular Docking. Sci. Rep. 2017, 7, 17315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Antonopoulou, I.; Hunt, C.; Cerullo, G.; Varriale, S.; Gerogianni, A.; Faraco, V.; Rova, U.; Christakopoulos, P. Tailoring the Specificity of the Type C Feruloyl Esterase FoFaeC from Fusarium Oxysporum towards Methyl Sinapate by Rational Redesign Based on Small Molecule Docking Simulations. PLoS ONE 2018, 13, e0198127. [Google Scholar] [CrossRef] [Green Version]
  70. Varriale, S.; Cerullo, G.; Antonopoulou, I.; Christakopoulos, P.; Rova, U.; Tron, T.; Fauré, R.; Jütten, P.; Piechot, A.; Brás, J.L.A.; et al. Evolution of the Feruloyl Esterase MtFae1a from Myceliophthora Thermophila towards Improved Catalysts for Antioxidants Synthesis. Appl. Microbiol. Biotechnol. 2018, 102, 5185–5196. [Google Scholar] [CrossRef]
  71. Grajales-Hernández, D.; Armendáriz-Ruiz, M.; Velasco-Lozano, S.; López-Gallego, F.; Mateos-Díaz, J.C. Chitosan-Based CLEAs from Aspergillus Niger Type A Feruloyl Esterase: High-Productivity Biocatalyst for Alkyl Ferulate Synthesis. Appl. Microbiol. Biotechnol. 2020, 104, 10033–10045. [Google Scholar] [CrossRef]
  72. Katsoura, M.H.; Polydera, A.C.; Tsironis, L.D.; Petraki, M.P.; Rajačić, S.K.; Tselepis, A.D.; Stamatis, H. Efficient Enzymatic Preparation of Hydroxycinnamates in Ionic Liquids Enhances Their Antioxidant Effect on Lipoproteins Oxidative Modification. New Biotechnol. 2009, 26, 83–91. [Google Scholar] [CrossRef]
  73. Schär, A.; Nyström, L. Enzymatic Synthesis of Steryl Ferulates. Eur. J. Lipid Sci. Technol. 2016, 118, 1557–1565. [Google Scholar] [CrossRef]
  74. Topakas, E.; Stamatis, H.; Biely, P.; Kekos, D.; Macris, B.J.; Christakopoulos, P. Purification and Characterization of a Feruloyl Esterase from Fusarium Oxysporum Catalyzing Esterification of Phenolic Acids in Ternary Water–Organic Solvent Mixtures. J. Biotechnol. 2003, 102, 33–44. [Google Scholar] [CrossRef]
  75. Guyot, B.; Bosquette, B.; Pina, M.; Graille, J. Esterification of Phenolic Acids from Green Coffee with an Immobilized Lipase from Candida antarctica in Solvent-Free Medium. Biotechnol. Lett. 1997, 19, 529–532. [Google Scholar] [CrossRef]
  76. Romero-Borbón, E.; Grajales-Hernández, D.; Armendáriz-Ruiz, M.; Ramírez-Velasco, L.; Rodríguez-González, J.A.; Cira-Chávez, L.A.; Estrada-Alvarado, M.I.; Mateos-Díaz, J.C. Type C Feruloyl Esterase from Aspergillus Ochraceus: A Butanol Specific Biocatalyst for the Synthesis of Hydroxycinnamates in a Ternary Solvent System. Electron. J. Biotechnol. 2018, 35, 1–9. [Google Scholar] [CrossRef]
  77. Hüttner, S.; Zezzi Do Valle Gomes, M.; Iancu, L.; Palmqvist, A.; Olsson, L. Immobilisation on Mesoporous Silica and Solvent Rinsing Improve the Transesterification Abilities of Feruloyl Esterases from Myceliophthora Thermophila. Bioresour. Technol. 2017, 239, 57–65. [Google Scholar] [CrossRef] [PubMed]
  78. Thörn, C.; Gustafsson, H.; Olsson, L. Immobilization of Feruloyl Esterases in Mesoporous Materials Leads to Improved Transesterification Yield. J. Mol. Catal. B Enzym. 2011, 72, 57–64. [Google Scholar] [CrossRef]
  79. Vafiadi, C.; Topakas, E.; Christakopoulos, P. Preparation of Multipurpose Cross-Linked Enzyme Aggregates and Their Application to Production of Alkyl Ferulates. J. Mol. Catal. B Enzym. 2008, 54, 35–41. [Google Scholar] [CrossRef]
  80. Topakas, E.; Stamatis, H.; Mastihubova, M.; Biely, P.; Kekos, D.; Macris, B.J.; Christakopoulos, P. Purification and Characterization of a Fusarium Oxysporum Feruloyl Esterase (FoFAE-I) Catalysing Transesterification of Phenolic Acid Esters. Enzym. Microb. Technol. 2003, 33, 729–737. [Google Scholar] [CrossRef]
  81. Giuliani, S.; Piana, C.; Setti, L.; Hochkoeppler, A.; Pifferi, P.G.; Williamson, G.; Faulds, C.B. Synthesis of Pentylferulate by a Feruloyl Esterase from Aspergillus Niger Using Water-in-Oil Microemulsions. Biotechnol. Lett. 2001, 23, 325–330. [Google Scholar] [CrossRef]
  82. Antonopoulou, I.; Iancu, L.; Jütten, P.; Piechot, A.; Rova, U.; Christakopoulos, P. Screening of Novel Feruloyl Esterases from Talaromyces Wortmannii for the Development of Efficient and Sustainable Syntheses of Feruloyl Derivatives. Enzym. Microb. Technol. 2019, 120, 124–135. [Google Scholar] [CrossRef]
  83. Zerva, A.; Antonopoulou, I.; Enman, J.; Iancu, L.; Jütten, P.; Rova, U.; Christakopoulos, P. Optimization of Transesterification Reactions with CLEA-Immobilized Feruloyl Esterases from Thermothelomyces Thermophila and Talaromyces Wortmannii. Molecules 2018, 23, 2403. [Google Scholar] [CrossRef] [Green Version]
  84. Antonopoulou, I.; Iancu, L.; Jütten, P.; Piechot, A.; Rova, U.; Christakopoulos, P. Optimized Enzymatic Synthesis of Feruloyl Derivatives Catalyzed by Three Novel Feruloyl Esterases from Talaromyces Wortmannii in Detergentless Microemulsions. Comput. Struct. Biotechnol. J. 2018, 16, 361–369. [Google Scholar] [CrossRef]
  85. Antonopoulou, I.; Leonov, L.; Jütten, P.; Cerullo, G.; Faraco, V.; Papadopoulou, A.; Kletsas, D.; Ralli, M.; Rova, U.; Christakopoulos, P. Optimized Synthesis of Novel Prenyl Ferulate Performed by Feruloyl Esterases from Myceliophthora Thermophila in Microemulsions. Appl. Microbiol. Biotechnol. 2017, 101, 3213–3226. [Google Scholar] [CrossRef] [Green Version]
  86. Huang, S.M.; Wu, P.Y.; Chen, J.H.; Kuo, C.H.; Shieh, C.J. Developing a High-Temperature Solvent-Free System for Efficient Biocatalysis of Octyl Ferulate. Catalysts 2018, 8, 338. [Google Scholar] [CrossRef] [Green Version]
  87. Chen, B.; Liu, H.; Guo, Z.; Huang, J.; Wang, M.; Xu, X.; Zheng, L. Lipase-Catalyzed Esterification of Ferulic Acid with Oleyl Alcohol in Ionic Liquid/Isooctane Binary Systems. J. Agric. Food Chem. 2011, 59, 1256–1263. [Google Scholar] [CrossRef] [PubMed]
  88. Sun, S.; Qin, F.; Bi, Y.; Chen, J.; Yang, G.; Liu, W. Enhanced Transesterification of Ethyl Ferulate with Glycerol for Preparing Glyceryl Diferulate Using a Lipase in Ionic Liquids as Reaction Medium. Biotechnol. Lett. 2013, 35, 1449–1454. [Google Scholar] [CrossRef] [PubMed]
  89. Matsuo, T.; Kobayashi, T.; Kimura, Y.; Hosoda, A.; Taniguchi, H.; Adachi, S. Continuous Synthesis of Glyceryl Ferulate Using Immobilized Candida antarctica Lipase. J. Oleo Sci. 2008, 57, 375–380. [Google Scholar] [CrossRef] [Green Version]
  90. Tsuchiyama, M.; Sakamoto, T.; Fujita, T.; Murata, S.; Kawasaki, H. Esterification of Ferulic Acid with Polyols Using a Ferulic Acid Esterase from Aspergillus Niger. Biochim. Biophys. Acta-Gen. Subj. 2006, 1760, 1071–1079. [Google Scholar] [CrossRef]
  91. Kikugawa, M.; Tsuchiyama, M.; Kai, K.; Sakamoto, T. Synthesis of Highly Water-Soluble Feruloyl Diglycerols by Esterification of an Aspergillus Niger Feruloyl Esterase. Appl. Microbiol. Biotechnol. 2012, 95, 615–622. [Google Scholar] [CrossRef]
  92. Xin, J.-y.; Chen, L.-l.; Zhang, Y.x.; Wen, R.-r.; Zhao, D.-m.; Xia, C.-g. Lipase-Catalyzed Synthesis of α-Tocopheryl Ferulate. Food Biotechnol. 2011, 25, 43–57. [Google Scholar] [CrossRef]
  93. Tan, Z.; Shahidi, F. Chemoenzymatic Synthesis of Phytosteryl Ferulates and Evaluation of Their Antioxidant Activity. J. Agric. Food Chem. 2011, 59, 12375–12383. [Google Scholar] [CrossRef]
  94. Antonopoulou, I.; Dilokpimol, A.; Iancu, L.; Mäkelä, M.R.; Varriale, S.; Cerullo, G.; Hüttner, S.; Uthoff, S.; Jütten, P.; Piechot, A.; et al. The Synthetic Potential of Fungal Feruloyl Esterases: A Correlation with Current Classification Systems and Predicted Structural Properties. Catalysts 2018, 8, 242. [Google Scholar] [CrossRef] [Green Version]
  95. Couto, J.; Karboune, S.; Mathew, R. Regioselective Synthesis of Feruloylated Glycosides Using the Feruloyl Esterases Expressed in Selected Commercial Multi-Enzymatic Preparations as Biocatalysts. Biocatal. Biotransformation 2010, 28, 235–244. [Google Scholar] [CrossRef]
  96. Antonopoulou, I.; Papadopoulou, A.; Iancu, L.; Cerullo, G.; Ralli, M.; Jütten, P.; Piechot, A.; Faraco, V.; Kletsas, D.; Rova, U.; et al. Optimization of Enzymatic Synthesis of L-Arabinose Ferulate Catalyzed by Feruloyl Esterases from Myceliophthora Thermophila in Detergentless Microemulsions and Assessment of Its Antioxidant and Cytotoxicity Activities. Process Biochem. 2018, 65, 100–108. [Google Scholar] [CrossRef]
  97. Vafiadi, C.; Topakas, E.; Alderwick, L.J.; Besra, G.S.; Christakopoulos, P. Chemoenzymatic Synthesis of Feruloyl D-Arabinose as a Potential Anti-Mycobacterial Agent. Biotechnol. Lett. 2007, 29, 1771–1774. [Google Scholar] [CrossRef] [PubMed]
  98. Topakas, E.; Vafiadi, C.; Stamatis, H.; Christakopoulos, P. Sporotrichum Thermophile Type C Feruloyl Esterase (StFaeC): Purification, Characterization, and Its Use for Phenolic Acid (Sugar) Ester Synthesis. Enzym. Microb. Technol. 2005, 36, 729–736. [Google Scholar] [CrossRef]
  99. Couto, J.; St-Louis, R.; Karboune, S. Optimization of Feruloyl Esterase-Catalyzed Synthesis of Feruloylated Oligosaccharides by Response Surface Methodology. J. Mol. Catal. B Enzym. 2011, 73, 53–62. [Google Scholar] [CrossRef]
  100. Fitriani, I.N.; Utami, W. Potential Phytochemical Inhibitor from Allium Cepa for the Medication of COVID-19. Alkimia J. Ilmu Kim. Dan Terap. 2021, 4, 80–87. [Google Scholar] [CrossRef]
  101. Bondhon, T.A.; Mahamud, R.A.; Jannat, K.; Hasan, A.; Jahan, R.; Rahmatullah, M. In Silico Binding Studies with B-Sitosterol and Some of Its Fatty Acid Esters to 3C-like Protease of SARS-CoV-2. J. Med. Plants Stud. 2020, 8, 86–90. [Google Scholar] [CrossRef]
  102. Sabater-Molina, M.; Larqué, E.; Torrella, F.; Zamora, S. Dietary Fructooligosaccharides and Potential Benefits on Health. J. Physiol. Biochem. 2009, 65, 315–328. [Google Scholar] [CrossRef]
  103. Cui, C.; Wang, Z.P.; Du, X.J.; Wang, L.Z.; Yu, S.J.; Liu, X.H.; Li, Z.M.; Zhao, W.G. Synthesis and Antiviral Activity of Hydrogenated Ferulic Acid Derivatives. J. Chem. 2013, 2013, 269434. [Google Scholar] [CrossRef]
  104. Lan, X.; Xie, D.; Yin, L.; Wang, Z.; Chen, J.; Zhang, A.; Song, B.; Hu, D. Novel α,β-Unsaturated Amide Derivatives Bearing α-Amino Phosphonate Moiety as Potential Antiviral Agents. Bioorganic Med. Chem. Lett. 2017, 27, 4270–4273. [Google Scholar] [CrossRef]
  105. Wu, Z.; Zhang, J.; Chen, J.; Pan, J.; Zhao, L.; Liu, D.; Zhang, A.; Chen, J.; Hu, D.; Song, B. Design, Synthesis, Antiviral Bioactivity and Three-Dimensional Quantitative Structure–Activity Relationship Study of Novel Ferulic Acid Ester Derivatives Containing Quinazoline Moiety. Pest Manag. Sci. 2017, 73, 2079–2089. [Google Scholar] [CrossRef]
  106. Spasova, M.; Philipov, S.; Nikolaeva-Glomb, L.; Galabov, A.S.; Milkova, T. Cinnamoyl- and Hydroxycinnamoyl Amides of Glaucine and Their Antioxidative and Antiviral Activities. Bioorg. Med. Chem. 2008, 16, 7457–7461. [Google Scholar] [CrossRef] [PubMed]
  107. Tang, X.; Zhang, C.; Chen, M.; Xue, Y.; Liu, T.; Xue, W. Synthesis and Antiviral Activity of Novel Myricetin Derivatives Containing Ferulic Acid Amide Scaffolds. New J. Chem. 2020, 44, 2374–2379. [Google Scholar] [CrossRef]
  108. Gan, X.; Hu, D.; Wang, Y.; Yu, L.; Song, B. Novel Trans-Ferulic Acid Derivatives Containing a Chalcone Moiety as Potential Activator for Plant Resistance Induction. J. Agric. Food Chem. 2017, 65, 4367–4377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Wang, Z.; Xie, D.; Gan, X.; Zeng, S.; Zhang, A.; Yin, L.; Song, B.; Jin, L.; Hu, D. Synthesis, Antiviral Activity, and Molecular Docking Study of Trans-Ferulic Acid Derivatives Containing Acylhydrazone Moiety. Bioorg. Med. Chem. Lett. 2017, 27, 4096–4100. [Google Scholar] [CrossRef]
  110. Ichimura, T.; Otake, T.; Mori, H.; Maruyama, S. HIV-1 Protease Inhibition and Anti-HIV Effect of Natural and Synthetic Water-Soluble Lignin-like Substances. Biosci. Biotechnol. Biochem. 1999, 63, 2202–2204. [Google Scholar] [CrossRef]
  111. Hariono, M.; Abdullah, N.; Damodaran, K.V.; Kamarulzaman, E.E.; Mohamed, N.; Hassan, S.S.; Shamsuddin, S.; Wahab, H.A. Potential New H1N1 Neuraminidase Inhibitors from Ferulic Acid and Vanillin: Molecular Modelling, Synthesis and in Vitro Assay. Sci. Rep. 2016, 6, 38692. [Google Scholar] [CrossRef] [Green Version]
  112. Nguyen, D.D.; Gao, K.; Chen, J.; Wang, R.; Wei, G.-W. Unveiling the Molecular Mechanism of SARS-CoV-2 Main Protease Inhibition from 137 Crystal Structures Using Algebraic Topology and Deep Learning. Chem. Sci. 2020, 11, 12036–12046. [Google Scholar] [CrossRef]
  113. Arnott, J.A.; Planey, S.L. The Influence of Lipophilicity in Drug Discovery and Design. Expert Opin. Drug Discov. 2012, 7, 863–875. [Google Scholar] [CrossRef]
  114. Coimbra, J.T.S.; Feghali, R.; Ribeiro, R.P.; Ramos, M.J.; Fernandes, P.A. The Importance of Intramolecular Hydrogen Bonds on the Translocation of the Small Drug Piracetam through a Lipid Bilayer. RSC Adv. 2021, 11, 899–908. [Google Scholar] [CrossRef]
  115. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-Likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [Green Version]
  116. Benet, L.Z.; Hosey, C.M.; Ursu, O.; Oprea, T.I. BDDCS, the Rule of 5 and Drugability. Adv. Drug Deliv. Rev. 2016, 101, 89–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Molsoft LLC About Molecular Properties Prediction. Available online: https://www.molsoft.com/mpropdesc.html (accessed on 1 May 2022).
  118. Zhang, L.; Zhou, L.; Bao, L.; Liu, J.; Zhu, H.; Lv, Q.; Liu, R.; Chen, W.; Tong, W.; Wei, Q.; et al. SARS-CoV-2 Crosses the Blood–Brain Barrier Accompanied with Basement Membrane Disruption without Tight Junctions Alteration. Signal Transduct. Target. Ther. 2021, 6, 337. [Google Scholar] [CrossRef] [PubMed]
  119. Ghahremanian, S.; Rashidi, M.M.; Raeisi, K.; Toghraie, D. Molecular Dynamics Simulation Approach for Discovering Potential Inhibitors against SARS-CoV-2: A Structural Review. J. Mol. Liq. 2022, 354, 118901. [Google Scholar] [CrossRef] [PubMed]
  120. Mahmud, S.; Mita, M.A.; Biswas, S.; Paul, G.K.; Promi, M.M.; Afrose, S.; Hasan, R.; Shimu, S.S.; Zaman, S.; Uddin, S.; et al. Molecular Docking and Dynamics Study to Explore Phytochemical Ligand Molecules against the Main Protease of SARS-CoV-2 from Extensive Phytochemical Datasets. Expert Rev. Clin. Pharmacol. 2021, 14, 1305–1315. [Google Scholar] [CrossRef] [PubMed]
  121. Khan, J.; Sakib, S.A.; Mahmud, S.; Khan, Z.; Islam, M.N.; Sakib, M.A.; Emran, T.B.; Simal-Gandara, J. Identification of Potential Phytochemicals from Citrus Limon against Main Protease of SARS-CoV-2: Molecular Docking, Molecular Dynamic Simulations and Quantum Computations. J. Biomol. Struct. Dyn. 2021, 1–12. [Google Scholar] [CrossRef]
  122. Biswas, S.; Mahmud, S.; Mita, M.A.; Afrose, S.; Hasan, R.; Sultana Shimu, M.S.; Saleh, A.; Mostafa-Hedeab, G.; Alqarni, M.; Obaidullah, A.J.; et al. Molecular Docking and Dynamics Studies to Explore Effective Inhibitory Peptides against the Spike Receptor Binding Domain of SARS-CoV-2. Front. Mol. Biosci. 2022, 8, 791642. [Google Scholar] [CrossRef] [PubMed]
Figure 1. SARS-CoV-2 Mpro in the active form of a homodimer (PDB ID: 7JKV). The three domains and their secondary structure are visible in the left monomer (domain I is in cyan, domain II in orange, and domain III in purple). The right monomer is shown as surface, with the catalytic residues His41 and Cys145 being highlighted in green and yellow, respectively.
Figure 1. SARS-CoV-2 Mpro in the active form of a homodimer (PDB ID: 7JKV). The three domains and their secondary structure are visible in the left monomer (domain I is in cyan, domain II in orange, and domain III in purple). The right monomer is shown as surface, with the catalytic residues His41 and Cys145 being highlighted in green and yellow, respectively.
Biomedicines 10 01787 g001
Figure 2. Superimposed binding modes of the inhibitors N3 (a) and GC376 (b) as occurring from the co-crystallization structure (PDB ID: 6LU7 and 7D1M, respectively) (petrol blue) and the molecular docking simulation (purple), respectively. The root-mean-square-deviation (RMSD) of the binding between the co-crystallized and docked complex is indicated.
Figure 2. Superimposed binding modes of the inhibitors N3 (a) and GC376 (b) as occurring from the co-crystallization structure (PDB ID: 6LU7 and 7D1M, respectively) (petrol blue) and the molecular docking simulation (purple), respectively. The root-mean-square-deviation (RMSD) of the binding between the co-crystallized and docked complex is indicated.
Biomedicines 10 01787 g002
Figure 3. Binding of FA to the active site of Mpro. The first three clusters resulting from the simulation are presented.
Figure 3. Binding of FA to the active site of Mpro. The first three clusters resulting from the simulation are presented.
Biomedicines 10 01787 g003
Figure 4. Structures and binding modes of alkyl and alkenyl FA esters in the active site of Mpro.
Figure 4. Structures and binding modes of alkyl and alkenyl FA esters in the active site of Mpro.
Biomedicines 10 01787 g004
Figure 5. Structures and binding modes of fatty acid, polyol, tocopherol, and sterol FA derivatives at the active site of Mpro.
Figure 5. Structures and binding modes of fatty acid, polyol, tocopherol, and sterol FA derivatives at the active site of Mpro.
Biomedicines 10 01787 g005
Figure 6. Structures and binding modes of the monosaccharide esters of FA at the active site of Mpro.
Figure 6. Structures and binding modes of the monosaccharide esters of FA at the active site of Mpro.
Biomedicines 10 01787 g006
Figure 7. Structures and binding modes of disaccharide esters of FA at the active site of Mpro.
Figure 7. Structures and binding modes of disaccharide esters of FA at the active site of Mpro.
Biomedicines 10 01787 g007
Figure 8. Structures and binding modes of trisaccharide and polyol esters of FA at the active site of Mpro.
Figure 8. Structures and binding modes of trisaccharide and polyol esters of FA at the active site of Mpro.
Biomedicines 10 01787 g008
Figure 9. Structure and binding of the three suggested FOS ferulate structures at the active site of Mpro.
Figure 9. Structure and binding of the three suggested FOS ferulate structures at the active site of Mpro.
Biomedicines 10 01787 g009
Figure 10. Structure and binding mode of FA rutinoside at the active site of Mpro.
Figure 10. Structure and binding mode of FA rutinoside at the active site of Mpro.
Biomedicines 10 01787 g010
Figure 11. Structures and binding modes of chemically synthesized FA derivatives in the active site of Mpro.
Figure 11. Structures and binding modes of chemically synthesized FA derivatives in the active site of Mpro.
Biomedicines 10 01787 g011aBiomedicines 10 01787 g011b
Figure 12. (a) RMSD values of the C-alpha atoms of the complexes of the ligands FA rutinoside, raffinose ferulate, compound e27, and reference inhibitor N3 with Mpro throughout the simulation time; (b) Radius of gyration of the complexes throughout the simulation time; (c) RMSF values of residues of Mpro in complex with the ligands; (d) Number of hydrogen bonds formed between the ligands and Mpro throughout the simulation time.
Figure 12. (a) RMSD values of the C-alpha atoms of the complexes of the ligands FA rutinoside, raffinose ferulate, compound e27, and reference inhibitor N3 with Mpro throughout the simulation time; (b) Radius of gyration of the complexes throughout the simulation time; (c) RMSF values of residues of Mpro in complex with the ligands; (d) Number of hydrogen bonds formed between the ligands and Mpro throughout the simulation time.
Biomedicines 10 01787 g012
Figure 13. Number of contacts of the ligands (a) FA rutinoside, (b) raffinose ferulate, (c) compound e27, and (d) N3 with Mpro residues throughout the simulation time. The residues marked with an asterisk (*) are residues that appeared to have contact with the ligand in the MD simulation but were not given as contacting residues in the molecular docking simulation output. However, they are neighboring contacting residues. This can be due to the fact that since MD trajectory analysis uses the distance as a parameter to calculate contacts, sometimes atoms from neighboring residues can be within a distance small enough to be regarded as contacts.
Figure 13. Number of contacts of the ligands (a) FA rutinoside, (b) raffinose ferulate, (c) compound e27, and (d) N3 with Mpro residues throughout the simulation time. The residues marked with an asterisk (*) are residues that appeared to have contact with the ligand in the MD simulation but were not given as contacting residues in the molecular docking simulation output. However, they are neighboring contacting residues. This can be due to the fact that since MD trajectory analysis uses the distance as a parameter to calculate contacts, sometimes atoms from neighboring residues can be within a distance small enough to be regarded as contacts.
Biomedicines 10 01787 g013
Table 1. Listing and synthesis methods of enzymatically synthesized FA derivatives.
Table 1. Listing and synthesis methods of enzymatically synthesized FA derivatives.
CompoundReactionDonorAcceptorEnzymeSolvent SystemYield (Time)T (°C)Reference
Methyl ferulateEsterificationFAMethanolCLEAs AnFaeASolvent-free (containing buffer)20.6% (24 h)30[71]
EsterificationMethanolFAImmobilized CALB[bmim]PF641.7% (72 h)60[72]
Ethyl ferulateEsterificationFAEthanolCLEAs AnFaeASolvent-free (containing buffer)50.5% (24 h)30[71]
EsterificationEthanolFARMLHexane76.2% (72 h)61[73]
EsterificationEthanolFAImmobilized CALB[bmim]PF640.7% (72 h)60[72]
Propyl ferulateEsterificationFAPropanolCLEAs AnFaeASolvent-free (containing buffer)98.8% (24 h)30[71]
EsterificationPropanolFAImmobilized RML[bmim]PF648.2% (72 h)60[72]
TransesterificationMFAPropanolFoFae-IIn-Hexane:1-propanol: water16% (224 h)30[74]
Butyl ferulateEsterificationFAButanolCLEAs AnFaeASolvent-free (containing buffer)99.5% (24 h)30[71]
EsterificationButanolFAImmobilized RML[bmim]PF652.6% (72 h)60[72]
EsterificationButanolFANovozym 435Solvent-freeTraces (15 d)60[75]
TransesterificationMFAButanolAocFaeCIsooctane: butanol: buffern.q.30[76]
TransesterificationMFA1-ButanolC1 FAEs immobilized on mesoporous silicaSolvent-free (containing buffer)n.q.30[77]
TransesterificationMFA1-ButanolDepol 740 LSolvent-free (containing buffer)Up to 90% (6 d)37[78]
TransesterificationMFA1-ButanolCLEAs Ultraflo LHexane: 1-butanol: buffer97% (144 h)37[79]
TransesterificationMFA1-ButanolFoFae-IHexane:1-butanol: buffer∼13% (144 h)35[80]
Isobutyl ferulateEsterificationFAIsobutanolCLEAs AnFaeASolvent-free (containing buffer)98.4% (24 h)30[71]
Pentyl ferulateEsterificationFAPentanolCLEAs AnFaeASolvent-free (containing buffer)99.4% (24 h)30[71]
EsterificationFA1-PentanolFAEACTAB: hexane: pentanol: buffer60% (n/q)40[81]
Isopentyl ferulateEsterificationFAIsopentanolCLEAs AnFaeASolvent-free (containing buffer)97.0% (24 h)30[71]
Prenyl ferulateTransesterificationVFAPrenolFae125n-Hexane: buffer: DMSO92.5% (24 h)25[82]
TransesterificationVFAPrenolCLEAs Fae125n-Hexane: buffer83.7% (34.3 h)32[83]
TransesterificationVFAPrenolFae125n-Hexane: t-butanol: buffer81.1% (24 h)40[84]
TransesterificationVFAPrenolC1FaeB2n-Hexane: t-butanol: buffer71.5% (24 h)30[85]
Hexyl ferulateEsterificationFAHexanolCLEAs AnFaeASolvent-free (containing buffer)98.5% (24 h)30[71]
EsterificationHexanolFAImmobilized RML[bmim]PF638.1% (72 h)60[72]
Octyl ferulateEsterificationFAOctanolCLEAs AnFaeASolvent-free (containing buffer)99.3% (24 h)30[71]
EsterificationOctanolFANovozym 435Solvent-free93.2% (72 h)92.2[86]
EsterificationOctanolFAImmobilized RML[bmim]PF634.9% (72 h)60[72]
EsterificationOctanolFANovozym 435Solvent free13% (15 d)60[75]
Dodecyl (or lauryl) ferulateEsterificationFADodecanolCLEAs AnFaeASolvent-free (containing buffer)96.6% (24 h)30[71]
EsterificationDodecanolFANovozym 435Solvent-free10% (15 d)60[75]
Octadecyl (or stearyl) ferulateEsterificationOctadecanolFAImmobilized RMLHexanen.q. (72 h)61[73]
Oleyl ferulateEsterificationFAOleyl alcoholCLEAs AnFaeASolvent-free (containing buffer)100% (24 h)30[71]
TransesterificationOleyl alcoholFANovozym 435Hexane99.17% (4 d)60[87]
Glyceryl ferulateTransesterificationGlycerolEFANovozym 435EMIMTF2N100% (12 h)70[88]
EsterificationFAGlycerolChirazyme L2 C-2Solvent-free80% (>3 h)80[89]
EsterificationFAGlycerolFAE-PLGlycerol: DMSO: buffer81% (n.q.)50[90]
Diglyceryl ferulateEsterificationFADiglycerin SFAE-PLDiglycerin S: DMSO: buffer95% (12 h)50[91]
Tocopheryl ferulateTransesterificationVitamin EEFANovozym 435Solvent-free25.2% (72 h)60[92]
Sitosteryl ferulateTransesterification
Esterification
SitosterolEFA
FA
CRLHexane55% (5 d)
35% (5 d)
63[73]
TransesterificationSitosterolVFACRLHexane: 2 butanone∼55% (10 d)45[93]
d-glucose ferulateTransesterificationVFAD-glucoseFae125n-Hexane: t-butanol: buffer22.5% (8 h)45[94]
d-galactose ferulateTransesterificationVFAD-galactoseC1FaeA1n-Hexane: t-butanol: buffer22.8% (8 h)45[94]
EsterificationFAD-galactoseFlavourzymeHexane: t-butanol: buffer41.9% (144 h)35[95]
d-mannose ferulateTransesterificationVFAD-mannoseC1FaeA1n-Hexane: t-butanol: buffer21.5% (8 h)45[94]
d-fructose ferulateTransesterificationVFAD-fructoseC1FaeA1n-Hexane: t-butanol: buffer29.4% (8 h)45[94]
Arabinose ferulateTransesterificationVFAL-ArabinoseFae125n Hexane: buffer: DMSO56.2% (24 h)40[82]
TransesterificationVFAL-ArabinoseFae125n-Hexane: t-butanol: buffer33.0%40[96]
TransesterificationVFAL-ArabinoseC1FaeA1n-Hexane: t-butanol: buffer52.2% (8 h)55[84]
TransesterificationVFAL-arabinoseCLEAs Fae125n-Hexane: buffer58.1% (10 h)32[83]
EsterificationFAD-ArabinoseMultifect P3000Hexane: t-butanol: buffer36.7% (144 h)35[95]
TransesterificationMFAD-arabinoseStFae-CHexane: t-butanol: buffer45% (n.q.)35[97]
TransesterificationMFA
EFA
L-arabinoseStFae-CHexane: t-butanol: bufferUp to 50% (120 h)
6.3% (n.q.)
35[98]
d-xylose ferulateTransesterificationVFAD-xyloseC1FaeA1n-Hexane: t-butanol: buffer7.5% (8 h)45[94]
EsterificationFAD-xyloseMultifect P3000Hexane: t-butanol: buffer30.8% (144 h)35[95]
d-lactose ferulateTransesterificationVFAD-lactoseC1FaeA1n-Hexane: t-butanol: buffer<2% (8 h)45[94]
EsterificationFALactoseDepol 740 Ln-Hexane: 2-butanone: buffer4.4% (n.q.)35[99]
d-sucrose ferulateTransesterificationVFAD-sucroseFaeA1n-Hexane: t-butanol: buffer8.2% (8 h)45[94]
EsterificationFAD-sucroseDepol 740 Ln-Hexane: 2-butanone: buffer13.2% (n.q.)35[99]
d-maltose ferulateTransesterificationVFAD-maltoseC1FaeA1n-Hexane: t-butanol: buffer8.9% (8 h)45[94]
d-cellobiose ferulateTransesterificationVFAD-cellobioseC1FaeA1n-Hexane: t-butanol: buffer<2% (8 h)45[94]
Xylobiose ferulateEsterificationFAXylobioseDepol 740 Ln-Hexane: 2-butanone: buffer9.4% (n.q.)35[99]
Galactobiose ferulateEsterificationFAGalactobioseDepol 740 Ln-Hexane: 2-butanone: buffer5.4% (n.q)35[99]
Arabinobiose ferulateEsterificationFAArabinobioseDepol 740 Ln-Hexane: 2-butanone: buffer7.9% (n.q)35[99]
Raffinose ferulateEsterificationFARaffinoseDepol 740 Ln-Hexane: 2-butanone: buffer11.9% (7 d)35[99]
FOS ferulateEsterificationFAFOSDepol 740 Ln-Hexane: 2-butanone: buffer9.6% (n.q.)35[99]
d-mannitol ferulateTransesterificationVFAD-mannitolC1FaeA1n-Hexane: t-butanol: buffer26.7% (8 h)45[94]
d-sorbitol ferulateTransesterificationVFAD-sorbitolC1FaeA1n-Hexane: t-butanol: buffer50.0% (8 h)45[94]
d-xylitol ferulateTransesterificationVFAD-xylitolC1FaeA1n-Hexane: t-butanol: buffer43.3% (8 h)45[94]
FA rutinosideTransglycosylationRutinFARutinase derived from tartary buckwheatBuffer∼4.5 μmol (48 h)40[35]
Yields were expressed based on the limiting reactant. FA: Ferulic acid; VFA: Vinyl ferulate; MFA: Methyl ferulate; EFA: Ethyl ferulate; CLEAs: Cross-linked enzyme aggregates; AnFaeA, FAEA: Feruloyl esterase from Aspergillus niger; RML: Lipase from Rhizomucor miehei; FoFae-I, FoFae-II: Feruloyl esterases from Fusarium oxysporum; Novozym 435: Lipase B from C. antarctica immobilized on a microporous acrylic resin (synonym CALB); AocFaeC: Feruloyl esterase from Aspergillus ochraceus; C1FAEs, e.g., C1FaeA1, C1FaeB2: Feruloyl esterases from Myceliophthora thermophila C1; Depol 740 L, Ultraflo L: Commercial multi-enzymatic preparation from Humicola spp. with side feruloyl esterase activity; FAEA: Fae125: Feruloyl esterase from Talaromyces wortmanni; Chirazyme L-2: Immobilized lipase from Candida antarctica; FAE-PL: FAE from Aspergillus niger purified from the commercial preparation ‘Amano’ Pectinase PL; CRL: Lipase from Candida rugosa; Flavourzyme: Commercial multi-enzymatic preparation from Aspergillus oryzae with side feruloyl esterase activity; Multifect P300: Commercial multi-enzymatic preparation from Bacillus amyloliquefaciens with side feruloyl esterase activity; St-FaeC: Feruloyl esterase from Sporotrichum thermophile ATCC 34628; Cetyltrimethylammoniumbromide (CTAB); n.q.: not quantified.
Table 2. Molecular docking simulation results for FA and its enzymatically synthesized derivatives.
Table 2. Molecular docking simulation results for FA and its enzymatically synthesized derivatives.
CompoundBinding Energy (kcal/mol) 1No of Interactions Total Contacting Residues
H-Bond 2Hydrophobic 3Pi-Pi
Ferulic acid−5.873 (ARG 188, THR 190 × 2)1 (GLN 189)1 (HIS 41)HIS 41, MET 49, TYR 54, CYS 145, HIS 164, MET 165, GLU 166, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
Methyl ferulate−5.734 (GLY 143 × 2, SER 144, CYS 145)1 (MET 165)1 (HIS 163)LEU 27, HIS 41, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, MET 165, GLU 166, ARG 188, GLN 189, GLN 192
Ethyl ferulate−5.877 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145, GLN 189)1 (GLN 189)1 (HIS 163)LEU 27, HIS 41, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, GLN 192
Propyl ferulate−5.986 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145)1 (MET 165)1 (HIS 163)LEU 27, HIS 41, CYS 44, MET 49, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, ASP 187, ARG 188, GLN 189
Butyl ferulate−6.026 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145)1 (MET 165)1 (HIS 163)HIS 41, CYS 44, MET 49, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, ASP 187, ARG 188, GLN 189
Isobutyl ferulate−6.307 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145, GLN 189)1 (MET 165)1 (HIS 163)HIS 41, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, GLN 192
Pentyl ferulate−5.922 (THR 190, GLN 192)1 (MET 165)1 (HIS 41)HIS 41, MET 49, TYR 54, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
Isopentyl ferulate−6.586 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145)1 (MET 49)1 (HIS 163)LEU 27, HIS 41, CYS 44, MET 49, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, ASP 187, ARG 188, GLN 189
Prenyl ferulate−6.756 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145)1 (GLN 189)1 (HIS 163)LEU 27, HIS 41, CYS 44, MET 49, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, ASP 187, ARG 188, GLN 189
Hexyl ferulate−6.477 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145, GLN 189)1 (GLN 189)1 (HIS 163)HIS 41, MET 49, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, ASP 187, ARG 188, GLN 189
Octyl ferulate−6.206 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145)1 (HIS 41)1 (HIS 163)HIS 41, MET 49, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, ASP 187, ARG 188, GLN 189, THR 190
Dodecyl ferulate−5.563 (ARG 188, THR 190 × 2)1 (GLU 166)1 (HIS 41)HIS 41, MET 49, PHE 140, LEU 141, ASN 142, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, PHE 181, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
Octadecyl ferulate−5.204 (ARG 188, THR 190 × 2, GLN 192)1 (MET 49)0THR 25, THR 26, LEU 27, HIS 41, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, PRO 168, HIS 172, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
Oleyl ferulate−5.174 (ARG 188 × 2, THR 190 × 2)1 (MET 49)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
Glyceryl ferulate−6.5512 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145, GLU 166, ARG 188, GLN 189, THR 190 × 2, GLN 192)1 (MET 165)1 (HIS 163)LEU 27, HIS 41, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, GLN 192
Diglyceryl ferulate−6.6110 (TYR 54, LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145, HIS 164, ARG 188, GLN 189)1 (GLN 189)1 (HIS 163)HIS 41, CYS 44, MET 49, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
Tocopheryl ferulate−6.912 (SER 46, GLN 189)1 (GLN 189)THR 25, LEU 27, HIS 41, SER 46, GLU 47, MET 49, LEU 50, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190
Sitosteryl ferulate−7.813 (THR 25, HIS 41, GLY 143)1 (PRO 168)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, CYS 44, THR 45, SER 46, MET 49, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-glucose ferulate−7.0911 (LEU 141, SER 144, HIS 163, GLU 166, ARG 188, GLN 189, THR 190 × 3, GLN 192 × 2)1 (GLU 166)1 (HIS 163)HIS 41, PHE 140, LEU 141, ASN 142, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-galactose ferulate−7.2413 (LEU 141 × 2, ASN 142, GLY 143, SER 144 × 3, CYS 145, HIS 163, ARG 188, THR 190 × 2, GLN 192)1 (GLN 189)0PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, MET 165, GLU 166, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-mannose ferulate−7.0911 (LEU 141, SER 144, HIS 163, GLU 166, ARG 188, GLN 189, THR 190 × 3, GLN 192 × 2)1 (GLU 166)1 (HIS 163)HIS 41, PHE 140, LEU 141, ASN 142, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-fructose ferulate−7.068 (THR 26 × 2, PHE 140, LEU 141, ASN 142, SER 144, HIS 163, GLU 166)1 (HIS 41)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, THR 45, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, GLN 189
l-arabinose ferulate−7.085 (HIS 41, MET 49, HIS 164, GLN 189, THR 190)1 (LEU 167)0HIS 41, MET 49, TYR 54, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-xylose ferulate−7.3713 (LEU 141 × 2, ASN 142, GLY 143, SER 144 × 3, CYS 145, HIS 163, ARG 188, THR 190 × 2, GLN 192)1 (ΜΕΤ 165)1 (HIS 163)PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
d-lactose ferulate−7.6312 (ASN 142 × 2, GLY 143, GLU 166 × 3, ARG 188, GLN 189, THR 190 × 3, GLN 192)1 (ΜΕΤ 165)1 (HIS 41)HIS 41, MET 49, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-sucrose ferulate−7.7714 (THR 26, LEU 141 × 2, GLY 143 × 2, SER 144 × 3, CYS 145, HIS 163 × 2, GLU 166, GLN 189 × 2)1 (GLN 189)0THR 25, THR 26, LEU 27, HIS 41, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, GLN 189, THR 190, ALA 191, GLN 192
d-maltose ferulate−7.4713 (SER 46 × 2, LEU 141 × 2, ASN 142, GLY 143 × 2, SER 144 × 2, CYS 145, GLU 166, GLN 189 × 2)1 (THR 25)0THR 24, THR 25, THR 26, LEU 27, HIS 41, THR 45, SER 46, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, GLN 189
d-cellobiose ferulate−7.509 (THR 24, LEU 141, ASN 142 × 2, GLY 143 × 2, SER 144, THR 190, GLN 192)1 (ΜΕΤ 165)0THR 24, THR 25, THR 26, LEU 27, HIS 41, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
Xylobiose ferulate−7.9711 (THR 24 × 2, THR 25, THR 26, THR 45 × 2, THR 46 × 2, HIS 41, PHE 140, GLU 166)1 (GLU 166)1 (HIS 163)THR 24, THR 25, THR 26, LEU 27, HIS 41, THR 45, SER 46, MET 49, PHE 140, LEU 141, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, GLN 189
Galactobiose ferulate−8.3612 (ASN 119 × 2, PHE 140, LEU 141, GLY 143, SER 144 × 2, CYS 145, GLU 166 × 2, GLN 189, THR 190)1 (GLN 189)0GLN 19, THR 25, THR 26, LEU 27, HIS 41, MET 49, TYR 118, ASN 119, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, GLN 189, THR 190, ALA 191, GLN 192
Arabinobiose ferulate−7.887 (THR 26, PHE 140, ASN 142, GLU 166 × 2, LEU 167, GLN 189)1 (GLN 189)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, MET 49, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
Raffinose ferulate−8.348 (THR 26, SER 144 × 2, CYS 145, HIS 163, GLU 166, THR 190, GLN 192)1 (MET 49)0THR 24, THR 25, THR 26, LEU 27, THR 45, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
FOS ferulate 1−8.189 (SER 46 × 2, PHE 140, LEU 141 × 2, GLY 143, SER 144, HIS 163, THR 190)1 (GLN 189)0THR 25, THR 26, HIS 41, THR 45, SER 46, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
FOS ferulate 2−7.348 (THR 26 × 3, GLY 143 × 2, HIS 164, GLU 166, THR 190)1 (LEU 27)0THR 25, THR 26, LEU 27, HIS 41, VAL 42, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, GLN 192
FOS ferulate 3−8.5214 (SER 46 × 3, LEU 141 × 2, ASN 142, GLY 143 × 2, SER 144 × 2, CYS 145, HIS 163, HIS 164, GLN 192)1 (LEU 50)0THR 24, THR 25, THR 26, LEU 27, HIS 41, CYS 44, THR 45, SER 46, GLU 47, MET 49, LEU 50, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-mannitol ferulate−6.3812 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145, GLU 166, ARG 188, GLN 189, THR 190 × 2, GLN 192)1 (ΜΕΤ 165)1 (HIS 163)PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-sorbitol ferulate−6.4313 (LEU 141, GLY 143 × 2, SER 144 × 2, CYS 145, GLU 166, ARG 188, GLN 189 × 2, THR 190 × 2, GLN 192)1 (ΜΕΤ 165)1 (HIS 163)PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
d-xylitol ferulate−6.6815 (PHE 140, LEU 141 × 2, ASN 142, GLY 143, SER 144 × 3, CYS 145, HIS 163, GLU 166, ARG 188, THR 190 × 2, GLN 192)1 (GLN 189)0PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, MET 165, GLU 166, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
FA rutinoside−8.4016 (THR 24 × 3, THR 25, THR 45, LEU 141 × 2, GLY 143 × 3, SER 144 × 2, CYS 145, HIS 163, GLU 166, ARG 188, GLN 189)1 (MET 49)0THR 24, THR 25, THR 26, LEU 27, HIS 41, CYS 44, THR 45, SER 46, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, ARG 188, GLN 189, THR 190, GLN 192
N3−8.264 (CYS 145, GLU 166, GLN 189)1 (MET 49)1(HIS 41)THR 25, LEU 27, HIS 41, MET 49, LEU 50, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
GC376−7.805 (HIS 41, PHE 140, HIS 163, GLU 166, GLN 189)1 (ASP 187)0HIS 41, MET 49, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
1: The binding energies are given as negative values and correspond to the best cluster for each compound. A lower binding energy corresponds to a higher binding affinity. 2: H-bonds were calculated by Pymol. 3: Hydrophobic and pi-pi interactions were calculated by the YASARA structure.
Table 3. Listing and synthesis method of chemically synthesized FA derivatives.
Table 3. Listing and synthesis method of chemically synthesized FA derivatives.
CategoryCode 1NameDerivatization MethodYield NoD 2Reference
FA amide7aNot givenSix steps: acetylation with acetic anhydride in aqueous sodium hydroxide solution, reaction with thionyl chloride, reaction with a substituted 2-amino-1-phenylethanone, reduction to the corresponding alcohol with sodium borohydride and hydrolyzation of the acetyl group with NaOH, alkylation of the phenolic hydroxyl group, and alkylation with a bromoalkane in the presence of NaH.Not given16[32]
Hydrogenated FA amides (A)13bNot givenFour steps: catalytic hydrogenation of FA using Pd/C and H2 in the presence of HCl, multi-step reaction involving an acid chloride intermediate (including hydrolysis, hydroxyl protection, acyl chloride formation, amidation, and deprotection) and microwave radiation, alkylation of the phenolic hydroxyl group with bromoalkene and NaOH, reaction with bromoalkane, and deprotonation with NaOH.71%9[32]
Hydrogenated FA amides (B)4fN-(2-(4-(Benzyloxy)phenyl)-2-(prop-2-yn-1-yloxy)ethyl)-3-(3,4-dimethoxyphenyl)propanamideThree steps: catalytic hydrogenation of FA with Pd/C and H2 towards ethyl ferulate, substitution with 2-amino-1-phenylethanol under microwave radiation at 130 oC, alkylation with bromoalkane.68%7[103]
FA sulfonamide2€-3-(4-(2-((4-acetamidophenyl)sulfonamido)ethoxy)-3-methoxyphenyl)acrylate methylTwo parallel steps: reaction of FA with alcohol catalyzed by sulfuric acid, and reaction of sulfonyl chloride and bromoethylamine hydrobromide in dichloromethane in the presence of triethylamine. Dissolvation of intermediate A in acetonitrile and potassium carbonate and combination with the other intermediate towards the target compound.45%16[33]
α,β-Unsaturated amide derivatives of FA with an α-aminophosphonate moietyg18(E)-((4-chlorophenyl)(3-(3-methoxy-4-((4-(trifluoromethyl)benzyl)oxy)phenyl)acrylamido)methyl)phosphonateTwo parallel routes towards two intermediates, which are then combined via dehydration condensation reaction towards the final product. The first intermediate involves treatment of an aromatic aldehyde with ammonia, reaction with diethyl phosphite, and then hydrolysis to diethyl 1-aminoarylmethylphosphonate. The second intermediate is also produced in three steps, starting from FA, which is methylated in with methyl alcohol in the presence of sulfuric acid, then esterification with benzyl halide in the presence of potassium carbonate and acetonitrile, and then hydrolyzation with NaOH.53.60%26[104]
FA derivatives with a quinazoline moiety (A)e272-methoxyphen€(E)-3-(3-methoxy-4-((4-oxoquinazolin-3(4H)-yl)methoxy)phenyl)acrylateThe first step of the derivatization involves esterification of FA, either with the appropriate alcohol in the presence of sulfuric acid, or by reaction with acetic anhydride, NaOH, and then with thionyl chloride towards an O-acetyl ferulic acid chloride, and finally mixture with tetrahydrofuran, triethylamine, and the appropriate phenol. Then, the intermediate was mixed with 3-chloromethyl-4(3H)-quinazolinone, potassium carbonate, potassium iodide, and acetronitrile toward the final product.50.80%28[105]
FA derivatives with a quinazoline moiety (B)e284-allyl-2-methoxyp€yl-(E)-3-(3-methoxy-4-((4-oxoquinazolin-3(4H)-yl)methoxy)phenyl)acrylateThe first step of the derivatization involves esterification of FA, either with the appropriate alcohol in the presence of sulfuric acid, or by reaction with acetic anhydride, NaOH, and then with thionyl chloride towards an O-acetyl ferulic acid chloride, and finally mixture with tetrahydrofuran, triethylamine, and the appropriate phenol. Then, the intermediate was mixed with 3-chloromethyl-4(3H)-quinazolinone, potassium carbonate, potassium iodide, and acetronitrile toward the final product.64.80%28[105]
FA amide of 3-aminomethyl glaucine5Feruloyl amide of 3-aminomethylglaucinePeptide chemistry methods using EDC/HOBt to link 3-aminomethylglaucine to FA. 3-aminomethylglaucine was produced from glaucine through reaction with N-(hydroxymethyl)acetamide in acidic media and subsequent hydrolyzation.61.60%1[106]
Myricetin derivatives with a FA amide scaffold4n(E)-N-(4-bromophenyl)-3-(4-(3-((5,7-dimethoxy-4-oxo-2-(3,4,5-trimethoxyphenyl)-4Hchromen-3-yl)oxy)propoxy)-3-methoxyphenyl)acrylamideSynthesis of two intermediates, which are then combined using DMF and potassium carbonate. The first one is derived from FA, involving reaction with acetic anhydride in the presence of NaOH, then amidation through a reaction with a phenylamine in the presence of HOBt and EDCl and lastly dissolution in acetonitrile and hydrazine hydrate. The second is a myricitrin derivative occurring from reaction with DMF, potassium carbonate, and methyl iodide in the presence of hydrochloride and then with DMF and dibromoalkanes.67.92%22[107]
FA derivatives containing dithioacetal moiety (A)2a4-(bis((2-Hydroxyethyl)thio)methyl)-2-methoxyphenyl(E)-3-(4-acetoxy-3-methoxyphenyl)acrylateReaction of FA with acetic anhydride and NaOH towards O-acetyl FA, then reaction with thionyl chloride and mixing of the respective chloride with 1,2 dioxane, triethylamine, and hydroxy aldehyde. The synthesized intermediate was mixed with thiol, NaHSO4·SiO2, and dichloromethane towards the final product.69.30%17[34]
FA derivatives containing dithioacetal moiety (B)2yNot givenReaction of FA with acetic anhydride and NaOH towards O-acetyl FA, then reaction with thionyl chloride and mixing of the respective chloride with 1,2 dioxane, triethylamine, and hydroxy aldehyde. The synthesized intermediate was mixed with thiol, NaHSO4·SiO2, and dichloromethane towards the final product.Not given8[34]
FA derivatives containing dithioacetal moiety (C)2sNot givenReaction of FA with acetic anhydride and NaOH towards O-acetyl FA, then reaction with thionyl chloride and mixing of the respective chloride with 1,2 dioxane, triethylamine, and hydroxy aldehyde. The synthesized intermediate was mixed with thiol, NaHSO4·SiO2, and dichloromethane towards the final product.Not given2[34]
Trans-FA esters with a chalcone groupF3(E)-methyl-3-(4-(2-(4-((E)-3-(2-fluorophenyl)acryloyl)phenoxy)ethoxy)-3-methoxyphenyl)acrylateEsterification of FA with the appropriate alcohol in the presence of sulfuric acid and reaction with potassium carbonate in butanone with the addition of 1,2, dibromoethane towards an intermediate compound. Reaction of 4-hydroxyacetophenone with the appropriate aromatic aldehyde and reaction of the product with the previously mentioned intermediate and potassium carbonate in dimethylformamide.92.60%35[108]
Trans-Fa derivatives containing acylhydrazone moietyD4(E)-3-(4-(benzyloxy)-3-methoxyphenyl)-N’-(thiophen-2-ylmethylene)acrylohydrazideStarting with trans-ferulic acid, through four steps, including substitution, using RX in potassium carbonate and dimethylformamide, hydrolysis, using KOH and methanol, hydrazinoly-sis, using hydroxybenzotriazole (HoBt/EDCl) in DMF, and condensation, involving an appropriate aldehyde and methanol.80.80%23[109]
DiFADiferulic acidFractionation of dehydrogenated polymers of FA by ultrafiltration (synthesized with horseradish peroxidase from FA).Not givenn.d.[110]
TriFATriferulic acidFractionation of dehydrogenated polymers of FA by ultrafiltration (synthesized with horseradish peroxidase from FA).Not givenn.d.[110]
FA 3-amino derivativesMY3(E)-ethyl 3-(4-isopropoxy-3-methoxy-5-nitrophenyl)acrylateNitration of FA with acetic and nitric acid, esterification of the product with the corresponding alcohol in sulfuric acid, and mixture of the derived compound with sodium carbonate in DMF. Addition of the corresponding alkyl bromide and tetra-butylammonium iodide in DMF to the previously described solution to yield the final product.9%23[111]
1: Name of the compound as it is coded in the respective publication; 2: Number of FA derivatives synthesized in the respective study, among which the ones used in this work were selected.
Table 4. Molecular docking simulation results for the chemically synthesized derivatives of FA.
Table 4. Molecular docking simulation results for the chemically synthesized derivatives of FA.
CompoundBinding Energy 1 (kcal/mol)No of Interactions Total Contacting Residues
H-Bond 2Hydrophobic 3Pi-Pi
7a−6.671 (HIS 41)1 (MET 165)1 (HIS 41)THR 25, THR 26, LEU 27, HIS 41, MET 49, LEU 141, ASN 142, GLY 143, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
13b−6.5401 (GLN 189)0THR 26, LEU 27, HIS 41, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
4f−6.273 (ASN 142, HIS 163, GLN 189)1 (GLN 189)1 (HIS 41)THR 25, THR 26, LEU 27, HIS 41, VAL 42, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, ASP 187, ARG 188, GLN 189
2−7.723 (THR 25, HIS 41, GLN 192)1 (THR 25)0THR 24, THR 25, THR 26, LEU 27, HIS 41, THR 45, MET 49, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, GLN 192
g18−6.9101 (GLY 143)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, MET 49, ASN 119, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
e27−8.112 (HIS 41, GLN 192)1 (GLN 189)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, CYS 44, MET 49, PRO 52, TYR 54, GLY 143, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
e28−7.762 (HIS 41, GLN 192)1 (GLN 189)1 (HIS 41)THR 25, THR 26, LEU 27, HIS 41, CYS 44, MET 49, PRO 52, TYR 54, GLY 143, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
5−7.164 (THR 26, GLY 143, SER 144, CYS 145)1 (MET 49)1 (HIS 41)THR 25, THR 26, LEU 27, HIS 41, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, PRO 168, HIS 172, ARG 188, GLN 189, THR 190, GLN 192
4n−7.822 (LEU 141, GLY 143)1 (MET 165)0LEU 27, HIS 41, SER 46, MET 49, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, ALA 191
2a−6.5210 (THR 24, THR 45, THR 46 × 2, GLY 143, GLU 166, ARG 188, THR 190 × 2, GLN 192)1 (THR 25)0THR 24, THR 25, THR 26, HIS 41, CYS 44, THR 45, SER 46, MET 49, ASN 142, GLY 143, SER 144, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190, GLN 192
2y−6.5411 (HIS 41, THR 45, SER 46 × 3, LEU 141, ASN 142, GLY 143, SER 144 × 2, CYS 145)1 (HIS 41)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, THR 45, SER 46, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, GLN 189
2s−6.6701 (MET 165)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, MET 49, TYR 54, ASN 142, GLY 143, CYS 145, HIS 164, MET 165, ASP 187, ARG 188, GLN 189
F3−7.805 (THR 24, THR 25, THR 45 × 2, SER 46)1 (MET 165)2 (HIS 41 × 2)THR 24, THR 25, THR 26, LEU 27, HIS 41, THR 45, SER 46, MET 49, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ARG 188, GLN 189, THR 190
D4−6.8901 (MET 165)1 (HIS 41)THR 25, THR 26, LEU 27, HIS 41, VAL 42, MET 49, LEU 50, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, PRO 168, HIS 172, GLN 189, THR 190, ALA 191
diFA−7.6401 (HIS 41)1 (HIS 41)THR 24, THR 25, THR 26, LEU 27, HIS 41, CYS 44, THR 45, SER 46, MET 49, TYR 54, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 164, MET 165, GLU 166, ASP 187, ARG 188, GLN 189
triFA−8.324 (THR 26, TYR 54 × 2, ASP 187)1 (MET 165)0THR 24, THR 25, THR 26, LEU 27, HIS 41, CYS 44, MET 49, PRO 52, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, ASP 187, ARG 188, GLN 189, THR 190, GLN 192
MY3−6.294 (GLY 143, SER 144 × 2, GLN 189)1 (MET 165)0HIS 41, MET 49, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, HIS 172, PHE 181, VAL 186, ASP 187, ARG 188, GLN 189
N3−8.264 (CYS 145, GLU 166, GLN 189)1 (MET 49)1(HIS 41)THR 25, LEU 27, HIS 41, MET 49, LEU 50, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
GC376−7.805 (HIS 41, PHE 140, HIS 163, GLU 166, GLN 189)1 (ASP 187)0HIS 41, MET 49, TYR 54, PHE 140, LEU 141, ASN 142, GLY 143, SER 144, CYS 145, HIS 163, HIS 164, MET 165, GLU 166, LEU 167, PRO 168, HIS 172, ASP 187, ARG 188, GLN 189, THR 190, ALA 191, GLN 192
1: The binding energies are given as negative values and correspond to the best cluster for each compound. A lower binding energy corresponds to a higher binding affinity. 2: H-bonds were calculated by Pymol. 3: Hydrophobic and pi-pi interactions were calculated by the YASARA structure.
Table 5. Physicochemical properties, lipophilicity, solubility, bioavailability, and druglikeness predictions for FA and its selected derivatives.
Table 5. Physicochemical properties, lipophilicity, solubility, bioavailability, and druglikeness predictions for FA and its selected derivatives.
Physicochemical PropertiesLipophilicity/SolubilityBioavailability and Druglikeness
Compound Name FormulaMW 1 (g/mol)RB 2HBA 3HBD 4Fraction C sp3 5TPSA 6
(Ų)
pKa of Most Basic/Acidic Group 7Log Po/w 8LogS 9Lipinski 10Bioavailability Score 11Druglikeness Score 12
FAC10H10O4194.183420.166.76<0./4.541.51−2.11Yes (0)0.85−0.61
Methyl ferulateC11H12O4208.214410.1855.76<0./9.691.84−2.32Yes (0)0.55−0.76
Ethyl ferulateC12H14O4222.245410.2555.76<0./9.692.2−2.55Yes (0)0.55−0.55
Propyl ferulateC13H16O4236.266410.3155.76<0./9.692.73−2.89Yes (0)0.55−0.34
Butyl ferulateC14H18O4250.297410.3655.76<0./9.693.09−3.12Yes (0)0.55−0.42
Isobutyl ferulateC14H18O4250.296410.3655.76<0./9.693.17−3.24Yes (0)0.55−0.1
Pentyl ferulateC15H20O4264.328410.455.76<0./9.693.63−3.47Yes (0)0.55−0.51
Isopentyl ferulateC15H20O4264.327410.455.76<0./9.693.52−3.47Yes (0)0.55−0.01
Prenyl ferulateC15H18O4262.36410.2755.76<0./9.693.34−3.41Yes (0)0.55−0.49
Hexyl ferulateC16H22O4278.349410.4455.76<0./9.694.17−3.82Yes (0)0.55−0.51
Octyl ferulateC18H26O4306.411410.555.76<0./9.695.25−4.52Yes (0)0.55−0.51
Dodecyl ferulateC22H34O4362.515410.5955.76<0./9.697.42−5.94Yes (0)0.55−0.51
Octadecyl ferulateC28H46O4446.6621410.6855.76<0./9.6810.67−8.08Yes; (1, logP)0.55−0.51
Oleyl ferulateC28H44O4444.6520410.6155.76<0./9.699.74−7.55Yes (1, logP)0.55−0.45
Glyceryl ferulateC13H16O6268.267630.3196.22<0./9.690.53−1.61Yes (0)0.55−0.05
Diglyceryl ferulateC16H22O8342.3411840.44125.68<0./9.70−0.23−1.28Yes (0)0.55−0.11
Tocopheryl ferulateC39H58O5606.8717510.6264.99<0./9.6912.51−10.56No (2, MW, logP)0.171.14
Sitosteryl ferulateC39H58O4590.8811410.7255.76<0./9.6911.61−10.19No (2, MW, logP)0.171.21
D-glucose ferulateC16H20O9356.326950.44145.91<0./9.69−0.87−1.28Yes (0)0.55−0.15
D-galactose ferulateC16H20O9356.326950.44145.91<0./9.69−0.87−1.28Yes (0)0.55−0.15
D-mannose ferulateC16H20O9356.326950.44145.91<0./9.69−0.87−1.28Yes (0)0.55−0.15
D-fructose ferulateC16H20O9356.327950.44145.91<0./9.69−0.56−1.41Yes (0)0.55−0.24
L-arabinose ferulateC15H18O8326.36840.4125.68<0./9.69−0.25−1.5Yes (0)0.55−0.18
D-xylose ferulateC15H18O8326.36840.4125.68<0./9.69−0.25−1.5Yes (0)0.55−0.18
FOS ferulate 1C26H28O12532.49111250.31181.44<0./9.691.71−3.73No (2; MW, HBD)0.170.58
D-lactose ferulateC21H28O15520.4491580.57234.29<0./9.69−2.87−0.79No (3; MW, HBD, HBA)0.17−0.02
D-sucrose ferulateC22H30O14518.47101480.59225.06<0./9.69−1.98−1.27No (3; MW, HBD, HBA)0.170.01
D-maltose ferulateC22H30O14518.47101480.59225.06<0./9.69−3.01−0.69No (3; MW, HBD, HBA)0.170.03
D-cellobiose ferulateC22H30O14518.4791480.59225.06<0./9.69−3.01−0.69No (3; MW, HBD, HBA)0.170.03
Xylobiose ferulateC20H26O12458.4171260.55184.6<0./9.69−1.78−1.24No
(2; HBD, HBA)
0.170.33
FOS ferulate 2C22H30O14518.47101480.59225.06<0./9.69−2.4−1.01No
(3; MW, HBD, HBA)
0.170.27
Galactobiose ferulateC32H38O17694.63141780.44260.59<0./9.69−0.74−2.94No (3; MW, HBD, HBA)0.170.03
Arabinobiose ferulateC31H36O14632.61141450.42199.9<0./9.691.22−3.8No
(2; MW, HBA)
0.170.07
Raffinose ferulateC29H42O18678.631318100.69283.98<0./12.89−3.58−1.03No; (3; MW, HBD, HBA)0.17−0.21
FOS ferulate 3C58H64O281209.12928110.38410.8<0./9.691.47−6.56No
(3; MW, HBD, HBA)
0.170.27
d-mannitol ferulateC16H22O9358.3410960.44156.91<0./9.69−1.32−0.75Yes
(1; HBD)
0.55−0.26
d-sorbitol ferulateC16H22O9358.3410960.44156.91<0./9.69−1.32−0.75Yes (1; HBD)0.55−0.26
d-xylitol ferulateC15H20O8328.319850.40136.68<0./9.69−0.7−1.03Yes (0)0.55−0.26
FA rutinosideC23H32O13516.4991370.61204.83<0./4.07−1.81−1.43No
(3; MW, HBD, HBA)
0.11−0.22
Compound 7aC24H27NO4393.4812410.2956.79−1.12/15.253.96−4.29Yes (0)0.550.81
Compound 13bC22H24ClNO4401.8811410.3256.79−1.89/13.893.5−4.13Yes (0)0.550.92
Compound 4fC29H31NO5473.5614510.2866.02−1.89/13.894.33−4.96Yes (0)0.550.42
Compound 2C21H23NO8S449.4712800.29133.1<0./17.653.63−4.41Yes (0)0.55−0.54
Compound g18C29H30ClF3NO6P611.9715910.2892.9−5.09/12.646.18−6.86No (2; MW, logP)0.170.37
Compound e27C26H22N2O6458.469700.1288.882.31/25.164.43−5.36Yes (0)0.550.59
Compound e28C29H26N2O6498.5311700.1488.882.31/19.295.5−6.11Yes (0)0.550.54
Compound 5C32H34N2O7558.6210720.2898.726.30/9.695.28−6.33Yes (1; MW)0.551.4
Compound 4nC39H38BrNO11776.62171110.23133.151.05/13.507.16−8.44No
(2; MW, HBA)
0.170.6
Compound 2aC26H32O8S2536.6617820.38162.12<0./15.393.44−4.46Yes (1; MW)0.550.84
Compound 2yC23H26O7S2478.5814720.30152.89<0./15.393.34−4.26Yes (0)0.550.45
Compound 2sC18H16O2S2328.455200.1776.9<0./25.904.76−4.95Yes (1; logP)0.550.05
Compound F3C28H25FO6476.4912700.1471.06<0./27.895.68−5.96Yes (0)0.550.02
Compound D4C22H20N2O3S392.479410.0988.161.11/13.724.71−5.1Yes (0)0.55−0.42
DiFAC20H18O8386.357840.10133.52<0./4.542.69−3.79Yes (0)0.560
TriFAC29H26O10534.51101030.17140.98<0./4.544.16−5.46Yes (1; MW)0.560.32
Compound MY3C16H21NO4291.347400.4472.12<0./20.144.32−4.12Yes (0)0.55−0.61
1: Molecular weight; 2: Number of rotatable bonds; 3: Number of hydrogen bond acceptors; 4: Number of hydrogen bond donors; 5: The ratio of sp3 hybridized carbons over the total carbon count of the molecule; 6: Topological polar surface area, as calculated by SwissADME; 7: As calculated by MOLSOFT; 8: The octanol water partition coefficient, as calculated by the program XLOGP through SwissADME; 9: LogS calculated by SwissADME, as a measure of solubility. Based on its value, the compounds are categorized into insoluble (logS < −10), poorly soluble (−10 < logS < −6), moderately soluble (−6 < logS < −4), soluble (−4 < logS < −2), very soluble (−2 < logS < 0), and highly soluble (logS > 0); 10: Lipinski´s rule of 5 sets 5 criteria, the violation 2 or more of which indicates low oral bioavailability of a compound. In the parentheses, the number and description of the criteria violated are given; 11: Probability of a compound to have a bioavailability of more than 10% in rats, given by SwissADME; 12: Druglikeness as calculated by MOLSOFT. Overall, positive values prompt a drug-like compound.
Table 6. Pharmacokinetic and toxicity predictions for FA and its selected derivatives.
Table 6. Pharmacokinetic and toxicity predictions for FA and its selected derivatives.
Compound Name GI Absorption 1BBB 2 Permeant P-gp Substrate 3CYP1A2 Inhibitor CYP2C19 Inhibitor CYP2C9 Inhibitor CYP2D6 Inhibitor CYP3A4 Inhibitor Log Kp 4 (cm/s)LD50 5 (mg/kg)HepatotoxicityCarcinogenicityMutagenicityCytotoxicity
A 6P 7APAPAP
FAHighYesNoNoNoNoNoNo−6.4117720.510.610.960.88
Methyl ferulateHighYesNoNoNoNoNoNo−6.269780.560.670.890.94
Ethyl ferulateHighYesNoYesNoNoNoNo−6.099780.670.730.820.91
Propyl ferulateHighYesNoYesYesNoNoNo−5.89780.60.770.810.87
Butyl ferulateHighYesNoYesYesNoNoNo−5.6396000.60.780.80.86
Isobutyl ferulateHighYesNoYesYesNoNoNo−5.589780.60.70.810.84
Pentyl ferulateHighYesNoYesYesNoNoNo−5.3496000.720.740.770.85
Isopentyl ferulateHighYesNoYesYesNoNoNo−5.4196000.530.690.790.81
Prenyl ferulateHighYesNoYesYesNoNoNo−5.539780.570.680.70.75
Hexyl ferulateHighYesNoYesYesNoNoNo−5.0496000.80.760.720.82
Octyl ferulateHighYesNoYesNoYesYesNo−4.4496000.80.760.720.82
Dodecyl ferulateHighNoNoYesNoYesYesNo−3.2496000.80.760.720.82
Octadecyl ferulateLowNoNoNoYesNoNoNo−1.4596000.80.760.720.82
Oleyl ferulateLowNoNoNoNoNoNoYes−2.196000.80.760.720.82
Glyceryl ferulateHighNoNoNoNoNoNoNo−7.569780.890.830.760.86
Diglyceryl ferulateHighNoNoNoNoNoNoNo−8.559780.90.830.770.83
Tocopheryl ferulateLowNoYesNoNoNoNoNo−1.1250000.740.680.730.81
Sitosteryl ferulateLowNoNoNoNoNoNoNo−1.6696000.740.660.930.79
D-glucose ferulateLowNoNoNoNoNoNoNo−9.0950000.780.80.770.83
D-galactose ferulateLowNoNoNoNoNoNoNo−9.0950000.780.80.770.83
D-mannose ferulateLowNoNoNoNoNoNoNo−9.0950000.780.80.770.83
D-fructose ferulateLowNoNoNoNoNoNoNo−8.8750000.890.840.740.82
L-arabinose ferulateHighNoNoNoNoNoNoNo−8.4750000.760.760.690.85
D-xylose ferulateHighNoNoNoNoNoNoNo−8.4750000.760.760.690.85
FOS ferulate 1LowNoYesNoNoNoNoNo−8.3350000.870.840.770.76
D-lactose ferulateLowNoNoNoNoNoNoNo−11.5150000.750.810.690.72
D-sucrose ferulateLowNoNoNoNoNoNoNo−10.8750000.870.830.770.79
D-maltose ferulateLowNoNoNoNoNoNoNo−11.650000.810.810.790.78
D-cellobiose ferulateLowNoNoNoNoNoNoNo−11.650000.810.810.790.78
Xylobiose ferulateLowNoNoNoNoNoNoNo−10.3650000.770.760.80.75
FOS ferulate 2LowNoNoNoNoNoNoNo−11.17n.a.0.910.840.770.77
Galactobiose ferulateLowNoYesNoNoNoNoNo−11.0650000.830.80.810.79
Arabinobiose ferulateLowNoYesNoNoNoNoNo−9.2950000.770.740.780.76
Raffinose ferulateLowNoNoNoNoNoNoNo−12.9850000.870.860.790.77
FOS ferulate 3LowNoYesNoNoNoNoNo−12.63n.a.0.880.830.760.73
D-mannitol ferulateLowNoNoNoNoNoNoNo−9.4296000.870.860.770.84
D-sorbitol ferulateLowNoNoNoNoNoNoNo−9.4296000.870.860.770.84
D-xylitol ferulateLowNoNoNoNoNoNoNo−8.896000.90.870.770.84
FA rutinosideLowNoNoNoNoNoNoNo−10.7440000.820.860.820.73
Compound 7aHighYesYesYesNoYesYesYes−5.8915000.760.530.690.78
Compound 13bHighYesYesYesYesYesYesYes−6.2712000.830.590.730.7
Compound 4fHighYesYesNoYesYesYesYes−6.1153000.880.560.720.74
Compound 2LowNoYesNoYesYesYesYes−6.4610000.510.660.690.72
Compound g18LowNoYesNoYesYesYesYes−5.6515000.670.650.660.72
Compound e27HighNoNoNoYesYesNoYes−5.9510000.760.520.530.63
Compound e28HighNoNoNoYesYesNoYes−5.4410000.750.510.550.62
Compound 5HighNoYesNoYesYesNoNo−5.966000.860.570.580.57
Compound 4nLowNoNoNoNoNoNoNo−5.9550000.510.520.60.59
Compound 2aLowNoNoNoYesNoYesNo−7.1317720.840.680.810.79
Compound 2yLowNoNoNoYesYesYesNo−6.8510000.760.750.840.79
Compound 2sHighNoNoYesYesYesNoNo−4.9231500.540.550.740.74
Compound F3HighNoYesNoNoYesNoYes−5.1721000.580.680.820.62
Compound D4HighNoNoYesYesYesNoYes−5.3535060.610.590.520.72
DiFAHighNoNoNoNoNoNoNo−6.7511000.520.670.830.63
TriFALowNoNoNoNoYesNoNo−6.6117720.650.510.580.79
Compound MY3HighYesNoYesYesNoNoNo−5.0150000.640.60.610.71
1: Gastrointestinal absorption; 2: Blood brain barrier; 3: P-glycoprotein; 4: Measure of skin permeation. The more negative the value of logKp, the lower the skin permeability indicated. All of the aforementioned pharmacokinetic properties were predicted by SwissADME; 5: Lethal dose, as calculated by ProToxII. Based on this value, chemicals are categorized in 6 toxicity classes. Compounds in this study fall under the three less toxic categories: Class IV: harmful if swallowed (300 < LD50 ≤ 2000); Class V: may be harmful if swallowed (2000 < LD50 ≤ 5000); and Class VI: non-toxic (LD50 > 5000); 6: Prediction for the inactivity (green) or activity (red) of the compound in the respective toxicity category, as given by ProToxII; 7: Probability of this prediction, given by ProToxII. An encouraging indicator is that most of the compounds are not P-glycoprotein or cytochrome inhibitors, meaning that they can be selective Mpro targets, while they also seem to have acceptable skin permeability, taking into consideration that more negative values of logKp are interpreted as lower skin permeability [115]. As far as blood brain barrier (BBB) permeability is concerned, most compounds are not estimated to be able to permeate the BBB. This means that unwanted side-effects in the central nervous system are avoided. Nevertheless, there have been indications that SARS-CoV-2 does cross the barrier, potentially causing brain damage [118], so it is not entirely clear whether BBB permeability is an undesired property.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Antonopoulou, I.; Sapountzaki, E.; Rova, U.; Christakopoulos, P. The Inhibitory Potential of Ferulic Acid Derivatives against the SARS-CoV-2 Main Protease: Molecular Docking, Molecular Dynamics, and ADMET Evaluation. Biomedicines 2022, 10, 1787. https://doi.org/10.3390/biomedicines10081787

AMA Style

Antonopoulou I, Sapountzaki E, Rova U, Christakopoulos P. The Inhibitory Potential of Ferulic Acid Derivatives against the SARS-CoV-2 Main Protease: Molecular Docking, Molecular Dynamics, and ADMET Evaluation. Biomedicines. 2022; 10(8):1787. https://doi.org/10.3390/biomedicines10081787

Chicago/Turabian Style

Antonopoulou, Io, Eleftheria Sapountzaki, Ulrika Rova, and Paul Christakopoulos. 2022. "The Inhibitory Potential of Ferulic Acid Derivatives against the SARS-CoV-2 Main Protease: Molecular Docking, Molecular Dynamics, and ADMET Evaluation" Biomedicines 10, no. 8: 1787. https://doi.org/10.3390/biomedicines10081787

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop