Get Permission Bhati and Dathar: Review on molecular docking analysis of herbal compounds and their activity against SARS and JEV using In-silico and In vitro approaches


Introduction

To bring out a new drug from the laboratory to the market, it takes approximately 10-12 years with an average cost US $1.2 to $1.4 billion or more per drug.1 Thus, there is a need for some fast processes and methods for the designing, discovery and development of a new chemical entity (NCE). There are many computational and Insilico tools such as genomics, proteomics, bioinformatics, and excellent technologies like, combinatorial chemistry, high throughput screening (HTS), virtual screening in vitro, in silico ADMET screening, de novo and structure-based drug design that plays an important role to speed up the modern era drug discovery and development process. 2 Computational drug design approaches are mainly concentrating on the design of ligands/molecules for active or target sites with identified three-dimensional structure. Computational methods are also helpful to check the Drug- likeness properties of the molecule, after that molecule is docked with the target, selected according to their binding affinities. These molecules are further optimized to enhance binding characteristics and the toxicity is predicted by using different online web servers. Computational techniques integrate biological, mathematical and computer-based models to predict the most favourable binding conformation of ligands in the active site of the particular receptor. The prediction power of these techniques is increasing day by day due to advancement in the field of Molecular Biology, Biotechnology, Bioinformatics, Mathematics, and Chemistry.3 The insilico drug discovery process is described in figure below-

Figure 1

A flowchart outlining a generalized structure-based insilico drug discovery strategy

https://s3-us-west-2.amazonaws.com/typeset-prod-media-server/8b75da92-684b-4f1e-93cb-6d714c65b098image1.png

To select a novel drug candidate, in-silico or computational methods play key role to design and develop new drugs and target proteins in the fields of pharmaceutics and biotechnology. These methods are used to investigate molecular modeling of gene, gene sequence, gene analysis and 3D structure of proteins which play a great role in target identification and prediction of new chemical entity.

Methods used in in-silico drug designing

Some important in-silico techniques which are helpful in drug design are follows.

Homology modelling

Homology modelling allows making an unidentified model of a target protein according to its amino acid arrangement and experimental three-dimensional structure of a related homologous protein and displays similarity with the sample sequence. 4

Molecular docking

Molecular docking includes the interaction of two or more molecules to form a stable complex. Stability of the complex depends upon the binding properties of ligand and target protein. 5 The most appropriate complexes are selected on the basis of scoring function in the software. The molecules are docked against the active site and then scored to identify the one which binds more tightly to the target protein. 6

Virtual screening

Virtual screening allows testing of large libraries of molecules and compounds for their potential to interact with specific site of target protein which can screen more than thousands of possible molecules to a practicable number that can be easily synthesized, purchased and tested in the laboratory. 7

Quantitative structure-activity relationship

Quantitative structure-activity relationship is widely used to demonstrate the relationship between the structural properties of molecules with their biological activities. 8

Comparative molecular field analysis

Comparative molecular field analysis is a well-recognized 3D QSAR method which gives the values of ClogP which shows the solvent repellent constraints of the ligands and also describes the steric and electrostatic values of the ligands. 9

Comparative molecular similarity indices analysis

Comparative Molecular Similarity Indices Analysis is used in the drug discovery process to recognize the common properties, which are important for the suitable biological receptor binding with ligands. 10

3D Pharmacophore mapping

3D Pharmacophore mapping quickly predicts lead compounds along with a preferred target which has been used to make it most powerful and successful insilico and computational method.11

Conformational analysis

Conformations of molecules play a key role in the prediction of the physio-chemical properties but also helpful in prediction of biological activity of the compound. 12

Monte carlo simulation

Monte Carlo simulation helps in the generation of suitable conformations of a system by using computer simulation to allow thermodynamic, structural, and numerical properties to be calculated as a weighted average of these properties over these conformations. 13

In this review we describe the different medicinal compounds which can be screened by the use of computational methods.

Materials and Methods

Selection and refinement of various targets of JEV protein

The reported three-dimensional (3D) crystal structures of different microbial proteins were retrieved from Protein Data Bank (PDB) available at (http://www.rcsb.org/pdb/home/home.do). Retrieved structures were downloaded in PDB format and exported into the Maestro software (Schrodinger, LLC, Cambridge, USA) and refined by mediating insertion of processing, optimization, and minimization steps indicated in protein preparation wizard.

Binding site detection

Site map tool from Schrodinger was used to find the active sites in protein which have not internal ligand in their structure. The site score and draggability scores were used to select potential binding sites. The sites having the draggability score near to 1 were considered for docking study. 14

Preparation of ligands

Chemical structures of the herbal compound were obtained from the PubChem chemistry database, and the structures drawn and optimized in ChemDraw (Perkin Elmer Informatics, Waltham, MA, USA). The low energy 3D conformation of ligand was executed with the support of LigPrep OPLS3e force field. The ionization states were also adjusted prior to docking simulations.

Molecular docking

Maestro suites (Schrodinger, LLC, Cambridge, USA) was used to dock herbal compounds against the target proteins of microbes. The receptor grid was generated by specifying the internal ligand and by doing the site mapping for the proteins which have not internal ligand in their structure. 15

3.5. Molecular dynamics simulations were performed for herbal compounds towards the target proteins by using Desmond module of Schrödinger with 100ns simulation time. The root mean square deviation (RMSD) for the protein backbone and root mean square fluctuation (RMSF) of the protein residues were plotted to examine the convergence of the ligands to equilibrium.

Review of Literature

In-silico study or computational screening of compounds against SARS

Shen et al.16 have screened 290 compounds against HCoV-OC43 strain and found 27 compounds to be effective against SARS.

Table 1

In- vitro study of herbals used against SARS

Herbal extract

Active Constituent

Family

Cells

Strain

Conc.

IC 50 value/EC 50 value

Mechanism

-

Lycorine

Amaryllidaceae

Vero E6, BHK-21, DBT, 293 FT, LLC-MK2, 17-Cl-1, and DPP4 expressing Huh 7.5 cells

HCoV-OC43

-

EC 50 value 0.15 µM

Inhibition of viral replication

16

Pectolinarin

α-L- rhamnopyranosyl, L- mannopyranosyl, β-D- glucopyranoside and β-D- glucopyranoside

-

E. Coli BL 21(DE3)

-

2-320 µM

IC 50 value 37.78 µM

-

17

Tetraandrine

-

Menispermaceae

MRC-5

HCOV-OC 43

0.2 µM

IC 50 value 0.33± 0.03 µM

By inhibiting the nucleocapsid protein and HCoV-OC43 spike expression

18

Juglanin (arabinose residue)

Kaempferol, kaempferol glycoside, acylated kaempferol glucoside

Polygonaceae

Xenopus oocytes

-

10 µM, 20 µM

IC 50 value 2.3 µM

Inhibit 3a ion channel of coronavirus

19

(ethanolic extract)

Phlorotannin

-

Vero cells (African green monkey cell line)

PEDV SM 98 strain

30 µM

IC 50 value 12.4 ±2.2 µg/ml

By inhibiting viral haemagglutination binding to SA receptor in host cells

20

-

Scutellarein

Lamiaceae

-

-

2 µm

IC50 value 0.86± 0.48µM

Inhibition of SARS CoV helicase and nsP13 by affecting the ATPase activity

21

Salvia miltiorrhiza (dried roots) (ethanol extract)

-

Laminaceae

E. Coli BL21 (DE3) Codon Plus RIL cells

-

30, 15, 7.5 µM

IC 50 value 0.7 µM

Inhibits cysteine protease

22

-

Gallocatechin gallate

-

-

-

-

IC 50 value 47 µM

By blocking the enzymatic activity of SARS CoV 3CL pro

23

Euphorbia neriifolia (ethanolic extract)

3-β-friedelanol, 3-β-acetoxyfriedelane, friedelin actinomycin D, epitaraxerol,

Euphorbiaceae

MRC-5 cells

Strain 229 E

-

-

-

24

Pelargonium sidoicks (aqueous extract)

Eps 7630

Geraniaceae

MDCK, Vero, Caco-2, Mel-Ho, Human foreskin fibroblast

Influenza virus strain H1N1, H3N2, H5N1

100 µg/ml

-

-

25

Sinomenium acutum

-

Menispermaceae

Mouse Spleenic lymphocytes

-

0-400 µg/ml

IC 50 value 198.6 µg/ml

By inhibiting SARS CoV RdRp and 3CL pro

26

Coriolus versicolor

-

Polyporaceae

Mouse Spleenic lymphocytes

-

0-400 µg/ml

IC 50 value 108.4 µg/ml

By inhibiting SARS CoV RdRp and 3CL pro

27

Torreya nucifera (leaf) (ethanolic extract)

Amentoflavone

Taxaceae

--

-

100 µg/ml

IC 50 value 8.3 µM

By inhibiting SARS CoV 3CL protease activity

28

Tylophorine compounds (methanolic extract)

phenanthroindozolidine and phenanthroquinolizidine

Apocynaceae

Vero 76 cells

SARS CoV Urbani strain

5, 50, 500 nm

less than 5 to 40 nm

29

-

Procyanidin A2 Procyanidin B1

-

-

SARS CoV PUMC 01F5

-

i)IC50 value 29.9±3.3µM ii)IC50 value 41.3±3.4 µM

Interfering with endocytosis

30

Rheum palmatum L. (root and rhizomes) (ethanolic extract)

Anthraquinone

Polygonaceae

Vero E6 cells

SARS CoV and SARS CoV 3CL protease

100, 50, 25, 12.5, 6.25, 3.12 and 1.56 µg/ml

IC 50 value 13.76 ±0.03 µg/ml (96%)

Inhibits SARS-3CL pro activity

31

Toona sinensis (aqueous leaf extract)

-

Meliaceae

African green monkey kidney cell line Vero (CCL-81)

i) HCoV 229 E strain ii) Sar CoV strain FFM1

i)5-20 µg/ml ii)50-200 µg/ml

-

Inhibit the cellular entry of SARS CoV

32

Houttuyria cordata (aqueous extract)

-

Saururaceae

Mouse splenic lymphocytes

-

0-400µg/ml

IC50 value 50-1000 µg/ml

By inhibiting SARS CoV 3CL protease activity, viral polymerase and RdRp activity

33

Sophora subprostateradix (methanolic extract)

Matrine, oxymatrine, sophoranone, sophocarpine

Fabaceae

Vero cells

Plaque cloned A59 strain of MHV Mouse DBT cells

1, 10, 50, 100 µg/ml

EC 50 value 27.5 ±1.1 µg/ml

-

34

Veronica linariifolia (ethanolic extract)

Luteolin

Plantaginaceae

Vero E6 cells

SARS CoV (BJ01 strain)

-

EC50 value 10.6 µm

By binding with the surface spike protein of the virus and interfering with the entry of the virus into the host cells

35

Boenninghausenia sessilicarpa (ethanolic extract)

Laptodactylone

-

Vero E6 cells

RPMI-1640

100, 20, 4 µg/ml

EC50 value 60%, 4% and 0% respectively

-

36

-

Saikosaponin B2

MRC-5, ATCC, CCL-171

HCoV 229E strain

6 µmol/L

IC50 value 1.7±0.1 µmol/L

By preventing viral attachment and penetration at the early stage of HCoV-229E infection

37

Glycyrrhiza

Glycyrrhizin, Glycyrrhizic acid derivative

Fabaceae

Vero E6 cells

SARS CoV strain FFM1

4000ug/ml

EC 50 value-40 µM (5- 50 µM)

Inhibition of viral replication, Induction of cellular NO -synthase and thus affecting viral adsorption and penetration

38

Lindera aggregate (root) (ethanolic extract)

-

Lauraceae

-

Viral Strain i) BJ001 ii) BJ006

--

EC50 value 88.2(±7.7) EC50 value 80.6(±5.2)

-

39

Isatis indigotica (root) (aqueous extract)

Indigo, indirubin, indicant, β-sitosterol γ-sitosterol and sinigrin

Cruciferae

Vero cells

-

-

IC50 value 90.1±4.2 (217 µM)

By inhibiting the 3CL protease enzyme

40

Calophyllum blancoi (acetone extract)

Blancoxanthone

Guttiferae

Human lung fibroblast (MRC-5)

HCOV-229 E strain

-

EC 50 value 3 µg/ml

--

41

Galla chinensis (ethanolic extract)

Tetra-O-galloyl-β-D glucose (TGG)

-

Vero E6 cells

SARS CoV (BJ01 strain)

-

EC50 value 4.5 µm

By binding with the surface spike protein of the virus and thus interfering entry of the virus into the host cells

42

Scutellaria baicalensis

Baicalin

Lamiaceae

i)frhK4 cell line ii) Vero E6 cells

i)SARS CoV ii)39849 strain

11 µg/ml

i)EC 50 value 12.5-25 (48 hr) 25-50 (72 hr) ii)EC 50 value 12.5(48 hr) 100 (72 hr)

Inhibit Angiotensin-converting enzyme (ACE)

43

Aescin (Horse chestnut)

-

Sapindaceae

Vero E6 cells

-

3 µM

EC 50 value 6.0 µM

-

44

Glycyrrhiza

Glycyrrhizin, Glycyrrhizic acid derivative

Fabaceae

Vero cells

SARS CoV

4000ug/ml

EC 50 value- 300 mg/L (316-62.5 mg/L)

Inhibition of viral replication, Induction of cellular NO -synthase and thus affecting viral adsorption and penetration

45

Table 2

List of compounds analyzed against JEV by using insilico techniques

Drug

Binding interactions with different targets of JEV.

Binding affinity (ΔG) Kcal/Mol

Target

Chemdiv-3

H-Bonding- ASN313, PRO314, ALA315, VAL323.

Envelope protein. (3P54)

46

4-hydroxypandurantin

H-Bonding- GLY80, ASP81, ILE60, VAL60, TRP62. Hydrophobic contact- ASP79, CYS101, LEU104, ALA105, THR108, ALA111, ILE112, ALA115, TYR119.

−9.95

NS3 helicase

47

kaempferol

Make complex with RNA by R1B, R2B, R3B.

R1b= -3.64 R2b= -3.70 R3b= -5.04

NS1 (5O36)

48

kanamycin

Vander waal force-LYS200, GLN457

-147.367

NS3 helicase/nucleoside triphosphatase (2z83)

49

CW-33

H-BOND- ASN152, GLU115. Π cation- ARG76.

Dock score= 42.021

NS2B‑NS3 protease (4R8T)

50

Rosameric acid

Hydrogen and weak vab der waal forces with LYS, TYR, PRO, ALA, PHE.

-126

Envelope Protein Domain III (3P54)

51

Niclosamide

H-BOND- LYS166: LYS166:123.

-5.43

Envelope protein (3P54)

52

Compound 1 and compound 2.

Compound-1- H-bond- HIS-288, THR451, ARG458. Compound-2- THR451, HIS288, ARG458, THR290.

_______

NS3 helicase (2z83)

53

Deoxynojirimycin

H-BOND- ASP10, ASN8, ASP10, CYS30, VAL24.

-6.55

envelope protein (3P54)

54

Phytolacca Americana L

ASP-542, CYS-82

NS3 (-655.1) NS5 (-782.7) envelope protein (633.4)

NS5 (4K6M) NS3 (2Z83) envelope protein(3p54)

55

Jo et al.17 have screened various compounds through induced fit docking studies and proteolytic assay and found that compounds such as Herbacetin, rhoifolin pectolinarin were found to be effective against SARS CoV.

Nguyen et al.23 have screened various flavonoids through molecular docking studies against 3CLpro of SARS CoV and found gallocatechin gallate (with binding energy -14kCal/mol) to be the best compound effective against SARS-CoV. Ryu et al.28 reported flavones such as apigenin, luteolin and quercetin were able to inhibit 3CLpro activity with IC50 values of 280.8, 20.2, and 23.8 µM respectively.

Chen et al. 56 performed molecular docking studies and SPR/FRET-based bioassay on quercetin-3-β-galactoside which might be effective against SARS CoV through inhibition of 3CLproactivity. Ho et al. 35 have performed computational screening of 312 Chinese medicinal herbs and found 3 compounds belonging to the family Polygonaceae to be effective against SARS virus.

Wang et al.57 screened 11 molecules from TCMD primarily based on template strain MDL 28170 and found MOL736 (derived from Artemisia annua) with the lowest binding energy and highest similarity reported to be effective against SARS CoV by inhibiting endosomal protease activity of SARS virus.

Toney et al. 58 have screened 14000 molecules and reported 1853 molecules to serve as 3CLpro inhibitors. Further molecular docking studies showed that the 10 compounds have the lowest docking energy (-11.6 kCal/mol) with desirable chemical properties as therapeutics.

Herbals used against SARS (in-vitro study)

Many of the natural compounds like lycorine, emetine, herbacetin, pectolinarin, rhoifolin, glycyrrhiza, lycoris radiata, aloe-emodin, baicalin and isatis indigotica are found to be effective against in SARS infection by inhibiting viral replication and nucleocapsid protein against SARS CoV strain. The relevant studies have been extracted from Pub Med and compiled in the below mentioned Table 1.

Correlation of In-silico data to Experimental Data

In order to screen the thousands of compounds in library the in-silico predictions are the best tools to select the compound which has potential affinity to inhibit the JE infection. However, the experimental data also must determine the activity of compound against specific target and the data obtained in the in-silico may not be reliable as compared to in-vitro or in-vivo.

The drugs show different activity in-vivo as compared to in-silico and in-vitro, this may be due to poor pharmacokinetic properties or physicochemical properties or inability to interact with the target at particular environment etc. So, in current review we have correlated the in-silico and experimental (in-vitro or in-vivo) activity of the molecule against JEV infection. However, we have found valid and similar results against JE infection under in-silico and also in-vitro and in-vivo except for some compounds.

By computer simulations, Chemdiv-3 was found to be effective against JEV, and at a concentration of 20µM has potential to inhibit the viral growth in animal (Balb-2 strain mice), it has shown moderate activity against JE infection and the mice survived 3 more days compared to control mice (which JE infected mice). 46 In case of Phytolacca-mother tincture, excellent results were shown in-silico as well as in-vivo (in JE infected human), at the concentration of 26.5µM upon daily administration. 55

Kaempferol and daidzein have also shown great activity by computational simulations. However, the experimental data has shown that moderate activity (in-vitro) towards JE infected BHK-21 cells which inhibitted 70% of viral replication at the concentration of 25.7 µM kaempferol and 29.7 µM for daidzein. 48 Doxycycline and kanamycin are capable of inhibiting JE infection and inflammation under in-vivo conditions similar to computational study. But, doxycycline has shown more activity than kanamycin intraperitoneally into Swiss albino mice. 59 CW-33 has shown similar interaction under in-silico and in-vitro by inhibiting the RNA replication against JEV. 50

Niclosamide, Cilnidipine and FGIN-1-27 and Rosameric acid have shown very effective against JEV by computational simulation. However, Niclosamide, Clinidipine have shown poor activity in-vivo (mice) due to poor penetration into blood brain barrier, whereas Rosameric acid and FGIN-1-27 have shown similar activity as in-silico which prevented increasing of viral load in JE infected mice. 51, 52 Fang et al. 53 have discovered 2 azole compounds which have shown similar interaction in-silico whereas, compound 1 has shown 71% inhibition and compound 2 has shown 95% inhibition in-vitro.

Other drugs like 4-hydroxypandurantin, Aminotetrahydroquinazoline derivative were tested only by in-silico approaches, further investigation is required for the exploration of those drugs for the treatment of JEV infection. 54

Conclusion

Studies conducted on treatment of viruses like SARS CoV and Japanese Encephalitits Virus with various herbal compounds. The efficacy of the drugs or compounds was tested by different In silico and In vitro methods. The drugs have shown varied binding efficacy targeting different viral proteins and few of them interacting with viral RNA. The inhibitory action of the drug also varied with the concentration. However, the behavior of the drug is different In silico and In vitro.

Conflict of Interest

The authors declare no relevant conflicts of interest.

Source of Funding

None.

References

1 

A Wadood N Ahmed L Shah A Ahmad H Hassan S Shams In-silico drug design: An approach which revolutionarised the drug discovery processDrug Des Deliv2013113

2 

J Kirchmair M J Williamson J D Tyzack L Tan P J Bond A Bender Computational prediction of metabolism: Sites, products, SAR, P450 enzyme dynamics, and mechanismsJ Chem Inf Model20125236174810.1021/ci200542m

3 

S Ekins J Mestres B Testa In silico pharmacology for drug discovery: Methods for virtual ligand screening and profilingBr J Pharmacol2007152192010.1038/sj.bjp.0707305

4 

S Kaczanowski P Zielenkiewicz Why similar protein sequences encode similar three-dimensional structures?Theor Chem Acc20101256435010.1007/s00214-009-0656-3

5 

G Maithri B Manasa SS Vani A Narendra T Harshitha Computational Drug Design and Molecular Dynamic Studies-A ReviewInt J Biomed Data Min20176112310.4172/2090-4924.1000123

6 

Y Tanaka M Kuroda Y Yasutake M Yao K Tsumoto N Watanabe Crystal structure analysis reveals a novel ESAT-6 secretion system C protein in Staphylococcus aureusProteins20077036596410.1002/prot.21302

7 

W Patrick W Matthew T Stahl MA Murcko Virtual screening - An overviewDrug Discov Today1998341607810.1016/S1359-6446(97)01163-X

8 

ME Suh SY Park HJ Lee Comparison of QSAR methods (CoMFA, CoMSIA, HQSAR) of anticancer 1-N-substituted imidazoquinoline-4,9-dione derivativesBull Korean Chem Soc200223341722

9 

ER Malinowski Statistical F-tests for abstract factor analysis and target testingJ Chemom198931496010.1002/cem.1180030107

10 

Y Kurogi O Guner Pharmacophore Modeling and Three-dimensional Database Searching for Drug Design Using CatalystCurr Med Chem20128910355510.2174/0929867013372481

11 

S Zobel-Roos A Schmidt F Mestmäcker M Mouellef M Huter L Uhlenbrock Accelerating biologics manufacturing by modeling or: Is Approval under the QbD and PAT approaches demanded by authorities acceptable without a digital-twin? Processes2019729410.3390/pr7020094

12 

H Alonso AA Bliznyuk JE Gready Combining docking and molecular dynamic simulations in drug designMed Res Rev20062655316810.1002/med.20067

13 

M E Tuckerman G J Martyna Understanding Modern Molecular Dynamics: Techniques and ApplicationsJ Phys Chem B200010421597810.1021/jp992433y

14 

V Navyashree K Kant A Kumar Natural chemical entities from Arisaema genus might be a promising break-through against Japanese encephalitis virus infection: a molecular docking and dynamics approachJ Biomol Struct Dyn202139414041610.1080/07391102.2020.1731603

15 

K Kant R Rawat V Bhati S Bhosale D Sharma S Banerje Computational identification of natural product leads that inhibit mast cell chymase: an exclusive plausible treatment for Japanese encephalitisJ Biomol Struct Dyn202139412031210.1080/07391102.2020.1726820

16 

L Shen J Niu C Wang B Huang W Wang N Zhu High-Throughput Screening and Identification of Potent Broad-Spectrum Inhibitors of CoronavirusesJ Virol20199312 e00023-1910.1128/JVI.00023-19

17 

DE Kim JS Min MS Jang JY Lee YS Shin CM Park Natural Bis-Benzylisoquinoline Alkaloids-Tetrandrine, Fangchinoline, and Cepharanthine, Inhibit Human Coronavirus OC43 Infection of MRC-5 Human Lung CellsBiomolecules201991169610.3390/biom9110696

18 

S Schwarz D Sauter W Lü K Wang B Sun T Efferth Coronaviral ion channels as target for Chinese herbal medicineFor Immunopathol Dis Therap20123111310.1615/ForumImmunDisTher.2012004378

19 

J Y Park J H Kim J M Kwon H J Kwon H J Jeong Y M Kim Dieckol, a SARS-CoV 3CLpro inhibitor, isolated from the edible brown algae Ecklonia cavaBioorganic Med Chem201321133730710.1016/j.bmc.2013.04.026

20 

MS Yu JLee JM Lee Y Kim YW Chin JG Jee Identification of myricetin and scutellarein as novel chemical inhibitors of the SARS coronavirus helicase, nsP13Bioorg Med Chem Lett2012221240495410.1016/j.bmcl.2012.04.081

21 

JY Park JH Kim YM Kim HJ Jeong DW Kim KH Park Tanshinones as selective and slow-binding inhibitors for SARS-CoV cysteine proteasesBioorg Med Chem2012201959283510.1016/j.bmc.2012.07.038

22 

TTH Nguyen HJ Woo HK Kang VD Nguyen YM Kim DW Kim Flavonoid-mediated inhibition of SARS coronavirus 3C-like protease expressed in Pichia pastorisBiotechnol Lett2012345831810.1007/s10529-011-0845-8

23 

FR Chang CT Yen M Ei-Shazly WH Lin MH Yen KH Lin Anti-human coronavirus (anti-HCoV) triterpenoids from the leaves of Euphorbia neriifoliaNat Prod Commun201271114157

24 

M Michaelis HW Doerr J Cinatl Investigation of the influence of EPs® 7630, a herbal drug preparation from Pelargonium sidoides, on replication of a broad panel of respiratory virusesPhytomedicine2011185384610.1016/j.phymed.2010.09.008

25 

KP Fung PC Leung KWS Tsui CCD Wan KB Wong MYM Waye Immunomodulatory activities of the herbal formula Kwan Du Bu Fei Dang in healthy subjects: A randomised, double-blind, placebo-controlled studyHong Kong Med J2011172413

26 

CC Wen LF Shyur JT Jan PH Liang CJ Kuo P Arulselvan Traditional chinese medicine herbal extracts of cibotium barometz, gentiana scabra, dioscorea batatas, cassia tora, and taxillus chinensis inhibit sars-cov replicationJ Tradit Complement Med201111415010.1016/s2225-4110(16)30055-4

27 

YB Ryu HJ Jeong JH Kim YM Kim JY Park D Kim Biflavonoids from Torreya nucifera displaying SARS-CoV 3CLpro inhibitionBioorganic Med Chem201018227940710.1016/j.bmc.2010.09.035

28 

CW Yang YZ Lee IJ Kang DL Barnard JT Jan D Lin Identification of phenanthroindolizines and phenanthroquinolizidines as novel potent anti-coronaviral agents for porcine enteropathogenic coronavirus transmissible gastroenteritis virus and human severe acute respiratory syndrome coronavirusAntiviral Res2010882160810.1016/j.antiviral.2010.08.009

29 

M Zhuang H Jiang Y Suzuki X Li P Xiao T Tanaka Procyanidins and butanol extract of Cinnamomi Cortex inhibit SARS-CoV infectionAntiviral Res2009821738110.1016/j.antiviral.2009.02.001

30 

W Luo X Su S Gong Y Qin Anti-SARS coronavirus 3C-like protease effects of Rheum palmatum L. ExtractsBioscience Trends2009341246

31 

CJ Chen M Michaelis HK Hsu CC Tsai KD Yang YC Wu Toona sinensis Roem tender leaf extract inhibits SARS coronavirus replicationJ Ethnopharmacol200812011081110.1016/j.jep.2008.07.048

32 

KM Lau KM Lee CM Koon CSF Cheung CP Lau HM Ho Immunomodulatory and anti-SARS activities of Houttuynia cordataJ. Ethnopharmacol20081181798510.1016/j.jep.2008.03.018

33 

HY Kim HS Shin H Park YC Kim YG Yun S Park In vitro inhibition of coronavirus replications by the traditionally used medicinal herbal extracts, Cimicifuga rhizoma, Meliae cortex, Coptidis rhizoma, and Phellodendron cortexJ Clin Virol2008412122810.1016/j.jcv.2007.10.011

34 

TY Ho SL Wu JC Chen CC Li CY Hsiang Emodin blocks the SARS coronavirus spike protein and angiotensin-converting enzyme 2 interactionAntiviral Res20077429210110.1016/j.antiviral.2006.04.014

35 

QY Yang XY Tian W Fang Bioactive coumarins from Boenninhausenia sessilicarpaJ Asian Nat Prod Res2007915965

36 

PW Cheng LT Ng LC Chiang CC Lin Antiviral effects of saikosaponins on human coronavirus 229E in vitroClin Exp Pharmacol Physiol2006337612610.1111/j.1440-1681.2006.04415.x

37 

G Hoever L Baltina M Michaelis R Kondratenko L Baltina GA Tolstikov Antiviral activity of glycyrrhizic acid derivatives against SARS-coronavirusJ Med Chem20054841256910.1021/jm0493008

38 

SY Li C Chen HQ Zhang HY Guo H Wang L Wang Identification of natural compounds with antiviral activities against SARS-associated coronavirusAntiviral Res2005671182310.1016/j.antiviral.2005.02.007

39 

CW Lin FJ Tsai CH Tsai CC Lai L Wan TY Ho Anti-SARS coronavirus 3C-like protease effects of Isatis indigotica root and plant-derived phenolic compoundsAntiviral Res2005681364210.1016/j.antiviral.2005.07.002

40 

YC Shen TW Li AT Khalil LC Chiang PW Cheng Bioactive Pyranoxanthones from the Roots of Calophyllum blancoiChem Pharm Bull2005532244710.1002/chin.200530115

41 

L Yi Z Li K Yuan X Qu J Chen G Wang Small Molecules Blocking the Entry of Severe Acute Respiratory Syndrome Coronavirus into Host CellsJ Virol2004782011334910.1128/JVI.78.20.11334-11339.2004

42 

F Chen KH Chan Y Jiang RYT Kao HT Lu KW Fan In vitro susceptibility of 10 clinical isolates of SARS coronavirus to selected antiviral compoundsJ Clin Virol2004311697510.1016/j.jcv.2004.03.003

43 

CY Wu JT Jan SH Ma CJ Kuo HF Juan YSE Cheng Small molecules targeting severe acute respiratory syndrome human coronavirusProc Natl Acad Sci U. S. A20041012710012710.1073/pnas.0403596101

44 

J Cinatl M Michaelis G Hoever W Preiser HW Doerr Development of antiviral therapy for severe acute respiratory syndromeAntiviral Res2005662819710.1016/j.antiviral.2005.03.002

45 

S Jo S Kim D H Shin M S Kim Inhibition of SARS-CoV 3CL protease by flavonoidsJ Enzyme Inhib Med Chem20203511455110.1080/14756366.2019.1690480

46 

T Zhang Z Wu J Du Y Hu L Liu F Yang Anti- Japanese-encephalitis-viral effects of kaempferol and daidzin and their RNA-binding characteristicsPLoS One201271e3025910.1371/journal.pone.0030259

47 

C Ye P Bian J Zhang H Xiao L Zhang W Ye Structure-based discovery of antiviral inhibitors targeting the E dimer interface of Japanese encephalitis virusBiochem Biophys Res Commun201951523667110.1016/j.bbrc.2019.05.148

48 

S Kulkarni L R Samant S Verma A Chowdhary Silico Analysis of Lutein and Rosmarinic Acid against Envelope Protein Domain III ofJapanese Encephalitis Virus20145311620

49 

M Rath MC Sahu SK Sahu Docking and Inhibition of Antiviral Ligands of Japanese Encephalitis Virus with Capsid Envelope Strain SA-14-14-2(3P54). Int J Sci Res2018712394210.21275/ART20179235

50 

J Fang H Li D Kong S Cao G Peng R Zhon Structure based discovery of two antiviral inhibitors targeting the NS3 helicase of Japanese encephalitis virusSci Rep201663455010.1038/srep34550

51 

SK Gupta S Singh A Nischal KK Pant PK Seth Molecular docking and simulation studies towards exploring antiviral compounds against envelope protein of Japanese encephalitis virusNetw Model Anal Health Inform Bioinforma20132231310.1007/s13721-013-0040-z

52 

HC Chaudhury AK Panda SK Behera M Mohanty BB Subudhi Case report on recovery from Japanese encephalitis virus infection by complementary use of phytolacca-mother tincture and in silico analysisJ Herb Med2018145228

53 

C Seniya H Mishra A Yadav N Sagar B Chaturvedi K Uchadia Antiviral potential of 4-hydroxypanduratin A, secondary metabolite of Fingerroot, Boesenbergia pandurata (Schult.), towards Japanese Encephalitis virus NS2B/NS3 proteBioinformation201391546010.6026/97320630009054

54 

KC Chen YF Lin AC Huang JY Gao CW Lin JC Lien Molecular interaction of the antiviral compound CW-33 and its analogues with the NS2B-NS3 protease of the Japanese encephalitis virusInt J Mol Med201943520243210.3892/ijmm.2019.4113

55 

M Nath B Deb A computational approach of antibiotics as novel drug target for Japanese encephalitis virus NS helicase/nucleoside triphosphataseMOJ Proteomics Bioinforma2018731849

56 

L Chen J Li C Luo H Liu W Xu G Chen Binding interaction of quercetin-3-β-galactoside and its synthetic derivatives with SARS-CoV 3CLpro: Structure-activity relationship studies reveal salient pharmacophore featuresBioorganic Med Chem200614248295306

57 

SQ Wang QS Du K Zhao AX Li DQ Wei KC Chou Virtual screening for finding natural inhibitor against cathepsin-L for SARS therapyAmino Acids20073311293510.1007/s00726-006-0403-1

58 

JH Toney S Navas-Martín SR Weiss A Koeller Sabadinine: A Potential Non-Peptide Anti-Severe Acute-Respiratory-Syndrome Agent Identified using Structure-Aided DesignJ Med Chem200447510798010.1021/jm034137m

59 

R Topno S A Khan Efficacy of antibiotics (Doxycycline and Kanamycin) against Japanese encephalitis virus infectionTrop Biomed201835123945



jats-html.xsl


This is an Open Access (OA) journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

  • Article highlights
  • Article tables
  • Article images

Article History

Received : 28-04-2022

Accepted : 23-05-2022


View Article

PDF File   Full Text Article


Copyright permission

Get article permission for commercial use

Downlaod

PDF File   XML File   ePub File


Digital Object Identifier (DOI)

Article DOI

https://doi.org/10.18231/j.ijmmtd.2022.022


Article Metrics






Article Access statistics

Viewed: 1287

PDF Downloaded: 328



Medical Abbreviation List