Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a pandemic cause of Corona Virus Disease (COVID-19), that has claimed numerous human lives across the globe. Main protease being the active protein of SARS-CoV-2 requires urgent mitigating effect against the spread of the virus. The therapeutic roles of the active compounds present in ten typical African medicinal plants were investigated in this study. Five active compounds Curcuma longa (Curcumin and Bisdethoxy curcumin), Garcinia kola (kolaviron), Zingiber officinale (Gingerol) and Vernonia amygdalina (Artemisinin) were selected and docked against Main protease through receptor grid generation, protein ligand docking, receptor ligand complex pharmacophore and binding free energy. The results obtained revealed Curcumin had the highest binding score of −8.628 kcal/mol while artermisinin presented the least with −4.123 kcal/mol. The outcome of the pharmacokinetic prediction in this study revealed high transport capacity across the gastrointestinal tract and high blood brain barrier permeability for curcumin, bisdemethoxy curcumin, gingerol and artemisinin. The exemption is gingerol with low LD50 value (250 mg/kg), the LD50 of all active compounds ranged from 2000 to 4228 mg/kg. Adsorption, distribution, metabolism, excretion and toxicity (ADMET) properties exhibited by all compounds portrayed them as non-hepatotoxic, non-cytotoxic, non-mutagenic and non-carcinogenic. The active compounds exhibited drug-likeness features against Main protease of Covid-19.
Keywords: COVID-19, Main protease, Active compounds, Molecular docking, Pharmacophore, Binding free energy
1. Introduction
The Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) as well as Middle East Respiratory Syndrome Coronavirus (MERS-CoV) are member of Coronaviridae family, which affects species ranging from human beings to animals, causing dreadful respiratory diseases [1]. Coronavirus 2019 (COVID-19) emerged from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a pandemic [2] discovered in Wuhan, Hubei province China, where it was spontaneously transferred from animal source like bats as possible sources to human being [[3], [4], [5]]. The infection leads to severe respiratory disease after an incubation period of 2–14 days [6]. The mode of transmission among human beings has been confirmed to be based on contaminated hands, infected surfaces and salivary or airway droplets [7]. The death rate has risen to 5,620,865 with infection cases at 360,578,392 as at 4:37 p.m. CET, January 27, 2022 [8]. When infecting human cells, SARS-CoV-2 attaches itself with angiotensin converting enzyme 2, ACE2 [9,10]. ACE2 functions through the decrease blood pressure by lowering the angiotensin 2 [11,12]. The inflammatory around the lung, through animal research, has been shown as been reversible by improving the expression of ACE2 [13]. Viral proteases enzymes are major drug target; they function essentially, in viral protein maturation through proproteins removal after translation processes in the cytosol of the host cell. SARS-CoV-2 has become medically important coronaviruses because of the resulting health challenges [14,15]. The viral particles of coronavirus contain 4 major structural proteins: spike, membrane, envelope and nucleocapsid protein. Spike is a vital target for virus entrance into human cells via interaction with the ACE2. Nonstructural proteins possess enzymatic activities like proteases alongside RNA polymerase that redirected its activities. The blocking of the enzymatic regulations is useful when developing antiviral drugs against SARS-CoV2. SARS-CoV-2 is a positive-stranded large RNA genome enveloped betacoronavirus consisting approximately 30 kb of encoded proteins [[16], [17], [18]]. One of them, Main protease (3 chymotrypsin-like protease) being a cysteine protease, aids maturation cleavage of repeating amino acid units linked by a peptide bond in the process of virus reprodution [[19], [20], [21]]. Main protease is a homodimer consisting of two promoters: papin-like cyctseine protease (PL pro) and 3 chymotrypsin-like protease (3CLpro) alongside three domains; domain I (residue 8–101), domain II (residue 102–184), and domain III (residue 201–303). Domains I and II, are made up of six antiparallel β-barrels. An antiparallel globular cluster of five α helices forms domain. Domain III is involved in indirect interaction with substrate crucial for enzymatic activity of proteins through the removal of inactive protease [22]. Main protease is an upstream enzyme that involves in the SARS-CoV-2 replication and transcription [23]. The presence of computational model through in-silico screening of potential inhibitory role against the Main protease becomes possible.
Applications of plant extracts in medicine have being historically common due to their effectiveness against numerous infections majorly in Africa [24]. Approximately 70% of medications are synthesized directly or indirectly from the active ingredient present in plant extracts nevertheless the high rate of consumption for health improvement is observed in rural environment [24,25]. Plant extracts are readily available, safe, natural and affordable with limited side effect as compared to synthesized drugs. Plants generally are composed of phytochemical constituents present in different sections ranging from flower, leaf, stem and roots which serves as bioactive compounds responsible for numerous therapeutic role like anti diabetic, anti-inflammatory, antiviral and anti-microbial effects [26]. There are common plants used in Nigeria due to their bioactive compounds and previous infection treatment history. Bitter kola (Garcinia kola) is found majorly in moist forests of Central and Western African countries belonging to the family Guttiferae [27]. The entire parts of the plant are of importance, it is orally applied to alleviate poor health status ranging from erectile dysfunction, cough, gastric problems and high blood pressure. The major biflavonoids present in Garcinia kola is kolaviron responsible for its anti-inflammatory, anti-microbial and wound healing features [28,29]. Bitter leaf (Vernonia amygdalina) is a small shrub of tropical Africa origin belonging to the daisy family. Vernonia amygdalina is major traditional dish in some region in Nigeria due to its nutritional composition. The presence of artemisinin a bioactive terpenoid present in bitter leaf could be responsible for its usefulness in orthodox medication, being responsible for treating chicken pox, stomach ache, measles, pneumonia and cancer [30]. Ginger (Zingiber officinale) and turmeric (Curcuma longa) are used as cuisines and medicinal spices globally [31,32]. They are perennial plants of tropical and subtropical Asia Origin. Zingiber officinale contains gingerol as the bioactive compound of phenolic compound and terpeniods while Curcuma longa, curcumin being its bioactive ingredient of polyphenol making them responsible for various therapeutic functions in aliments like cough, cold, fever, arthritis and cancer [30,31]. This study was aimed at characterizing the potency of these medicinal plants against SARS-CoV2 protease through molecular docking, pharmacophore modelling and ADMET studies.
2. Methods
2.1. Protein preparation
The crystal structure of Main protease (PDB ID: 6LU7) resolution 2.16 Å was retrieved from Protein Data Bank (PDB) repository. The sample was prepared employing protein preparation wizard panel of Glide [33] to assign bond orders, add hydrogen, create disulfide bonds and, fill missing loops and side chains with prime. Water molecules outside 3.0 Å of the heteroatoms were detached and the structure minimized and optimized employing OPLS3 and PROPKA respectively [33,34]. Afterwards, the receptor grid file was created to define the binding pocket of the ligands.
2.2. Ligand preparation
About sixteen compounds from reviews on phytochemistry and medicinal importance of selected herbs commonly used in treating various infections and ailments in Nigeria [30]. Their structures including that of the standard inhibitor also known as (1s,2s)-2-({n-[(Benzyloxy)carbonyl]-L-Leucyl}amino)-1-Hydroxy-3-[(3s)-2-Oxopyrrolidin-3-Yl]propane-1-Sulfonic Acid (K36) were downloaded from the PubChem database were prepared for molecular docking using Ligprep module [35]. Low-energy 3D structures having appropriate chiralities were generated. Possible ionization states for each ligand structure were generated at physiological pH of 7.2 ± 0.2. Also generated were stereoisomers for each ligand by maintaining specified chiralities and varying others.
2.3. Receptor grid generation
Receptor grid generation permits specifying the size and position of the protein's active site for ligand docking. The scoring grid was specified with respect to the co-crystalized ligand (inhibitors N3) applying the receptor grid generation tool of Schrödinger Maestro 12.5. The van der Waals (vdW) radius scaling factor of nonpolar receptor atoms of 1.0 and partial charge cut-off of 0.25 were applied.
2.4. Protein-ligand docking
Molecular docking studies were undertaken with the generated receptor grid file using Glide tool of Schrödinger Maestro 12.5. The ligands already prepared were docked applying standard precision (SP), setting ligand sampling to flexible, with the ligand sampling set to none (refine only). The vdW radius scaling factor and partial charge cut-off for ligand atoms were 0.80 and 0.15 respectively.
2.5. Receptor-ligand complex pharmacophore modelling
A receptor-ligand complex pharmacophore model was developed with PHASE using the first 3 compounds having the top binding affinity towards the target protein. Auto (E-pharmacophore) method was applied; hypothesis set at 7 as the highest number of features being generated, 2.0 as lowest feature to feature distance, 4.0 as lowest feature to feature distance for feature of the same type and donor as vector.
2.6. Binding free energy calculation
The Prime MM-GBSA panel was used to determine the binding free energy for ligand–protein complexes employing the MM-GBSA technology available with Prime [36]. The binding free energy of the protein-ligand complexes was then used to obtain stability of their complexes via Prime MM-GBSA program (Schrödinger suite version 2020–3). Prior to this, the ligands were prepared by ligprep, while the respective proteins were prepared using the protein preparation wizard methods as discussed earlier. The active sites of the proteins were predicted by sitemap. Hence, the compounds were docked with proteins using glide SP docking. With OPLS3 force field selected and VSGB employed as the continuum solvent model, others were set as default [36].1
2.7. Pharmacology parameters
The absorption, distribution, metabolism, excretion and toxicity (ADMET) features of the test compounds were obtained employing in silico integrative model predictions on the SwissADME and PROTOX-II software respectively.
3. Results
3.1. 2D structures of some active compounds present in the medicinal plants
The 2D structures of the five most bioactive constituents of African medicinal plants, namely, curcumin, kolaviron, bisdemethoxycurcumin, gingerol, and artemisinin were modeled and used as ligands for docking studies against Main protease of SARS-CoV-2 (Fig. 1 ). In the molecular docking of the plants active compound against SARS-CoV-2 Main protease, several active compounds exhibited numerous level of binding affinity against the protein of interest, as represented in Table 1 . The binding affinities ranging from −8.628 to −2.236 kcal/mol of the change in Gibbs free energy (ΔG) against SARS-CoV-2 Main protease. Curcumin has the greatest binding affinity with −8.628 kcal/mol. The binding affinity of kolaviron is −7.027 kcal/mol, (1s,2s)-2-({n-[(Benzyloxy)carbonyl]-L-Leucyl}amino)-1-Hydroxy-3-[(3s)-2-Oxopyrrolidin-3-Yl]propane-1-Sulfonic Acid the standard ligand is −6.541 kcal/mol, bis-demethoxy curcumin is −4.975 kcal/mol, gingerol is −4.252 kcal/mol and arteminsin with the least binding affinity of −4.123 kcal/mol among the top five compounds.
Fig. 1.
2D Structures of active compound present in medicinal plants.
Table 1.
The docking score (kcal/mol) of the active compound of medicinal plants against main proteinase of SARS-CoV-2.
Active compound | Main proteinase of SARS-CoV-2 | PubChem ID |
---|---|---|
Curcumin | −8.628 | 969516 |
Kolaviron | −7.027 | 155169 |
(1s,2s)-2-({n-[(Benzyloxy)carbonyl]-L-Leucyl}amino)-1-Hydroxy-3-[(3s)-2-Oxopyrrolidin-3-Yl]propane-1-Sulfonic Acid (standard inhibitor) (K36) |
−6.541 | 118737648 |
Bis-demethoxycurcumin | −4.975 | 5315472 |
Gingerol | −4.252 | 442793 |
Artemisinin | −4.123 | 68827 |
Nimbic acid | −3.965 | 25446 |
Thymoquinone | −3.931 | 10281 |
Paradol | −3.924 | 94378 |
Nimbolide | −3.883 | 12313376 |
Ibuprofen | −3.727 | 3672 |
Geraniol | −3.42 | 637566 |
Beta-Pinene | −3.188 | 14896 |
Citral | −3.061 | 638011 |
Limonene | −2.9 | 22311 |
Allicin | −2.236 | 65036 |
Alpha-Pinene | 6654 |
3.2. Molecular modeling of biological interactions
The molecular interaction among the standard inhibitor (K36), Vernonia amygdaline, Garcinia kola, Zingiber officinale and Curcuma longa with the SARS-CoV-2 Main protease, showing the binding pocket of the enzyme comprising of various amino acid residue as shown in Fig. 2a, Fig. 2b, Fig. 2c, Fig. 2d, Fig. 2e, Fig. 2f . Artemisinin formed hydrogen bond with GLN 189 and GLU 166; the hydrogen bond was formed in gingerol at THR 190 and GLN 189; bis-demethoxycurcumin formed hydrogen bond with THR 26, THR 190, GLN 189 and GLU 166; moreso, curcumin bonded at the position THR 26, GLY 143, GLN 189 and THR 190 with hydrogen; kolaviron formed hydrogen bond also at GLU 166 and GLY 143 while (1s,2s)-2-({n-[(Benzyloxy)carbonyl]-L-Leucyl}amino)-1-Hydroxy-3-[(3s)-2-Oxopyrrolidin-3-Yl]propane-1-Sulfonic Acid (standard K36) binds with hydrogen bond at GLN 189 and HIS 41.
Fig. 2a.
3D representations of SARS-CoV-2 3C-like Protease-kolaviron.
Fig. 2b.
2D and 3D representations of SARS-CoV-2 3C-like Protease-curcumin.
Fig. 2c.
2D and 3D representations of SARS-CoV-2 3C-like Protease-Bisdemethoxycurcumin.
Fig. 2d.
2D and 3D representations of SARS-CoV-2 3C-like Protease-Artemisinin.
Fig. 2e.
2D and 3D representations of SARS-CoV-2 3C-like Protease-Gingerol.
Fig. 2f.
2D and 3D representations of SARS-CoV-2 3C-like Protease-standard inhibitor.
3.3. Adsorption, distribution, metabolism, excretion and toxicity (ADMET) properties
The SwissADMET predictions of lipophilicity, solubility, drug-likeness and oral bioavailability of the selected bio active compounds are presented as Table 2 , the pharmacokinetic features as Table 3 while the protox II predicted toxicity profile are as presented in Table 4 . The water solubility, Log S, of curcumin, kolaviron, bisdemethoxy curcumin and -Gingerol were predicted to be moderately soluble while artemisinin is soluble. Additionally, the Lipohilicity (Log P) are of the range −0.48 for kolaviron to 2.62 for artemisinin. In terms of drug-likeness, the outcome of this study indicated that kolaviron violated three of the Linpinski rules, while curcumin, bisdemethoxy curcumin, gingerol and artemisinin fully obeyed the rules. Additionally, the bioavailability scores of curcumin, bisdemethoxy curcumin, gingerol and artemisinin is 0.55 while that of artemisinin is 0.17 (see Table 5).
Table 2.
SwissADMET prediction outputs of selected active compounds.
AC | Molecular weight | Mean logp (0–3) | Silicos-IT Log SW (−0.7 to +6.0) | Silicos-IT class | Lipinski violations (>500 g/mol) | Veber violations (<140 Å2) | Bioavailability Score (100%) |
---|---|---|---|---|---|---|---|
A | 368.38 | 1.47 | −4.45 | MS | 0 | 0 | 0.55 |
B | 588.52 | −0.48 | −5.76 | MS | 3 | 1 | 0.17 |
C | 308.33 | 2.13 | −4.23 | MS | 0 | 0 | 0.55 |
D | 294.39 | 2.14 | −4.58 | MS | 0 | 0 | 0.55 |
E | 282.33 | 2.62 | −2.03 | S | 0 | 0 | 0.55 |
A = Curcumin, B=Kolaviron, C=Bisdemethoxycurcumin, D = Gingerol, E = Artemisinin, AC = Active compound; S=Soluble; MS = Moderately Soluble.
Table 3.
Pharmacokinetics prediction output of selected active compounds.
AC | GI A | BBBP | Pgp | CYP1A2I | CYP2C19I | CYP2C9I | CYP2D6I | CYP3A4I |
---|---|---|---|---|---|---|---|---|
A | High | No | No | No | No | Yes | No | Yes |
B | Low | No | No | No | No | Yes | No | Yes |
C | High | Yes | No | Yes | No | Yes | No | Yes |
D | High | Yes | No | Yes | No | No | No | No |
E | High | Yes | No | Yes | No | No | No | No |
A = Curcumin; B=Kolaviron; C=Bisdemethoxy curcumin; D = Gingerol; E = Artemisinin AC = Active compound; GIA = GI Absorption; BBB BBB permeant; Pgp = Pgp substrate; CYP1A2I = CYP1A2 inhibitor; CYP2C19I = CYP2C19 inhibitor; CYP2C9I = CYP2C9 inhibitor; CYP2D6I = CYP2D6 inhibitor; CYP3A4I = CYP3A4 inhibitor.
Table 4.
Protoxll-predicated toxicity profile of selected active compound.
AC | H T | C T | IT | MT | CT | LD50 (mg/kg) | PTC |
---|---|---|---|---|---|---|---|
A | Inactive | Inactive | Inactive | Inactive | Inactive | 2000 | 4 |
B | Inactive | Inactive | Inactive | Inactive | Inactive | 2000 | 4 |
C | Inactive | Inactive | Active | Inactive | Inactive | 2560 | 5 |
D | Inactive | Inactive | Inactive | Inactive | Inactive | 250 | 3 |
E | Inactive | Inactive | Inactive | Inactive | Inactive | 4228 | 5 |
A = Curcumin; B=Kolaviron; C-Bisdemethoxy curcumin, D-Gingerol, E− Arteminsinin
HT=Hepatotoxicity; CT=Carcinogenicity; IT=Immunotoxicity, CT=Cytotoxicity; PTC=Predicted Toxicity Class.
Table 5.
Binding free energy calculations of top five hit compounds against main protease.
AC | ΔGBinda | ΔG_Coulombb | ΔG_Covalentc | ΔG_Hbondd | ΔG_Lipoe | ΔG_Packingf | ΔG_vdWg |
---|---|---|---|---|---|---|---|
A | −59.50 | −36.72 | 7.61 | −2.49 | −17.82 | −0.81 | −41.05 |
B | −61.33 | −41.99 | 6.91 | −3.84 | −7.34 | −4.52 | −39.90 |
C | −42.99 | 28.22 | 6.75 | −1.32 | −20.49 | −0.34 | −53.66 |
D | −52.87 | −28.69 | 4.06 | −2.43 | −13.31 | −1.33 | −29.12 |
E | −45.10 | −17.89 | 7.47 | −1.62 | −16.75 | −0.51 | −33.12 |
F | −22.12 | 1.34 | −0.01 | −1.13 | −7.20 | 0 | −26.67 |
AC-Active compound A-Curcumin B-Kolaviron C- (1s,2s)-2-({n-[(Benzyloxy)carbonyl]-L-Leucyl}amino)-1-Hydroxy-3-[(3s)-2-Oxopyrrolidin-3-Yl]propane-1-Sulfonic Acid D-Bis Demethoxy curcumin E-Gingerol F-Artemisinin.
MM-GBSA free energy (kcal/mol) of binding.
Contribution to the MM-GBSA free energy of binding (kcal/mol) from the Coulomb energy.
Contribution to the MM-GBSA free energy of binding (kcal/mol) from hydrogen bonding.
Contribution to the MM-GBSA free energy of binding (kcal/mol) from lipophilic binding.
Contribution to the MM-GBSA free energy of binding (kcal/mol) from packing binding.
Contribution to the MM-GBSA free energy of binding (kcal/mol) from solvent GB binding.
The pharmakokinetic prediction in Table 3 portrayed high transport capacity across the gastrointestinal tract and high blood brain barrier (BBB) for curcumin, bisdemethoxy curcumin, gingerol and artemisinin. However, none of the active compounds is substrate to permeability of the glycoprotein (P-gp). Furthermore, bisdemethoxy curcumin, gingerol and artemisinin are predicted to be able to inhibit CYP1A2; curcumin, kolaviron and bisdemethoxy curcumin are predicted to inhibit CYP2C9 and CYP3A4. Moreover, curcumin, kolaviron, bisdemethoxy curcumin gingerol and artemisinin were predicted not to inhibit CYP2C19 and CYP2D6.
As presented in Table 4, the protoxll-predicated toxicity profile of the compounds showed that curcumin, kolaviron, bisdemethoxy curcumin, gingerol and artemisinin do not tend to be hepatotoxic, carcinogenic, mutagenic and cytotoxic. However, curcumin, kolaviron, gingerol and artemisinin are predicted to have immunotoxic potentials. The exemption of gingerol with low LD50 value (250 mg/kg), the LD50 of the active compound ranged from 2000 to 4228 mg/kg. Moreso, curcumin and kolaviron belong to the acute oral toxicity class 4, bisdemethoxy curcumin and artemisinin to class 5, and gingerol to class 3.
3.4. Receptor-ligand pharmacophore modelling
The active compounds produced pharmacophore models against SARS-CoV-2 Main protease. The model revealed four sorts of characteristics:D: Hydrogen Acceptor, A: Hydrogen Donor, H: hydrophobic, and R: Aromatic ring which are presented in figure below: Kolaviron formed two hydrogen bond donor, two hydrogen bond acceptor and one aromatic ring with the enzyme. Curcumin and bisdemethoxy curcumin uses two hydrogen donor, hydrogen acceptor and aromatic rings; the standard inhibitor requires one hydrogen donor, one hydrogen acceptor and one aromatic ring (see Fig. 3 ).
Fig. 3.
Pharmacophore models of kolaviron, curcumin, Bisdemethoxy curcumin and (1s,2s)-2-({n-[(Benzyloxy)carbonyl]-L-Leucyl}amino)-1-Hydroxy-3-[(3s)-2-Oxopyrrolidin-3-Yl]propane-1-Sulfonic Acid on SARS-CoV-2 Main proteinase.
3.5. Binding free energy calculation
The binding free energy of the protein-ligand complexes was employed to determine the stability of their complexes via Prime MM-GBSA program (Schrödinger suite version 2020–3) (see Fig. 4 ).
Fig. 4.
Binding free energy MMGBSA dG Bind (ΔGbind) versus docking score (kcal/mol).
4. Discussion
The Coronaviruses are the virus with positive-polarity RNA genome, making them to depend less on the host cell during replication. The replication occurs in the cytoplasm of the epithelial cells of the respiratory system and the gastrointestinal system [16,18]. The computational approach of drug design against covid-19 is essential to reduce cost, save time and improve output. NADPH and dTDP-4-dehydro-6-deoxy-l-mannose show a significant interaction in silico with the active site of Mpro, with a binding energy of 8.5 and 8.6 kcal/mol, respectively [37]. This study involved five approaches comprising of protein and ligand preparation, receptor grid generation, protein-ligand docking, receptor ligand pharmacophore, binding free energy and pharmacology parameters. In this study, 5 major bioactive compounds of plant extract in addition with standard ligand were docked against SARS-CoV-2 Main protease, curcumin had the highest docking score of −8.62 kcal/mol, followed by kolaviron with −7.027 kcal/mol as compared with the standard ligand (K36) −6.541 kcal/mol. The presence of flavonoinds and curcumin in Curcuma longa has been shown to be responsible for its chemopreventive and physiological effects in many tumor bioassays and the decreased tumor cell growth [38]. Curcumin has been reported to possess antioxidant, anti-inflammatory and antibacterial properties [38]. In-silico approach recently found curcumin as safe, being able to also interact in molecular mechanisms with proteins [39]. The docking score obtained for curcumin being higher than the standard ligand K36 may inform its level of interaction with the active site of Main protease of Sars-Cov-2 by forming hydrogen bonds [40,41]. The protoxll-predicted toxicity profile of curcumin inferred is non-hepatotoxic, non-cytotoxic, non-mutagenic with the LD50 prediction at 2000 mg/kg body weight suggesting that curcumin have good therapeutic properties with drug-likeness for oral drug development [[41], [42], [43]]. Kolaviron is the active ingredient of Garcinia kola known to possess biflavonoids responsible for its numerous health benefits; the high docking score may account for its interactions with Main protease of covid-19 making it pharmacologically active with numerous pharmacokinetic properties both in-vivo and in-silico [44,45]. Bisdemethoxy curcumin is a derivative of curcumin, composing of polyphenol and possess anti-cancer and hepatoprotective effect in-vivo [46]. Furthermore, the in-silico ADMET studies have predicted its role as antiviral among many therapeutic functions [47]. Gingerol the active ingredient of Zingiber officinale serves as anti-inflammatory agent; gingerol intreacts with main protease of covid-19 on the carboxyl and hydroxyl ends of the chains which can serve as potential drug target [48]. Artemisinin the active component of Vernonia amygdalina and an active anti-malaria component [4,49,50], though had the least docking score, interact with SARS-CoV-2 Main protease on Glu 166 and Gln 189 making it a potential drug target [51].
These six compounds interacted with important active site amino acids residues of the enzyme; curcumin, kolaviron, bisdemethoxy curcumin, gingerol, artemisinin and the standard ligand formed one or more hydrogen bonds with Gln 189. Additionally, curcumin, bisdemethoxy curcumin, gingerol, artemisinin, standard K36 formed hydrogen bond with amino acid GLN 189 [52]. There were hydrogen interaction with curcumin and kolavion on amino acid GLY143, bisdemethoxy curcumin and artemisinin on GLU 166 [53,54]. Curcumin and bisdemethoxy curcumin formed hydrogen bonds on THR26, finally bisdemethoxy curcumin and gingerol interacted with hydrogen bonds at the THR 190 amino acid pocket of the 6LU7 protein of the Main protease of covid 19.
The drug-likeness features which include flexibility, lipophilicity, water solubility, molecular size, plasma protein binding, and saturation of the compound polarity, determine the orally bioavailability of a compound [55,56]. Curcumin, bisdemethoxy curcumin, gingerol and artemisinin possess high water solubility, vital transport factor within the blood, however kolaviron is poorly soluble. Furthermore, curcumin, bisdemethoxy curcumin, gingerol and artemisinin obeyed both Vebers and Lipinski rule of five making them orally active. In Veber's rule, compounds that meet only the two criteria of ≤10 rotatable bonds and polar surface area ≤140 Å are projected to have good oral bioavailability while the Lipinski rule constitutes octanol/water partition coefficient (C logP) ≤ 5, number of hydrogen bond acceptors (HBA) ≤ 10, the criteria of molecular weight (MW) ≤ 500 with an orally active drug not violating beyond one of these criteria [11]. The toxicity profile of active compounds of curcumin, bisdemethoxy curcumin, gingerol and artemisinin are not likely to produce any toxic effect on the Hepatocyte and cytosol. Moreso, of all the active compounds gingerol is the most toxic compound with the LD50 of 250 mg/kg, while other active compounds belonging to oral toxicity class 5 are relatively safe, with LD50 ranging from 2000 to 4228 mg/kg. The pharmacokinetic properties of curcumin, bisdemethoxy curcumin, gingerol and artemisinin include the underlying role offered by drug-metabolising enzymes like cytochrome P-450 thereby inhibiting the metabolism of drugs being substrates of one or more of the enzymes, resulting in certain degrees of drug-drug interaction [54,57,58].
Binding free energy determines the stability of the protein-ligand complexes [58]; as the binding free energy increases, the ligand-bound protein becomes more stable and favorable. Curcumin exhibited the highest stability of Main protease, followed by kolaviron, the standard, bisdemethoxy curcumin, gingerol and artemisinin. The visualization in the scatter plot is thus a validation of how reliable the docking procedure is in predicting the active site of the protein. Moreso, the main donors to the free binding energy are covalent energy, Coulomb energy, lipophilic bonding, hydrogen bonding and van der Waals energy.
5. Conclusion
The potency of the active ingredients from ten medicinal plants common in Southwest Nigeria against Covid-19 virus has been determined. Five out of sixteen active compounds from the ten medicinal plants demonstrated positive inhibitory role against Main protease of Covid-19. These include curcumin, kolaviron, bisdemethoxy curcumin, gingerol and artemisinin. These active compounds recorded various docking scores against main protease of Covid-19 with curcumin being the highest having −8.62 kcal/mol followed by kolaviron with −7.027 kcal/mol, the standard ligand with −6.541 kcal/mol, bisdemethoxy curcumin with −5.641 kcal/mol, gingerol with −4.975 kcal/mol and artemisinin with −4.252 kcal/mol. These five active compounds retain a favorable ADMET profile with none exhibiting any affinity towards cytotoxicity, hepatotoxicity, mutagenicity and carcinogenicity. As a result, these active compounds may be investigated further through experimental studies and possibly developed into novel drugs or supplements for the treatment of Covid-19.
Ethics approval and consent to participate
Not applicable.
Sources of funding
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Consent for publication
Not applicable.
Availability of data and materials
The data that support the findings of this study are available from the corresponding author upon reasonable request.
Funding
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Authors’ contributions
Conception and design of the study: OP and OF. Acquisition of data, analysis and interpretation of data: OP, OF and AE. Drafting the article, revising it critically for important intellectual content: OP, OF and AE. Final approval of the version to be submitted: OP, OF and AE.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
Not applicable.
Footnotes
The authors contributed equally to this work.
References
- 1.Cava C., Bertoli G., Castiglioni I. In silico discovery of candidate drugs against Covid-19. Viruses. 2020;12(4):404. doi: 10.3390/v12040404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zhou F., Yu T., Du R., Fan G., Liu Y., Liu Z., et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Huang L.L., Shen S.P., Yu P., Wei Y.Y. Dynamic basic reproduction number based evaluation for current prevention and control of COVID-19 outbreak in China. Zhonghua Liuxingbingxue Zazhi. 2020;41:466–469. doi: 10.3760/cma.j.cn112338-20200209-00080. [DOI] [PubMed] [Google Scholar]
- 4.Wang Y., Liu M., Gao J. Enhanced receptor binding of SARS-CoV-2 through networks of hydrogen-bonding and hydrophobic interactions. Proc Natl Acad Sci USA. 2020;117:13967–13974. doi: 10.1073/pnas.2008209117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhu Z.B., Zhong C.K., Zhang K.X., Dong C., Peng H., Xu T., Wang A.L., Guo Z.R., Zhang Y.H. Epidemic trend of corona virus disease 2019 (COVID-19) in mainland China. Zhonghua Yufang Yixue Zazhi. 2020;54(6):620–624. doi: 10.3760/cma.j.cn112150-20200222-00163. [DOI] [PubMed] [Google Scholar]
- 6.Ahn D.G., Shin H.J., Kim M.H., Lee S., Kim H.S., Myoung J., Kim B.T., Kim S.J. Current status of epidemiology, diagnosis, therapeutics, and vaccines for novel coronavirus disease 2019 (COVID-19) J Microbiol Biotechnol. 2020;30(3):313–324. doi: 10.4014/jmb.2003.03011. Epub 2020/04/03, PubMed PMID: 32238757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Park M., Cook A.R., Lim J.T., Sun Y., Dickens B.L. A systematic review of COVID-19 epidemiology based on current evidence. J Clin Med. 2020;9(4) doi: 10.3390/jcm9040967. Epub 2020/04/05, PubMed PMID: 32244365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.WHO Health Emergency Dashboard WHO (COVID-19) homepage. 2021. https://covid19.who.int/situation report
- 9.Krishna S., Augustin Y., Wang J., Xu C., Staines H.M., Platteeuw H., et al. Repurposing antimalarial to tackle the COVID-19 pandemic. Trends Parasitol. 2021;37(1):8–11. doi: 10.1016/j.pt.2020.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wu F., Zhao S., Yu B., Chen Y., Wang W., Hu Y. Complete genome characterisation of a novel coronavirus associated with severe human respiratory disease in Wuhan, China. bioRxiv. 2020 doi: 10.1101/2020.01.24.919183. [DOI] [Google Scholar]
- 11.Patel V.B., Zhong J.C., Grant M.B., Oudi G.Y. Role of the ACE2/angiotensin 1–7 Axis of the renin-angiotensin system in heart failure. Circ Res. 2016;118(8):1313–1326. doi: 10.1161/CIRCRESAHA.116.307708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Santos R.A.S., Sampaio W.O., Alzamora A.C., Motta-Santos D., Alenina N., Bader M., et al. The ACE2/angiotensin-(1–7)/MAS Axis of the renin-angiotensin system: focus on angiotensin-(1–7) Physiol Rev. 2018;98(1):505–553. doi: 10.1152/physrev.00023.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zambelli V., Bellani G., Borsa R., Pozzi F., Grassi A., Scanziani M., et al. Angiotensin-(1–7) improves oxygenation, while reducing cellular infiltrate and fibrosis in experimental Acute Respiratory Distress Syndrome. Intensive Care Med Exp. 2015;3(1):44. doi: 10.1186/s40635-015-0044-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cui J., Li F., Shi Z.L. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 2019;17:181–192. doi: 10.1038/s41579-018-0118-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gates B. Responding to covid-19—a once-in-a-century pandemic? N Engl J Med. 2020;382(18):1677–1679. doi: 10.1056/NEJMp2003762. [DOI] [PubMed] [Google Scholar]
- 16.Fehr A., Perlman S. Coronaviruses: an overview of their replication and pathogenesis. Methods Mol Biol. 2015;1282:1–23. doi: 10.1007/978-1-4939-2438-7_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dömling A., Gao L. Chemistry and biology of SARS-CoV-2. Inside Chem. 2020;6:1283–1295. doi: 10.1016/j.chempr.2020.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lu R., Zhao X., Li J., Niu P., Yang B., Wu H., et al. Genomic characterization and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395(10224):565–574. doi: 10.1016/S0140-6736(20)30251-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gorbalenya A.E., Koonin E.V., Donchenko A.P., Blinov V.M. Coronavirus genome: prediction of putative functional domains in the nonstructural polyprotein by comparative amino acid sequence analysis. Nucleic Acids Res. 1989;17(12):4847–4861. doi: 10.1093/nar/17.12.4847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lee H.J., Shieh C.K., Gorbalenya A.E., Koonin E.V., La Monica N., Tuler J., et al. The complete sequence (22 kilobases) of murine coronavirus gene1 encoding the putative proteases and RNA polymerase. Virology. 1991;180(2):567–582. doi: 10.1016/0042-6822(91)90071-I. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ziebuhr J., Snijder E.J., Gorbalenya A.E. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J Gen Virol. 2000;81(4):853–879. doi: 10.1099/0022-1317-81-4-853. [DOI] [PubMed] [Google Scholar]
- 22.Shi J., et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science. 2020;368:1016–1020. doi: 10.1126/science.abb7015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Asghari A., Naseri M., Safari H., Saboory E., Parsamanesh N. The novel insight of SARS-CoV-2 molecular biology and pathogenesis and therapeutic options. DNA Cell Biol. 2020;39:1741–1753. doi: 10.1089/dna.2020.5703. 2020. [DOI] [PubMed] [Google Scholar]
- 24.Abd El-Gani M.M. Traditional medicinal plants of Nigeria: an overview. Agric Biol J N Am. 2016;7(5):220–247. [Google Scholar]
- 25.Akinyemi K.O., Oladapo O., Okwara C.E., Ibe C.C., Fasure K.E. Screening of crude extracts of six medicinal plants used in South-West Nigerian unorthodox medicine for anti-methicillin resistant Staphylococcus aureus activity. BMC Compl Alternative Med. 2005;5:6. doi: 10.1186/1472-6882-5-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Aye M.M., Aung H.T., Sein M.M., Armijos C. A review on the phytochemistry, medicinal properties and pharmacological activities of 15 selected Myanmar. Medicinal Plants Molecules. 2019;24:293. doi: 10.3390/molecules24020293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Peprah T., Keyereh B., Owusu K., Adu-Bredu S. Drought tolerance of Garcinia kola and Garcinia afzelia at the seedling stage. Ghana J For. 2009;25:13–27. [Google Scholar]
- 28.Nwaehujor C.O., Udegbunam R.I., Ode J.O., Udegbunam S.O. Analgesic, anti-inflammatory anti-pyretic activities of Garcinia hydroxybiflavanonol (GB1) from Garcinia kola. J. Korean Soc. Appl. Biol. Chem. 2015;58:91–96. [Google Scholar]
- 29.Tshibangu P.T., Kapepula P.M., Kapinga M.J.K., Lupona H.K., Ngombe N.K., Kalenda D.T., Jansen O., Wauters J.N., Tits M., Angenot L., et al. Fingerprinting and validation of a LC-DAD method for the analysis of biflavanones in Garcinia kola -based antimalarial improvedtraditional medicines. J Pharmaceut Biomed Anal. 2016;128:382–390. doi: 10.1016/j.jpba.2016.04.042. [DOI] [PubMed] [Google Scholar]
- 30.Azeez A.A., Akeredolu O.A., Igata D.F., Akomolede L.A., Ojokunle A.M., Ogundoyin A.A. A review on the phytochemistry and medicinal values of ten common herbs used in Nigeria. Journal of Research in Forestry, Wildlife & Environment. 2020;12(3) [September] [Google Scholar]
- 31.Gang D.R., Ma X. Ginger and turmeric ancient spices and modern medicines in Plant genetics and genomics: crops and models. Genomics of Tropical Crop Plants. 2008;1:299–311. [Google Scholar]
- 32.Ernst M., Durbin K. Center for Crop Diversification, University of Kentucky; Lexington, KY: 2019. Ginger and turmeric. CCD-CP-138.http://www.uky.edu/ccd/sites/www.uky.edu.ccd/files/ginger_tumeric.pdf College of Agriculture, Food and Environment. Available: [Google Scholar]
- 33.Schrödinger release. 2020-3. Schrödinger Software release 2020 Schrödinger; New York, NY, USA: 2020. LLC. [Google Scholar]
- 34.Olsson M.H. Protein electrostatics and pKa blind predictions; contribution from empirical predictions of internal ionizable residues. Proteins: Structure, Function, and Bioinformatics. 2011;79:3333–3345. doi: 10.1002/prot.23113. [DOI] [PubMed] [Google Scholar]
- 35.Schrödinger release. 2020-1: LigPrep 2020 schrödinger. LLC; New York, NY, USA: 2020. [Google Scholar]
- 36.Ojo O.A., Adegboyega A.E., Johnson G.I., Umedum N.L., Onuh K., Adeduro M.N., et al. Deciphering the interactions of compounds from Allium sativum targeted towards identification of novel PTP 1B inhibitors in diabetes treatment: a computational approach. Informatics Med Unlocked [Internet. 2021;26 doi: 10.1016/j.imu.2021.100719. Available from: [DOI] [Google Scholar]
- 37.Sardanelli A.M., Isgr C., Palese L.L. SARS-CoV-2 main protease active site ligands in the human metabolome. Molecules. 2021;26(1409) doi: 10.3390/molecules26051409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Oghenejobo M., Opajobi O.A., Bethel O.U.S., et al. Antibacterial evaluation, phytochemical screening and ascorbic acid assay of turmeric (Curcuma longa) MOJ Bioequiv Availab. 2017;4(2):232–239. doi: 10.15406/mojbb.2017.04.00063. [DOI] [Google Scholar]
- 39.Ya'u Ibrahim Z., Uzairu A., Shallangwa G., Abechi S. Molecular docking studies, drug-likeness and in-silico ADMET prediction of some novel β-Amino alcohol grafted 1,4,5-trisubstituted 1,2,3-triazoles derivatives as elevators of p53 protein levels. Scientific African. 2020;10 [Google Scholar]
- 40.Maurya V.K., Kumar S., Prasad A.K., Bhatt M.L.B., Saxena S.K. Structure-based drug designing for potential antiviral activity of selected natural products from ayurveda against SARS-CoV-2 spike glycoprotein and its cellular receptor. Virus (Tokyo) 2020;31:179–193. doi: 10.1007/s13337-020-00598-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Moraes B.C.Q., Ciofi-Silva C.L., de Paula A.V., et al. SARS-CoV-2 aerosol generation during respiratory equipment reprocessing. Antimicrob Resist Infect Control. 2021;10(82) doi: 10.1186/s13756-021-00955-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Alam M.S., Lee D.U. Synthesis, biological evaluation, drug-likeness, and in silico screening of novel benzylidene-hydrazone analogues as small molecule anticancer agents. Arch Pharm Res (Seoul) 2016;39:191–201. doi: 10.1007/s12272-015-0699-z. [DOI] [PubMed] [Google Scholar]
- 43.Oshevire D.B., Mustapha A., Alozieuwa B.U., Badeggi H.H., Ismail A., Hassan O.N., Ugwunnaji P.I., Ibrahim J., Lawal B., Berinyu E.B. In-silico investigation of curcumin drug-likeness, gene-targets and prognostic relevance of the targets in panels of human cancer cohorts. GSC Biolog Pharm Sci. 2021;14 01): 037–046. [Google Scholar]
- 44.Omole J.G., Ayoka O.A., Alabi Q.K., Adefisayo M.A., Asafa M.A., Olubunmi B.O., Fadeyi B.A. Protective effect of kolaviron on cyclophosphamide-induced cardiac toxicity in rats. J Evid Based Integ Med. 2018;23:1–11. doi: 10.1177/2156587218757649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Erukainure O.L., Salau1 V.F., Chukwuma C.I., Islam Md S. Kolaviron: a biflavonoid with numerous health benefits. Curr Pharmaceut Des. 2021;27(4):490–504. doi: 10.2174/1381612826666201113094303. [DOI] [PubMed] [Google Scholar]
- 46.Kumaravel M., Sankar P., Latha P., Benson C.S., Rukkumani R. Antiproliferative effects of an analog of curcumin in Hep-2 cells: acomparative study with curcumin. Nat Prod Commun. 2013;8(2):183–186. [PubMed] [Google Scholar]
- 47.Patel K.P., Vunnam S.R., Patel P.A., Krill K.L., Korbitz P.M., Gallagher J.P. Transmission of SARS-CoV-2: an update of current literature. Eur J Clin Microbiol Infect Dis. 2020;39(1):1–7. doi: 10.1007/s10096-020-03961-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Rathinavel Thirumalaisamy, Srinivasan Palanisamy, Thangaswamy Selvankumar. Phytochemical 6-Gingerol -A promising Drug of choice for COVID-19. Int. J. Adv. Sci. Eng. 2020;6(4):1482–1489. [Google Scholar]
- 49.Hien T.T., Hanpithakpong W., Truong N.T., Toi P.V., Farrar J. Orally formulated artemisinin in healthy fasting Vietnamese male subjects: a randomized, four-sequence, open-label, pharmacokinetic crossover study. Clin Therapeut. 2011;33(5):644–654. doi: 10.1016/j.clinthera.2011.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Li G., Yuan M., Li H., Deng C., Wang Q., Tang Y. Safety and efficacy of artemisinin-piperaquine for treatment of COVID-19: an open-label, non-randomised and controlled trial. Int J Antimicrob Agents. 2021;57:106216. doi: 10.1016/j.ijantimicag.2020.106216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Uckun F.M., Trieu V.N. Medical-scientific rationale for a randomized, placebo-controlled, Phase 2 study of trabedersen/OT-101 in COVID-19 patients with hypoxemic respiratory failure. Ann. Pulm. Crit. Care Med. 2020;3(1):1–9. [Google Scholar]
- 52.Coelho C., Gallo G., Campos C.B., Hardy L., Würtele M. Biochemical screening for SARS-CoV-2 main protease inhibitors. PLoS One. 2020;15(10) doi: 10.1371/journal.pone.0240079. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cheng S.C., Chang G.G., Chou C.Y. Mutation of Glu-166 blocks the substrate-induced dimerization of SARS coronavirus main prote-ase. Biophys J. 2010;98:1327–1336. doi: 10.1016/j.bpj.2009.12.4272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Johnson T.O., Abolaji A.O., Omale S., Longdet I.Y., Kutshik R.J., et al. Benzo [a] pyrene and Benzo [a] pyrene-7, 8-dihydrodiol-9, 10-epoxide induced locomotor and reproductive senescence and altered biochemical parameters of oxidative damage in Canton-S Drosophila melanogaster. Toxicol Rep. 2021;8:571–580. doi: 10.1016/j.toxrep.2021.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Pires D.E., Blundell T.L., Ascher D.B. pkCSM: predicting small-mole-cule pharmacokinetic and toxicity properties using graph-based signatures. J Med Chem. 2015;58:4066–4072. doi: 10.1021/acs.jmedchem.5b00104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.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. doi: 10.1038/srep42717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Shweta M., Rashmi D. In-vitro ADME studies of TUG-891, a GPR-120 inhibitor using Swiss ADME. J Drug Deliv Therapeut. 2019;9:266–369. [Google Scholar]
- 58.Iwaloye O., Elekofehinti O., Kikiowo B., Fadipe T., Orimoloye A., Ariyo E. Discovery of traditional Chinese medicine derived compounds as wild type and mutant Plasmodium falciparum dihydrofolate reductase inhibitors: induced fit docking and ADME studies. Curr Drug Discov Technol. 2021;18(4):554–569. doi: 10.2174/1570163817999200729122753. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.