Next Article in Journal
A Probable Anti-COVID Phytochemical (1,7-Bis-(4-hydroxyphenyl)-1-heptene-3,5-dione) Screened Computationally from the Rhizome of Curcuma longa 
Previous Article in Journal
Age-Related Features in Systemic Inflammatory Response in Male Wistar Rats with Different Hypoxia Tolerance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

Naturally Occurring Green Tea Polyphenols as Anti-Mycobacterial Agents †

Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra 835215, Jharkhand, India
*
Author to whom correspondence should be addressed.
Presented at the 1st International Electronic Conference on Molecular Sciences: Druggable Targets of Emerging Infectious Diseases (ECMS 2021), 1–14 September 2021; Available online: https://ecms2021.sciforum.net/.
These authors contributed equally to this work.
Med. Sci. Forum 2021, 7(1), 5; https://doi.org/10.3390/ECMS2021-10844
Published: 31 August 2021

Abstract

:
Tuberculosis (TB) is a global health burden especially in tropical countries. Extensive increments in MDR (Multidrug resistance (MDR): Resistance to at least both isoniazid and rifampicin.) and XDR (Extensive drug resistance (XDR): Resistance to any fluoroquinolone, and at least one of three second-line injectable drugs (capreomycin, kanamycin, and amikacin), in addition to multidrug resistance) tuberculosis highlights the ineffectiveness of established anti-TB agents. There is an urgent necessity to identify potent anti-TB agents with unique mechanisms. Green tea and Black tea polyphenols have great potential to inhibit viruses including SARS-COV-2, bacterial strains, etc. In this context, we have screened and identified 65 Green tea bioactive compounds against four mycobacterial pantothenate synthetase and enoyl acyl carrier enzymes. Our molecular docking results revealed that Theaflavin-3-gallate had a higher binding affinity against 2X22 and 3IVX targets with docking scores of −134.13 and −135.592 Kcal/mol, respectively. Furthermore, our molecular dynamics simulations for 10 ns resulted better stabilities of these complexes. We also evaluated in silico drug-likeness and toxicity profiles for the studied polyphenols. Our in silico toxicity analysis suggested that these polyphenols would exhibit lesser toxicity such as eye corrosion, skin irritations, etc. Thus, our present study would provide better insights on studying naturally occurring polyphenols as potential anti-TB agents.

1. Introduction

Tuberculosis (TB), which is a communicable disease, is one of the top 10 causes of death worldwide, especially in low-income tropical countries, where there is a scarcity of healthcare facilities. As per the WHO estimates for the year 2019, a total of 1.4 million people died due to TB [1]. The rising cases of multidrug-resistant TB (MDR-TB) are alarming and present a global health security threat (206,030 people were found to have multidrug- or rifampicin-resistant TB (MDR/RR-TB) strains) [1,2]. The unusual cell wall, made up of α-alkyl-β-hydroxy fatty acids or mycolic acid (MA), acts as a major barrier for therapeutic drugs to reach inside mycobacterial cells. It is noteworthy to mention that the MA serve key roles in maintaining structural integrity and to provide protection against an oxidative stress. It is also worth noting, that targeting a 2-trans-enoyl-acyl carrier protein reductase, called InhA is not always a good idea. Although, it is a good target, which is vital, and is the target for isoniazid, but resistance to isoniazid is one of the criteria for classifying M. tb as MDR, though most of the mutations occur in katG gene, activating isoniazid. Therefore, new drugs active on InhA could only partly overcome MDR [3]. Green tea and Black tea are the most popular beverages consumed. These are particularly derived from the plant Camellia sinensis [4]. In vitro and animal studies provide strong evidence that polyphenols derived from tea (polyphenols (the green tea polyphenols (GTPs)), especially flavanols, flavandiols, flavonoids, and phenolic acids, etc.) may possess the bioactivity that can affect the pathogenesis of several chronic diseases (Figure 1). The GTPs are also known for their wide pharmacological potentials, including anticarcinogenic, antioxidant, antituberculosis (anti-TB), and also, very recently, anti-SARS-Cov-2 properties [4,5]. It is interesting to note that these health-enhancing effects of GTPs were mainly attributed to the phytoconstituent present called ‘(−)-epigallocatechin-3-gallate’ (EGCG). In a very recent study, GTP epigallocatechin-3-gallate was demonstrated to inhibit InhA, the enoyl-ACP reductase of mycobacterium. This has prompted us to screen in silico a set of GTPs against various pivotal targets of mycobacterium including InhA [6,7]. For the best-docked top three hits with higher docking scores, we listed down their drug-likeness assessment, and ADMET (absorption, distribution, metabolism, excretion, toxicity) properties. Furthermore, we examined molecular dynamics simulations for the best docked hit, i.e., target complexes for the duration of 10 ns each.
Herein, we had three objectives to screen a set of known 65 bioactive molecules from tea against known anti-TB targets. Secondly, we also compared molecular docking simulation and molecular dynamics results with standard anti-TB drugs (Pyrazinamide, Ethambutol and Isoniazid) against mycobacterial targets. Lastly, we signified a probable lead that could be developed as a drug candidate against mycobacterial targets.

2. Materials and Methods

2.1. Molecular Docking Analysis

A set of 65 reported tea bioactive compounds was retrieved from the study reported by Bhardwaj et al. 2021 [7]. All the structures were then drawn using ‘ChemDraw V. 12.1’. All the 3D crystal structures of 4 mycobacterial proteins (the enoyl reductase receptor protein (PDB IDs:2NV6; 2X22 (crystal structure of M. tuberculosis InhA inhibited by PT70); 1BVR; and the pantothenate synthetase, Crystal structure of pantothenate synthetase in complex with 2-(2-(benzofuran-2-ylsulfonylcarbamoyl)-5-methoxy-1H-indol-1-yl)acetic acid, i.e., 3IVX) were downloaded from the protein database bank (PDB database, www.rcsb.org accessed on 20 April 2021). For the protein preparations and ligand preparations, we followed the known protocols. Finally, molecular docking simulations were performed with popular software, including, ‘Molegro Virtual Docker v. 6.0.1’ and ‘iGemDock’ as per standard procedures. The best docked hits were identified via higher docking scores and were then visualized with Discovery Studio 2020 Visualizer (BIOVIA, Dassault Systèmes) or with Pymol (GLSL version 4.60, for educational use).

2.2. In Silico Drug-Likeness and ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) Analysis

For the best docked top three hits, we predicted their ADME properties using SWISS tools (http://www.swissadme.ch accessed on 20 April 2022). In order to access a drug-likeness nature of obtained best docked hits, we used Lipinski’s rule of five criteria. The assessments for toxicities were predicted by using online platform, ‘admetSAR’(http://lmmd.ecust.edu.cn:8000/ accessed on 20 April 2022).

2.3. Normal Mode Analysis

To gain more insights into the conformational flexibilities [8] of proteins with their best docked hits, we performed the Normal Mode Analysis (NMA) with internal coordinates (IC) using a fast and easy server, iMODS (http://imods.chaconlab.org/ accessed on 20 April 2022). This server also guides medicinal chemists by providing more details on co-variance map, eigenvalues, deformability, variance, the collective motions of proteins, B-factor, etc. Deformations in proteins were depicted by the term deformability, while mobility profile was denoted by the B-factor.

2.4. Molecular Dynamics Analysis

Molecular dynamics (MD) simulation for a period of 10 ns was performed for best docked hit with Theaflavin-3-gallate: target protein, 2X22 complex and it was achieved with Desmond module implemented in a Schrödinger package, 2020. For setting up initial systems, we used the OPLS-2005 molecular mechanic’s force field. We kept ensemble class at NPT (temperature: 300 k, pressure: 1.01325 bar). Then, system was simulated further through the multistep MD protocols.

3. Results and Discussion

3.1. Molecular Docking Simulations

In order to gain more insights on binding mechanisms, we docked a set of 65 green tea bioactive compounds into 4 mycobacterial target proteins using ‘iGemDock’ tool. The docking protocol was validated via a redocking approach and was obtained with RMSD below 2 Å [9,10,11]. A dataset molecule, Theaflavin-3-gallate interacted with target proteins 2X22 and 3IVX with highest binding scores of −134.13 and −135.592 Kcal/mol, respectively. Compound, Theaflavin-3-gallate interacted with key amino acid residues, TRP A:160; MET A:103; GLN A:100; ASN A:159; MET A:155; THR A:162; PRO A:156, etc. (Figure 2). The results for the remaining green tea/black tea biomolecules are listed in Table 1 and Table 2.

3.2. Molecular Dynamics Simulation and Normal Mode Analysis

The highest scored biomolecule, Theaflavin-3-gallate with protein 2X22 was simulated for molecular dynamics and normal mode analysis. MD simulations depicted that Root Mean Square Fluctuation (RMSF) values were obtained within tolerable ranges. The Root mean square deviation (RMSD) value was obtained below 3 Å, suggesting stability of complex (Figure 3). From our NMA results, we noticed that Theaflavin-3-gallate with protein 2X22 complex was retained with good deformability, and eigenvalue value profiles (Figure 3).

3.3. In Silico ADME Studies

Cytochrome P450 (CYPs) enzymes are a family metabolic enzymes responsible for bio-transformations of almost ~90% FDA approved drugs. Phase I and Phase II are two important pathways involved in the metabolism of xenobiosis. Our in silico calculated ADMET (absorption, distribution, metabolism, excretion, toxicity) properties for the top best-docked three hits are represented in Table 3. Compounds, Theaflavin-3-gallate, Epigallocatechin and Epigallocatechin Gallate (EGCG) exhibited non-carcinogenic, non-AMES toxic, and class IV acute oral toxicity profiles. All 3 of our proposed hits were found to have positive human intestinal absorption profiles and negative the Blood–brain barrier passage profiles.

4. Conclusions

It is noteworthy to mention that green tea polyphenols have significant prooxidant properties and a great potential to inhibit in vitro SARS-Cov-2, bacterial and mycobacterial growths. However, our in silico methodology used herein indicates four probable therapeutic targets involved in anti-TB potentials. Furthermore, we also wish to note that apart from the reported potential of EGCG, Theaflavin-3-gallate may have strong interaction with InhA target. The tea extract containing Theaflavin-3-gallate could also be tested in vitro for anti-TB assessments. Moreover, we believe that the core structure of Theaflavin-3-gallate could also be explored further to develop more potent synthetic analogues for TB. Our in silico ADMET analysis suggested safer probable pharmacokinetics for GTPs.
Many of the virtually screened compounds are usually inactive on mycobacterial cells due to their cell wall permeability. For better screening of virtually screening hits, a deeper understanding of the cell biology of mycobacteria and a thorough structure analysis of selected hits is required. Indeed, major limitations characterizing docking include a restricted sampling of both ligand and receptor conformations in pose prediction, and the use of approximated scoring functions, which very often provide results that do not correlate with the experimental binding affinities. Thus, the proper selection of a protein and binding site along with the best docking software will increase the likelihood of retaining the correct hits.
However, despite the success of molecular docking or drug repurposing via in silico methodologies, one must take into considerations the usage of an appropriate scoring function and algorithm, which may otherwise jeopardize molecular screening.

Author Contributions

Conceptualization, S.N.M. and A.P.; methodology, S.N.M.; software, S.N.M.; writing—review and editing, S.N.M. and A.P.; visualization, S.N.M. and A.P.; supervision, S.N.M. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

We wish to thank the Dept. of Pharmaceutical Sciences, Birla Institute of Technology, Mesra, India for financial assistance. One of the authors SM is thankful to Schrodinger Team, Banglore for providing trial license. SM is also thankful for the provision of IRF (PHD/PH/10006/20) (Ref. No. GO/Estb/Ph.D./IRF/2020-21/2484A) provided by BIT, Mesra, India.

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Acknowledgments

The authors would like to thank the Head, Department of Pharmaceutical Sciences and Technology, BIT, Mesra for providing the research facilities for performing the current study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Poro, K.E.; Hoekou, Y.; Pissang, P.; Kpabi, I.; Novidzro, K.M.; Dagnra, A.Y.; Tchacondo, T.; Batawila, K. In vitro Antimycobacterial Activity of Selected Medicinal Plants against Mycobacterium tuberculosis. Int. J. Curr. Microbiol. Appl. Sci. 2021, 10, 3201–3208. [Google Scholar]
  2. WHO TB Factsheet. Available online: https://www.who.int/news-room/fact-sheets/detail/tuberculosis (accessed on 17 July 2021).
  3. Banerjee, A.; Dubnau, E.; Quemard, A.; Balasubramanian, V.; Um, K.S.; Wilson, T.; Collins, D.; de Lisle, G.; Jacobs, W.R., Jr. inhA, a gene encoding a target for isoniazid and ethionamide in Mycobacterium tuberculosis. Science 1994, 263, 227–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Henss, L.; Auste, A.; Schürmann, C.; Schmidt, C.; von Rhein, C.; Mühlebach, M.D.; Schnierle, B.S. The green tea catechin epigallocatechin gallate inhibits SARS-CoV-2 infection. J. Gen. Virol. 2021, 102, 001574. [Google Scholar] [CrossRef] [PubMed]
  5. Anand, P.K.; Kaul, D.; Sharma, M. Green tea polyphenol inhibits Mycobacterium tuberculosis survival within human macrophages. Int. J. Biochem. Cell Biol. 2006, 38, 600–609. [Google Scholar] [CrossRef] [PubMed]
  6. Narayanan, S.; Ramesh, K.V. Epigallocatechin gallate, a green tea polyphenol inhibits Mycobacterium smegmatis: In silico and in vitro studies. Indian J. Pharm. Sci. 2017, 79, 625–632. [Google Scholar] [CrossRef] [Green Version]
  7. Bhardwaj, V.K.; Singh, R.; Sharma, J.; Rajendran, V.; Purohit, R.; Kumar, S. Identification of bioactive molecules from tea plant as SARS-CoV-2 main protease inhibitors. J. Biomol. Struct. Dyn. 2021, 39, 3449–3458. [Google Scholar] [CrossRef] [PubMed]
  8. Kovacs, J.; Chacón, P.; Abagyan, R. Predictions of Protein Flexibility: First Order Measures. PROTEINS: Structure, Function, and Bioinformatics. Proteins 2004, 56, 661–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Mali, S.N.; Chaudhari, H.K. Computational studies on imidazo [1,2-a] pyridine-3-carboxamide analogues as antimycobacterial agents: Common pharmacophore generation, atom-based 3D-QSAR, molecular dynamics simulation, QikProp, molecular docking and prime MMGBSA approaches. Open Pharm. Sci. J. 2018, 5, 12–23. [Google Scholar] [CrossRef] [Green Version]
  10. Hsu, K.C.; Chen, Y.F.; Lin, S.R.; Yang, J.M. iGEMDOCK: A graphical environment of enhancing GEMDOCK using pharmacological interactions and post-screening analysis. BMC Bioinform. 2011, 12, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Mali, S.N.; Pandey, A.; Thorat, B.R. Multiple 3D- and 2D-quantitative structure–activity relationship models (QSAR), theoretical study and molecular modeling to identify structural requirements of imidazopyridine analogues as anti-infective agents against tuberculosis. Struct. Chem. 2022. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of Green Tea bioactive compounds (representative).
Figure 1. Chemical structures of Green Tea bioactive compounds (representative).
Msf 07 00005 g001
Figure 2. 2D and 3D-interaction profiles for best docked Theaflavin-3-gallate with 2X22.
Figure 2. 2D and 3D-interaction profiles for best docked Theaflavin-3-gallate with 2X22.
Msf 07 00005 g002
Figure 3. (a) The Root Mean Square Deviations (RMSD) of backbone atoms relative to the starting complexes during 10 ns MD; (b) Protein RMSF plot (On this plot, peaks indicate areas of the protein that fluctuate the most during the simulation and Protein residues Table 3. gallate with 2X22, respectively; (c) Normal mode analysis-Deformability; (d) B-factor analysis.
Figure 3. (a) The Root Mean Square Deviations (RMSD) of backbone atoms relative to the starting complexes during 10 ns MD; (b) Protein RMSF plot (On this plot, peaks indicate areas of the protein that fluctuate the most during the simulation and Protein residues Table 3. gallate with 2X22, respectively; (c) Normal mode analysis-Deformability; (d) B-factor analysis.
Msf 07 00005 g003
Table 1. Docking interaction energies * of selected 65 bio-active molecules and 3 FDA approved drugs for target protein 2X22.
Table 1. Docking interaction energies * of selected 65 bio-active molecules and 3 FDA approved drugs for target protein 2X22.
MoleculesiGemDock Interaction EnergyMolecules-iGemDock Interaction Energy
Oolonghomobisflavan A−66.2219Theaflavic Acid−84.4934
Theasinensin D−72.1619Barrigenol R1−86.4843
Theaflavin-3-gallate−134.13Barringtogenol−89.0693
Isotheaflavin−72.621Camelliagenin−95.1799
Epigallocatechin-3,5-Di-O-Gallate−72.0176Gallocatechin−86.7374
Oolonghomobisflavan B−75.4779Catechin−102.992
Cis-3-Hexenol−63.5566Epicatechin−98.6033
Epigallocatechin-3,4-Di-O-Gallate−92.6784Epiafzelechin−91.5357
Vicenin 2−96.9806Quercetin−102.834
Epicatechin-3,5-Di-O-Gallate−101.495Cryptoxanthin−95.1799
Rutin−87.1416Myricetin−83.5936
Proanthocyanidin−84.8129Apigenin−83.6163
Pheophytin−90.2865Nerolidol−84.584
Benzaldehyde−91.9877Kaempferol−89.1838
Epitheaflavic Acid 3’-Gallate−65.361Theanine−83.9851
Epigallocatechin Gallate−122.3403Ascorbic Acid−80.1271
Theasinensin E−62.6409Quinic Acid−85.3299
Myricitrin−61.915Succinic Acid−85.5696
Theaflavin−65.9704Methyl Salicylate−81.1848
Epicatechin Gallate−75.5287Theobromine−84.7269
Kaempferitrin−72.7401Caffeine−84.4502
Isoquercetin−89.9058Xanthine−86.7595
Epiafzelechin 3-O-Gallate−79.4119Linalool Oxide−83.9907
Pheophorbide−71.1657Phenylacetaldehyde−87.8044
Epigallocatechin 3-O-P-Coumarate−78.8643Methylxanthine−79.6185
Pheophorbide−68.9266Theophylline−88.1319
Oxalic Acid−87.9277Geraniol−95.2378
Cryptoxanthin−81.2634Hexanal−95.8974
Isovitexin−82.924Diphenylamine−93.4455
Vitexin−85.6638Trans−2-Hexenal−94.076
Chlorogenic Acid−89.7604Linalool−86.4307
Coumaroyl Quinic Acid−94.7189Phenylethanol−101.468
Epigallocatechin−115.6776Ciprofloxacin *−108.9558
* Docking scores have been provided only for the higher affinity scored target protein.
Table 2. Energy contribution of the key residues computed by docking methodology.
Table 2. Energy contribution of the key residues computed by docking methodology.
Sr. No.MoleculesResidues with Contribution Energy (kcal/mol)
1IsoniazideTYR A:158 (PI-PI STACKING); VAL A:203; MET A:199; LYS A:165
2PyrazinamideTYR A:158; MET A:161; ALA A:198
3CiprofloxacinPRO A:156; MET A:199; TYR A:158; VAL A:203
4Theaflavin-3-gallate
(Best docked)
TRP A:160; MET A:103; GLN A:100; ASN A:159; MET A:155; THR A:162; PRO A:156
5EpigallocatechinALA A:198; MET A:162; PRO A:193; PHE A:149; MET A:199; TYR A:158
6Epigallocatechin Gallate (EGCG)ALA A:198; MET A:162; PRO A:193; PHE A:149; MET A:199; TYR A:158
7Inbound ligandALA A:198; MET A:162; PRO A:193; PHE A:149; MET A:199; TYR A:158
Table 3. In silico ADMET profiling for top 3 best docked hits against target 2X22.
Table 3. In silico ADMET profiling for top 3 best docked hits against target 2X22.
PropertiesTheaflavin-3-GallateEpigallocatechinEpigallocatechin Gallate (EGCG)
CYP450 2C9 SubstrateNon-substrateNon-substrateNon-substrate
CYP450 2D6 SubstrateNon-substrateNon-substrateNon-substrate
CYP450 3A4 SubstrateNon-substrateNon-substrateNon-substrate
Human Ether-a-go-go-Related Gene InhibitionWeak inhibitorWeak inhibitorWeak inhibitor
AMES ToxicityNon-AMES toxicNon-AMES toxicNon-AMES toxic
CarcinogensNoneNoneNone
Acute Oral ToxicityIVIVIV
P-glycoprotein InhibitorNon-inhibitorNon-inhibitorNon-inhibitor
Rat Acute Toxicity (LD50, mol/kg)2.66931.87002.6643
Human Intestinal Absorption+++
Blood–brain barrier---
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mali, S.N.; Pandey, A. Naturally Occurring Green Tea Polyphenols as Anti-Mycobacterial Agents. Med. Sci. Forum 2021, 7, 5. https://doi.org/10.3390/ECMS2021-10844

AMA Style

Mali SN, Pandey A. Naturally Occurring Green Tea Polyphenols as Anti-Mycobacterial Agents. Medical Sciences Forum. 2021; 7(1):5. https://doi.org/10.3390/ECMS2021-10844

Chicago/Turabian Style

Mali, Suraj N., and Anima Pandey. 2021. "Naturally Occurring Green Tea Polyphenols as Anti-Mycobacterial Agents" Medical Sciences Forum 7, no. 1: 5. https://doi.org/10.3390/ECMS2021-10844

Article Metrics

Back to TopTop