Next Article in Journal
The Effect of a Single Freeze–Thaw Cycle on Matrix Metalloproteinases in Different Human Platelet-Rich Plasma Formulations
Previous Article in Journal
The Risk of Colorectal Adenoma in Nonalcoholic or Metabolic-Associated Fatty Liver Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Phytocompounds from Himalayan Medicinal Plants as Potential Drugs to Treat Multidrug-Resistant Salmonella typhimurium: An In Silico Approach

1
Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh 173212, India
2
Department of Pharmaceutical and Medical Chemistry, University of Uyo, Uyo 520003, Nigeria
3
Organic Chemistry Research Lab, Department of Chemistry, Obafemi Awolowo University, Osun 220282, Nigeria
4
Laboratory for Translational Chemistry and Drug Discovery, Hansraj College, University of Delhi, Delhi 110007, India
5
Laboratory of Computational Modelling of Drugs, South Ural State University, 454080 Chelyabinsk, Russia
6
Department of Immunology, University of Oslo, 0315 Oslo, Norway
7
Department of Biotechnology, College of Engineering, Suwon University, Hwaseong-si 18323, Korea
8
Plasma Bioscience Research Center & Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Biomedicines 2021, 9(10), 1402; https://doi.org/10.3390/biomedicines9101402
Submission received: 15 September 2021 / Revised: 30 September 2021 / Accepted: 1 October 2021 / Published: 5 October 2021
(This article belongs to the Section Drug Discovery, Development and Delivery)

Abstract

:
Medicinal plants can be used as natural therapeutics to treat diseases in humans. Enteric bacteria possess efflux pumps to remove bile salts from cells to avoid potential membrane damage. Resistance to bile and antibiotics is associated with the survival of Salmonella enterica subspecies enterica serovar Typhimurium (S. typhimurium) within a host. The present study aimed to investigate the binding affinity of major phytocompounds derived from 35 medicinal plants of the North Western Himalayas with the RamR protein (PDB ID 6IE9) of S. typhimurium. Proteins and ligands were prepared using AutoDock software 1.5.6. Molecular docking was performed using AutoDock Vina and MD simulation was performed at 100 ns. Drug likeness and toxicity predictions of hit phytocompounds were evaluated using molinspiration and ProTox II online servers. Moreover, docking, drug likeness, and toxicity results revealed that among all the selected phytocompounds, beta-sitosterol exhibited the most efficacious binding affinity with RamR protein (PDB ID 6IE9) and was nontoxic in nature. MD simulation data revealed that beta-sitosterol in complex with 6IE9 can be used as an antimicrobial. Furthermore, beta-sitosterol is stable in the binding pocket of the target protein; hence, it can be further explored as a drug to inhibit resistance-nodulation-division efflux pumps.

1. Introduction

Salmonella is a bacterial pathogen that infects the intestinal tract and gallbladder and causes numerous foodborne illnesses in humans. Enteric bacteria, such as Salmonella, tolerate the existence of bile acids for their survival in the gastrointestinal transit and gallbladder [1,2]. Nontyphoidal serovars (NTSs) of Salmonella enterica are the major causes of foodborne illnesses and diarrhea occurring worldwide [3,4]. In S. enterica, the resistance-nodulation-division (RND) pump is translated from the acrAB gene regulated by RamA, a transcriptional activator. RamR inhibits the expression of the ramA gene involved in multidrug resistance in Salmonella enterica subspecies enterica serovar Typhimurium (S. typhimurium). S. typhimurium is one of the NTSs causing severe human infections and results in more hospitalizations and mortality worldwide [5]. Additionally, the treatment choices are limited because antibiotics may lead to enhanced shedding of S. typhimurium and its emergence as multidrug-resistant bacteria [6,7]. Presently, it is no longer considered as the first choice of antimicrobial drug due to its resistance. The advent of new resistance mechanisms exists in S. typhimurium, leading to challenges in treating infections. Therefore, alternative therapeutic approaches are required. S. typhimurium comprises of at least nine multidrug efflux systems [8]; among these, the AcrAB-TolC system, containing the AcrB transporter of the RND family, is particularly effective in developing resistance to bile acid [8,9]. RamR is a local transcriptional repressor, which belongs to the TetR family of regulatory proteins [10]; it helps in impairing the ramA gene expression that affects ramA gene transcription resulting in multidrug resistance. Therefore, it is crucial to identify novel pharmacological targets against drug-resistant S. typhimurium.
Medicinal plants have played a pivotal role in treating diseases since the prehistoric period. These plants comprise various phytoconstituents in every part (bark, leaves, flowers, roots, fruits, and seeds), and exhibit high therapeutic value [11].
Herbal medications have recently gained immense interest as they are safe and economic and have been widely used for several years to treat diseases. Bioactive components are secondary metabolites of plants that produce pharmacological and toxicological issues in living organisms. It is difficult to screen each phytoconstituent for toxicity. In drug design, computational techniques play a crucial role in studying the toxicity of chemical and natural compounds as well as their properties [12]. In silico studies, with specific reference to toxicity prediction and molecular docking for each phytochemical in order to determine their therapeutic efficiency, require less time, are economic, and can harm animals [13,14]. Therefore, the present study was designed to investigate the binding affinity of phytocompounds of 35 important medicinal plants of the Northwestern Himalaya with S. typhimurium protein (RamR transcriptional repressor of TetR family) with PDB ID: 6IE9 to prevent inflammatory gastroenteritis.

2. Methodology

2.1. Bioinformatics Tools

Open Babel GUI [15], UCSF Chimera 1.8.1, Pubchem (www.pubchem.com (accessed on 15 May 2021), RCSB PDB (http://www.rscb.org/pdb (accessed on 15 May 2021)), Autodock/vina software [16], and Discovery Studio were used in the present investigation.

2.2. Ligand Preparation

Seventy major phytocompounds of 35 medicinal plants from Himachal Pradesh, India, were selected for molecular docking analysis. The three-dimensional structures of all the phytocompounds and resistant bile components of S. typhimurium (chenodeoxycholic acid) were downloaded from Pubchem (www.pubchem.com (accessed on 15 May 2021)) in .sdf format, which was finally converted into a PDB file. Each selected ligand (phytocompounds and bile component) was prepared using the open Babel software from the command line on an Ubuntu terminal. Table 1 lists the names of the phytocompounds selected for this study, their plant sources, pharmacological properties, and ethnomedicinal uses.

2.3. Protein Preparation

RamR of S. typhimurium [67] was used for molecular docking with major phytocompounds from 35 important medicinal plants (Table 1) found in the northwestern Himalayas of Himachal Pradesh to identify potential inhibitors of S. typhimurium. The 3-D structure protein (PDB ID = 6IE9) was downloaded from the protein databank (http://www.rscb.org/pdb (accessed on 15 May 2021)) as a pentamer, and chain A was extracted for docking using PyMol. Chain A was prepared for docking, and a grid box was set to cover the entire protein (grid box dimensions = 40, 40, 40 Å) and was centered at x, y, z = 11.029, 33.324, 12.359 Å, respectively.

2.4. Molecular Docking of Major Phytocompounds of Thirty-Five Medicinal Plants

The AutoDock tool was used to dock the selected ligands to the catalytic triad of proteins, which was further stored as a pdbqt file. Docking was carried out to estimate the population of possible ligand conformations/orientations at the binding site. To align the ligands in the same spatial coordinates, a vina perl script was used [16]. The best conformation was selected with the minimum docked energy after completing the docking search. The pdb complex of protein and ligands was analyzed using Discovery Studio (https://discover.3ds.com/d (accessed on 15 May 2021)) to study the interactions between proteins and ligands. The binding strength of the ligand was calculated as a negative score (kcal/mol).

2.5. Drug Likeness Calculations

The drugs were scanned to assess whether the selected phytochemicals met the drug-likeness criteria. Lipinski’s rule of 5 using Molinspiration (http://www.molinspiration.com (accessed on 15 May 2021)) was used to verify drug likeness attributes, such as the number of hydrogen acceptors <10, number of hydrogen donors <5, molecular weight <500 Da, and partition coefficient log P > 5. The smiles format of all major phytocompounds was uploaded for further screening [68].

2.6. ADMET Screening and Toxicity Prediction of Phytocompounds

Absorption, distribution, metabolism, excretion, and toxicity (ADMET) screening was performed to evaluate the absorption, toxicity, and drug-likeness properties of the selected phytocompounds. The 3-D structures of 11 phytocompounds (asiaticoside, beta-sitosterol, bryophyllin A, madecassoside, Mahanimbine, Pennogenin, Rutin, Solasonine, Solamargine, Withaferin A, and Withanone) were saved in smiles format and uploaded on the SWISSADME (http://www.swissadme.ch/ (accessed on 15 May 2021)) (Molecular Modeling Group of the SIB (Swiss Institute of Bioinformatics) and PROTOX-II (https://tox-new.charite.de/protox_II/) web servers (Charite University of Medicine, Institute for Physiology, Structural Bioinformatics Group, Berlin, Germany) [69,70,71,72] for ADMET screening. SWISSADME is an online tool used to predict ADME and pharmacokinetic and physicochemical features of a molecule, which are the main determinants for clinical trials. Toxicity was evaluated in compounds with LD50 values ≤50 mg/kg (Class I), >50 mg/kg but <500 mg/kg (Class II), 500 < LD50 ≤ 5000 mg/kg (Class III), and LD50 > 5000 mg/kg (Class IV). Classes I, II, and III exhibited less toxicity, whereas Class IV revealed no toxicity [73,74]. Moreover, PROTOX is a rodent oral toxicity server that determines the LD50 value and the toxicity class of a target molecule [69]. A schematic of the experiment is illustrated in Figure 1.

2.7. MD Simulation of Protein Ligand Complexes

The ligand–protein complex structure was prepared before MD simulation to remove the structural errors. Extensive 100 ns MD simulation was performed on the Desmond platform to analyze the ligand behavior within the complex [75]. The complex was solvated in a TIP3P (8018 molecules) water model and 0.15 M NaCl (Na: 54.42 mMol and Cl: 49.88 mMol) to mimic a physiological ionic concentration. The molecular mechanics/generalized born surface area (MM/GBSA) was used for binding free energy calculations [75]. MD simulation trajectories were used as inputs to calculate the MM/GBSA of the binding free energies of the ligands and to investigate their binding mechanisms. The thermodynamic binding energy was calculated for every 1000th frame, as the complete MD simulation includes 10,000 frames. The OPLS 2005 force field was used for the MD simulations.

3. Results

3.1. Molecular Docking of 70 Major Phytocompounds from 35 Medicinal Plants with Ram R Protein of S. typhimurium

Molecular docking was performed with the RamR protein of S. typhimurium and AutoDock vina software to study the interactions of the major phytocompounds of 35 medicinal plants of the northwestern Himalayas in efflux pump inhibition. The docking results revealed that out of 70 phytocompounds from 35 medicinal plants, only 11 phytocompounds from 8 medicinal plants revealed binding energy comparable to that of the bile component chenodeoxycholic acid. Asiaticoside from Centella asiatica exhibited the highest binding energy (−10.9 KJ/mol), followed by the bile component chenodeoxycholic acid (−10.8 KJ/mol), bryophyllin A (−10.6 KJ/mol), pennogenin (−10.3 KJ/mol), withaferin A (−10.2 KJ/mol), madecassoside and solasonine (−9.7 KJ/mol), solamargine (−9.5 KJ/mol), mahanimbine (−9.4 KJ/mol), withanone (−9.3 KJ/mol), rutin (−9.2 KJ/mol), and beta-sitosterol (−9.2 KJ/mol). Interactive amino acids are listed in Table 2 and Figure 2.
Asiaticoside in complex with 6IE9 revealed hydrogen bonding with Ser137, Glu113, Leu115, Asp152, Ala110, and Arg148 and hydrophobic interactions with Ile106, Tyr59, Lys63, Asp124, Glu120, Ala149, Lys114, Ser112, Val111, Lys117, Cys134, Leu130, Arg136, Thr85, Ile88, Met70, Leu139, Phe155, Leu66, Tyr92, Leu156, and Met140. Similarly, other phytocompounds, such as madecassoside, exhibited hydrogen bonding with Tyr59, Asp124, Ala110, Ala123, and Cys67 and hydrophobic interactions with Trp95, Leu66, Phe155, Thr85, Met70, Tyr92, Ser137, Leu139, Arg136, Leu130, Leu156, Ile88, Phe127, Lys63, Arg131, Pro128, Leu115, Asp152, Val11, Ala149, Glu120, Arg148, Met140, and Ile106. Beta-sitosterol revealed hydrogen bonding with Thr85 and Ser137 and hydrophobic interactions with Val138, Ala81, Ile88, Leu156, Met70, Tyr92, Phe155, Leu66, and Lys63. Bryophyllin A exhibited hydrogen bonding with Thr85 and Cys67 and hydrophobic interactions with Phe155, Arg148, Met140, Cys134, Ile88, Leu139, Ser137, Arg136, Ala81, Met84, Lys63, Leu66, Met70, Ile106, Tyr59, Leu156, Ala110, and Asp152. Moreover, mahanimbine revealed hydrophobic interactions with Arg136, Ser137, Thr85, Cys67, Tyr59, Ile106, Ala110, Leu156, Lys63, Tyr92, Leu66, Phe155, Met70, Ile88, Leu139, and Leu130. Pennogenin exhibit hydrogen bonding with Arg148 and hydrophobic interactions with Cys134, Met84, Ile88, Met70, Tyr92, Arg107, Phe155, Ile106, Leu156, Ala110, Leu66, Lys63, Met140, Tyr59, Leu139, Ser137, Arg136, Thr85, and Ala81. Rutin presented hydrogen bonding with Ser137, Thr85, and Tyr59 and hydrophobic interactions with Cys134, Met84, Glu120, Asp152, Arg148, Ala110, Leu156, Ala123, Asp124, Tyr92, Leu66, Phe155, Met70, Lys63, Cys67, Phe127, Arg136, Leu130, Ile88, and Leu139. Furthermore, solasonine revealed hydrogen bonding with Arg148 and Ser137 and hydrophobic interaction with Phe127, Ala123, Leu130, Arg136, Glu120, Lys117, Tyr59, Leu139, Ser137, Met140, Thr85, and Ile88. Solamargine exhibited hydrogen bonding with Arg131, Lys63, Asp124, Asp145, and Arg148 and hydrophobic interactions with Glu146, Asp145, Lys117, Glu120, Met70, Tyr92, Leu66, Phe155, Asp152, Met140, Arg136, Leu139, Leu130, Lys68, Ile88, Arg131, Asp124, and Ala149. Withaferin A presented hydrogen bonding with Tyr59 and Tyr92 and hydrophobic interactions with Ile106, Ala110, Leu66, Lys63, Phe155, Arg148, Leu139, Val138, Thr85, Trp185, Ser137, Met84, Cys134, Arg136, Ile88, Leu130, Met140, and Leu156. Withanone exhibited hydrophobic interactions with Ile106, Leu66, Lys63, Arg148, Tyr92, Met70, Leu130, Arg136, Ile88, Cys134, Thr85, Met84, Ser137, Leu139, Met140, Phe155, Asp152, Ala110, Leu156, and Tyr59. Chenodeoxycholic acid revealed hydrogen bonding with Ser137, Thr85, Asp152, and Tyr59 and hydrophobic interaction with Val138, Cys134, Met84, Leu139, Ile88, Arg148, Met140, Phe155, Leu156, Ile106, Ala110, Lys63, Tyr92, Leu66, Met70, and Arg136. The ribbon and 3-dimensional structure of 11 major phytocompounds and bile components in complex with 6IE9 are illustrated in Figure 3 and Figure 4. Furthermore, all the selected phytocompounds were screened for drug likeness and ADME/T.

3.2. Drug Likeness Prediction of Active Phytocompounds of Eight Medicinal Plants

Drug likeness was predicted using Molinspiration (server) to study the drug-likeness properties of active phytocompounds, which are based on the Lipinski rule of 5. Lipinski’s rule of five was followed by all selected phytocompounds and the bile component, chenodeoxycholic acid, which revealed no violation. Among all the phytocompounds, bryophyllin A, pennogenin, withaferin A, and withanone followed all the rules of drug likeness, whereas beta-sitosterol and mahanimbine exhibited one violation, which was acceptable (Table 3).

3.3. Toxicity Prediction of Active Phytocompound and Chenodeoxycholic Acid

The toxicity of phytocompounds was predicted using ProTox-II server and the results are summarized in Table 4. Among all the phytocompounds, only beta-sitosterol exhibited one violation and immune toxicity. Based on the molecular drug likeness and toxicity data, beta-sitosterol was found to be the best phytocompound, which can be used for efflux pump inhibition, and it was further selected for molecular dynamics studies.

3.4. MD Simulation of Protein–Ligand Complexes

Molecular dynamics simulation provides insight into the protein–ligand stability and protein structural flexibility of the docked complexes. The root-mean-square deviation (RMSD) plot of beta-sitasterol and 6IE9 complex exhibited significant stability in the protein pocket. These compounds fluctuate within the acceptable range between 3.2 and 5.6 Å, whereas protein Cα RMSD became stable after 25 ns and fluctuates in the range between 4.8 and 6.0 Å (Figure 5). The trajectory analysis revealed that a sharp change in the ligand RMSD at approximately 45 ns mainly occurred due to the aliphatic chain; this was also observed in the ligand RMSF plot, where fluctuation occurs in atoms 26–30 (Figure 5 and Figure 6). Several interactions were responsible for the conformational stability of the compound within the binding pocket, where hydrophobic interactions developed with residues L66, I88, Y92, M126, L130, Val141, Phe155, and L156 (Figure 7). The hydroxyl group revealed hydrogen bonding and water interactions with residues R136, S137, and R131. Moreover, MD simulation supported the docking results, where the compound interacted with residues that were linked with the molecule (Figure 2B and Figure 7). Furthermore, the thermodynamic energy analysis revealed that the average binding free energy was 138.65 ± 19.84 kcal/mol, whereas that of the docked complex was 109.18 kcal/mol (Table 5).

4. Discussion

Poor pharmacological characteristics are the major cause of late-stage failure in drug discovery. Thus, early determination of the inherent medicinal activities of the target compounds is crucial [76]. Moreover, medicinal plant species are abundant in Asia’s Himalayan woodlands, and they play a pivotal role in rural livelihoods by producing various valuable food and pharmaceutical commodities [77]. In recent years, the WHO estimated a remarkable increase in the multidrug resistance rate worldwide due to Salmonella strains [78]. Salmonella infections are gaining importance worldwide owing to their socioeconomic impact. Salmonella Typhimurium is one of the most common serovars predominantly associated with clinically reported human salmonellosis in several countries, accounting for at least 15% of infections worldwide [5]. Moreover, at least nine multidrug efflux pumps confer drug resistance in Salmonella; among these, AcrAB is constitutively expressed and is the most potent drug for intrinsic drug resistance [8]. AcrAB is a member of the RND family transporter that cooperates with TolC, an outer membrane component [79]. The AcrAB-TolC system comprising RND transporters can accumulate substrates in the periplasm rather than in the membrane or cytoplasm [80]. A common mechanism of intrinsic resistance to antimicrobial agents in Gram-negative bacteria is represented by the RND family efflux systems, which extrude a broad spectrum of antibiotics and biocides from the periplasm to the exterior of the cell [81].
The present study explored some medicinal plants, including Girardinia diversifolia, which we reported earlier for synergistic and efflux pump inhibitory activity against different strains of S. typhimurium and Staphylococcus aureus [82,83]. In contrast to our study, Mehta et al. [84] reported that methanolic extracts of Pistacia integerrima, Ocimum sanctum, C. asiatica, Momordica charantia, Zingiber officinale, and Withania somnifera exhibited synergistic activity in combination with ciprofloxacin and tetracycline against multidrug resistance. AcrAB-TolC in Salmonella Typhimurium acts as an efflux pump inhibitor. Furthermore, they reported the binding affinity (−8.2 kcal mol−1) of lariciresinol with 6EI9 (RamR). Similarly, Luhata et al. [85] reported the antibacterial activity of beta-sitosterol against S. aureus. Sen et al. [86] reported the antibacterial activity of beta-sitosterol against Escherichia coli, Pseudomonas aeruginosa, S. aureus, and Klebsiella pneumoniae. Rolta et al. [87] studied the antibacterial and antifungal activities of phytocompounds of Rheum emodin (emodin, rhein-13c6, and chrysophenodimethy ether) by molecular docking and MD simulations and found that phytocompounds of R. emodin exhibited the best interaction with bacterial and fungal targets. Similarly, Rolta et al. [71] studied the interactions of phytocompounds with the N-protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) 2020, and reported that emodin, aloe-emodin, anthrarufin, alizarine, and dantron phytocompounds efficiently inhibit SARS-CoV-2 N-protein. Salaria et al. [88] studied the in vitro and in silico antibacterial and antifungal activities of essential oil, thymol derived from Thymus serpyllum, and validated the docking results via MD simulations. The conformational changes during protein–ligand interactions have been extensively studied via MD simulation methods [89].

5. Conclusions

The major phytocompounds of 30 fine medicinal plants of the northwestern Himalayas were selected for molecular docking study with the 6EI9 (RamR) target protein of S. typhimurium. Among all the selected phytocompounds, 11 phytocompounds exhibited the best activity compared to the standard drugs. Drug likeness and toxicity data revealed that beta-sitosterol, a major phytocompound of G. diversifolia (Link) Friis, is nontoxic in nature and follows the drug likeness rule. Moreover, MD simulation of beta-sitosterol in complex with 6EI9 was found to be stable between 0 and 100 ns time period. In this study, we found that beta-sitosterol is a potential plant-based drug for treating S. typhimurium infection. Furthermore, this study needs to be validated through in vitro and in vivo experiments.

Author Contributions

Conceptualization, R.R., D.S., J.M.; software, O.A.F., P.P.S. and B.R.; formal analysis, R.R., J.M., D.S., O.A.; writing—original draft preparation, N.K., A.C., E.H.C. and N.K.K.; writing—review and editing, N.K.K. and E.H.C.; supervision, N.K.K. and E.H.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Research Foundation (NRF) of Korea, funded by the Korean government (NRF-2021R1A6A1A03038785, 2021R1F1A1055694, 2021R1C1C1013875) and by Kwangwoon University in 2021.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are included in this manuscript.

Acknowledgments

The authors acknowledge Shoolini University, Solan (India) to support this research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Prouty, A.M.; Van Velkinburgh, J.C.; Gunn, J.S. Salmonella enterica serovar typhimurium resistance to bile: Identification and characterization of the tolQRA cluster. J. Bacteriol. 2002, 184, 1270–1276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Begley, M.; Sleator, R.D.; Gahan, C.G.; Hill, C. Contribution of three bile-associated loci, bsh, pva, and btlB, to gastrointestinal persistence and bile tolerance of Listeria monocytogenes. Infect. Immun. 2005, 73, 894–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kabir, M.R.; Hossain, M.A.; Paul, S.K.; Mahmud, C.; Ahmad, S.; Mahmud, N.U.; Sultana, S.; Yesmin, T.; Hoque, S.M.; Habiba, U.; et al. Enteropathogens associated with acute diarrhea in a tertiary hospital of Bangladesh. Mymensingh Med. J. 2012, 21, 618–623. [Google Scholar]
  4. Kozak, G.K.; MacDonald, D.; Landry, L.; Farber, J.M. Foodborne outbreaks in Canada linked to produce: 2001 through 2009. J. Food Prot. 2013, 76, 173–183. [Google Scholar] [CrossRef]
  5. Scallan, E.; Hoekstra, R.M.; Angulo, F.J.; Tauxe, R.V.; Widdowson, M.A.; Roy, S.L.; Jones, J.L.; Griffin, P.M. Foodborne illness acquired in the United States—Major pathogens. Emerg. Infect. Dis. 2011, 17, 7–15. [Google Scholar] [CrossRef] [PubMed]
  6. Gopinath, S.; Lichtman, J.S.; Bouley, D.M.; Elias, J.E.; Monack, D.M. Role of disease-associated tolerance in infectious superspreaders. Proc. Natl. Acad. Sci. USA 2014, 111, 15780–15785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Strugnell, R.A.; Scott, T.A.; Wang, N.; Yang, C.; Peres, N.; Bedoui, S.; Kupz, A. Salmonella vaccines: Lessons from the mouse model or bad teaching? Curr. Opin. Microbiol. 2014, 17, 99–105. [Google Scholar] [CrossRef] [PubMed]
  8. Nishino, K.; Latifi, T.; Groisman, E.A. Virulence and drug resistance roles of multidrug efflux systems of Salmonella enterica serovar Typhimurium. Mol. Microbiol. 2006, 59, 126–141. [Google Scholar] [CrossRef]
  9. Prouty, A.M.; Brodsky, I.E.; Falkow, S.; Gunn, J.S. Bile-salt-mediated induction of antimicrobial and bile resistance in Salmonella typhimurium. Microbiology 2004, 150, 775–783. [Google Scholar] [CrossRef]
  10. Abouzeed, Y.M.; Baucheron, S.; Cloeckaert, A. RamR mutations involved in efflux-mediated multidrug resistance in Salmonella enterica serovar Typhimurium. Antimicrob. Agents Chemother. 2008, 52, 2428–2434. [Google Scholar] [CrossRef] [Green Version]
  11. Gordon, M.C.; David, J.N. Natural product drug discovery in the next millennium. Pharm. Biol. 2001, 39, 8–17. [Google Scholar]
  12. Lopez-Vallejo, F.; Caulfield, T.; Martínez-Mayorga, K.; Giulianotti, M.A.; Houghten, R.A.; Nefzi, A.; Medina-Franco, J.L. Integrating virtual screening and combinatorial chemistry for accelerated drug discovery. Comb. Chem. High Throughput Screen. 2011, 14, 475–487. [Google Scholar] [CrossRef] [PubMed]
  13. Gupta, M.; Sharma, R.; Kumar, A. Docking techniques in pharmacology: How much promising? Comput. Biol. Chem. 2018, 76, 210–217. [Google Scholar] [CrossRef] [PubMed]
  14. War, A.R.; Taggar, G.K.; Hussain, B.; Taggar, M.S.; Nair, R.M.; Sharma, H.C. Plant defence against herbivory and insect adaptations. AoB Plants 2018, 10, ply037. [Google Scholar]
  15. O’Boyle, N.M.; Banck, M.; James, C.A.; Morley, C.; Vandermeersch, T.; Hutchison, G.R. Open Babel: An open chemical toolbox. J. Cheminform. 2011, 3, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [Green Version]
  17. Boktapa, N.R.; Sharma, A.K. Wild medicinal plants used by local communities of Manali, Himachal Pradesh, India. Ethnobot. Leafl. 2010, 14, 259–267. [Google Scholar]
  18. Sharma, R.; Manhas, R.K.; Magotra, R. Ethnoveterinary remedies of diseases among milk yielding animals in Kathua, Jammu and Kashmir, India. J. Ethnopharmacol. 2012, 141, 265–272. [Google Scholar] [CrossRef]
  19. Baba, M.; Asano, R.; Takigami, I.; Takahashi, T.; Ohmura, M.; Okada, Y.; Sugimoto, H.; Arika, T.; Nishino, H.; Okuyama, T. Studies on Cancer Chemoprevention by Traditional Folk Medicines XXV.-Inhibitory Effect of Isoliquiritigenin on Azoxymethane-Induced Murine Colon Aberrant Crypt Focus Formation and Carcinogenesis. Biol. Pharm. Bull. 2002, 25, 247–250. [Google Scholar] [CrossRef] [Green Version]
  20. Kupeli, E.; Kosar, M.; Yesilada, E.; Baser, K.H.C. A comparative study on the anti-inflammatory, antinociceptive and antipyretic effects of isoquinoline alkaloids from the roots of Turkish Berberis species. Life Sci. 2002, 72, 645–657. [Google Scholar] [CrossRef]
  21. Duke, J.A.; Ayensu, E.S. Medicinal Plants of China; Reference Publications, Inc.: Algonac, MI, USA, 1985; ISBN 0-917256-20-4. [Google Scholar]
  22. Phillips, R.; Foy, N. Herbs; Pan Books Ltd.: London, UK, 1990; ISBN 0-330-30725-8. [Google Scholar]
  23. Kumar, S.; Chand, G.; Sankhyan, P. Herbal folk remedies for curing various ailments in lug valley of district kullu, himachal pradesh (n.w. himalaya). Int. J. Ayurvedic Herbmed. 2013, 3, 1308–1314. [Google Scholar]
  24. Dhananjay, J.; Deshpande, A. Hand Book of Medicinal Herbs; Agrobios: Jodhpur, India, 2006. [Google Scholar]
  25. Hasimi, N.; Ertas, A.; Oral, E.V.; Yener, I.; Alkan, H.; Boga, M.; Yılmaz, M.A.; Yener, I.; Gazioglu, I.; Ozaslan, C.; et al. Chemical profile of Malva neglecta and Malvellasherardiana by Lc-MS/MS, GC/MS and their anticholinesterase, antimicrobial and antioxidant properties with aflatoxin-contents. Marmara Pharm. J. 2017, 21, 471–484. [Google Scholar] [CrossRef] [Green Version]
  26. Taherian, R.; Taherian, M.; Maghsoudi, H.; Haj-alahyari, S. The effect of aqueous extract of Malva neglecta on expression of inflammatory biomarkers involved in pain in synoviocytes and THP -1 cells as a model of monocyte/macrophage and human cartilage cells in osteoarthritis. J. Cell. Mol. Anesth. 2017, 2, 149–156. [Google Scholar]
  27. Duke, J.A.; Bugenschutz-godwin, M.J.; Du collier, J.; Duke, P.K. Hand Book of Medicinal Herbs, 2nd ed.; CRC Press: Boca Raton, FL, USA, 2002. [Google Scholar]
  28. Caliskan, O.; Polat, A.A. Phytochemical and antioxidant properties of selected fig (Ficus carica L.) accessions from the eastern Mediterranean region of Turkey. Sci. Hortic. 2011, 128, 473–478. [Google Scholar] [CrossRef]
  29. Sirohi, B.; Sagar, R. Antipyretic activity of hydroalcholic extract of D. hatagirea roots and Lavandula stoechas flowers on Brewers yeast induced Pyrexia in Wistar rats. J. Drug Discov. Ther. 2019, 9, 701–704. [Google Scholar]
  30. Sharma, S.; Jain, P.K.; Parkhe, G. Extraction, phytochemical screening and anti-inflammatory activity of hydro ethanolic extracts of roots of D. hatagirea. J. Drug Discov. Ther. 2020, 19, 86–90. [Google Scholar]
  31. Kumar, N.; Shikha, S.; George, V.C.; Suresh, P.K.; Kumar, R.A. Anticancer and anti-metastatic activities of rheum emodi rhizome chloroform extracts. Asian J. Pharm. Clin. Res. 2012, 5, 189–194. [Google Scholar]
  32. Arvindekar, A.U.; Laddha, K.S. An efficient microwave-assisted extraction of anthraquinones from Rheum emodi: Optimisation using RSM, UV and HPLC analysis and antioxidant studies. Ind. Crop. Prod. 2016, 83, 587–595. [Google Scholar] [CrossRef]
  33. Ahmad, W.; Zaidi, S.M.; Mujeeb, M.; Ansari, S.H.; Ahmad, S. HPLC and HPTLC methods by design for quantitative characterization and in vitro anti-oxidant activity of polyherbal formulation containing Rheum emodi. J. Chromatogr. Sci. 2014, 52, 911–918. [Google Scholar] [CrossRef] [Green Version]
  34. Bhattacharjee, S.; Bhattacharya, S.; Jana, S.; Singh, D. Review on medicinally important species of picrorhiza. Int. J. Pharm. Res. Biosci. 2013, 2, 1–16. [Google Scholar]
  35. Bhattacharyya, P.; Kumaria, S.; Bose, B.; Paul, P.; Tandon, P. Evaluation of genetic stability and analysis of phytomedicinal potential in micro propagated plants of Rumex nepalensis—A medicinally important source of pharmaceutical biomolecules. J. Appl. Res. Med. Aromat. Plants 2017, 6, 80–91. [Google Scholar]
  36. Pandey, Y.; Bhatt, S.S. Overview of Himalayan yellow raspberry (Rubus ellipticus Smith): A nutraceutical plant. J. Appl. Nat. Sci. 2016, 8, 494–499. [Google Scholar] [CrossRef] [Green Version]
  37. Prajapati, S.N.; Parmar, K.A. Anti-viral and in-vitro free radical scavenging activity of leaves of Rubia cordifolia. Int. J. Phytomed. 2011, 3, 98–107. [Google Scholar]
  38. Grieve, M. A Modern Herbal; Dover Publications Inc.: New York, NY, USA, 1981; Volume 2, pp. 562–566. [Google Scholar]
  39. Mabey, R. The New Age Herbalist; Macmillan Publishing Company: New York, NY, USA, 1988; p. 113. [Google Scholar]
  40. Junior, G.M.V.; Rocha, C.Q.; Rodrigues, T.S.; Hiruma-Lima, C.A.; Vilegas, W. New steroidal saponins and antiulcer activity from Solanum paniculatum L. Food Chem. 2015, 186, 160–167. [Google Scholar] [CrossRef]
  41. Kadima, J.N.; Kasali, F.M.; Bavhure, B.; Mahano, A.O.; Bwironde, F.M. Comparative antidiabetic potential and survival function of Harungana madagascariensis, Physalis peruviana, Solanum Americanum and Tithonia diversifolia extracts on alloxan-induced diabetes in guinea-pigs. Int. J. Pharm. Pharm. Res. 2016, 5, 196–206. [Google Scholar]
  42. Pant, S.; Samant, S.S. Ethnobotanical observations in the Mornaula reserve forest of Komoun, West Himalaya, India. Ethnobot. Leafl. 2010, 14, 193–217. [Google Scholar]
  43. Rani, S.; Rana, J.C.; Rana, P.K. Ethnomedicinal plants of Chamba district, Himachal Pradesh, India. J. Med. Plant Res. 2013, 7, 3147–3157. [Google Scholar]
  44. Jugran, A.K.; Rawat, S.; Bhatt, I.D.; Rawal, R.S. Valeriana jatamansi: An herbaceous plant with multiple medicinal uses. Phytother. Res. 2019, 33, 482–503. [Google Scholar] [CrossRef]
  45. Li, Y.B.; Chen, C.; Mao, S.; Guo, C.; Zhao, T.T.; Wu, L.L.; Wang, X.Y.; Liu, A.; Yang, Z.Y. Anti-depression Effect and Mechanism of Valerianae Jatamansi Rhizoma et Radix. Chin. J. Exp. Tradit. Med. Formulae 2020, 26, 235–240. [Google Scholar]
  46. Verma, G.; Dua, V.K.; Agarwal, D.D.; Atul, P.K. Anti-malarial activity of Holarrhenaantidysenterica and Viola canes-cens, plants traditionally used against malaria in the Garhwal region of north-west Himalaya. Malar. J. 2011, 10, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Masood, M.; Arshad, M.; Asif, S.; Chaudhari, S.K. Viola canescens: Herbal wealth to be conserved. J. Bot. 2014, 2014, 1–6. [Google Scholar] [CrossRef] [Green Version]
  48. Bhatti, V.; Vashishtha, D. Indigenous plants in traditional healthcare system in Kedarnath valley of western Himalaya. Indian J. Tradit. Knowl. 2008, 7, 300–310. [Google Scholar]
  49. Rokaya, M.B.; Münzbergová, Z.; Timsina, B. Ethnobotanical study of medicinal plants from the Humla district of western Nepal. J. Ethnopharmacol. 2010, 130, 485–504. [Google Scholar] [CrossRef]
  50. Shrestha, S.S.; Sut, S.; Ferrarese, I.; Marco, B.D.S.; Zengin, G.; De Franco, M.; Pant, D.R.; Mahomoodally, M.F.; Ferri, N.; Biancorosso, N.; et al. Himalayan Nettle Girardinia diversifolia as a Candidate Ingredient for Pharmaceutical and Nutraceutical Applications—Phytochemical Analysis and In Vitro Bioassays. Molecules 2020, 25, 1563. [Google Scholar] [CrossRef] [Green Version]
  51. Biswas, K.R.; Khan, T.; Monalisa, M.N.; Swarna, A.; Ishika, T.; Rahman, M. Medicinal plants used by folk medicinal practitioners of four adjoining villages of Narail and Jessore Districts, Bangladesh. Am. Eurasian J. Sustain. Agric. 2011, 5, 23–33. [Google Scholar]
  52. Pattanayak, P.; Behera, P.; Das, D.; Panda, S.K. Ocimum sanctum Linn. A reservoir plant for therapeutic applications: An overview. Pharmacogn. Rev. 2010, 4, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Scartezzini, P.; Speroni, E. Review on some plants of Indian traditional medicine with antioxidant activity. J. Ethnopharmacol. 2000, 71, 23–43. [Google Scholar] [CrossRef]
  54. Pitipanapong, J.; Chitprasert, S.; Goto, M.; Jiratchariyakul, W.; Sasaki, M.; Shotipruk, A. New approach for extraction of charantin from Momordicacharantia with pressurized liquid extraction. Sep. Purif. Technol. 2007, 52, 416–422. [Google Scholar] [CrossRef]
  55. Omoya, F.O.; Akharaiyi, F.C. Mixture of honey and ginger extract for antibacterial assessment on some clinical isolates. Int. J. Pharm. Biomed. Res. 2011, 2, 39–47. [Google Scholar]
  56. Montalván, V.; Gallo, M.; Rojas, E. A 25 years-old woman with a postvaccine thalamic pseudotumoral lesion. Rev. Clin. Esp. 2015, 215, 468–472. [Google Scholar] [CrossRef] [PubMed]
  57. Priya, G.; Parminder, N.; Jaspreet, S. Antimicrobial and antioxidant activity on Emblica officinalis seed extract. Int. J. Res. Ayurveda Pharm. 2012, 3, 591–596. [Google Scholar]
  58. Mahata, S.; Pandey, A.; Shukla, S.; Tyagi, A.; Husain, S.A.; Das, B.C.; Bharti, A.C. Anticancer Activity of Phyllanthus emblica Linn. (Indian Gooseberry): Inhibition of Transcription Factor AP-1 and HPV Gene Expression in Cervical Cancer Cells. Nutr. Cancer 2013, 65, 88–97. [Google Scholar] [CrossRef]
  59. Parvu, M.; Moţ, C.A.; Parvu, A.E.; Mircea, C.; Stoeber, L.; Rosca-Casian, O.; Ţigu, A.B. Allium sativum extract chemical composition, antioxidant activity and antifungal effect against Meyerozymaguillier mondii and Rhodotorula mucilaginosa causing onychomycosis. Molecules 2019, 24, 3958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Mahata, S.; Maru, S.; Shukla, S.; Pandey, A.; Mugesh, G.; Das, B.C.; Bharti, A.C. Anticancer property of Bryophyllum pinnata (Lam.) Oken. leaf on human cervical cancer cells. BMC Complement. Altern. Med. 2012, 12, 15. [Google Scholar] [CrossRef] [Green Version]
  61. Satyal, P.; Paudel, P.; Raut, J.; Deo, A.; Dosoky, N.S.; Setzer, W.N. Volatile constitu-ents of Pinus roxburghii from Nepal. Pharmacogn. Res. 2013, 5, 43–48. [Google Scholar]
  62. Lutterodt, H.; Luther, M.; Slavin, M.; Yin, J.J.; Parry, J.; Gao, J.-M.; Yu, L. Fatty acid profile, thymoquinone content, oxidative stability, and antioxidant properties of cold-pressed black cumin seed oils. Food Sci. Technol. 2010, 43, 1409–1413. [Google Scholar]
  63. Ali, A.A.; Al-Rahwi, K.; Lindequist, U. Some medicinal plants used in Yemeni herbal medicine to treat malaria. Afr. J. Tradit. Complementary Altern. Med. 2004, 1, 72–76. [Google Scholar] [CrossRef]
  64. Desai, S.N.; Patel, D.K.; Devkar, R.V.; Patel, P.V.; Ramachandran, A.V. Hepatoprotective potential of polyphenol rich extract of Murrayakoenigii, L.: An in vivo study. Food Chem. Toxicol. 2012, 50, 310–314. [Google Scholar] [CrossRef] [PubMed]
  65. Mahipal, P.; Pawar, R.S. Nephroprotective effect of Murraya koenigii on cyclophosphamide induced nephrotoxicity in rats. Asian Pac. J. Trop. Med. 2017, 10, 808–812. [Google Scholar] [CrossRef]
  66. Defillipo, P.P.; Raposo, A.H.; Fedoce, A.G.; Ferreira, A.S.; Polonini, H.C.; Gattaz, W.F.; Raposo, N.R. Inhibition of cPLA2 and sPLA2 activities in primary cultures of rat cortical neurons by Centella asiatica water extract. Nat. Prod. Commun. 2012, 7, 841–843. [Google Scholar] [CrossRef]
  67. Yamasaki, S.; Nakashima, R.; Sakurai, K.; Baucheron, S.; Giraud, E.; Doublet, B.; Cloeckaert, A.; Nishino, K. Crystal structure of the multidrug resistance regulator RamR complexed with bile acids. Sci. Rep. 2019, 9, 1–8. [Google Scholar] [CrossRef] [PubMed]
  68. Rosell, R.; Crinó, L. Pemetrexed combination therapy in the treatment of non–small cell lung cancer. Semin. Oncol. 2002, 29, 23–29. [Google Scholar]
  69. Banerjee, P.; Eckert, A.O.; Schrey, A.K.; Preissner, R. ProTox-II: A webserver for the prediction of toxicity of chemicals. Nucleic Acids Res. 2018, 46, 257–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Rolta, R.; Yadav, R.; Salaria, D.; Trivedi, S.; Imran, M.; Sourirajan, A.; Baumler, D.J.; Dev, K. In silico screening of hundred phytocompounds of ten medicinal plants as potential inhibitors of nucleocapsid phosphoprotein of COVID-19: An approach to prevent virus assembly. J. Biomol. Struct. Dyn. 2020, 1–8. [Google Scholar] [CrossRef]
  71. Rolta, R.; Salaria, D.; Kumar, V.; Sourirajan, A.; Dev, K. Phytocompounds of Rheum emodi, Thymus serpyllum and Artemisia annua inhibit COVID-19 binding to ACE2 receptor: In silico approach. Curr. Pharmacol. Rep. 2021, 7, 135–149. [Google Scholar] [CrossRef]
  72. Salaria, D.; Rolta, R.; Sharma, N.; Dev, K.; Sourirajan, A.; Kumar, V. In silico and In vitro evaluation of the anti-inflammatory and antioxidant potential of Cymbopogon citratus from North-western Himalayas. BioRxiv 2020. [Google Scholar] [CrossRef]
  73. Cheng, F.; Li, W.; Zhou, Y.; Shen, J.; Wu, Z.; Liu, G.; Lee, P.W.; Tang, Y. AdmetSAR: A comprehensive source and free tool for assessment of chemical ADMET properties. J. Chem. Inf. Model. 2012, 52, 3099–3105. [Google Scholar] [CrossRef]
  74. Yang, H.; Lou, C.; Sun, L.; Li, J.; Cai, Y.; Wang, Z.; Tang, Y. AdmetSAR 2.0: Web-service for prediction and optimization of chemical ADMET properties. Bioinformatics 2019, 35, 1067–1069. [Google Scholar]
  75. Schrodinger, S. Release 2020-1: Protein Preparation Wizard; Epik, LLC: New York, NY, USA; Impact, Schrödinger, LLC: New York, NY, USA; Prime, Schrodinger, LLC: New York, NY, USA, 2020. [Google Scholar]
  76. Kennedy, T. Managing the drug discovery/development interface. Drug Discov. Today 1997, 2, 436–444. [Google Scholar] [CrossRef]
  77. Kala, C.P. Ethnomedicinal botany of the Apatani in the eastern Himalayan region of India. J. Ethnobiol. Ethnomed. 2005, 1, 11–18. [Google Scholar] [CrossRef] [Green Version]
  78. World Health Organization: Drug-Resistant Salmonella. 2016. Available online: http://www.who.int/mediacentre/factsheets/fs139/en/ (accessed on 15 May 2021).
  79. Koronakis, V.; Sharff, A.; Koronakis, E.; Luisi, B.; Hughes, C. Crystal structure of the bacterial membrane protein TolC central to multidrug efflux and protein export. Nature 2000, 405, 914–919. [Google Scholar] [CrossRef]
  80. Nikaido, H. Multidrug efflux pumps of gram-negative bacteria. J. Bacteriol. 1996, 178, 5853–5859. [Google Scholar] [CrossRef] [Green Version]
  81. Nakae, T.; Saito, K.; Nakajima, A. Effect of sulbactam on anti-pseudomonal activity of beta-lactam antibiotics in cells producing various levels of the MexAB-OprM efflux pump and beta-lactamase. Microbiol. Immunol. 2000, 44, 997–1001. [Google Scholar] [CrossRef] [Green Version]
  82. Mehta, J.; Jandaik, S.U. Evaluation of phytochemicals and synergistic interaction between plant extracts and antibiotics for efflux pump inhibitory activity against salmonella enterica serovar typhimurium strains. Int. J. Pharm. Pharm. Sci. 2016, 8, 217–223. [Google Scholar] [CrossRef] [Green Version]
  83. Jandaik, S.U.; Mehta, J.; Mohan, M. Synergistic and efflux pump inhibitory activity of plant extracts and antibiotics on staphylococcus aureus strains. Asian J. Pharm. Clin. Res. 2016, 9, 277–282. [Google Scholar]
  84. Mehta, J.; Rolta, R.; Dev, K. Role of medicinal plants from Northwestern Himalayas as an efflux pump inhibitor against MDR AcrAB-TolC Salmonella enterica serovar typhimurium: In vitro and In silico studies. J. Ethanopharmacol. 2021, 114589. [Google Scholar] [CrossRef] [PubMed]
  85. Luhata, L.P.; Usuki, T. Antibacterial activity of β-sitosterol isolated from the leaves of Odontonema strictum (Acanthaceae). Bioorg. Med. Chem. Lett. 2021, 48, 128248. [Google Scholar] [CrossRef]
  86. Sen, A.; Dhavan, P.; Shukla, K.K.; Singh, S.; Tejovathi, G. Analysis of IR, NMR and antimicrobial activity of β-sitosterol isolated from Momordica charantia. Sci. Secur. J. Biotechnol. 2012, 1, 9–13. [Google Scholar]
  87. Rolta, R.; Salaria, D.; Kumar, V.; Patel, C.N.; Sourirajan, A.; Baumler, D.J.; Dev, K. Molecular docking studies of phytocompounds of Rheum emodi Wall with proteins responsible for antibiotic resistance in bacterial and fungal pathogens: In silico approach to enhance the bio-availability of antibiotics. J. Biomol. Struct. Dyn. 2020, 1–5. [Google Scholar] [CrossRef]
  88. Salaria, D.; Rolta, R.; Patel, C.N.; Dev, K.; Saurirajan, A.; Kumar, V. In vitro and in silico analysis of Thymus serpyllum essential oil as bioactivity enhancer of antibacterial and antifungal agents. J. Biomol. Struct. Dyn. 2021, 1–20. [Google Scholar] [CrossRef]
  89. Li, W.; Shen, J.; Liu, G.; Tang, Y.; Hoshino, T. Exploring coumarin egress channels in human cytochrome p450 2a6 by random acceleration and steered molecular dynamics simulations. Proteins 2011, 79, 271–281. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic of experimentation.
Figure 1. Schematic of experimentation.
Biomedicines 09 01402 g001
Figure 2. Two-dimensional structure of 11 phytocompounds and bile components in complex with RamR protein of Salmonella Typhimurium: (A) asiaticoside, (B) beta-sitosterol, (C) bryophyllin A, (D) madecassoside, (E) mahanimbine, (F) pennogenin, (G) rutin, (H) solamargine, (I) solasonine, (J) withaferin A, (K) withanone, and (L) chenodeoxycholic acid.
Figure 2. Two-dimensional structure of 11 phytocompounds and bile components in complex with RamR protein of Salmonella Typhimurium: (A) asiaticoside, (B) beta-sitosterol, (C) bryophyllin A, (D) madecassoside, (E) mahanimbine, (F) pennogenin, (G) rutin, (H) solamargine, (I) solasonine, (J) withaferin A, (K) withanone, and (L) chenodeoxycholic acid.
Biomedicines 09 01402 g002
Figure 3. Ribbon structure of 11 active phytocompounds and bile component in complex with RamR protein of Salmonella Typhimurium: (A) asiaticoside, (B) beta-sitosterol, (C) bryophyllin A, (D) madecassoside, (E) mahanimbine, (F) pennogenin, (G) rutin, (H) solamargine, (I) solasonine, (J) withaferin A, (K) withanone, and (L) chenodeoxycholic acid.
Figure 3. Ribbon structure of 11 active phytocompounds and bile component in complex with RamR protein of Salmonella Typhimurium: (A) asiaticoside, (B) beta-sitosterol, (C) bryophyllin A, (D) madecassoside, (E) mahanimbine, (F) pennogenin, (G) rutin, (H) solamargine, (I) solasonine, (J) withaferin A, (K) withanone, and (L) chenodeoxycholic acid.
Biomedicines 09 01402 g003
Figure 4. Three-dimensional interactions of active phytocompounds and standard drugs in complex with RamR protein of Salmonella Typhimurium: (A) asiaticoside, (B) beta-sitosterol, (C) bryophyllin A, (D) madecassoside, (E) mahanimbine, (F) pennogenin, (G) rutin, (H) solamargine, (I) solasonine, (J) withaferin A, (K) withanone, and (L) chenodeoxycholic acid.
Figure 4. Three-dimensional interactions of active phytocompounds and standard drugs in complex with RamR protein of Salmonella Typhimurium: (A) asiaticoside, (B) beta-sitosterol, (C) bryophyllin A, (D) madecassoside, (E) mahanimbine, (F) pennogenin, (G) rutin, (H) solamargine, (I) solasonine, (J) withaferin A, (K) withanone, and (L) chenodeoxycholic acid.
Biomedicines 09 01402 g004
Figure 5. Root-mean-square deviation plot of C-alpha of beta-sitasterol in complex with 6IE9at 100 ns depicting the quality of the pose with respect to time.
Figure 5. Root-mean-square deviation plot of C-alpha of beta-sitasterol in complex with 6IE9at 100 ns depicting the quality of the pose with respect to time.
Biomedicines 09 01402 g005
Figure 6. Ligand RMSF plot of beta-sitasterol in complex with 6IE9: X-axis depicts atom index and Y-axis reveals RMSF (Å).20 stands for atom number which is oxygen.
Figure 6. Ligand RMSF plot of beta-sitasterol in complex with 6IE9: X-axis depicts atom index and Y-axis reveals RMSF (Å).20 stands for atom number which is oxygen.
Biomedicines 09 01402 g006
Figure 7. Histogram plot of beta-sitasterol in complex with 6IE9: X-axis depicts interactive amino acids and Y-axis reveals interaction fractions. (Gray color indicates Hydrophobic interactions, Blue color indicates water bridges and Green color indicates Hydrogen bonding).
Figure 7. Histogram plot of beta-sitasterol in complex with 6IE9: X-axis depicts interactive amino acids and Y-axis reveals interaction fractions. (Gray color indicates Hydrophobic interactions, Blue color indicates water bridges and Green color indicates Hydrogen bonding).
Biomedicines 09 01402 g007
Table 1. Medicinal plants used for molecular docking with their uses in various ailments.
Table 1. Medicinal plants used for molecular docking with their uses in various ailments.
S. No.PhytocompoundsBotanical Names (Family)Common/Local NameAilments
1.(Z)-Ligustilide Angelica glauca
Edgew. (Apiaceae)
ChoraStimulant, appetizer, arthritis, carminative, diaphoretic, diuretic, constipation, debility, joint problems, bronchitis, dysentery, menorrhea, stomach disorders, vomiting [17,18].
Angelicide
2.P-coumaric acid Heracleum lanatum Michx (Apiaceae) PatralaFever, abdominal cramps, leukoderma, aphrodisiac, digestive, mildly expectorant and sedative, nausea, tumor [19].
Scopoletin
3.Palmatine Berberis aristata DC.
(Berberidaceae)
KashmalAcidity, eye infection, microbes, fever, hepatotoxic, hyperglycemic, lipidemic, cancer, oxidative stress diarrhea, hemorrhoids, osteoporosis, HIV-AIDS, diabetes, jaundice, wound healing [20].
Rutin
4.PodophyllotoxinSinopodophyllum hexandrum (Royle) T.S. Ying (Berberidaceae)BankakdiCancer, snakebite, jaundice, stomachache, intestinal purgative, vomiting, necrotic wounds, tumor, arthritis [21,22].
Quercetin
5.AstragalinChenopodium album L.
(Amaranthaceae)
BaathuParasitic worms, inflammation, fever, arthritis, constipation, toothache, bug bites, sunstroke, tooth decay [21].
Kaempferol
6.ApigeninSedum glaucophyllum R.T. Clausen (Crassulaceae)Mochu-gha, ludruBurn, cut, abscesses, blisters [23].
Luteolin
7.PhytolSolena amplexicaulis (Lam.) Gandhi (Cucurbitaceae)KakdiCancer, oxidative stress, inflammation, tumor, antimicrobial, diuretic, fever, jaundice [24].
Carane
8.HydroxytyrosolMalva neglecta Wall
(Malvaceae)
SonchalConstipation, women sterility, wound healing, hemorrhoids, asthma, diarrhea, rheumatic pain, stomachache, abdominal pain, renal diseases, throat infection, common cold, stomachache, antimicrobial, oxidative stress, inflammation, stress, liver damage, ulcer, cancer, bronchitis, kidney stone [25,26].
Hexatriacontane
9.CyanidinFicus carica L. (Moraceae)Common figOxidative stress, Cancer, colic, indigestion, loss of appetite, diarrhea, sore throats, coughs, bronchial problems, heart disease, liver problem, lowering of blood sugar, cholesterol-lowering, inflammation, Antimicrobial, relieve spasm of involuntary muscle, fever, TB, platelet aggregation inhibitor, mutagen [27,28].
Psoralen
10.MilitarineDactylorhiza hatagirea (D.Don) Soo (Orchidaceae)Panja, SalampanjaWound healing, inflammation, bleeding, fever, cancer, diabetes, neurological function, burns, and bronchitis [29,30].
Resveratrol
11.Gallic acidRheum australe D.Don
(Polygonaceae)
Chuchi, ChukariDiabetes, inflammation, oxidative stress, cancer, gastric disorder, cuts & wounds, fractured bones, liver damage, immune-enhancing, lower blood glucose, smallpox, muscle sprain [31,32].
Rhein
12.NepodinRumex hastatus D.Don (Polygonaceae)AlmoruJaundice, hepatitis, Blood purification, Scurvy, Diuretic, cooling, astringent, constipation, oxidative stress, snakebites, foot and mouth infections, asthma, cough, headache, diarrhea, dysentery, fever, weakness, and scabies [33].
Rumexoside
13.KutkosidePicrorrhizakurooa Royle (Plantaginaceae)KaruLiver damage, oxidative stress, cancer, asthma, stimulate immune system, neuritogenic, neuron degeneration, jaundice, allergy, piles, leukoderma, snake bite, liver disease, fever, parasitic worms, improving heart muscle contraction, high blood pressure, diabetes, cold, cough, stomach ache [34].
Picroside III
14.AloesinRumex nepalensis Meisn (Polygonaceae)Nepal dockPurgative, oxidative stress, fever, inflammation, tumor, diabetic, mental disorder, Wound healing, analgesic and CNS depressant, skeletal muscle relaxant [35].
Orcinol glucoside
15.CatechinRubus ellipticus Sm. (Rosaceae)Akhe, Yellow Himalayan raspberryDysentery, oxidative stress, diabetes, tumor, Nephroprotective, sore throats, cold, colic, constipation, gastritis, dysentery, diarrhea [36].
Caffeic acid
16.RubiadinRubia cordifolia L. (Rubiaceae)MishtuImmune-related diseases inflammation, urinary infections, bone ache, skin diseases, vertigo, insomnia, rheumatism, tuberculosis, hematemesis, menstrual disorders, contusions [37].
Mollugin
17.Verbascoside Verbascum thapsus L. (Scrophulariaceae)JanglitamakuPain, muscle spasm, bleeding, nerve tonic, wounds, allergy, cancer, oxidative stress, blood pressure, anxiety, inflammation, sepsis, diuretic, cough, skin diseases, cuts, wounds and swelling, diarrhea [38,39].
Aucubin
18.SolasonineSolanum americanum Mill. (Solanaceae)Bara lianchuHealing, dental caries, bladder spasm, joint pains, cooling, cough, gastric ulcer, protozoal infections, diabetes, inflammation [40,41].
Solamargine
19.PennogeninTrilliumgovanianum Wall. Ex D. Don (Melanthiaceae)Nag ChhatriDysentery, wounds, inflammation, antiseptic, boils, menstrual and sexual disorders, pain, inflammation, Leishmanial infection, cancer, wound [42,43].
2,4-Decadienal
20.Protocatechuic acidValeriana jatamansi Jones (Caprifoliaceae)NihaniCuts, wounds, skin disorders, analgesic, anxiety disorder, tranquilizing hypnotic, irritable bowel syndrome, epilepsy, snake poisoning, hyperlipidemia, depressive insomnia, rotavirus enteritis [44,45].
Valtrate
21.Methyl salicylateViola canescens Wall. (Violaceae)BankshaCough, cold, fever, jaundice, malaria, protozoa infection, cancer, flatulence, inflammation or irritation, bleeding abrations, fever, respiratory problems, sepsis, fever [46,47].
Emetine
22.β-sitosterolGirardinia diversifolia (Link) Friis
(Urticaceae)
ZaranCytotoxic, Snake bite, Muscles sprain, constipation, headaches, fever, ringworm, gastric troubles, eczema, chest and joint pain, rheumatism, tuberculosis, headache, joint aches, diabetes, asthma, stomach inflammation, gonorrhea, delivery problems, bone fracture, internal injury, blood purification [48,49,50].
Scopoletin
23.AtropineDatura stramonium L.
(Solanaceae)
DhaturaAsthma, inflammation, pain and spasm in irritable bowel, gout, madness, epilepsy, depression, burns, rheumatism Parkinson’s disease, piles, pain [51].
Scopolamine
24.Eugenol Ocimum sanctum L.
(Labiatae)
TulsiBronchial asthma, fever, cold, cough, malaria, dysentery, convulsions, diarrhea, arthritis, skin diseases, insect bites, gastric, liver and heart disorder, diabetes stomachache, headache, inflammation, tuberculosis, stress, poisoning, leukoderma [52].
Cirsilineol
25.CharantinMomordica charantia L.
(Cucurbitaceae)
Bitter GourdCholesterol, HIV, gout, jaundice, abdominal pain, kidney (stone), rheumatism, fever, scabies, ulcer, inflammation, leukemia, diabetes, tumor, diabetes [53,54].
Momordicine
26.GingerolZingiber officinale Roscoe (Zingiberaceae)GingerInflammation, nausea, analgesic, fever, dysentery, heartburn, flatulence, diarrhea, diabetes, carminative, stimulant to GIT, relieve spasm of involuntary muscle, digestion, vasodilation, cough, asthma, pain, flatulence, constipation [55].
Lariciresinol
27.WithanoneWithania somnifera (L.) Dunal (Solanaceae)AshwagandhaAbortion, clear or open the natural ducts of the fluids and secretions, pain, promoting calm and sleep, miscarriage, post-partum difficulties, inflammation, tumor, stress, oxidative stress, mind-booster, rejuvenation [56].
Withaferin A
28.GeraniinPhyllanthus emblica L. (Phyllanthaceae)Indian gooseberryTumor, pain, fever, stress, inflammation, oxidative stress, depression, liver damage, ulcer, radioprotective, diabetes, cancer, wound healing, cytotoxic [57,58].
Phyllanthin
29.AllicinAllium sativum L. (Amaryllidaceae)GarlicCold, influenza, dyspepsia, loss of appetite, snake bites, stress, inflammation, diabetes, aging effects, cancer, lung disorders, whooping cough, stomach disorders, cold, earache, cardiovascular disorder, Alzheimer’s disease [59].
Pyrogallol
30.Quercitrin Bryophyllum pinnatum (Lam.) Oken
(Crassulaceae)
PattharcaṭṭaUlcer, inflammation, analgesic, jaundice, kidney stones, respiratory tract infections, boils, insect bites, hypertension, diabetes, cancer, HPV [60].
Bryophyllin A
31.Alpha-pinenePinus roxburghii Sarg.
(Pinaceae)
Chir pineDyslipidemia, oxidative stress, wound healing, analgesic, inflammation, cytotoxic [61].
Abietic acid
32.ThymoquinoneNigella sativa L.
(Ranunculaceae)
Black cuminAsthma, hypertension, diabetes, inflammation, cough, bronchitis, headache, eczema, fever, dizziness, influenza, carminative, stimulant, diuretic [62].
Thymol
33.Aloe-emodinAloe barbadensis Miller
(Asphodelaceae)
(Aloe vera ) Burn injury, eczema, cosmetics, inflammation, fever, malaria [63].
Emodin
34.KoenimbineMurraya koenigii (L.) Spreng. (Rutaceae)Curry treePiles, inflammation, itching, fresh cuts, dysentery, bruises, and edema, helminth infection, analgesics, digestives, and appetizers, oxidative stress, inflammation, nephroprotective [64,65].
Mahanimbine
35.AsiaticosideCentella asiatica (L.) Urb. (Apiaceae)Brahma mandukiUlcerous skin, weakness, burns, duodenal, stomach ulcers, lupus, antinociceptive, inflammation, scleroderma, leprosy vein disorder, neuroprotection, wound healing, eczema, dermatitis, psoriasis [66].
Madecassoside
Table 2. Table describing the active phytocompounds, plant source, binding energy, and interactive amino acids.
Table 2. Table describing the active phytocompounds, plant source, binding energy, and interactive amino acids.
Name of CompoundPlant SourceBinding
Energy (KJ/Mol)
No. of Hydrogen BondsHydrogen BondsInteractive Amino Acids
AsiaticosideCentella asiatica (L.) Urb.−10.96Ser137, Glu113, Leu115, Asp152, Ala110, Arg148Ile106, Tyr59, Lys63, Asp124, Glu120, Ala149, Lys114, Ser112, Val111, Lys117, Cys134, Leu130, Arg136, Thr85, Ile88, Met70, Leu139, Phe155, Leu66, Tyr92, Leu156, Met140
MadecassosideCentella asiatica (L.) Urb.−9.75Tyr59, Asp124, Ala110, Ala123, Cys67Trp95, Leu66, Phe155, Thr85, Met70, Tyr92, Ser137, Leu139, Arg136, Leu130, Leu156, Ile88, Phe127, Lys63, Arg131, Pro128, Leu115, Asp152, Val11, Ala149, Glu120, Arg148, Met140, Ile106
Beta-sitosterolGirardinia diversifolia (Link) Friis−9.12Thr85, Ser137Tyr59, Ile106, Ala110, Asp152, Met140, Arg148, Leu139, Arg136, Val138, Ala81, Ile88, Leu156, Met70, Tyr92, Phe155, Leu66, Lys63
Bryophyllin ABryophyllum pinnatum (Lam.) Oken−10.62Thr85, Cys67Phe155, Arg148, Met140, Cys134, Ile88, Leu139, Ser137, Arg136, Ala81, Met84, Lys63, Leu66, Met70, Ile106, Tyr59, Leu156, Ala110, Asp152
MahanimbineMurraya koenigii (L.) Spreng−9.4--Arg136, Ser137, Thr85, Cys67, Tyr59, Ile106, Ala110, Leu156, Lys63, Tyr92, Leu66, Phe155, Met70, Ile88, Leu139, Leu130
PennogeninTrilliumgovanianum Wall. Ex D.Don−10.31Arg148Cys134, Met84, Ile88, Met70, Tyr92, Arg107, Phe155, Ile106, Leu156, Ala110, Leu66, Lys63, Met140, Tyr59, Leu139, Ser137, Arg136, Thr85, Ala81
RutinBerberis aristata DC.−9.23Ser137, Thr85, Tyr59Cys134, Met84, Glu120, Asp152, Arg148, Ala110, Leu156, Ala123, Asp124, Tyr92, Leu66, Phe155, Met70, Lys63, Cys67, Phe127, Arg136, Leu130, Ile88, Leu139
SolasonineSolanum americanum Mill.−9.72Arg148, Ser137Glu146, Asp145, Lys117, Glu120, Met70, Tyr92, Leu66, Phe155, Asp152, Met140, Arg136, Leu139, Leu130, Lys68, Ile88, Arg131, Asp124, Ala149
SolamargineSolanum americanum Mill.−9.55Arg131, Lys63, Asp124, Asp145, Arg148Phe127, Ala123, Leu130, Arg136, Glu120, Lys117, Tyr59, Leu139, Ser137, Met140, Thr85, Ile88
Withaferin AWithania somnifera (L.) Dunal−10.22Tyr59, Tyr92Ile106, Ala110, Leu66, Lys63, Phe155, Arg148, Leu139, Val138, Thr85, Trp185, Ser137, Met84, Cys134, Arg136, Ile88, Leu130, Met140, Leu156
WithanoneWithania somnifera (L.) Dunal−9.3--Ile106, Leu66, Lys63, Arg148, Tyr92, Met70, Leu130, Arg136, Ile88, Cys134, Thr85, Met84, Ser137, Leu139, Met140, Phe155, Asp152, Ala110, Leu156, Tyr59
Chenodeoxycholic acidBile component−10.84Ser137, Thr85, Asp152, Tyr59Val138, Cys134, Met84, Leu139, Ile88, Arg148, Met140, Phe155, Leu156, Ile106, Ala110, Lys63, Tyr92, Leu66, Met70, Arg136
Table 3. Drug likeness prediction of 11 active phytocompounds of 8 medicinal plants.
Table 3. Drug likeness prediction of 11 active phytocompounds of 8 medicinal plants.
PhytocompoundsmiLogPTPSA MWnONnOHNHNviolations
Chenodeoxycholic acid4.2577.75392.28430
Asiaticoside0.37315.21959.1319123
Beta-sitosterol8.6220.23414.72111
Bryophyllin A2.09115.44472.53820
Madecassoside−0.55335.44975.1320133
Mahanimbine7.1025.02331.46211
Pennogenin4.9958.92430.63420
Rutin−1.06269.43610.5216103
Solasonine1.40258.72884.0717103
Solamargine2.41238.49868.071693
Withaferin A3.8696.36470.61620
Withanone4.1596.36470.61620
miLogP—Molinspiration LogP (To measure lipophilicity), TPSA—topological polar surface area, MW—Molecular wait, nON—hydrogenbonds acceptor, nOHNH—hydrogen bonds donors, nviolations—Number of violations.
Table 4. Toxicity prediction of active phytocompounds and bile component.
Table 4. Toxicity prediction of active phytocompounds and bile component.
PhytocompoundsProTox-II
LD50(mg/kg)Hepato-ToxicityCarcino-GenecityImmuno
Toxicity
Muta-GenicityCyto-Toxicity
Chenodeoxycholic acid2000
(Class 4)
ActiveInactiveInactiveInactiveInactive
Asiaticoside4000
(Class 5)
InactiveInactiveActiveInactiveInactive
Beta-sitosterol890
(Class 4)
InactiveInactiveActiveInactiveInactive
Bryophyllin A31
(Class 2)
InactiveInactiveActiveInactiveActive
Madecassoside1190
(Class 4)
ActiveInactiveActiveInactiveInactive
Mahanimbine4000
(Class 5)
InactiveInactiveActiveInactiveInactive
Pennogenin1190
(Class 4)
ActiveInactiveActiveInactiveInactive
Rutin1190
(Class 4)
ActiveInactiveActiveInactiveActive
Solasonine500
(Class4)
InactiveInactiveActiveInactiveActive
Solamargine1190
(Class 4)
InactiveInactiveActiveInactiveActive
Withaferin A300
(Class 3)
InactiveInactiveActiveInactiveActive
Withanone7
(Class 2)
InactiveInactiveActiveInactiveActive
Table 5. Thermodynamic binding energy of Beta-sitasterol in complex with 6IE9.
Table 5. Thermodynamic binding energy of Beta-sitasterol in complex with 6IE9.
TitleMMGBSA (kcal/mol)
Frame 1−105.8395994
Frame 2−122.4871042
Frame 3−131.2524436
Frame 4−113.052146
Frame 5−139.4301793
Frame 6−154.7243297
Frame 7−149.2144921
Frame 8−157.1657804
Frame 9−164.8064715
Frame 10−148.5722473
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mehta, J.; Rolta, R.; Salaria, D.; Awofisayo, O.; Fadare, O.A.; Sharma, P.P.; Rathi, B.; Chopra, A.; Kaushik, N.; Choi, E.H.; et al. Phytocompounds from Himalayan Medicinal Plants as Potential Drugs to Treat Multidrug-Resistant Salmonella typhimurium: An In Silico Approach. Biomedicines 2021, 9, 1402. https://doi.org/10.3390/biomedicines9101402

AMA Style

Mehta J, Rolta R, Salaria D, Awofisayo O, Fadare OA, Sharma PP, Rathi B, Chopra A, Kaushik N, Choi EH, et al. Phytocompounds from Himalayan Medicinal Plants as Potential Drugs to Treat Multidrug-Resistant Salmonella typhimurium: An In Silico Approach. Biomedicines. 2021; 9(10):1402. https://doi.org/10.3390/biomedicines9101402

Chicago/Turabian Style

Mehta, Jyoti, Rajan Rolta, Deeksha Salaria, Oladoja Awofisayo, Olatomide A. Fadare, Prem Prakash Sharma, Brijesh Rathi, Adity Chopra, Neha Kaushik, Eun Ha Choi, and et al. 2021. "Phytocompounds from Himalayan Medicinal Plants as Potential Drugs to Treat Multidrug-Resistant Salmonella typhimurium: An In Silico Approach" Biomedicines 9, no. 10: 1402. https://doi.org/10.3390/biomedicines9101402

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

Article Metrics

Back to TopTop