Kousar 2020
Kousar 2020
Research Article
Phytochemicals from Selective Plants Have Promising
Potential against SARS-CoV-2: Investigation and Corroboration
through Molecular Docking, MD Simulations, and
Quantum Computations
Received 6 June 2020; Revised 4 August 2020; Accepted 12 August 2020; Published 13 October 2020
Copyright © 2020 Kafila Kousar et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Coronaviruses have been reported previously due to their association with the severe acute respiratory syndrome (SARS). After
SARS, these viruses were known to be causing Middle East respiratory syndrome (MERS) and caused 35% evanescence amid
victims pursuing remedial care. Nowadays, beta coronaviruses, members of Coronaviridae, family order Nidovirales, have
become subjects of great importance due to their latest pandemic originating from Wuhan, China. The virus named as human-
SARS-like coronavirus-2 contains four structural as well as sixteen nonstructural proteins encoded by single-stranded
ribonucleic acid of positive polarity. As there is no vaccine available to treat the infection caused by these viruses, there is a dire
need for taking necessary steps against this virus. Herein, we have targeted two nonstructural proteins of SARS-CoV-2, namely,
methyltransferase (nsp16) and helicase (nsp13), respectively, due to their substantial activity in viral pathogenesis. A total of
2035 compounds were analyzed for their pharmacokinetics and pharmacological properties. The screened 108 compounds were
docked against both targeted proteins and were compared with previously reported known compounds. Compounds with high
binding affinity were analyzed for their reactivity through DFT analysis, and binding was analyzed using molecular dynamics
simulations. Through the analyses performed in this study, it is concluded that EryvarinM, Silydianin, Osajin, and Raddeanine
can be considered potential inhibitors for MTase, while TomentodiplaconeB, Osajin, Sesquiterpene Glycoside, Rhamnetin, and
Silydianin for helicase after these compounds are validated thoroughly using in vitro and in vivo protocols.
1. Introduction agents for SARS-CoV, so the use of face mask, hand washing,
and careful disposal of medical equipment and other
Coronaviruses have become a source of multiple systematic materials infected with nasal secretions are preliminary pre-
infections in various animals [1]. These viruses have previ- ventive measures which must be followed to avoid the spread
ously caused respiratory tract infections in humans, includ- of the pandemic virus [3]. The initial clinical manifestations
ing severe acute respiratory syndrome and the Middle East in the SARS-CoV infected patients observed were fever,
respiratory syndrome [2]. As there are no specific therapeutic cough, headache, myalgia, diarrhoea, dyspnea, leucopenia,
2 BioMed Research International
lymphopenia, thrombocytopenia, hypoxaemia, pulmonary They provide an operative platform, where scientists can ana-
infiltration, and disturbed hepatic and renal function leading lyze a wide range of biological phenomena, different pathways,
to death in some individuals [4]. and molecular interactions. These methods are primarily cost-
The genome of SARS-CoV is 30 kb encoding 27 proteins effective and consist of authentic methods which predict the
and has 14 ORFs. These proteins are further classified as results with the highest accuracy [21, 22].
structural proteins and nonstructural proteins. The structural The present study aims at the in silico analysis of two
proteins like nucleocapsid protein (N), pike surface glycopro- nonstructural proteins of novel coronavirus (SARS-CoV-2),
tein (S), a small envelope protein (E), and matrix protein (M) i.e., helicase (nsp13) and methyltransferase (nsp16). The
are known to play a role in binding to host receptors. The study is performed opting standard protocols of computer-
genome of the virus encodes 16 nonstructural proteins like aided drug discovery, as previously mentioned in a series of
Helicase (nsp13), RNA-dependent RNA polymerase publication [23–35]. The analyses performed included
(nsp12), Papain-like protease (nsp3), main protease (nsp5) ADMET analysis of phytochemicals, molecular docking of
also known as 3C-like protease (3CLpro), and 2 ′ O methyl- phytochemicals with targeted proteins of a novel coronavi-
transferase (nsp16). The nonstructural proteins are likely to rus, binding analysis through molecular dynamics (MD)
be involved in transcription, replication, and pathogenesis simulations, and reactivity analysis through density func-
and play a vital role in the life cycle of the pathogen [5–9]. tional theory (DFT) calculations.
Owing to the critical role of nonstructural proteins of the
virus in virulence, these proteins are potential targets for 2. Materials and Methods
antiviral drugs [9].
Helicase of coronavirus is a member of superfamily-I Herein, the MTase and helicase of the novel coronavirus were
helicase comprising of seven conserved regions. This protein targeted for the identification of candidate inhibitors; thus, a
winds down partially duplex ribonucleic acid in a 5 ′ to 3 ′ series of analyses were performed, and the flowchart is shown
way to make it open [10]. The helicase (nps13) of SARS- in Figure 1.
CoV belongs to superfamily 1 (SF1) of the six helicase super-
families; the division is based upon various specified regions. 2.1. Obtaining the Phytochemicals. The datasets comprising
This enzyme can open up both ribo- and deoxyribonucleic of a wide range of phytochemicals (2035) were collected by
acid duplexes in 5 ′ to 3 ′ direction [11]. The associated using two different databases [36, 37] and also have been
NTPase action can target all-natural nucleotides and deoxy- reported previously in [35] (Table S1). The set comprised
nucleotides as substrates [12]. It is also reported that SARS- of a variety of phytochemical groups, i.e., 322 alkaloids, 113
CoV-nsp12 can improve the helicase action of SARS-nsp13 terpenoids, 105 aurones, 101 chalcones, 378 flavonoids, 211
by cumulative action on nucleic acid (dsRNA or dsDNA) lignans, 255 carboxylic acids, 301 polyphenols, and 249
unwinding by 2-folds [13]. quinones [35]. The 3D structures for the phytochemicals
There are almost a dozen conserved motifs in SF1 heli- were obtained from PubChem (https://pubchem.ncbi.nlm
cases, those involved in direct binding with nucleic acid tri- .nih.gov/). The pharmacological and pharmacokinetic
phosphates and nucleic acids. Two motifs known as Walker properties based on parameters of ADMET (Absorption,
A and B are common in all of members of this super family. Distribution, Metabolism, Excretion and Toxicity) were
By investigating the structure of the enzyme, it is revealed evaluated with the help of the SwissADME web server [38]
that the two RecA-like domains (1A and 2A) constitute the and PreADMET server [39], as reported in [23, 26, 28, 29,
catalytic site of SF1 helicases. This enzyme is known to be 31]. SwissADME server was utilized for the determination
engaged in synthesis of viral RNA and also manipulates the of ADME properties of the phytochemicals, while
DNA of host [13–15]. 2 ′ -O-MTase (2’O methyltransferase) PreADMET was utilized to assess the druglikeness features
of coronaviruses is highly conserved and is known to play and toxicity level of the drug. The structural file (.SDF) of
an indispensable role in viral replication and evasion from the 2035 phytochemicals was utilized for the prediction
innate immunity [16]. It is also reported that the methyl- purposes.
transferases are vital for viral replication in cell cultures.
Another protein nsp10 is also crucial for the proper function- 2.2. Homology Modelling. The crystal structure of MTase was
ing of MTase activity in the host cells for proliferation [17, available at RCSB with PDB ID: 6W61; however, helicase
18]. Due to the conserved nature of this enzyme and its role structure was unavailable. For protein structure prediction
in virulence, this enzyme is an ideal target for potential anti- and homology modelling, the polypeptide sequence of
viral agents and vaccines against SARS-CoV-2, SARS-CoV, SARS-CoV-2 (GenBank ID: QHD43415.1) was used, com-
MERS-CoV, or other RNA and DNA viruses [19]. prising 7096 residues. For mapping helicase sequence, multi-
Phytochemicals have been known from ancient times for ple sequence alignment was performed between targeted
their immense potential and beneficial properties against sev- sequence and SARS-CoV sequence (UniProt Acc ID:
eral infectious diseases and health-related complications [20]. P0C6X7). Later on, homology modelling was performed.
The phytochemicals constitute magnificent potentials, specifi- PSI-BLAST method was performed for template structure
cally antiviral properties, and can be used in treating viral which was homologous to the helicase. Thus, it was observed
infections. With time, in silico approaches are gaining much that the sequence of helicase showed 96.08% similarity with
attention around the world for their advanced strategies and 6YJT. The Modeller 9.18 was used to perform the homology
effective techniques related to the field of medical sciences. modelling with help of 6YJT structure [40]. Opting
BioMed Research International 3
Protein structure
Phytochemicals retrival and Binding analysis Reactivity
collection and initial prediction through MD analysis through
screening through through simulations DFT
ADMET homology
modelling
homology modelling, 100 models were predicted and evalu- Table 1: Grid box dimensions for receptors (Å3).
ated based on Discrete Optimized Protein Energy (DOPE)
Receptor Grid box dimensions
score. The structure quality assessment was performed by
the Ramachandran Plot using RAMPAGE tool [41]. For MTase 26 × 20 × 18
identifying pocket, POVME 2.0 was used [42]. Helicase 20 × 18 × 18
Figure 2: Predicted 3D model for helicase. Red depicts α-helices; β-purple depicts the strands while green depicts random coil.
180
0
Ψ
–180
–180 0 180
Table 2: Results of molecular docking of Remdesivir, Prulifloxacin, and Nelfinavir with MTase of SARS-CoV-2.
Sr. no Compound name Binding affinity (kcal/mol) K i value (μM) Interactions with MTase of SARS-CoV-2
suggest host switches. SARS-CoV switched hosts from herein, another set of phytochemicals has been used to target
Rhinolophid bats to palm civets and human [54]. The evolu- MTase and helicase.
tionary studies also showed that bats also host MERS-like The criteria for evaluating the compounds based on their
virus suggesting bats as MERS-CoV source; infecting ADMET profiles was “Violations from Lipinski’s rule = Zero;
humans had evidence that camels might also be its likely Soluble = High/Very High; Absorption in Gastrointestinal
source [55]. These viruses can transmit to humans, directly (GI) tract = Efficient or Moderate; Blood-brain barrier
or indirectly [56]. (BBB) permeability = No; and Toxicity/carcinogenicity =0”
Currently, there is no suitable vaccine available which can [58]. Through the screening criteria for their ADMET prop-
be used to cure infections caused by the novel coronavirus erties, 108 phytochemicals out of 2035 qualified for perform-
currently named as COVID-19. It is highly needed to design ing the further analysis (Table S2). These selected
a drug or a vaccine to treat this virus as well as to reduce the phytochemicals were prepared for molecular docking with
viral deportment so that its spread could be limited. Repur- our targeted proteins.
posing of drugs is a remarkable approach to opt antiviral
compounds as COVID-19 drug candidates [57]. Recently, a 3.1. Structure Evaluation for Helicase. The structure of heli-
study has been reported targeting the main protease of case was modelled using homology modelling due to unavail-
SARS-CoV-2, using a set of phytochemicals [30]. However, ability of X-ray crystallographic structure. As the predicted
6 BioMed Research International
Table 3: Results of molecular docking of Remdesivir, Prulifloxacin, and Nelfinavir with helicase of SARS-CoV-2.
Sr. no Compound name Binding affinity (kcal/mol) K i value (μM) Interactions with helicase of SARS-CoV-2
model with the lowest DOPE score was used further, it was residues in helicase model, it was observed that 98.0% resi-
observed to be -24913.17, and the predicted model is shown dues were in the favoured region, and 1.9% residues were in
in Figure 2. the allowed region while only 0.1% residues were in outlier
For quality assessment of the structure, the region (Figure 3).
Ramachandran plot was generated for the predicted model
using RAMPAGE tool [41]. As per the analysis of possible 3.2. Molecular Docking and Binding Stability. Initially,
conformations of φ and ψ angles for individual amino acid Remdesivir, Prulifloxacin, and Nelfinavir were docked with
BioMed Research International 7
Sr. no Compound name Binding affinity (kcal/mol) K i value (μM) Interactions with MTase of SARS-CoV-2
Table 4: Continued.
Sr. no Compound name Binding affinity (kcal/mol) K i value (μM) Interactions with MTase of SARS-CoV-2
H-Bonds
Donor
Acceptor
MTase and helicase, and the binding affinities of these com- A recent paper reported an inhibitor effect of Remdesivir (a
pounds were used as a threshold for screening phytochemi- new antiviral drug) on the growth of SARS-CoV-2 in vitro,
cals (Tables 2 and 3). Based on the results, it was observed and an early clinical trial conducted in SARS-CoV-2 Chinese
that threshold for screening phytochemicals for MTase was patients [61]. When Remdesivir interacted with MTase and
-8.2 kcal/mol, and for helicase, it was -8.1 kcal/mol. Using helicase, it showed -7.0 kcal/mol and -6.8 kcal/mol binding
these thresholds, phytochemicals were screened and further affinity, respectively. Recent studies proposed a few drugs
analyzed for stability and reactivity. that target COVID-19 and suggested these compounds could
Herein, we selected Nelfinavir, Prulifloxacin, and be used to treat COVID-19 [62]. Prulifloxacin gave -7.6 kcal/-
Remdesivir from three different drug repurposing studies mol with MTase and -8.1 kcal/mol with helicase. Nelfinavir, a
and docked them as controls in the present study [59, 60]. repurposed drug, is previously known for its selectively
BioMed Research International 9
Sesquiterpene
3 -8.2 0.961
Glycoside
10 BioMed Research International
Table 5: Continued.
inhibitory properties against HIV protease. This enzyme EryvarinM docked with MTase with a binding affinity of
performs the posttranslational treatment of HIV propep- -8.6 kcal/mol with a K i value of 0.489 μM while forming a
tides. Undeveloped, noninfectious viral particles form in conventional hydrogen bond with ASN6841, ASP6873,
the cells containing this drug [63]. When Nelfinavir was ASN6899, CYS6913, and LYS6968. Formation of pi-cation and
docked against MTase, it showed binding affinity of pi-anion was observed with LYS6844 and ASP6897. Further-
-8.2 kcal/mol, while for helicase, it gave binding affinity value more, along with these interactions, pi-alkyl interactions with
of -6.2 kcal/mol. MET6929 and LEU6898 were observed (Figure 4).
Silydianin docked with binding affinity -8.5 kcal/mol and
3.2.1. Results of Molecular Docking of MTase of SARS-CoV-2. K i value of 0.579 μM while interacting with GLY6911,
All 108 compounds, initially screened through ADMET, ASP6928, CYS6913, and ASN6899, by forming conventional
were docked with MTase of SARS-CoV-2, and the threshold hydrogen bonds. Furthermore, it formed a carbon-
used for screening phytochemicals was -8.2 kcal/mol. Thus, hydrogen bond and pi-acceptor-acceptor interactions with
by applying the threshold, it was observed that only four MET6929 and ASP6897, while pi-alkyl and alkyl interactions
compounds passed the threshold which were EryvarinM, with LEU6898, PHE6947, and LYS6944 were observed.
Silydianin, Osajin, and Raddeanine and showed similar or Osajin and Raddeanine docked with binding affinity
better results than those of Remdesivir, Prulifloxacin, and -8.2 kcal/mol and K i value of 0.961 μM. Osajin formed a con-
Nelfinavir (Table 4). ventional hydrogen bond with ASP6928; while Raddeanine
BioMed Research International 11
H-Bonds
Donor
Acceptor
3.00
2.50
2.00
1.50
1.00
0.50
0.00
0 5000 10000 15000 20000 25000 30000 35000 40000 45000 50000
TomentodiplaconeB Rhamnetin
Osajin Silydianin
Sesquiterpene glycoside
(a)
Rg graph for MTase complexes at 300 K
3.00
2.50
2.00
1.50
1.00
0.50
0.00
0 5000 10000 15000 20000 25000 30000 35000 40000 45000 50000
EryvarinM Osajin
Silydianin Raddeanine
(b)
Figure 6: MD simulations based Rg graphs of complexes for strongly binding phytochemicals: (a) complexes of helicase; (b) complexes of
MTase.
simulations. Average RMSD values are reported in Table 6, of gyration. These values depict high stability, compactness,
while graphs of the radius of gyration are shown in Figure 3. and stable folding of protein tertiary structure, as well as
According to the results shown in Table 6, the RMSD stability in protein-ligand complexes. The changes and fluc-
values were observed to have mere changes, while looking tuations were observed in all complexes and not in any spe-
in complexes of same receptors; however, these values were cific complex. The complexes with less binding affinity
very low, i.e., less than 3.50 Å. Furthermore, the graphs showed a decrease in stability and compactness, as these
shown in Figure 6 depicted fewer fluctuations in the radius compounds were not bound strongly. These trends and
BioMed Research International 13
Table 7: Reactivity of phytochemicals with helicase and MTase depicted by band energy gaps.
Complexes ELUMO (kcal/mol) EHOMO (kcal/mol) Band energy gap (ΔE) (kcal/mol)
MTase-EryvarinM -0.274 -0.391 0.112
MTase-Silydianin -0.236 -0.351 0.115
MTase-Osajin -0.271 -0.400 0.129
MTase-Raddeanine -0.242 -0.375 0.133
Helicase-TomentodiplaconeB -0.280 -0.395 0.116
Helicase-Osajin -0.218 -0.335 0.117
Helicase-Sesquiterpene Glycoside -0.198 -0.321 0.123
Helicase-Rhamnetin -0.303 -0.428 0.125
Helicase-Silydianin -0.118 -0.246 0.128
results are also in accordance with various previously International Concern on 30 January 2020. However, still,
reported results [33, 60]. there exists no remedy, drug, or vaccine for the treatment
of COVID-19. This study provides insights into the mecha-
3.3. Reactivity Studies for Phytochemicals and Targeted nism of selective phytochemicals, when docked against two
Proteins’ Complexes. Reactivity of bound phytochemicals main targets of a novel coronavirus, MTase and helicase, by
with helicase and MTase was analyzed through density func- showing their pharmacological properties, binding and its
tional theory- (DFT-) based computations, which works on stability, and the reactivity. Through analysis, it is concluded
the principles of quantum mechanics and its descriptors. that phytochemicals such as EryvarinM, Raddeanine,
The result showed that the phytochemicals with the broad TomentodiplaconeB, Osajin, Sesquiterpene Glycoside,
range of chemical diversity exhibited good binding interac- Rhamnetin, and Silydianin can be considered as candidate
tions with both the proteins. EryvarinM, Silydianin, Osajin, inhibitors for targeted proteins and as drugs, after their
and Raddeanine, which mainly exhibited best docking results in vitro and in vivo examinations.
for MTase, were further selected for reactivity analysis, while
for helicase, TomentodiplaconeB, Osajin, Sesquiterpene
Glycoside, Rhamnetin, and Silydianin were selected. For
Data Availability
studying reactivity, band energy gap was computed using The data used to support the findings of this study are
molecular orbital energy descriptors, and the results are included within the supplementary information files.
shown in Table 7.
Lower band energy gaps show high reactivity; thus, these
results exhibited in Table 7 exhibited high reactivity of phy- Conflicts of Interest
tochemicals with targeted receptors. The band energy gap
The authors declare that there is no conflict of interest
values for these phytochemicals ranged from 0.112 kcal/mol
to 0.133 kcal/mol and 0.116 kcal/mol to 0.128 kcal/mol, for regarding the publication of this paper.
MTase and helicase, respectively, showing narrow energy
gaps and proving their high reactivity properties. B3LYP Supplementary Materials
function from DFT was applied to analyze the molecular
orbital energies, and it is well established in the literature Table S1: list of 2035 phytochemicals and their plants.
that the lower band energy gap depicts higher reactivity of Table S2: ADMET of Screened 108 Phytochemicals.
compounds. The reason is that band energy gaps are com- (Supplementary Materials)
puted through molecular descriptors, and these descriptors
are responsible for the charges transferred in a chemical References
reaction [64]. These energies can characterize the electro-
[1] S. R. Weiss and S. Navas-Martin, “Coronavirus pathogenesis
philic or nucleophilic nature of a compound. Therefore,
and the emerging pathogen severe acute respiratory syndrome
the screened phytochemicals illustrated the higher reactivity
coronavirus,” Microbiology and Molecular Biology Reviews,
of these phytochemicals, as reported in various previous vol. 69, no. 4, pp. 635–664, 2005.
studies [23–25, 27, 30–33, 35, 52, 65]. [2] C. Chu, V. Cheng, I. Hung et al., “Role of lopinavir/ritonavir in
the treatment of SARS: initial virological and clinical findings,”
4. Conclusion Thorax, vol. 59, no. 3, pp. 252–256, 2004.
[3] Z. Song, Y. Xu, L. Bao et al., “From SARS to MERS, thrusting
The novel coronavirus is causing the COVID-19 worldwide, coronaviruses into the spotlight,” Viruses, vol. 11, no. 1,
a disease which has high morbidity and a significant mortal- p. 59, 2019.
ity rate. The virus, sweeping across the whole world, is pan- [4] J. Sizun, N. Arbour, and P. J. Talbot, “Comparison of immuno-
demic as declared by the World Health Organization, and fluorescence with monoclonal antibodies and RT-PCR for the
the disease was declared as a Public Health Emergency of detection of human coronaviruses 229E and OC43 in cell
14 BioMed Research International
culture,” Journal of Virological Methods, vol. 72, no. 2, pp. 145– [20] C. J. Dillard and J. B. German, “Phytochemicals: nutraceuticals
152, 1998. and human health,” Journal of the Science of Food and Agricul-
[5] J. F.-W. Chan, K.-H. Kok, Z. Zhu et al., “Genomic characteri- ture, vol. 80, no. 12, pp. 1744–1756, 2000.
zation of the 2019 novel human-pathogenic coronavirus iso- [21] X. H. Ma, Z. Shi, C. Tan et al., “In-silico approaches to multi-
lated from a patient with atypical pneumonia after visiting target drug discovery,” Pharmaceutical Research, vol. 27, no. 5,
Wuhan,” Emerging Microbes & Infections, vol. 9, no. 1, pp. 739–749, 2010.
pp. 221–236, 2020. [22] M. Mukhtar, M. Arshad, M. Ahmad, R. J. Pomerantz,
[6] Y. Zhou, Y. Hou, J. Shen, Y. Huang, W. Martin, and F. Cheng, B. Wigdahl, and Z. Parveen, “Antiviral potentials of medicinal
“Network-based drug repurposing for novel coronavirus 2019- plants,” Virus Research, vol. 131, no. 2, pp. 111–120, 2008.
nCoV/SARS-CoV-2,” Cell Discovery, vol. 6, no. 1, pp. 1–18, [23] A. Akhtar, A. Amir, W. Hussain, A. Ghaffar, and N. Rasool,
2020. “In silico computations of selective phytochemicals as poten-
[7] D. E. Gordon, G. M. Jang, M. Bouhaddou et al., “A SARS-CoV- tial inhibitors against major biological targets of diabetes mel-
2 protein interaction map reveals targets for drug repurpos- litus,” Current Computer-Aided Drug Design, vol. 15, no. 5,
ing,” Nature, vol. 583, pp. 1–13, 2020. pp. 401–408, 2019.
[8] C S G of the International, “The species severe acute respira- [24] A. Akhtar, W. Hussain, and N. Rasool, “Probing the pharma-
tory syndrome-related coronavirus: classifying 2019-nCoV cological binding properties, and reactivity of selective phyto-
and naming it SARS-CoV-2,” Nature Microbiology, vol. 5, chemicals as potential HIV-1 protease inhibitors,”
no. 4, p. 536, 2020. Universitas Scientiarum, vol. 24, no. 3, pp. 441–464, 2019.
[9] M. Cascella, M. Rajnik, A. Cuomo, S. C. Dulebohn, and R. Di [25] N. Arif, A. Subhani, W. Hussain, and N. Rasool, “In silico inhi-
Napoli, “Features, evaluation and treatment coronavirus bition of BACE-1 by selective phytochemicals as novel poten-
(COVID-19),” in Statpearls, StatPearls Publishing, 2020. tial inhibitors: molecular docking and DFT studies,” Current
[10] A. O. Adedeji and H. Lazarus, “Biochemical characterization Drug Discovery Technologies, vol. 17, 2019.
of Middle East respiratory syndrome coronavirus helicase,” [26] W. Hussain, M. Ali, M. Sohail Afzal, and N. Rasool, “Penta-
mSphere, vol. 1, no. 5, article e00235, 2016. 1,4-diene-3-one oxime derivatives strongly inhibit the repli-
case domain of tobacco mosaic virus: elucidation through
[11] M. E. Fairman-Williams, U.-P. Guenther, and E. Jankowsky,
molecular docking and density functional theory mechanistic
“SF1 and SF2 helicases: family matters,” Current Opinion in
computations,” Journal of Antivirals & Antiretrovirals,
Structural Biology, vol. 20, no. 3, pp. 313–324, 2010.
vol. 10, no. 3, 2018.
[12] J. A. Tanner, R. M. Watt, Y.-B. Chai et al., “The severe acute
[27] W. Hussain, A. Amir, and N. Rasool, “Computer-aided study
respiratory syndrome (SARS) coronavirus NTPase/helicase
of selective flavonoids against chikungunya virus replication
belongs to a distinct class of 5 ′ to 3 ′ viral helicases,” Journal of
using molecular docking and DFT-based approach,” Struc-
Biological Chemistry, vol. 278, no. 41, pp. 39578–39582, 2003.
tural Chemistry, vol. 1, pp. 1–12, 2020.
[13] A. O. Adedeji, B. Marchand, A. J. te Velthuis et al., “Mecha-
[28] W. Hussain, I. Qaddir, S. Mahmood, and N. Rasool, “In silico
nism of nucleic acid unwinding by SARS-CoV helicase,” PLoS
targeting of non-structural 4B protein from dengue virus 4
One, vol. 7, no. 5, 2012.
with spiropyrazolopyridone: study of molecular dynamics
[14] A. Seybert, A. Hegyi, S. G. Siddell, and J. Ziebuhr, “The human simulation, ADMET and virtual screening,” Virus Disease,
coronavirus 229E superfamily 1 helicase has RNA and DNA vol. 29, pp. 1–10, 2018.
duplex-unwinding activities with 5 ′ -to-3 ′ polarity,” RNA, [29] I. Qaddir, N. Rasool, W. Hussain, and S. Mahmood, “Com-
vol. 6, no. 7, pp. 1056–1068, 2000. puter-aided analysis of phytochemicals as potential dengue
[15] A. Seybert, L. C. Van Dinten, E. J. Snijder, and J. Ziebuhr, “Bio- virus inhibitors based on molecular docking, ADMET and
chemical characterization of the equine arteritis virus helicase DFT studies,” Journal of Vector Borne Diseases, vol. 54, no. 3,
suggests a close functional relationship between arterivirus p. 255, 2017.
and coronavirus helicases,” Journal of Virology, vol. 74, [30] N. Rasool, A. Akhtar, and W. Hussain, “Insights into the
no. 20, pp. 9586–9593, 2000. inhibitory potential of selective phytochemicals against Mpro
[16] Y. Wang, Y. Sun, A. Wu et al., “Coronavirus nsp10/nsp16 of 2019-nCoV: a computer-aided study,” Structural Chemistry,
methyltransferase can be targeted by nsp10-derived peptide vol. 31, no. 5, pp. 1777–1783, 2020.
in vitro and in vivo to reduce replication and pathogenesis,” [31] N. Rasool, A. Ashraf, M. Waseem, W. Hussain, and
Journal of Virology, vol. 89, no. 16, pp. 8416–8427, 2015. S. Mahmood, “Computational exploration of antiviral activity
[17] A. Gorbalenya, S. Baker, R. Baric et al., “Coronaviridae Study of phytochemicals against NS2B/NS3 proteases from dengue
Group of the International Committee on Taxonomy of virus,” Turkish Journal of Biochemistry, vol. 44, no. 3,
Viruses. The species severe acute respiratory syndrome- pp. 261–277, 2019.
related coronavirus: classifying 2019-nCoV and naming it [32] N. Rasool, A. Bakht, and W. Hussain, “Analysis of inhibitor
SARS-CoV-2,” Nature Microbiology, vol. 2020, pp. 03-04, binding combined with reactivity studies to discover the
2020. potentially inhibiting phytochemicals targeting Chikungunya
[18] A. Wu, Y. Peng, B. Huang et al., “Genome composition and viral replication,” Current Drug Discovery Technologies,
divergence of the novel coronavirus (2019-nCoV) originating vol. 17, 2020.
in China,” Cell Host & Microbe, vol. 27, no. 3, pp. 325–328, [33] N. Rasool and W. Hussain, “Three major phosphoacceptor
2020. sites in HIV-1 capsid protein enhances its structural stability
[19] P. Krafcikova, J. Silhan, R. Nencka, and E. Boura, “Structural and resistance against inhibitor: explication through molecular
analysis of the SARS-CoV-2 methyltransferase complex dynamics simulation, molecular docking and DFT analysis,”
involved in RNA cap creation bound to sinefungin,” Nature Combinatorial Chemistry & High Throughput Screening,
Communications, vol. 11, no. 1, pp. 1–7, 2020. vol. 23, 2019.
BioMed Research International 15
[34] N. Rasool, W. Hussain, and S. Mahmood, “Prediction of pro- [50] P. Turner, XMGRACE, version 5.1. 19, Center for Coastal and
tein solubility using primary structure compositional features: Land-Margin Research, Oregon Graduate Institute of Science
a machine learning perspective,” Journal of Proteomics & Bio- and Technology, Beaverton, OR, 2005.
informatics, vol. 10, no. 12, pp. 324–328, 2017. [51] F. Neese, “The ORCA program system,” Wiley Interdisciplin-
[35] N. Rasool, A. Jalal, A. Amjad, and W. Hussain, “Probing the ary Reviews: Computational Molecular Science, vol. 2, no. 1,
pharmacological parameters, molecular docking and quantum pp. 73–78, 2012.
computations of plant derived compounds exhibiting strong [52] N. Rasool, S. Iftikhar, A. Amir, and W. Hussain, “Structural
inhibitory potential against NS5 from Zika virus,” Brazilian and quantum mechanical computations to elucidate the
Archives of Biology and Technology, vol. 61, 2018. altered binding mechanism of metal and drug with pyrazina-
[36] U. A. Ashfaq, A. Mumtaz, T. ul Qamar, and T. J. B. Fatima, midase from Mycobacterium tuberculosis due to mutagenic-
“MAPS Database: medicinal plant activities, phytochemical ity,” Journal of Molecular Graphics & Modelling, vol. 80,
and structural database,” Bioinformation, vol. 9, no. 19, pp. 126–131, 2017.
p. 993, 2013. [53] J. F. Drexler, V. M. Corman, and C. Drosten, “Ecology, evolu-
[37] A. Mumtaz, U. A. Ashfaq, M. T. ul Qamar et al., “MPD3: a use- tion and classification of bat coronaviruses in the aftermath of
ful medicinal plants database for drug designing,” vol. 31, SARS,” Antiviral Research, vol. 101, pp. 45–56, 2014.
no. 11, pp. 1228–1236, 2017. [54] A. Balboni, A. Palladini, G. Bogliani, and M. Battilani, “Detec-
[38] A. Daina, O. Michielin, and V. Zoete, “SwissADME: a free web tion of a virus related to betacoronaviruses in Italian greater
tool to evaluate pharmacokinetics, drug-likeness and medici- horseshoe bats,” Epidemiology and Infection, vol. 139, no. 2,
nal chemistry friendliness of small molecules,” Scientific pp. 216–219, 2011.
Reports, vol. 7, no. 1, article 42717, 2017. [55] C. B. Reusken, B. L. Haagmans, M. A. Müller et al., “Middle
[39] J. V. Cruz, R. B. Serafim, G. M. da Silva et al., “Computational East respiratory syndrome coronavirus neutralising serum
design of new protein kinase 2 inhibitors for the treatment of antibodies in dromedary camels: a comparative serological
inflammatory diseases using QSAR, pharmacophore- study,” The Lancet Infectious Diseases, vol. 13, no. 10,
structure-based virtual screening, and molecular dynamics,” pp. 859–866, 2013.
Journal of Molecular Modeling, vol. 24, no. 9, p. 225, 2018. [56] E. J. Snijder, E. Decroly, and J. Ziebuhr, “The nonstructural
[40] B. Webb and A. Sali, “Protein structure modeling with MOD- proteins directing coronavirus RNA synthesis and processing,”
ELLER,” in Protein Structure Prediction, pp. 1–15, Springer, Advances in Virus Research, vol. 96, pp. 59–126, 2016.
2014. [57] P. Colson, J.-M. Rolain, and D. Raoult, “Chloroquine for the
[41] M. Prisant, J. Richardson, and D. Richardson, “Structure vali- 2019 novel coronavirus SARS-CoV-2,” International Journal
dation by Calpha geometry: phi, psi and Cbeta deviation,” Pro- of Antimicrobial Agents, vol. 55, no. 3, p. 105923, 2020.
teins, vol. 50, article 437450, 2003. [58] A. N. Singh, M. M. Baruah, and N. Sharma, “Structure based
[42] J. D. Durrant, L. Votapka, J. Sørensen, and R. E. Amaro, docking studies towards exploring potential anti-androgen
“POVME 2.0: an enhanced tool for determining pocket shape activity of selected phytochemicals against prostate cancer,”
and volume characteristics,” Journal of Chemical Theory and Scientific Reports, vol. 7, no. 1, article 1955, 2017.
Computation, vol. 10, no. 11, pp. 5047–5056, 2014. [59] Y. Li, J. Zhang, N. Wang et al., “Therapeutic drugs targeting
[43] M. R. Koebel, G. Schmadeke, R. G. Posner, and S. Sirimulla, 2019-nCoV main protease by high-throughput screening,”
“AutoDock VinaXB: implementation of XBSF, new empirical bioRxiv, 2020.
halogen bond scoring function, into AutoDock Vina,” Journal [60] M. T. ul Qamar, S. M. Alqahtani, M. A. Alamri, and L.-
of Cheminformatics, vol. 8, no. 1, p. 27, 2016. L. Chen, “Structural basis of SARS-CoV-2 3CLpro and anti-
[44] G. M. Morris, R. Huey, W. Lindstrom et al., “AutoDock4 and COVID-19 drug discovery from medicinal plants,” Journal of
AutoDockTools4: automated docking with selective receptor Pharmaceutical Analysis, vol. 10, no. 4, pp. 313–319, 2020.
flexibility,” Journal of Computational Chemistry, vol. 30, [61] M. Wang, R. Cao, L. Zhang et al., “Remdesivir and chloroquine
no. 16, pp. 2785–2791, 2009. effectively inhibit the recently emerged novel coronavirus
[45] O. Trott and A. J. Olson, “AutoDock Vina: improving the (2019-nCoV) in vitro,” Cell Research, vol. 30, no. 3, pp. 269–
speed and accuracy of docking with a new scoring function, 271, 2020.
efficient optimization, and multithreading,” Journal of Compu- [62] Z. Xu, C. Peng, Y. Shi et al., “Nelfinavir was predicted to be a
tational Chemistry, vol. 31, no. 2, pp. 455–461, 2010. potential inhibitor of 2019-nCov main protease by an integra-
[46] D. Studio, Version 2.5, Accelrys Inc., San Diego, CA, USA, 2009. tive approach combining homology modelling, Mmolecular
[47] M. J. Abraham, T. Murtola, R. Schulz et al., “GROMACS: high docking and binding free energy alculation,” bioRxiv, 2020.
performance molecular simulations through multi-level paral- [63] B. Jarvis and D. Faulds, “Nelfinavir,” Drugs, vol. 56, no. 1,
lelism from laptops to supercomputers,” SoftwareX, vol. 1, pp. 147–167, 1998.
pp. 19–25, 2015. [64] E. Eroglu and H. Türkmen, “A DFT-based quantum theoretic
[48] T. I. Cheatham, J. Miller, T. Fox, T. Darden, and P. Kollman, QSAR study of aromatic and heterocyclic sulfonamides as car-
“Molecular dynamics simulations on solvated biomolecular bonic anhydrase inhibitors against isozyme, CA-II,” Journal of
systems: the particle mesh Ewald method leads to stable trajec- Molecular Graphics and Modelling, vol. 26, no. 4, pp. 701–708,
tories of DNA, RNA, and proteins,” Journal of the American 2007.
Chemical Society, vol. 117, no. 14, pp. 4193-4194, 1995. [65] N. Rasool, W. Husssain, and Y. D. Khan, “Revelation of
[49] B. Hess, H. Bekker, H. J. C. Berendsen, and J. G. E. M. Fraaije, enzyme activity of mutant pyrazinamidases from Mycobacte-
“LINCS: a linear constraint solver for molecular simulations,” rium tuberculosis upon binding with various metals using
Journal of Computational Chemistry, vol. 18, no. 12, pp. 1463– quantum mechanical approach,” Computational Biology and
1472, 1997. Chemistry, vol. 83, article 107108, 2019.