Asmaa Abo Elgoud Saida,
Ahmed H. Afifib,
Taha F. S. Alic,
Mamdouh Nabil Samya,
Usama Ramadan Abdelmohsen*ad,
Mostafa A. Fouada and
Eman Zekry Attiaa
aDepartment of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt. E-mail: usama.ramadan@mu.edu.eg; Fax: +20-86-2369075; Tel: +20-86-2347759
bDepartment of Pharmacognosy, Division of Pharmaceutical Industries, National Research Center, Dokki, 12622, Giza, Egypt
cDepartment of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
dDepartment of Pharmacognosy, Faculty of Pharmacy, Deraya University, Universities Zone, 61111 New Minia City, Egypt
First published on 6th October 2021
Chemical investigation of Aptenia cordifolia roots extract, using chromatographic and spectroscopic techniques, resulted in isolation and identification of eight known compounds. The basic ethyl acetate fraction (alkaloidal fraction) afforded O-methylsceletenone, epinine, 4-methoxy phenethylamine, and N-methyl tyramine while, the acidic ethyl acetate fraction (non-alkaloidal fraction) afforded only cis-N-coumaroyl tyramine. Moreover, the petroleum ether fraction afforded capric acid, tricosanol, and a mixture of β-sitosterol & stigma sterol. Upon screening of anti HCV activity of these three fractions, only the basic ethyl acetate fraction had high activity against HCV with an IC50 value equal to 2.4 μg mL−1 which provoked us to carry out structure based in silico virtual screening on the drug targets of HCV of isolated alkaloidal compounds as well as the previously dereplicated alkaloids through metabolomics from the antiviral active fraction. The tortuosamine compound exhibited the strongest binding to the active site of NS3/4A helicase with a binding affinity (−7.1 kcal mol−1) which is very close to the native ligand (−7.7 kcal mol−1).
Medicinal plants contain a variety of compounds that have the potential to treat ailments, particularly infectious disorders. Different studies declared beneficial effects of secondary metabolites derived from medicinal plants against viral diseases. A broad variety of these secondary metabolites, such as alkaloids, coumarins, lignans and polyphenolic compounds affect cellular functions and replication of various infectious viruses. Medicinal plants' history dates back to the origin of human civilization on this planet. Inhibitory activity of medicinal plants extracts against replication of several viruses was reported latterly, such as, herpes simplex virus type2 (HSV-2),8 HIV,9,10 hepatitis B virus (HBV),11,12 emerging viral infections associated with poxvirus, and severe acute respiratory syndrome (SARS) virus.13 Recently, various studies have been conducted to investigate the antiviral activity of a variety of plants. As the methanolic extract of both Terminalia bellerica seeds and Enicostemma axillare showed antiviral activity against hepatitis B.14 On the other hand, Stixis scandens Lour leafe extract showed powerful antiviral activity against porcine epidemic diarrhea virus.15 Moreover, silymarin, epigallocatechin gallate, naringenin,16 phenolic compounds of grape seed and aqueous extract of Eclibta alba leaves exhibited anti HCV activity.14 The alcoholic and/or water soluble extracts of medicinal plants were utilized during furthermost of these antiviral studies. Genus Aptenia is endemic to South Africa and includes four species, which are currently recognized as Aptenia cordifolia L.F. Schwantes, Aptenia geniculiflora L. Bittrich ex Gerbaulet, Aptenia haeckeliana A. Berger Bittrich ex Gerbaulet, and Aptenia lancifolia L. Bolus.17 Aptenia cordifolia, a well-known groundcover, was named heart-leaved ice plant and has only one synonym “Mesembryanthemum cordifolium”.18 So as to provide a complementary therapy for previously existing remedies, the antiviral activity of different fractions (petroleum ether, acidic ethyl acetate and basic ethyl acetate) derived from the total ethanol extract of A. cordifolia roots were examined against HCV by in vitro cells culture using luciferase assay and the cytotoxic effect was accessed by MTT assay. Additionally, we aimed to identify which compound/s in the basic ethyl acetate fraction responsible for the anti HCV activity as well as the mechanism of action by undergoing docking studies on the drug targets of HCV. Although there are many HCV targets, our docking study will focus on the most attractive viral proteins required for HCV replication. These targets are NS5B HCV RNA-dependent RNA polymerase, NS3/4A protease, NS3/4A helicase, and a new allosteric pocket in the HCV NS3–NS4A protein located at the interface between the protease and helicase domains. The compounds used for docking studies were the isolated alkaloids from the active fraction (basic ethyl acetate fraction), as well as, the previously nine dereplicated alkaloids from the basic ethyl acetate fraction through metabolomic analysis.19
Tricosanol alcohol (5 mg) was isolated as white powder and the complete assignment was confirmed by investigation of 1H-NMR, DEPT-Q and ESI-MS analyses. The molecular formula was determined to be C23H48O by ESI-MS that showed a pseudo molecular ion peak at 339 [M–H]−. The compound was identified by comparison of the ESI-MS and NMR data, with the reported data21 and this is the first time for its isolation from family Aizoaceae.
Capric acid was isolated as white powder (4 mg), the molecular formula was confirmed to be C10H20O2 by ESI-MS that showed a molecular ion peak at 172.8 [M]+. The compound was identified by comparison of the ESI-MS and 1H-NMR data with the reported data,22 and it was previously identified by GC-MS in Attalea dubia family Arecaceae, but this is the first time for its isolation from family Aizoaceae.
Cis-N-coumaroyl tyramine (3),which was isolated as white powder (5 mg), HR-ESI-MS showed a pesudo molecular ion peak at m/z 284.128 [M + H]+, (calculated mass at m/z 284.128) corresponding to the molecular formula C17H18NO3 in addition to, a characteristic fragment peak at m/z 147.057 corresponding to loss of [M–C8H11NO]. It was identified by comparison 1H-NMR data with the literature,23 and this is the first report for its isolation from family Aizoaceae.
4-Methoxyphenethylamine (1) was isolated as white powder (3 mg, purity 97%) HR-ESI-MS of compound 1 showed a pseudo molecular ion peak at m/z 152.1066 [M + H]+ (calculated mas 152.1075), corresponding to the molecular formula C9H14NO. From the HR-ESI-MS, 1H-NMR data and from the reported data in literature24 compound 1 was identified as 4-methoxyphenethylamine which was previously isolated from Coryphantha pectinata25 and this is the first report for its isolation from family Aizoaceae.
N-Methyltyramine (2) was isolated as white powder (4 mg). HR-ESI-MS of compound 2 showed a pseudo molecular ion peak at m/z 152.0697 [M + H]+ (calculated mas 152.1031), corresponding to the molecular formula C9H14NO. From ESI-MS, 1H-NMR data and from the data reported in literature26 compound 2 was identified as N-methyltyramine which was previously isolated from Sawa millet seeds26 and this is the first report for its isolation from family Aizoaceae.
O-methylsceletenone (4) was isolated as reddish brown powder (20 mg, purity 98%), the complete assignment of compound 4 was confirmed by investigation of 1H-NMR, DEPT and HR-ESI-MS. Positive HR-ESI-MS of compound 4 showed a quassi molecular ion peak at m/z 258.1462 [M + H]+, (calculated mass at m/z 258.1494) corresponding to the molecular formula C16H20NO2. From the HR-ESI-MS, NMR data and in comparison with the literature,27 compound 4 was identified as O-methyl sceletenone. It was previously isolated from different organs of the investigated plant.28
Epinine (5) was isolated as yellow powder (4 mg, purity 97%) HR-ESI-MS of compound 5 showed a pseudo molecular ion peak at m/z 168.9802 [M + H]+, (calculated mass at m/z 167.1024) corresponding to the molecular formula C9H14NO2. Comparison of the HR-ESI-MS and 1H-NMR data of compound 5, with the data reported in literature29,30 revealed that compound 5 is epinine alkaloid which was previously isolated from Cytisus scoparius Family Leguminosae31 and this is the first report for its isolation from family Aizoaceae.
Before docking of the screened compounds, we assessed our MOE induced docking protocol by re-docking the co-crystallized ligand within the active site of NS5B HCV RNA-dependent RNA polymerase (PDB code: 3H2L), NS3/4A protease (PDB code: 6NZT), NS3/4A helicase (PDB code: 4OKS), and a new allosteric pocket in the HCV NS3–NS4A protein located at the interface between the protease and helicase domains (PDB code: 4B73).
As seen in Fig. 3–6, MOE successfully re-docked the native ligand in its original pose with acceptable RMSD values of 0.28, 0.62, 0.31, and 0.16 Å respectively. The binding free energies of the re-docked ligand, as well as the screened compounds, were summarized in Table 1.
Fig. 3 The top-scoring docked pose of compound 6 to the HCV NS5B polymerase active site (PDB code: 3H2L) as predicted by MOE 2019.01. (A) An overview of the HCV NS5B polymerase active site (B) comparison of modeled binding mode of the co-crystallized ligand (a novel bicyclic dihydro-pyridinone inhibitor) (magenta stick model) and its superposed docking conformation (blue stick model). (C) Detailed binding mode of compound 6 (magenta stick model) displaying hydrogen bonds (black dashed line) and H–π interactions (red dashed line) with the key amino acid residue (blue stick model). (D) 2D depiction of compound 6 binding interactions with the critical amino acid residue. |
Fig. 4 The top-scoring docked pose of compound 8 to the HCV NS3/4A protease active site (PDB code 6NZT) as predicted by MOE 2019.01. (A) An overview of the HCV NS3/4A protease active site (B) comparison of modeled binding mode of the co-crystallized ligand Voxilaprevir (magenta stick model) and its superposed docking conformation (cyan stick model). (C) Detailed binding mode of compound 8 (green stick model) displaying H–π bond (red dashed line) with the key amino acid residue (cyan stick model). (D) 2D depiction of compound 8 binding interactions with the key amino acid residue. |
Fig. 5 The top-scoring docked pose of compound 6 to the HCV NS3 helicase active site (PDB code 4OKS) as predicted by MOE 2019.01. (A) An overview of the HCV NS3 helicase active site (B) comparison of modeled binding mode of the co-crystallized ligand (magenta stick model) and its superposed docking conformation (blue stick model). (C) Detailed binding mode of compound 6 (magenta stick model) displaying hydrogen bonds (black dashed line) with the key amino acid residue (blue stick model). (D) 2D depiction of compound 6 binding interactions with the key amino acid residues. |
Fig. 6 The top-scoring docked pose of compound 6 to the allosteric site on the HCV NS3–NS4A protein, located between the protease and helicase domains of the HCV NS3 protein (PDB code: 4B73) as predicted by MOE 2019.01. (A) An overview of the allosteric site on the HCV NS3–NS4A protein (B) comparison of modeled binding mode of the co-crystallized ligand (magenta stick model) and its superposed docking conformation (blue stick model). (C) Detailed binding mode of compound 6 (green stick model) displaying hydrogen and ionic bonds (black dashed line) with the key amino acid residue (blue stick model). (D) 2D depiction of compound 6 binding interactions with the key amino acid residue. |
Ligand | Binding affinity ΔG (kcal mol−1) | |||
---|---|---|---|---|
Polymerase | Protease | Helicase | Protease–helicase allosteric | |
Native ligand (RMSD) | −9.3 (0.28) | −11.3 (0.62) | −7.7 (0.31) | −9.4 (0.16) |
1 | −5.1 | −4.8 | −4.7 | −5.5 |
2 | −5.1 | −4.8 | −4.9 | −5.7 |
3 | −5.3 | −5.0 | −5.1 | −5.9 |
4 | −6.6 | −5.9 | −6.5 | −7.0 |
5 | −5.2 | −4.8 | −5.3 | −5.7 |
6 | −7.4 | −6.7 | −7.1 | −7.9 |
7 | −6.1 | −5.6 | −5.7 | −6.7 |
8 | −7.1 | −6.9 | −6.0 | −7.4 |
9 | −6.3 | −6.4 | −5.7 | −7.3 |
10 | −6.4 | −5.9 | −6.0 | −7.2 |
11 | −7.1 | −6.4 | −6.4 | −7.1 |
12 | −6.8 | −6.9 | −6.4 | −7.3 |
Among the screened compounds, compound 6 showed the highest binding affinity (7.4 kcal mol−1) to the active site of NS5B HCV RNA-dependent RNA polymerase. Fig. 3C demonstrated that compound 6 binds within the palm I region, where the phenyl ring makes three H–π interactions with three important amino acid residues (Tyr 415, Met 414 and Cys 366) in addition to two important hydrogen bonds made by NH atom to two key amino acid residues (Tyr 415 and Ser 367). These binding interactions explain the strong binding affinity of compound 6 to the active site of NS5B HCV RNA-dependent RNA polymerase (Table 2).
Ligand | Binding affinity (ΔG in kcal mol−1) | Interaction parameters | |||
---|---|---|---|---|---|
Interaction | AA residue | δ (Å) | E (kcal mol−1) | ||
Native ligand | −9.3 | H-Donor | Asp 318 (side chain) | 2.71 | −7.2 |
H-Acceptor | Ser 556 (side chain) | 3.11 | −1.0 | ||
H-Acceptor | Asn 291 (side chain) | 2.88 | −3.5 | ||
H–π | Met 414 (side chain) | 4.77 | −0.5 | ||
6 | −7.4 | H-Donor | Tyr 415 (side chain) | 3.10 | −0.2 |
H-Acceptor | Ser 367 (side chain) | 3.40 | −0.2 | ||
H–π | Tyr 415 (side chain) | 4.59 | −0.6 | ||
H–π | Met 414 (side chain) | 4.81 | −0.3 | ||
H–π | Cys 366 (side chain) | 4.23 | −0.2 |
Compounds 6, 8 and 12 demonstrated the best binding affinities (−6.7 and −6.9 and −6.9 kcal mol−1, respectively), among the screened compounds, to the active site of NS3/4A protease. Fig. 4C revealed that the phenyl ring of compound 8 binds within protease active site via a single H–π interaction with amino acid residue (Lys 1136). This binding interaction elucidates the weak binding affinity of compound 8 to the active site of NS3/4A protease (Table 3). Therefore, NS3/4A HCV protease protein is less likely to be a considerable target for our screened compounds.
Ligand | Binding affinity (ΔG in kcal mol−1) | Interaction parameters | |||
---|---|---|---|---|---|
Interaction | AA residue | δ (Å) | E (kcal mol−1) | ||
Native ligand | −11.3 | H-Donor | Arg 1155 (backbone) | 2.82 | −4.6 |
H-Donor | His 1057 (side chain) | 3.01 | −7.5 | ||
H-Donor | Ala 1157 (backbone) | 2.88 | −4.3 | ||
H-Acceptor | Gly 1137 (backbone) | 3.00 | −1.1 | ||
H-Acceptor | Gly 1137 (backbone) | 3.02 | −2.8 | ||
H-Acceptor | Ala 1157 (backbone) | 2.95 | −3.7 | ||
H–π | His 1057 (side chain) | 3.68 | −1.6 | ||
8 | −6.9 | H–π | Lys 1136 (side chain) | 4.36 | −0.6 |
As depicted in Fig. 5C, compound 6 binds strongly to the active site of NS3/4A helicase with binding affinity (−7.1 kcal mol−1) very close to the native ligand (−7.7 kcal mol−1). This strong binding affinity could be explained by two reasons. Firstly, compound 6 binds mainly through two strong hydrogen bonds to the key amino acid residues (Asp 496 and Glu 493) as well as a strong ionic bond to Glu 493 (Table 4). Secondly, the alkylamine side chain of compound 6 extended deeply in the lipophilic pocket of helicase active site which also increases the binding affinity of this compound to the target helicase protein. Consequently, NS3/4A helicase protein is more likely to be a potential target for compound 6, among our screened compounds.
Ligand | Binding affinity (ΔG in kcal mol−1) | Interaction parameters | |||
---|---|---|---|---|---|
Interaction | AA residue | δ (Å) | E (kcal mol−1) | ||
Native ligand | −7.7 | H-Donor | Trp 501 (backbone) | 2.81 | −2.3 |
H-Acceptor | Gly 255 (backbone) | 2.91 | −3.5 | ||
H-Acceptor | Gly 255 (backbone) | 2.92 | −4.6 | ||
H-Acceptor | Thr 269 (side chain) | 3.52 | −1.9 | ||
6 | −7.1 | H-Donor | Asp 496 (side chain) | 3.35 | −0.7 |
H-Donor | Glu 493 (side chain) | 2.91 | −13.1 | ||
Ionic | Glu 493 (side chain) | 2.91 | −5.1 |
Compound 6 displayed the highest binding affinity (−7.9 kcal mol−1) to the allosteric site of the HCV NS3–NS4A protein. As summarized in Table 5, compound 6 binds to the curial amino acid residue Asp 81 forms two bonds, one hydrogen bond and one ionic bond. Fig. 6C and D showed the 3D and 2D depiction of compound 6 within the allosteric site of the HCV NS3–NS4A protein displaying hydrogen and ionic bonds with the key amino acid residue (Asp 81). Subsequently, the new allosteric site of the HCV NS3–NS4A protein is considered as a plausible target for compound 6.
Ligand | Binding affinity (ΔG in kcal mol−1) | Interaction parameters | |||
---|---|---|---|---|---|
Interaction | AA residue | δ (Å) | E (kcal mol−1) | ||
Native ligand | −9.4 | H-Donor | Asp 79 (backbone) | 3.46 | −0.7 |
H-Donor | Cys 525 (backbone) | 3.00 | −4.2 | ||
H-Donor | H2O (glu 628) | 3.04 | −5.3 | ||
H-Donor | Leu 517 (backbone) | 2.99 | −1.1 | ||
H-Acceptor | Cys 525 (backbone) | 2.97 | −4.8 | ||
6 | −7.9 | H-Donor | Asp 81 (side chain) | 2.97 | −4.9 |
Ionic | Asp 81 (side chain) | 3.47 | −2.0 |
According to our molecular docking simulation study, compound 6 was the most promising anti-HCV candidate among the twelve members of the alkaloidal fraction of A. cordifolia roots as it gives an excellent binding affinity; specifically against NS3/4A helicase and a new allosteric pocket in the HCV NS3–NS4a protein located at the interface between the protease and helicase domains with a relative good binding energy and distance values from the amino acids of the active binding site compared with the native ligands in these proteins.
The ADME analysis has revealed that all the studied compounds show no violations toward Lipinski's rule and hence prove acceptable drug-likeness and pharmacokeintic properties with no potential toxicity (Tables 6 and 7).
Molecule | Hydrogen bond donors | Hydrogen bond acceptors | Number of rotatable bonds | MlogP | Molecular weight | Number of violations |
---|---|---|---|---|---|---|
1 | 1 | 2 | 3 | 1.53 | 151.21 | 0 |
2 | 2 | 2 | 3 | 1.53 | 151.21 | 0 |
3 | 1 | 2 | 3 | 1.83 | 165.23 | 0 |
4 | 0 | 3 | 2 | 2.00 | 257.33 | 0 |
5 | 3 | 3 | 3 | 0.92 | 167.21 | 0 |
6 | 1 | 4 | 6 | 2.27 | 326.44 | 0 |
7 | 1 | 3 | 1 | 1.75 | 243.31 | 0 |
8 | 0 | 3 | 3 | 2.68 | 275.39 | 0 |
9 | 0 | 3 | 2 | 2.08 | 259.35 | 0 |
10 | 1 | 3 | 4 | 2.08 | 261.37 | 0 |
11 | 0 | 3 | 5 | 2.24 | 273.38 | 0 |
12 | 0 | 3 | 5 | 2.33 | 275.39 | 0 |
Molecule | Bioavilability score | GI absorption | BBB permeation | Pgp substrate | PAINS alerts | Synthetic accessibility |
---|---|---|---|---|---|---|
1 | 0.55 | High | Yes | No | 0 | 1.00 |
2 | 0.55 | High | Yes | No | 0 | 1.00 |
3 | 0.55 | High | Yes | No | 0 | 1.00 |
4 | 0.55 | High | Yes | No | 0 | 3.32 |
5 | 0.55 | High | Yes | No | 1, catechol | 1.09 |
6 | 0.55 | High | Yes | Yes | 0 | 3.32 |
7 | 0.55 | High | Yes | No | 0 | 3.25 |
8 | 0.55 | High | Yes | No | 0 | 2.99 |
9 | 0.55 | High | Yes | No | 0 | 2.66 |
10 | 0.55 | High | Yes | No | 0 | 1.80 |
11 | 0.55 | High | Yes | No | 0 | 3.02 |
12 | 0.55 | High | Yes | No | 0 | 1.95 |
Footnote |
† Electronic supplementary information (ESI) available. See DOI: 10.1039/d1ra06139a |
This journal is © The Royal Society of Chemistry 2021 |