Next Article in Journal
Metabolomics as a Crucial Tool to Develop New Therapeutic Strategies for Neurodegenerative Diseases
Next Article in Special Issue
Metabolomic Strategies to Improve Chemical Information from OSMAC Studies of Endophytic Fungi
Previous Article in Journal
Sugarcane Metabolome Compositional Stability in Pretreatment Processes for NMR Measurements
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Identification of Plant-Derived Bioactive Compounds Using Affinity Mass Spectrometry and Molecular Networking

1
Research Centre for Plant Metabolomics, Department of Biochemistry, University of Johannesburg, P.O. Box 524, Auckland Park, Johannesburg 2006, South Africa
2
Department of Agriculture and Animal Health, Florida Campus, University of South Africa, Florida 1710, South Africa
*
Author to whom correspondence should be addressed.
Metabolites 2022, 12(9), 863; https://doi.org/10.3390/metabo12090863
Submission received: 6 August 2022 / Revised: 9 September 2022 / Accepted: 9 September 2022 / Published: 14 September 2022
(This article belongs to the Special Issue Metabolomics in Natural Products)

Abstract

:
Affinity selection-mass spectrometry (AS-MS) is a label-free binding assay system that uses UHPLC-MS size-based separation methods to separate target-compound complexes from unbound compounds, identify bound compounds, classify compound binding sites, quantify the dissociation rate constant of compounds, and characterize affinity-extracted ligands. This label-free binding assay, in contrast to conventional biochemical (i.e., high-throughput screening (HTS)) approaches, is applicable to any drug target, and is also concise, accurate, and adaptable. Although AS-MS is an innovative approach for identifying lead compounds, the possibilities of finding bioactive compounds are limited by competitive binding, which occurs during the equilibration of extracts with the target protein(s). Here, we discuss the potential for metabolite profiling complemented with molecular networking to be used alongside AS-MS to improve the identification of bioactive compounds in plant extracts. AS-MS has gained significant prominence in HTS labs and shows potential to emerge as the driving force behind novel drug development in the future.

1. Introduction

Natural products are secondary metabolites produced by organisms, particularly microorganisms and plants. As rich sources of bioactive compounds that are both medically and industrially relevant, natural products are key sources of novel medications and good lead compounds that can be modified extensively during the drug development process, notably as anticancer and antibacterial agents [1,2,3]. Secondary metabolites derived from natural sources are often regarded as having greater “drug-likeness and biological friendliness” than synthetically produced compounds, making them potential candidates for future drug development [4]. In addition to a wide range of scaffold variety and structural complexity [5], natural product extracts have diverse pharmacophores and a great degree of stereochemistry. Consequently, the varied forms and complicated carbon skeletons of natural products have resulted in a substantial fraction of natural products being used in drug discovery [4,6,7].
Previously, culture broths or extracts of microorganisms and plants were screened for desired activities, followed by compound isolation, activity screening, and structural analysis. The functional activity of the target is the constraint of most traditional high-throughput screening (HTS) assays, such as enzymatic or G-protein-coupled receptor (GPCR) signaling [8]. Although an effective approach to finding natural products with desired activities, the increased rediscovery rate of the same compounds following the tedious screening, purification, and identification stages make the discovery of new natural products increasingly difficult. Furthermore, each stage is time-consuming, labor-intensive, and, more importantly, compound-untargeted. As a result, developing an efficient and selective screening strategy for active compounds is essential.

2. Affinity Selection-Mass Spectrometry-Based Drug Discovery versus the Conventional Method

Recent advances in genomics technologies have revealed a wealth of natural compounds that are yet to be found and defined [3,5,9,10]. To identify new lead compounds for receptors that are known or suspected to be involved in a disease pathway, target-based screening approaches are performed [11]. In this regard, affinity selection-mass spectrometry (AS-MS), allows for the simultaneous characterization and dereplication of active ingredients in complex mixtures, such as extracts of botanicals, fungi, and microbial cultures [3]. AS-MS is a label-free binding assay system that uses UHPLC-MS size-based separation methods, such as ultrafiltration, gel permeation, or size-exclusion chromatography, to separate target-compound complexes from unbound compounds, identify bound compounds, classify compound binding sites, quantify the dissociation rate constant of compounds, and characterize affinity-extracted ligands [3,11,12,13]. The AS-MS method entails the expression of target proteins which are later immobilized onto the chromatographic column, followed by the equilibration of target proteins with the crude extract. This leaves bioactive compounds bound to the target protein, which are then characterized using LC-MS (Figure 1). Different types of AS-MS have been used to identify bioactive compounds from plant extracts, including pulsed ultrafiltration (PUF) AS-MS, size-exclusion chromatography (SEC) AS-MS, and magnetic microbead affinity selection screening (MagMASS) [14,15]. Table 1 summarizes native and affinity MS methods used for the identification of bioactive compounds in plants.
AS-MS can be either direct, in which the protein–ligand complexes are measured by MS, or indirect, in which the occurrence of a complex is inferred by detecting the ligand after it has dissociated from the protein target [18]. These label-free binding assay approaches are ideal for identifying ligands to target receptors because of their speed, selectivity, and sensitivity. As such, AS-MS allows for the rapid isolation of pharmacologically active molecules from complex mixtures for mass spectrometric characterization and identification [3,19,20,21]. During the AS-MS incubation of targets at a precise stage, the drug targets are frequently present in molar excess relative to probable ligands. This adds to the merits of AS-MS over conventional methods as it reduces competition, which could lead to the detection of ligands with lower affinity for the target [21]. Studies utilizing AS-MS to identify new bioactive metabolites from plant extracts are presented in Table 2 and their main findings are summarized. Furthermore, Table 2 shows docking studies conducted on AS-MS isolated compounds to confirm their interaction with targeted proteins. Molecular docking predicts ligand-target complex’s preferred confirmation and strength of association or binding affinity [16].
In comparison to conventional drug discovery (Figure 1), AS-MS offers greater advantages. This label-free binding assay, in contrast to typical biochemical HTS approaches, is applicable to any drug target and is concise, accurate, and adaptable without the requirement for compound or target modification [3,20,21,27]. As a result, AS-MS does not require radiolabels, UV, or fluorescent chromophores, and is compatible with all receptors, enzymes, incubation buffers, cofactors, and ligands [20]. Whereas conventional HTS is designed to discover only orthosteric ligands, AS-MS can also detect allosteric ligands. Moreover, natural product screens generally contain a library of natural extracts, which may not be amenable to conventional target-based assays [27,28,29]. As a result, AS-MS provides access to molecules with far greater chemical diversity than synthetic chemical libraries and can provide direct confirmation of the protein–ligand complex’s presence and stoichiometric information [3,21].

Metabolite Profiling Meeting AS-MS

Although AS-MS is innovative in identifying lead compounds, competitive binding occurs during the equilibration of extracts with the target protein(s) where only the compounds with high binding affinity could be identified as potential leads in drug discovery. However, this phenomenon limits the possibility of finding a wider range of bioactive compounds. The molecular networking (MN) approach has emerged as a useful tool to analyze tandem mass spectrometry (MS2) data to further identify compounds in MS-based metabolite profiling methods. The MN concept is based on organizing and visualizing tandem MS data using a spectral similarity map to identify the presence of homologous MS2 fragmentations. The nodes of structurally related compounds tend to cluster and generate clusters of analogues since they share similar fragmentation spectra [30,31,32]. Thus, mapping AS-MS isolated compounds over a molecular network of different chemical clusters will allow efficient selection of chemical clusters of potential bioactive compounds against a specific protein target (Figure 1). Furthermore, merging AS-MS isolated compounds over a molecular network can help in compound annotation/structural elucidation and activity relationships within a specific chemical cluster. Such compounds can be studied further to explore the binding affinity to the target protein using virtual screening (Figure 1). Virtual screening evaluates vast libraries of chemical compounds using computer technology and software to identify potential drug candidates based on biological structures [33]. The primary purpose of virtual screening is to condense the vast virtual chemical space of small organic molecules that can be synthesized or screened against a single target protein down to a manageable number of compounds that have the best probability of becoming a therapeutic candidate [32,33].
For example, in research conducted by Wang and colleagues [24], molecular networking and virtual screening coupled with AS-MS was used to discover two compounds, kurarinol and kurarinone, which were successfully identified from 11 traditional Chinese medicine (TCM) herbs and were confirmed to interact with GTPase of Ras. Initially, Ras protein was expressed; then, affinity mass selection was employed to find compounds active against the GTPase from crude extracts of TCM crude extracts. Thereafter, molecular networking was used to identify the two compounds, kurarinol and kurarinone, in the extracts confirmed to interact with Ras protein by virtual screening [24].

3. Concluding Remarks

AS-MS approaches can be used investigate the binding of candidate compounds to immobilized targets and serve as supplements to traditional drug development methods. These methods rely on target-based screening strategies to discover alternative lead compounds for receptors that are known or possibly involved in a disease pathway. As the first step in drug discovery, affinity selection-mass spectrometry (AS-MS) is useful for identifying lead compounds in plant extracts. The progress of instrumentation automation, particularly of LC and MS components, would naturally allow for more diverse and innovative applications, potentially expanding the already significant impact of AS-MS on drug discovery. As a result, AS-MS has gained significant prominence in high-throughput screening labs and may be the driving force behind novel small molecule medication development, and it is expected that these applications will continue and expand in the future. Furthermore, mapping of AS-MS isolated bioactive compounds over molecular networks will allow efficient selection of clusters of potential bioactive compounds. The mapping of AS-MS bioactive compounds on molecular networks also offers additional advantages, such as structural elucidation and activity relativity relationships within a chemical cluster to which the isolated compounds belong.

Author Contributions

Conceptualization, E.N.N. and M.I.M.; writing—original draft preparation, T.R., A.M. and P.W.M.; writing—review and editing, T.R., A.M., P.W.M., E.N.N., N.N. and M.I.M.; supervision, M.I.M.; project administration, E.N.N. and M.I.M.; funding acquisition, M.I.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was partially funded by the University of Johannesburg GES 4.0 COVID-19 awarded to M.I.M., grant number: 051155.25.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The University of Johannesburg is thanked for fellowship support to T.R., A.M. and P.W.M.; the South African National Research Foundation (NRF) is thanked for fellowship support to T.R. and P.W.M.; The University of South Africa is thanked for fellowship support to E.N.N.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harvey, A.L. Natural products in drug discovery. Drug Discov. Today 2008, 13, 894–901. [Google Scholar] [CrossRef] [PubMed]
  2. Lahlou, M. The Success of Natural Products in Drug Discovery. J. Pharm. Pharmacol. 2013, 4, 17–31. [Google Scholar] [CrossRef]
  3. Muchiri, R.N.; van Breemen, R.B. Affinity selection–mass spectrometry for the discovery of pharmacologically active compounds from combinatorial libraries and natural products. J. Mass. Spectrom. 2021, 56, e4647. [Google Scholar] [CrossRef] [PubMed]
  4. Chin, Y.W.; Balunas, M.J.; Chai, H.B.; Kinghorn, A.D. Drug discovery from natural sources. AAPS J. 2006, 8, 239–242. [Google Scholar] [CrossRef] [PubMed]
  5. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–221. [Google Scholar] [CrossRef] [PubMed]
  6. Rishton, G.M. Natural products as a robust source of new drugs and drug leads past successes and present-day issues. Am. J. Cardiol. 2008, 101, 43D–49D. [Google Scholar] [CrossRef]
  7. Dias, D.A.; Urban, S.; Roessner, U. A historical overview of natural products in drug discovery. Metabolites 2012, 2, 303–336. [Google Scholar] [CrossRef]
  8. Lu, Y.; Qin, S.; Zhang, B.; Dai, A.; Cai, X.; Ma, M.; Gao, Z.; Yang, D.; Stevens, R.C.; Jacobson, K.A.; et al. Accelerating the throughput of affinity mass spectrometry-based ligand screening towards a G protein-coupled receptor. Anal. Chem. 2019, 91, 8162–8169. [Google Scholar] [CrossRef]
  9. Rao, S.V.; Srinivas, K. Modern drug discovery process: An in silico approach. JBSA 2011, 2, 89–94. [Google Scholar]
  10. Louie, K.B. Comprehensive Natural Products III: Mass Spectrometry for Natural Product Discovery; Lawrence Berkeley National Laboratory (LBNL): Berkeley, CA, USA, 2020; pp. 63–306. [Google Scholar]
  11. Annis, A.; Chuang, C.C.; Nazef, N. Methods and Principles in Medicinal Chemistry Mass Spectrometry in Medicinal Chemistry || ALIS: An Affinity Selection–Mass Spectrometry System for the Discovery and Characterization of Protein–Ligand Interactions; Wiley-VCH Verlag GmbH & Co.KgaA: Hoboken, NJ, USA, 2007; Chapter 3,; pp. 121–156. Available online: https://onlinelibrary.wiley.com/doi/book/10.1002/9783527610907 (accessed on 8 September 2022).
  12. Annis, D.; Athanasopoulos, J.; Curran, P.; Felsch, J.; Kalghatgi, K.; Lee, W.; Nash, H.; Orminati, J.-P.; Rosner, K.; Shipps, G.; et al. An affinity selection-mass spectrometry method for the identification of small molecule ligands from self-encoded combinatorial libraries—Discovery of a novel antagonist of E-coli dihydrofolate reductase. Int. J. Mass Spectrom. 2004, 238, 77–83. [Google Scholar]
  13. Zehender, H.; Mayr, L.M. Application of high-throughput affinity-selection mass spectrometry for screening of chemical compound libraries in lead discovery. Expert Opin. Drug Discov. 2007, 2, 285–294. [Google Scholar] [CrossRef] [PubMed]
  14. Muchiri, R.N.; van Breemen, R.B. Drug discovery from natural products using affinity selection-mass spectrometry. Drug Discov. Today Technol. 2021, 40, 59–63. [Google Scholar] [CrossRef] [PubMed]
  15. Mak, T.; Rossojohn, J.; Littler, D.E.; Liu, M.; Quinn, R.J. Collision-induced affinity selection mass spectrometry for identification of ligands. ACS Bio. Med. Chem. Au. 2022. [Google Scholar] [CrossRef]
  16. Van Breemen, R.B.; Muchiri, R.N.; Bates, T.A.; Weinstein, J.B.; Leier, H.C.; Farley, S.; Tafesse, F.G. Cannabinoids Block Cellular Entry of SARS-CoV-2 and the Emerging Variants. J. Nat. Prod. 2022, 85, 176–184. [Google Scholar] [CrossRef]
  17. Tamara, S.; den Boer, M.A.; Heck, A.J.R. High-resolution native mass spectrometry. Chem Rev. 2022, 122, 7269–7326. [Google Scholar] [CrossRef] [PubMed]
  18. Whitehurst, C.; Annis, D. Affinity selection-mass spectrometry and its emerging application to the high throughput screening of G protein-coupled receptors. Comb. Chem. High Throughput Screen. 2008, 11, 427–438. [Google Scholar] [CrossRef]
  19. Prudent, R.; Annis, D.A.; Dandliker, P.J.; Ortholand, J.Y.; Roche, D. Exploring new targets and chemical space with affinity selection-mass spectrometry. Nat. Rev. Chem. 2020, 5, 62–71. [Google Scholar] [CrossRef]
  20. Motoyaji, T. Revolution of small molecule drug discovery by affinity selection-mass spectrometry technology. Chem. Pharm. Bull. 2020, 68, 191–193. [Google Scholar] [CrossRef]
  21. Zhu, D.; Su, H.; Ke, C.; Tang, C.; Witt, M.; Quinn, R.J.; Xu, Y.; Liu, J.; Ye, Y. Efficient discovery of potential inhibitors for SARS-CoV-2 3C-like protease from herbal extracts using a native MS-based affinity-selection method. J. Pharm. Biomed. Anal. 2022, 209, 114538. [Google Scholar] [CrossRef]
  22. Fu, Y.; Mo, H.-Y.; Gao, W.; Hong, J.-Y.; Lu, J.; Li, P.; Chen, J. Affinity selection-based two-dimensional chromatography coupled with high-performance liquid chromatography-mass spectrometry for discovering xanthine oxidase inhibitors from Radix Salviae miltiorrhizae. Anal. Bioanal. Chem. 2014, 406, 4987–4995. [Google Scholar] [CrossRef]
  23. Fu, X.; Wang, Z.; Li, L.; Dong, S.; Li, Z.; Jiang, Z.; Shui, W. Novel chemical ligands to ebola virus and marburg virus nucleoproteins identified by combining affinity mass spectrometry and metabolomics approaches. Sci. Rep. 2016, 6, 29680. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, Z.; Kim, U.; Liu, J.; Cheng, C.; Wu, W.; Guo, S.; Bai, G. Comprehensive TCM molecular networking based on MS/MS in silico spectra with the integration of virtual screening and affinity MS screening for discovering functional ligands from natural herbs. Anal. Bioanal. Chem. 2019, 411, 5785–5797. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, Z.; Liang, H.; Cao, H.; Zhang, B.; Li, J.; Wang, W.; Qin, S.; Wang, F.; Xuan, L.; Lai, L.; et al. Efficient ligand discovery from natural herbs by integrating virtual screening, affinity mass spectrometry and targeted metabolomics. Analyst 2019, 144, 2881–2890. [Google Scholar] [CrossRef]
  26. Tao, Y.; Chen, Z.; Zhang, Y.; Wang, Y.; Cheng, Y. Immobilised magnetic beads based multi-target affinity selection coupled with high performance liquid chromatography–mass spectrometry for screening anti-diabetic compounds from a Chinese medicine “Tang-Zhi-Qing”. J. Pharm. Biomed. Anal. 2013, 78–79, 190–201. [Google Scholar] [CrossRef]
  27. Imaduwage, K. High-Throughput Screening (HTS) of Potential Lead Compounds for Target Proteins with no False Identifications Using LC/MS. Bachelor’s Thesis, University of Kansas, Lawrence, KS, USA, 2017. [Google Scholar] [CrossRef]
  28. Henrich, C.J.; Beutler, J.A. Matching the power of high throughput screening to the chemical diversity of natural products. Nat. Prod. Rep. 2013, 30, 1284. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, M.; Carver, J.J.; Phelan, V.V.; Sanchez, L.M.; Garg, N.; Peng, Y.; Luzzatto-Knaan, T. Sharing and community curation of mass spectrometry data with global natural products social molecular networking. Nat. Biotechnol. 2016, 34, 828–837. [Google Scholar] [CrossRef] [PubMed]
  30. Da Silva, R.R.; Wang, M.; Nothias, L.-F.; van der Hooft, J.J.J.; Caraballo-Rodríguez, A.M.; Fox, E.; Dorrestein, P.C. Propagating annotations of molecular networks using in silico fragmentation. PLOS Comput. Biol. 2018, 14, e1006089. [Google Scholar] [CrossRef]
  31. Olivon, F.; Allard, P.M.; Koval, A.; Righi, D.; Genta-Jouve, G.; Neyts, J.; Apel, C.; Pannecouque, C.; Nothias, L.F.; Cachet, X.; et al. Bioactive natural products prioritisation using massive multi-informational molecular networks. ACS Chem. Biol. 2017, 12, 2644–2651. [Google Scholar] [CrossRef]
  32. Vyas, V. Virtual screening: A fast tool for drug design. Sci. Pharm. 2008, 76, 333–360. [Google Scholar] [CrossRef] [Green Version]
  33. Ingelheim, B. Virtual screening - What does it give us? Curr. Opin. Drug Discov. Dev. 2009, 12, 397–407. [Google Scholar]
Figure 1. Comparative illustration of conventional drug discovery (on the left) vs. AS-MS-based drug discovery (on the right) and how AS-MS is explored further to increase the possibilities of finding bioactive compounds using molecular networking (in the middle). In the molecular network, compounds that were initially identified using AS-MS (colored red and purple) will appear as nodes with other surrounding nodes (colored green) representing compounds with similar structures; these compounds can be virtually screened for binding affinity to target proteins, increasing the chances of finding more drug lead compounds. Green arrows indicate mapping of the LC-MS characterized ligands into molecular networking.
Figure 1. Comparative illustration of conventional drug discovery (on the left) vs. AS-MS-based drug discovery (on the right) and how AS-MS is explored further to increase the possibilities of finding bioactive compounds using molecular networking (in the middle). In the molecular network, compounds that were initially identified using AS-MS (colored red and purple) will appear as nodes with other surrounding nodes (colored green) representing compounds with similar structures; these compounds can be virtually screened for binding affinity to target proteins, increasing the chances of finding more drug lead compounds. Green arrows indicate mapping of the LC-MS characterized ligands into molecular networking.
Metabolites 12 00863 g001
Table 1. Short description of native and affinity MS methods.
Table 1. Short description of native and affinity MS methods.
DescriptionReferences
1. Affinity selection
Magnetic microbead affinity selection screening (MagMASS)MagMASS is a solid-phase alternative that complements the solution-phase screening approaches. MagMASS involves tethering the target to magnetic microbeads, incubating the immobilized protein with a natural product mixture, using magnetism to separate the ligand-protein/bead complexes from unbound compounds, and then releasing the bound ligands for UHPLC-MS analysis.[16]
Pulsed ultrafiltration (PUF) AS-MS.PUF AS-MS screening begins with the incubation of a mixture of compounds, such as a natural product extract with a solution-phase macromolecular receptor (protein, enzyme, or RNA). After equilibrium is achieved, ultrafiltration is used to separate the large ligand-receptor complexes from the unbound low-mass compounds. Because large pore sizes enable faster ultrafiltration separation, the pore size of the ultrafiltration membrane should be as large as possible while still retaining the macromolecular receptor.[16]
Collision-induced affinity selection mass spectrometry (CIAS-MS)Collision-induced affinity selection mass spectrometry (CIAS-MS) is a new method that relies on the affinity between a protein and its ligand for the identification of ligands.[15]
Size exclusion chromatography (SEC) AS-MSSEC AS-MS is a solution-phase screening approach like PUF AS-MS that begins with the incubation of a mixture of possible ligands with a macromolecular receptor. After equilibrium is achieved, SEC is used to separate the large ligand-receptor complexes from smaller, unbound compounds. The high mass complexes elute first during SEC and are then denatured using an organic solvent to release the ligands for reversed-phase LC-MS analysis.[16]
2. Native MS
Bioassay-guided fractionation-MSThis involves the analysis and characterization of molecules whereby the native structural features of the analytes are retained as much as possible. It provides binding informationabout each compound towards the protein of interest.[17]
Table 2. Identification of plant-derived bioactive compounds using AS-MS.
Table 2. Identification of plant-derived bioactive compounds using AS-MS.
PlantCompoundTargetDockingMain ResultsReferences
Cannabis sativa
Cannabigerolic acid
Cannabidiolic acid
Tetrahydrocannabinolic acid
COVID-19 Spike protein
Cannabigerolic acid binds to the anallosteric site of S1 with −6.6 kcal/mol binding energy.Cannabidiolic acid also binds at the orthosteric site with −6.3 kcal/mol.THCA-A bind at the orthosteric site with −6.5 kcal/mol binding energy.Bound to the spike protein thus preventing entry into the cell.[16]
Radix salvia miltiorrhiza
Salvianolic acid C
Salvianolic acid A
Xanthine oxidase (XOD)
NoSalvianolic acid C exhibited potent XOD inhibitory activity with an IC50 of 9.07 μM.[22]
Scutellaria baicalensis
Baicalein
Scutellarein
Ganhuangenin
3C-like protease (3CLpro)
NoThree flavonoids were identified as potential noncovalent inhibitors against 3CLpro with IC50 values of 0.94, 3.02, and 0.84 µM, respectively.[21]
Gancao (licorice root)
18β-glycyrrhetinic acid
Licochalcone A
Ebola virus (EBOV) nucleoprotein
Marburg virus (MARV) nucleoprotein
In silico docking analysis was employed to create a potential model for binding of GC7 and GC13 to EBOV nucleoprotein.By combining affinity mass spectrometry and metabolomics approaches, two compounds were identified from a traditional Chinese medicine Gancao (licorice root) that binds to nucleoproteins (NPs). These two ligands, 18β-glycyrrhetinic acid, and licochalcone A were verified by defined compound mixture screens and further characterized with individual ligand binding assays.[23]
Rhizoma atractylodis macrocephalaeRhizoma pinelliaeBulbus fritillariaRhizoma paridisRhizoma curcumaeFructus trichosanthisRhizoma dioscoreae bulbiferaeRadix sophorae flavescentisRadix ginsengRadix notoginsengRadix asparagi
Kurarinol
Kurarinone
Ras protein
For the docking analysis, ligands kurarinol, kurarinone, 20(s)-Rg3, and 20(s)-Rh2 were inserted into the GTP-binding pocket and the results demonstrated that kurarinol and kurarinone competed with GTP.Molecular networking and virtual screening coupled with affinity selection-mass spectrometry discovered two compounds, kurarinol and kurarinone, were confirmed to interact with GTPase of Ras and were successfully identified from 11 traditional Chinese medicine (TCM) herbs.[24]
Piper kadsuraPiper nigrumOphiopogon japonicusSalvia miltiorrhiza
HJ-1
HJ-4
HJ-6
EBOV nucleoprotein
Molecular docking studies were performed to create a docking model of HJ-4 interacting with the hydrophobic pocket in the C-lobe of the nucleoprotein.Through affinity selection-mass spectrometry approach, three compounds isolated from Piper nigrum (HJ-1, HJ-4, and HJ-6) strongly promoted the formation of large nucleoprotein oligomers and reduced the protein thermal stability, and docking studies were performed to show the interaction of HJ-4 to EBOV nucleoprotein.[25]
Glycyrrhiza inflata
Licochalcone A
Spike protein
No Small molecule ligands to the spike protein were discovered in extracts of the licorice species, Glycyrrhiza inflata. In particular, two hits were detected during screening of Glycyrrhiza inflata, and hit one was identified as licochalcone A while hit 2 corresponded to licoflavone B and glyinflanin K. However, in the absence of authentic standards, the conformation of this ligand (hit 2) is still ongoing. [3]
Rabdosia rubescensOridoninNsp9 ProteinNoA known SARS-CoV-2 Nsp9 ligand, oridonin, was successfully detected when it was mixed with Nsp9[15]
Tang-zhi-qing
2,3,4,6-Tetra-O-galloyl-d-glucose
1,2,3,4-Tetra-O-galloyl-d-glucose
1,2,3,4,6-Penta-O-galloyl-d-glucose
Quercetin-3-O-β-d-glucuronide
Quercetin-3-O-β-d-glucoside
Maltase
Invertase
Lipase
NoThrough the use of multiple target-immobilized magnetic beads coupled with high-performance liquid chromatography–mass spectrometry, five active compounds, namely, 2,3,4,6-tetra-O-galloyl-d-glucose, 1,2,3,4-tetra-O-galloyl-d-glucose, 1,2,3,4,6-penta-O-galloyl-d-glucose, quercetin-3-O-β-d-glucuronide, and quercetin-3-O-β-d-glucoside, were identified and their activities were validated by conventional inhibitory assay.[26]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ramatapa, T.; Msobo, A.; Maphari, P.W.; Ncube, E.N.; Nogemane, N.; Mhlongo, M.I. Identification of Plant-Derived Bioactive Compounds Using Affinity Mass Spectrometry and Molecular Networking. Metabolites 2022, 12, 863. https://doi.org/10.3390/metabo12090863

AMA Style

Ramatapa T, Msobo A, Maphari PW, Ncube EN, Nogemane N, Mhlongo MI. Identification of Plant-Derived Bioactive Compounds Using Affinity Mass Spectrometry and Molecular Networking. Metabolites. 2022; 12(9):863. https://doi.org/10.3390/metabo12090863

Chicago/Turabian Style

Ramatapa, Thabo, Anathi Msobo, Pfano W. Maphari, Efficient N. Ncube, Noluyolo Nogemane, and Msizi I. Mhlongo. 2022. "Identification of Plant-Derived Bioactive Compounds Using Affinity Mass Spectrometry and Molecular Networking" Metabolites 12, no. 9: 863. https://doi.org/10.3390/metabo12090863

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