Next Article in Journal
Increased Mobile Zinc Regulates Retinal Ganglion Cell Survival via Activating Mitochondrial OMA1 and Integrated Stress Response
Previous Article in Journal
Roles of Hydrogen Gas in Plants under Abiotic Stress: Current Knowledge and Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Ultrasound-Assisted Extraction, LC–MS/MS Analysis, Anticholinesterase, and Antioxidant Activities of Valuable Natural Metabolites from Astragalus armatus Willd.: In Silico Molecular Docking and In Vitro Enzymatic Studies

1
Faculty of Natural and Life Sciences, Department of Biology, Larbi Tébessi University, Tebessa 12000, Algeria
2
Laboratory of Functional and Evolutionary Ecology, Department of Biology, Faculty of Natural Sciences and Life, Research, Chadli Bendjedid University, El Tarf 36000, Algeria
3
Laboratory of Natural Substances, Biomolecules, and Biotechnological Applications, Department of Natural and Life Sciences, Larbi Ben M’Hidi University, Oum El Bouaghi 04000, Algeria
4
Estación Experimental del Zaidín (CSIC) Profesor Albareda 1, 18008 Granada, Spain
5
Biotechnology, Water, Environment and Health Laboratory, Abbes Laghrour University, Khenchela 40004, Algeria
6
Valorization of Natural Resources, Bioactive Molecules and Biological Analysis Unit, Department of Chemistry, University of Mentouri Constantine1, Constantine 1, Constantine 25000, Algeria
7
Department of Food Technology, Food Science and Nutrition, Faculty of Veterinary Sciences, University of Murcia, Regional Campus of International Excellence “Campus Mare Nostrum”, Espinardo, 30071 Murcia, Spain
8
Department of Food Science and Nutrition, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
9
Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
10
Department of Chemistry, Preparatory Year Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
11
Clinical Nutrition Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, P.O. Box 715, Makkah 24382, Saudi Arabia
12
Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
13
Toxicology and Forensic Sciences Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
*
Author to whom correspondence should be addressed.
Antioxidants 2022, 11(10), 2000; https://doi.org/10.3390/antiox11102000
Submission received: 22 September 2022 / Revised: 3 October 2022 / Accepted: 6 October 2022 / Published: 9 October 2022

Abstract

:
The Astragalus armatus Willd. plant’s phenolic constituent extraction and identification were optimized using the ultrasound-assisted extraction (UAE) method and the LC–MS/MS analysis, respectively. Additionally, cupric reducing antioxidant capacity (CUPRAC), beta carotene, reducing power, DMSO alcalin, silver nanoparticle (SNP)-based method, phenanthroline, and hydroxyl radical tests were utilized to assess the extract’s antioxidant capacity, while the neuroprotective effect was examined in vitro against acetylcholinesterase enzyme. This study accurately estimated the chemical bonding between the identified phenolic molecules derived from LC–MS/MS and the AChE. The extract was found to contain sixteen phenolic substances, and rosmarinic, protocatechuic, and chlorogenic acids, as well as 4-hydroxybenzoic, hyperoside, and hesperidin, were the most abundant substances in the extract. In all antioxidant experiments, the plant extract demonstrated strong antioxidant activity and a significant inhibitory impact against AChE (40.25 ± 1.41 μg/mL). According to molecular docking affinity to the enzyme AChE, the top-five molecules were found to be luteolin, quercetin, naringenin, rosmarinic acid, and kaempferol. Furthermore, these tested polyphenols satisfy the essential requirements for drug-like characteristics and Lipinski’s rule of five. These results highlight the significance of the A. armatus plant in cosmetics, as food additives, and in the pharmaceutical industry due to its rosmarinic and chlorogenic acid content.

1. Introduction

Synthetic compounds, notably tert-butylhydroquinone (TBHQ), propyl gallate (PG), and butylated hydroxytoluene (BHT), have been widely employed in the nutraceutical industries as the off-flavor development of foods as well as in pharmaceuticals to fight lipid oxidation [1].
Due to the negative impacts of these synthetic molecules, numerous research studies have concentrated on bioactive molecules from plants, which are used as safe antioxidant compounds [2,3]. These biomolecules have significant importance in reducing oxidative stress, which could destroy the biological mechanisms in the human body [3]. In reality, the preference for natural substances is also a consequence of the lack of adverse effects where numerous studies have shown that natural compounds counteract the negative effects of pharmaceutical treatments, implying that these natural molecules have greater medicinal value [4].
Various illnesses have been treated using plants [5] because of the existence of polyphenols, which have many biological activities [5]. Furthermore, the studies on enzyme inhibitors are regarded as the most effective in vitro strategies for a variety of diseases [6]. However, a complete understanding of the underlying physiological systems is required for the creation of therapeutic approaches [6]. Alzheimer’s disease (AD), or neurodegenerative disease, is currently affecting older adults more than ever before. AD patients’ brains were shown to have hyperactivity of acetylcholinesterase (AChE) [7]. The depletion of the acetylcholine (ACh) neurotransmitter by cholinesterase is linked to Alzheimer’s etiology [7]. Currently, the most successful therapeutic strategy is the application of ChE inhibitors [8], particularly natural anticholinesterases and antioxidants. As most of these medicines have numerous adverse effects, it is desirable to use and manage them effectively.
Legumes are typically a significant source of high-quality protein, vitamins, minerals, and bioactive compounds [9]. The growing demand for leguminous plants as a result of the scarcity of agricultural land and water resources is turning into a severe problem. Thus, food plants with substantial levels of bioactive substances are becoming more difficult to acquire and much more expensive. As a result, an important research tendency in recent years has been the discovery of additional raw and environmentally safe sources of essential nutrients, particularly from non-conventional plants [10], while still able of offering excellent nutrients for industrial use under salinity and dry situations [10].
The plants from the Astragalus genus, members of the Fabaceae family, are often utilized in food and medicine as well as feed for livestock [9,10]. Among these herbs, we are interested in Astragalus armatus Willd., which belongs to the Leguminosae plant and is prevalent in Mediterranean climate zones in Europe and North Africa [9]. This species is also found in Algeria’s Sahara Desert, Morocco, and Tunisia [10]. Aromatic compounds and essential oils are found in several Astragalus species, with a variety of medicinal and economic applications [11]. The capacity of this species to inhibit the enzymes associated with a variety of chronic illnesses has been demonstrated by several studies [10,11,12]. It has been demonstrated that several bioactive substances from this genus are effective in cancer treatment [12], including saponins, which had a substantial anticancer impact on hamsters [12]. Several polysaccharides and phenolic compounds with immunostimulatory properties are also found in this species [12].
However, a variety of solvents and methods have been employed to extract bioactive compounds from plants [12]. For this purpose, the current study aimed to extract the phenolic compound using an ultrasound-assisted extraction method, investigates the metabolite profiling of A. armatus using LC–MS/MS analyses, and assesses both its in vitro antioxidant potential and anticholinesterase effects. This study also reports a molecular docking analysis, where the identified polyphenols were used as ligands to test their inhibitory activity against AChE receptors to learn more about their mechanisms of action.

2. Materials and Methods

2.1. Chemicals and Instruments

The chemical profile was performed by using LC–MS/MS (Shimadzu, Kyoto, Japan). The activity assays were evaluated by using a Shimadzu UV spectrophotometer and a BioTek Power Wave XS microplate reader (USA). All the standard chemicals utilized in the antioxidant activities and LC–MS/MS were purchased from Merck. Germany Sigma provided the AChE as well as the reactive products of antioxidant tests (Germany). All solvents were of analytical grade [13].

2.2. Chemical Identification and Measurement of Phenolic Compound

2.2.1. Plant Extract Preparation

The aerial parts of A. armatus were taken from Algeria’s Sahara Desert (EL Oued). Therefore, the ultrasound-assisted extraction method was used in this investigation to perform the plant extraction. An ethanol–water mixture (70:30 v/v) was used to extract the plant. Following concentration with a rotary evaporator, the residual material was dissolved in water and then extracted using petroleum ether and ethanol, respectively [14]. The ethanolic fraction is the focus of our investigation.

2.2.2. Equipment and Chromatographic Parameters

The tandem MS system used in conjunction with the UHPLC (Nexera type Shimadzu, USA) was used for the LC–MS/MS technique [15]. Furthermore, The LC–MS analysis was also performed using LC-30AD binary pumps, a CTO-10ASvp column oven, a DGU-20A3R degasser, and a SIL-30AC autosampler, and the separation was carried out using a reversed-phase C18 Inertsil ODS-4 analytical column of 150 mm × 4.6 mm × 3 m at 40 °C. Mobile phase A (H2O, ammonium formate (5 mM), and formic acid 0.1%) and mobile phase B (methanol, ammonium formate (5 mM), and formic acid 0.1%) were used in the elution gradient. The solvent flow rate was kept at 0.5 mL/min, and the injection was fixed at 4 µL. The ESI source was used in air pressure ionization, where the optimal ESI circumstances were determined to be a DL temperature of 250 °C, an interface temperature of 350 °C, a heat block temperature of 400 °C, a drying gas flow rate of 15 L/min, and a nebulizing gas flow rate of 3 L/min.

2.2.3. MS Instrumentation

The MS identification was performed using a Shimadzu LCMS 8040 triple quadrupole mass spectrometer equipped with an ESI source that could operate in both positive and negative ionization mechanisms. The data from LC–MS/MS was collected and processed using Lab Solutions software (Shimadzu, Kyoto, Japan). The multiple reaction monitoring (MRM) strategy was used to quantify the analyses. The assay of the explored substances was carried out after two or three shifts per substance. The first one was for quantification, whereas the second and/or third were for confirmation.

2.2.4. Method Validation Parameters for LC–MS/MS

The detailed analytical characteristics of LC–MS/MS reference compounds have already been documented in the literature [13,14,15], including the linearity ranges and rectilinear regression estimates for the standard chemicals examined. All the calibration curves for all chemical compounds were linear and reproducible, with a correlation coefficient of more than 0.991. Ertas and Yener [13,14,15] depict, furthermore, the disclosed analytical method’s limit of detection (LOD) and limit of quantitation (LOQ). LOD varied from 0.05 to 25.8 g/L, while LOQ varied from 0.17 to 85.9 g/L. Furthermore, phenolic compound recoveries ranged from 96.9% to 106.2%.

2.3. Antioxidant Activity

The details of all biological activity studies are provided as Supplementary Material. A superoxide alkaline DMSO test, reducing power test, βeta-carotene bleaching test, cupric reducing assay (CUPRAC) measuring antioxidant capacity, the test of scavenging of hydroxyl radicals, o-phenanthroline assay, and the silver-nanoparticle-based method were used to estimate the antioxidant potential [15,16,17,18,19,20,21,22].

2.4. Acetylcholinesterase Inhibitory Assay

The anti-AChE inhibitory assay of A. armatus extract was performed using the technique outlined in Ellman’s work [23]. Briefly, 150 μL of a sodium phosphate buffer (100 Mm, pH 8.0), 10 μL of the extract, and a volume of 20 μL AChE were mixed. Then, 15 min after incubation at a suitable temperature for enzyme activation, 10 μL DTNB (0.5 mM) was introduced to the reactive mix. After that, 20 L of acetylthiocholine iodide was added to begin the reaction (0.71 mM). A 96-well microplate reader was used to measure the absorbance, and the percent inhibition was calculated as follows:
(E − S)/(E) × 100
where E is the enzyme’s activity without the extracts, and S is the enzyme’s activity with the extracts.
The experiments were carried out in triplicate. The standard compound used in the experiments was galantamine. A 50% inhibitory concentration was taken to represent the results (IC50).

2.5. Docking Molecular Analysis

This study used docking molecular analysis to measure the binding degree affinity between AChE and the polyphenols of LC–MS/MS results. First, the crystal structure of the AChE enzyme with ID 4EY6 was loaded from the PDB database (https://www.rcsb.org, accessed on 23 August 2022). The 3D structure of this receptor was prepared using the Chimera 1.15 program by removing the ligands and the co-crystallized solvents, as well as the water molecules. The global atomic charge of the protein was also corrected, and the H-atoms were added. On the other hand, the structures of the polyphenols of LC–MS/MS serving as ligands were obtained from the PubChem database (https://pubchem.ncbi.nlm.nih.gov/, accessed on 23 August 2022) in their SMILES formats, where they were converted into 3D structures using the Chimera package, which was also used to assign them Gasteiger charges and hydrogen atoms. The program AutoDock Vina was used to perform molecular docking. The outcomes were expressed in terms of binding affinity, where the ligands with the highest binding affinity were selected for the investigation of their interactions with the AChE active site.

2.6. Drug Likeness and ADMET Profiling

The drug-like qualities of the chosen phenolic substances were measured with the Swiss ADME Web Service and using the rules of Lipinski and Veber [24,25], the different biochemical attributes were determined. In addition, the two web applications ProTox-II (https://tox-new.charite.de, accessed on 26 August 2022) [26] and ADMETlab 2.0 (https://admetmesh.scbdd.com/, accessed on 26 August 2022) [27] were used to predict the toxicity of the selected ligands.

2.7. Statistical Analysis

The measurements were carried out in triplicate for each sample. The findings are expressed as mean ±standard deviation (SD). A one-way analysis of variance (ANOVA) of the bioassays was conducted using the PRISM GraphPad V: 5.00 (Trial), followed by the Tukey test at p 0.05.

3. Results and Discussion

3.1. Chemical Identification and Measurement of Phenolic Compounds

Several plants ingested as food have attracted increased interest due to the preventive impact of their phenolic compounds on cellular oxidative damage [13,14] and their availability in the human diet [28].
Numerous studies have been conducted on LC–MS/MS utilization for sensitive biomolecule characterization in plant extracts [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28]. As a result, the developed method of a triple quadrupole detector was used in this study. This method is considered more effective than other LC methods due to its high selectivity and sensitivity [5]. The base peak chromatogram (BPC) of A. armatus extract is presented in Figure 1, while the quantitative results are presented in Table 1.
In total, 16 molecules were identified and quantified in the tested plant. The differences were highly significant between the concentrations of phenolic compounds contained in the ethanolic extract (p < 0.05). It was clear that the major identified phenolic compounds were rosmarinic acid, chlorogenic acid, 4-hydroxybenzoic, protocatechuic acid, hyperoside, and hesperidin. Their 3D structures are presented in Figure 2. There was no significant variation in rosmarinic and chlorogenic acid concentrations. (p < 0.05).
In terms of the flavonoids, it was also remarkable to find a considerable level of quercetin (21.9 ± 2.1 µg/g), naringenin (5.3 ± 0.3 µg/g), luteolin (29.5 ± 2.0 µg/g), and kaempferol (35.0 ± 2.3 µg/g) in the plant extract. Additionally, a moderate amount of rutin (79.3 ± 22.7 µg/g), vanillin (65.3 ± 3.6 µg/g), salicylic acid (27.2 ± 1.4 µg/g), and low levels of tr-caffeic acid (9.8 ± 0.8 µg/g) were also observed.
In terms of non-phenolic compounds, A. armatus was found to contain a significant amount of quinic acid (12214.7 ± 664.3 µg/g) and a low value of malic acid (30.6 ± 2.3 µg/g) (Figure 1 and Table 1). Among the 27 standard chemicals used, gallic acid, tr-acotinic acid, myricetin, tannic acid, rhamnetin, coumarin, apigenin, chrysin, and fisetin were not found in the A. armatus extract.
Similar to the present work, and by using LC–ESI–MS, it has been documented that the A. armatus ethanolic fraction contains quercetin and kaempferol in low concentrations [12]. It was also mentioned that the A. armatus ethanolic fraction produced through sample extraction contained significant levels of cirsiliol (37.1 ± 1 µg/g DM).
The high concentration of phenolic content in the Astragalus genus has been observed previously [29]. Their structures were elucidated by the nuclear magnetic resonance (NMR) technical analysis in the form of 4′-dimethoxy isoflavane, 7,2′-dihydroxy-3′, isoliquiritigenin, formononetin, quercetin, ononin, kaempferol, vanillic, and p-hydroxybenzoic acids [29]. In addition, luteolin and its glucoside have been also isolated and described from A. galegiformis L., which confirms the richness of this genus in phenolic compounds. These advantageous compounds found in the extract explain the effectiveness of the ultrasound-assisted extraction technique on the determination of phenolic concentrations [1].
Additionally, this study provided the first significant findings on the high concentration of rosmarinic and chlorogenic acids in A. armatus. It may be suggested that this plant is a potential source of these two major substances. Several plant species have reportedly been tested to lengthen the shelf life of cheese and increase its quality, as well as for potential application in bread and cupcakes because of their high rosmarinic acid concentration [30]. Our results reinforce the possibility of also using this plant in this commercial context.

3.2. Antioxidant Activity

The search for bio-antioxidants with potential applications in the food, cosmetics, and medical sectors is now gaining more interest among researchers [5]. The antioxidant activity of plant extracts has an important impact on a variety of processes, including their ability to reduce and scavenge free radicals as well as their capacity to absorb oxygen radicals [5]. As a consequence, the antioxidant potential of the ethanolic extract of A. armatus was estimated using seven antioxidant tests (Table 2).
To our knowledge, there is no published research in the literature on A. armatus using these antioxidant techniques. As shown in Table 2, the terms “IC50” and “A0.50 levels” refer to the concentration at 0.50 absorbance and the 50% inhibitory percent quantity, respectively. These two values were calculated using a linear regression approach and are presented as mean SD (n = 3). The ethanolic extract was found to have a significant antioxidant activity, drawing more attention to this leguminous plant as a very promising natural antioxidant that could potentially be engaged in the treatment of various physiological pathologies caused by imbalances in oxidative systems. The observed variances in antioxidant effects that are influenced by the extracting solvent utilized have previously been examined [5]. For this reason, we can confirm that the method of extraction can significantly contributes to increasing the yield of polyphenolic content and antioxidant capacity.
Additionally, antioxidants are very essential for preventing lipid peroxidation and cellular damage caused by free radicals [28]. Lipid peroxidation is a chain reaction involving free radicals, which are associated with a wide range of biological diseases [31]. As indicated by the results, the A. armatus extract demonstrated a potent anti-lipid peroxidation activity with a high half-maximal inhibitory concentration of 5.12 ±1.2 µg/mL more than α-Tocopherol and BHT (11.43 ± 0.23 and 9.65 ± 1.1 µg/mL), which served as the standards for the β-Carotene bleaching method. As shown in Table 2, rosmarinic and chlorogenic acids are both present in very high concentrations in the A. armatus extract (80,695.32 and 76,635.0 µg/g dry extract, respectively). Therefore, it may be suggested that the observed activity is caused by rosmarinic and chlorogenic acids and their synergic effects with other phenolic compounds.

3.3. Acetylcholinesterase Inhibitory Assay

Plants are the most important source to create novel AChE inhibitor drugs for the treatment of degenerative disorders, including AD, which is considered the most common disease worldwide.
A variety of endogenous enzymes must be inhibited in order to reduce this neuropathology [32]. There have been reports that a variety of plant compounds in extracts are effective alternatives for treating Alzheimer’s disease. These bioactive substances have potent antioxidant activity that combats free radicals and reduces brain cell destruction [33,34].
There are currently few studies on the Astragalus genus and the ability of its species to inhibit enzyme function. Through this research, we aimed to demonstrate this plant’s capacity to obstruct AChE’s catalytic site.
As seen in Table 3, the AChE inhibitory activity of A. armatus was compared with that of galantamine. The plant’s IC50 value for blocking AChE was 40.25 µg/mL. The analysis using statistics demonstrates that the values were significantly different (p < 0.05).
These findings can be explained by the existence of chemicals in the extracts that are responsible for the inhibition of the AChE [4] and particularly by the ethanolic extract’s flavonoids. Our results agree with those of the investigations conducted by Teyeb et al. [35]. Additionally, the A. setulosus plant exhibited a good inhibition against BChE, according to a previous study by Zengin [36], while A. leporinus Boiss. var. hirsutus extract demonstrated a potent inhibitory activity, with an IC50 value of 66.15 ± 4.08 µg/mL.

3.4. Binding Mode Analysis Using a Molecular Docking Approach

Molecular docking has become a much more frequently used technique in the computer-aided drug development process. This groundbreaking strategy can significantly reduce energy use, costs, and time in drug discovery by screening large pharmaceutical libraries for future drug substances [37]. In the current work, we used a molecular docking method to screen the inhibitory effects of the polyphenolic components of LC–MS/MS.
It is well-established from crystal studies that Ser203 and His447 are the two major amino acids important for AChE’s catalytic properties. These two are located in the catalytic and oxyanion hole residues, which also include Glu202, Tyr33, and Trp86, located in the choline-binding pocket, and Trp236, Phe338, Phe297, and Phe295, composing the acyl-binding pocket of the enzyme [38,39]. The binding mode of the co-crystallized inhibitor galantamine, as shown in Table 4, clearly identified these key residues.
The docked reference molecule (galantamine) gave a binding energy value of −10.3 Kcal/mol and showed four hydrogen bonds, including one with the key amino acid Glu202. In addition, this molecule interacts by forming a P-Stacked amide bond with Gly121 and six Pi–alkyl bonds with Tyr337, Phe338, Phe295, Phe297, His447, and Trp86. Based on the high scores given by the top-five compounds (Table 4), it is evident that they all had a very strong affinity with the catalytic site of the enzyme. In addition, they all had interactions with at least one of the key amino acids (Table 4). With a docked score of −10.8 Kcal/mol, luteolin was determined as the ligand with the greatest inhibition potential. This molecule interacts by forming five hydrogen bonds with Asn87, Tyr133, Gly448, and Ser125. The key amino acid Trp86 provides two Pi–Pi-stacked interactions with these ligands. Moreover, several Van der Waals attractions reinforce the stability of the luteolin–AChE complex (Figure 3).

3.5. Drug-Likeness and ADMET Profiling

In Table 5, the ADME results of the top-five ligands measured using the SWISS-ADME server are displayed. Luteolin, quercetin, naringenin, rosmarinic acid, and kaempferol have molecular weights of 286.24, 302.24, 272.25, 360.31, and 286.24 g/mol, respectively; the results of these phenolic substances suggested that they could be easily transported, distributed, and absorbed through the biological membranes [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40]. In addition, luteolin, quercetin, naringenin, rosmarinic acid, and kaempferol were found to have LogP values of 1.73, 1.23, 1.84, 1.52, and 1.58, respectively, which are consistent with Lipinski’s rule of five. Additionally, the hydrogen bond donor and acceptor numbers of these ligands were less than five and less than ten, respectively, which satisfies the ADME standards of H-bond donors and acceptors. The ADME results showed that luteolin, quercetin, naringenin, rosmarinic acid, and kaempferol had topological polar surface (TPSA) values of 111.13, 131.36, 86.99, 144.52, and 111.13 Å2, respectively. These values fall within the acceptable range not exceeding Å2, as outlined earlier by Cecchelli [41]. Furthermore, the atom molar refractivity (AMR) values of the selected components varied between 71.57 for naringenin and 91.40 for rosmarinic acid, both of which fall within the margins designated for this criterion between 40 and 130 [42]. Compound solubility is another important biochemical criterion for the choice of a biologically active molecule. LogS is the measurement that represents this parameter. For the five best polyphenolic compounds, this measurement was between −3.71 for luteolin and −3.16 for quercetin, reflecting good solubility, which corresponds to fairly good absorption and distribution. The drug-likeness analysis of the top-five ligands of LC–MS/MS revealed that these metabolites have an advantageous pharmacological profile and can thus be classified as drug-like substances. The toxic behavior of the five ligands was also studied, and its results are reported in Table 6. The results from this analysis are more or less reassuring of the safety of these molecules. None of the ligands are hepatotoxic or androgenic disruptors. However, quercetin showed potential for carcinogenic and mutagenic activity, and similar to galantamine, chlorogenic acid may pose an immunotoxicological risk.

4. Conclusions

The findings of this study provide the first details on the effects of the ultrasound-assisted method of extraction on the chemical profiles as well as the antioxidants of the A. armatus extract collected from EL Oued, Algeria. As a consequence, the analysis of phytochemical constituents using the LC–MS/MS instrument attested to the presence of rosmarinic and cholinergic acids, with a high amount in the ethanolic fraction of A. armatus. Our findings also provide additional information to the literature data about the high antioxidant potential of the A. armatus plant by analyzing the results of reducing power, CUPRAC, beta carotene, DMSO alcalin, silver nanoparticle (SNP)-based method, phenanthroline, and hydroxyl radicals.
The present comprehensive study contends that five phytoconstituents, i.e., luteolin, quercetin, naringenin, rosmarinic acid, and kaempferol, are promising AChE inhibitors. Integrated molecular docking disclosed that these polyphenols establish a stable structure with the enzyme AChE with strong affinities. Furthermore, their potential inhibitors adhere to the requirements for drug-likeness and ADME assets, relying on Lipinski’s rule of five. As a result, the A. armatus plant can be used in food additives and preservatives due to its high concentration of chlorogenic and rosmarinic acids, as well as to replace synthetic antioxidants in cosmetic and pharmaceutical products.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antiox11102000/s1. In our case the supplementary material is neither a table, nor a figure or video. it is rather a part of the material and method including the detailed protocols of the different techniques used to evaluate the antioxidant activity of the extract studied.

Author Contributions

Methodology and writing, S.L., S.B. (Samira Bendjedid) and O.B.; writing—original draft preparation, R.S., A.A.M.A.-M., M.M.B., A.M.A., S.A.A., A.I.M.-G. and G.N.; writing—review and editing, visualization, K.K., S.B. (Samira Boussekine) and S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available upon request from the corresponding author.

Acknowledgments

The authors wish to express their thanks to the Algerian Ministry of Higher Education and Scientific Research (MESRS, DGRSDT) and The National Centre for Biotechnology Research (C.R.B.T) for their respective financial and material support. Additionally, special thanks to members of the Estación Experimental del Zaidín (CSIC) Profesor Albareda, Taif, Prince Sattam Bin Abdulaziz, Umm Al-Qura, King Abdulaziz, Universities, and Batterjee Medical College at Jeddah, for their scientific contributions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sarikurkcu, C.; Ozer, M.S.; Tlili, N. LC–ESI–MS/MS characterization of phytochemical and enzyme inhibitory effects of different solvent extract of Symphytum anatolicum. Ind. Crops Prod. 2019, 140, 111666. [Google Scholar] [CrossRef]
  2. Yang, C.S.; Ho, C.T.; Zhang, J.; Wan, X.; Zhang, K.; Lim, J. Antioxidants: Differing meanings in food science and health science. J. Agric. Food Chem. 2018, 66, 3063–3068. [Google Scholar] [CrossRef] [PubMed]
  3. Farhat, M.B.; Landoulsi, A.; Chaouch-Hamada, R.; Sotomayor, J.A.; Jordan, M.J. Characterization and quantification of phenolic compounds and antioxidant properties of Salvia species growing in different habitats. Ind. Crops Prod. 2013, 49, 904–914. [Google Scholar] [CrossRef]
  4. Bendjedid, S.; Djelloul, R.; Tadjine, A.; Bensouici, C.; Boukhari, A. In vitro Assessment of Total Bioactive Contents, Antioxidant, Anti-Alzheimer and Anti-diabetic Activities of Leaves Extracts and Fractions of Aloe vera. Chiang Mai Univ. J. Nat. Sci. 2020, 19, 469–485. [Google Scholar]
  5. Tlili, N.; Kirkan, B.; Sarikurkcu, C. LC–ESI–MS/MS characterization, antioxidant power and inhibitory effects on α-amylase and tyrosinase of bioactive compounds from hulls of Amygdalus communis: The influence of the extracting solvents. Ind. Crops Prod. 2019, 128, 147–152. [Google Scholar] [CrossRef]
  6. Yoon, J.-C.; Lee, S.-R.; Jung, I.-C. The Effects of Hyung Bang Sa Baek-San (Jing Fang Xie Bai San) on the Alzheimer’s Disease Model Induced by βA. J. Orient. Neuropsychiatry 2010, 21, 171–189. [Google Scholar]
  7. Mimori, Y.; Nakamura, S.; Yukawa, M. Abnormalities of acetylcholinesterase in Alzheimer’s disease with special reference to effect of acetylcholinesterase inhibitor. Behav. Brain Res. 1997, 83, 25–30. [Google Scholar] [CrossRef]
  8. Khattabi, L.; Boudiar, T.; Bouhenna, M.M.; Chettoum, A.; Chebrouk, F.; Chader, H.; Lozano-Sánchez, J.; Segura-Carretero, A.; Nieto, G.; Akkal, S. RP-HPLC-ESI-QTOF-MS Qualitative Profiling, Antioxidant, Anti-Enzymatic, Anti-Inflammatory and Non-Cytotoxic Properties of Ephedra alata Monjauzeana. Foods. 2022, 11, 145. [Google Scholar] [CrossRef] [PubMed]
  9. Magalhaes, S.C.; Taveira, M.; Cabrita, A.R.; Fonseca, A.J.; Valentão, P.; Andrade, P.B. European marketable grain legume seeds: Further insight into phenolic compounds profiles. Food Chem. 2017, 215, 177–184. [Google Scholar] [CrossRef]
  10. Mahmoudi, M.; Abdellaoui, R.; Boughalleb, F.; Yahia, B.; Mabrouk, M.; Nasria, N. Characterization of lipids, proteins, and bioactive compounds in the seeds of three Astragalus species. Food Chem. 2021, 339, 127824. [Google Scholar] [CrossRef]
  11. Agyemang, K.; Han, L.; Liu, E.; Zhang, Y.; Wang, T.; Gao, X. Recent Advances in Astragalus membranaceus Anti-Diabetic Research: Pharmacological Effects of Its Phytochemical Constituents. Evid.-Based Complement. Altern. Med. 2013, 2013, 654643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Lekmine, S.; Boussekine, S.; Akkal, S.; Martín-García, A.I.; Boumegoura, A.; Kadi, K.; Djeghim, H.; Mekersi, N.; Bendjedid, S.; Bensouici, C.; et al. Investigation of Photoprotective, Anti-Inflammatory, Antioxidant Capacities and LC–ESI–MS Phenolic Profile of Astragalus gombiformis Pomel. Foods. 2021, 10, 1937. [Google Scholar] [CrossRef] [PubMed]
  13. Ertas, A.; Yener, I. A comprehensive study on chemical and biological profiles of three herbal teas in Anatolia; rosmarinic and chlorogenic acids. S. Afr. J. Bot. 2020, 130, 274–281. [Google Scholar] [CrossRef]
  14. Lekmine, S.; Boussekine, S.; Kadi, K.; Martín-García, A.I.; Kheddouma, A.; Nagaz, K.; Bensouici, C. A comparative study on chemical profile and biological activities of aerial parts (stems, flowers, leaves, pods and seeds) of Astragalus gombiformis. Biocatal. Agric. Biotechnol. 2020, 27, 101668. [Google Scholar] [CrossRef]
  15. Kunchandy, E.; Rao, M.N.A. Oxygen radical scavenging activity of curcumin. Int. J. Pharm. 1990, 58, 237–240. [Google Scholar] [CrossRef]
  16. Oyaizu, M. Studies on products of browning reaction. Antioxidative activities of products of browning reaction prepared from glucosamine. Jpn. J. Nutr. Diet. 1986, 44, 307–315. [Google Scholar] [CrossRef] [Green Version]
  17. Marco, G.J. A rapid method for evaluation of antioxidants. J. Am. Oil Chem. Soc. 1968, 45, 594–598. [Google Scholar] [CrossRef]
  18. Apak, R.; Güçlü, K.; Özyürek, M.; Karademir, S.E. Novel Total Antioxidant Capacity Index for Dietary Polyphenols and Vitamins C and E, Using Their Cupric Ion Reducing Capability in the Presence of Neocuproine: CUPRAC Method. J. Agric. Food Chem. 2004, 52, 7970–7981. [Google Scholar] [CrossRef] [PubMed]
  19. Smirnoff, N.; Cumbes, Q.J. Hydroxyl radical scavenging activity of compatible solutes. Phytochemistry 1989, 28, 1057–1060. [Google Scholar] [CrossRef]
  20. Szydlowskaczerniak, A.; Dianoczki, C.; Recseg, K.; Karlovits, G.; Szlyk, E. Determination of antioxidant capacities of vegetable oils by ferric-ion spectrophotometric methods. Talanta 2008, 76, 899–905. [Google Scholar] [CrossRef] [PubMed]
  21. Özyürek, M.; Güngör, N.; Baki, S.; Güçlü, K.; APAK, R. Development of a Silver Nanoparticle-Based Method for the Antioxidant Capacity Measurement of Polyphenols. Anal. Chem. 2012, 84, 8052–8059. [Google Scholar] [CrossRef] [PubMed]
  22. Milica, K.; Zlatković, B.; Miladinović, B.; Živanović, S.; Mihajilov-Krstev, T.; Pavlović, D.; Kitić, D. Rosmarinic Acid Levels, Phenolic Contents, Antioxidant and Antimicrobial Activities of the Extracts from Salvia verbenaca L. Obtained with Different Solvents and Procedures: Bioeffects of Extracts from Salvia verbenaca L. J. Food Biochem. 2015, 39, 199–208. [Google Scholar]
  23. Ellman, G.L.; Courtney, K.D.; Andres, V., Jr.; Featherstone, R.M. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  24. Veber, D.F.; Johnson, S.R.; Cheng, H.Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef] [PubMed]
  25. Lipinski, C.A. Lead- and Drug-like Compounds: The Rule-Of-Five Revolution. Drug Discov. Today Tech. 2004, 1, 337–341. [Google Scholar] [CrossRef]
  26. 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] [Green Version]
  27. Xiong, G.; Wu, Z.; Yi, J.; Fu, L.; Yang, Z.; Hsieh, C.; Yin, M.; Zeng, X.; Wu, C.; Chen, X.; et al. ADMETlab2.0: An integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Res. 2021, 49, 5–14. [Google Scholar] [CrossRef]
  28. Gulcin, I.; Bursal, E.; Sehitoglu, H.M.; Goren, A.C. Polyphenol contents and antioxidant activity of lyophilized aqueous extract of propolis from Erzurum, Turkey. Food Chem. Toxicol. 2010, 48, 2227–2238. [Google Scholar] [CrossRef]
  29. Amarowicz, R.; Troszyńska, A.; Shahidi, F. Antioxidant activity of almond seed extract and its fractions. J. Food Lipids 2005, 12, 344–358. [Google Scholar] [CrossRef]
  30. Yilmaz, M.A. Simultaneous quantitative screening of 53 phytochemicals in 33 species of medicinal and aromatic plants: A detailed, robust and comprehensive LC–MS/MS method validation. Ind. Crops Prod. 2020, 149, 112347. [Google Scholar] [CrossRef]
  31. Dargel, R. Lipid peroxidation—A common pathogenetic mechanism. Exp. Toxicol. Pathol. 1992, 44, 169–181. [Google Scholar] [CrossRef]
  32. Phan, H.T.; Samarat, K.; Takamura, Y.; Azo-Oussou, A.F.; Nakazono, Y.; Vestergaard, M.C. Polyphenols modulate alzheimer’s amyloid beta aggregation in a structure-dependent manner. Nutrients 2019, 11, 756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bensaad, M.S.; Dassamiour, S.; Hambaba, L.; Bensouici, C.; Hambaba, L. In vitro assessment of antioxidant, anti-inflammatory, neuroprotective and antimicrobial activities of Centaurea tougourensis boiss. & reut. J. Pharm. Pharmacol. 2021, 9, 790–802. [Google Scholar]
  34. Mahfoudhi, A.; Prencipe, F.P.; Mighri, Z.; Pellati, F. Metabolite profiling of polyphenols in the tunisian plant Tamarix aphylla (L.) karst. J. Pharm. Biomed. Anal. 2014, 99, 97–105. [Google Scholar] [CrossRef] [PubMed]
  35. Teyeb, H.; Zanina, N.; Neffati, M.; Douki, W.; Najjar, M.F. Cytotoxic and antibacterial activities of leaf extracts of Astragalus gombiformis Pomel (Fabaceae) growing wild in Tunisia. Turk. J. Biol. 2012, 36, 53–58. [Google Scholar] [CrossRef]
  36. Zengin, G.; Uba, A.I.; Ocal, M.; Sharifi-Rad, M.; Caprioli, G.; Angeloni, S.; Altunoglu, Y.C.; Baloglu, M.C.; Yıldıztugay, E. Integration of in vitro and in silico approaches to assess three Astragalus species from Turkey flora: A novel spotlight from lab bench to functional applications. Food Biosci. 2022, 49, 101858. [Google Scholar] [CrossRef]
  37. Kumar, A.; Mishra, D.C.; Angadi, U.B.; Yadav, R.; Rai, A.; Kumar, D. Inhibition Potencies of Phytochemicals Derived from Sesame Against SARSCoV-2 Main Protease: A Molecular Docking and Simulation Study. Front. Chem. 2021, 9, 744376. [Google Scholar] [CrossRef]
  38. Brus, B.; Košak, U.; Turk, S.; Pišlar, A.; Coquelle, N.; Kos, J.; Stojan, J.; Colletier, J.P.; Gobec, S. Discovery, Biological Evaluation, and Crystal Structure of a Novel Nanomolar Selective Butyrylcholinesterase Inhibitor. J. Med. Chem. 2014, 57, 8167–8179. [Google Scholar] [CrossRef]
  39. Li, S.; Li, A.J.; Travers, J.; Xu, T.; Sakamuru, S.; Klumpp-Thomas, C.; Huang, R.; Xia, M. Identification of Compounds for Butyrylcholinesterase Inhibition. SLAS Discov. 2021, 26, 1355–1364. [Google Scholar] [CrossRef]
  40. Lipinski, C.A.; Lombardo, F.; Dominy, B.M.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 2001, 46, 3–26. [Google Scholar] [CrossRef]
  41. Cecchelli, R.; Berezowski, V.; Lundquist, S.; Culot, M.; Renftel, M.; Dehouck, M.P.; Fenart, L. Modelling of the blood-brain barrier in drug discovery and development. Nat. Rev. Drug Discov. 2007, 6, 650–661. [Google Scholar] [CrossRef] [PubMed]
  42. Benslama, O.; Benserradj, O.; Ghorri, S.; Mihoubi, I. Identification and virtual based screening of the bioinsecticidal potential of Metarhizium anisopliae destruxins as inhibitors of Culex quinquefasciatus chitinase activity. Biologia. 2022. [Google Scholar] [CrossRef]
Figure 1. Chromatogram of LC–MS/MS-analyzed A. armatus extract.
Figure 1. Chromatogram of LC–MS/MS-analyzed A. armatus extract.
Antioxidants 11 02000 g001
Figure 2. The 3D structures of the most abundant compounds in A. armatus extract visualized using Discovery Studio.
Figure 2. The 3D structures of the most abundant compounds in A. armatus extract visualized using Discovery Studio.
Antioxidants 11 02000 g002
Figure 3. Predictions of the top-five ligands’ binding modes with AChE (4EY6). Profile view and 2D schematic diagrams of (A) luteolin, (B) quercetin, (C) naringenin, (D) rosmarinic acid, and (E) kaempferol, trapped in AChE’s gorge pocket.
Figure 3. Predictions of the top-five ligands’ binding modes with AChE (4EY6). Profile view and 2D schematic diagrams of (A) luteolin, (B) quercetin, (C) naringenin, (D) rosmarinic acid, and (E) kaempferol, trapped in AChE’s gorge pocket.
Antioxidants 11 02000 g003aAntioxidants 11 02000 g003b
Table 1. LC–MS/MS data of polyphenols in A. armatus extract.
Table 1. LC–MS/MS data of polyphenols in A. armatus extract.
Peak and MoleculesRt
Retention Time
MS2
(Collision Energy)
Quantification
(µg Analyte/g Extract)
A. armatus
1Quinic acid3.3285 (22), 93 (22)963.12 ± 22.3 g
2Malic acid3.54115 (14), 71 (17)30.6 ± 2.3 i
3tr-Aconitic acid4.1385 (12), 129 (9)/
4Gallic acid4.29125 (14), 79 (25)/
5Chlorogenic acid5.43191 (17)76,635.0 ± 6.2 a
6Protocatechuic acid5.63109 (16), 108 (26)43,986.0 ± 1.2 b
7Tannic acid6.46124 (22), 78 (34)/
8tr-Caffeic acid7.37135 (15), 134 (24), 89 (31)86.71 ± 1.3 i
9Vanillin8.77136 (17), 92 (21)456.7 ± 2.4 h
10p-Coumaric acid9.53119 (15), 93 (31)/
11Rosmarinic acid9.57161 (17), 133 (42)80,695.32 ± 12.3 a
12Rutin10.18300 (37), 271 (51), 301 (38)856.5 ± 1.6 g
13Hesperidin9.69303, 46519,676.0 ± 3.2 c
14Hyperoside10.43300, 30123,976.0 ± 1.8 c
154-OH Benzoic acid11.7293, 6559,986.0 ± 6.3 b
16Salicylic acid11.7293, 65, 75433.71 ± 3.3 h
17Myricetin11.94179, 151, 137/
18Fisetin12.61135, 121/
19Coumarin12.52103, 91, 77/
20Quercetin14.48179, 151, 1216253.3 ± 2.1 d
21Naringenin14.66151, 119, 1074254.7 ± 3.1 de
22Hesperetin15.29164, 136, 108/
23Luteolin15.43175, 151, 1333221.5 ± 6.3 e
24Kaempferol15.43217, 133, 1512451.5 ± 1.8 ef
25Apigenin17.31151, 117/
26Rhamnetin18.94165, 121, 300/
27Chrysin21.18143, 119, 107/
/: Not found. MS2: segments of the MRM for the associated molecular ions. Significant differences exist between the values with various letters (a, b, c, d, e, f, g, h, and i) (p 0.05).
Table 2. Antioxidant activity the ethanolic fraction of A. armatus.
Table 2. Antioxidant activity the ethanolic fraction of A. armatus.
ProductsCUPRAC (A0.5)Reducing Power
(A0.5)
Beta Carotene
(IC50)
DMSO Alcalin
(IC50)
SNP (IC50)Phenanthroline
(A0.5)
Hydroxyl Radical
(IC50)
A. armatus14.58 ± 4.56 c25 ± 1.12 b5.12 ±1.2 a13 ± 1.25 b10.2 ± 1.7 b66 ± 1.14 d25 ± 1.2 b
BHT *8.86 ± 2.8 b/9.65 ± 1.1 b//2.33 ± 1.7 b/
BHA *5.26 ± 1.6 a/9.82 ± 2.1 b//2.84 ± 2.7 b/
A-tocopherol */34.93 ± 2.38 c11.43 ± 0.23 c4.2 ± 0.95 a///
Ascorbic acid *8.31 ± 0.1 b6.77 ± 1.15 a//7.14 ± 0.05 a3.08 ± 0.02 a12.33 ± 1.17 a
Tannic acid */5.17 ± 1.2 a/3.3 ± 0.91 a///
Trolox *8.69 ± 0.1 b5.23 ± 1.2 a//33.26 ± 2.1 c5.21 ± 0.27 c/
* Reference compounds. /, not tested.
Table 3. Galantamine standard and A. armatus fraction IC50 values (g/mL) for inhibiting acetylcholinesterase activity.
Table 3. Galantamine standard and A. armatus fraction IC50 values (g/mL) for inhibiting acetylcholinesterase activity.
ExtractIC50 Values (g/mL) for Inhibiting Acetylcholinesterase Activity
A. armatus40.25 ± 1.3 a
Galantamine34.75 ± 1.1 b
Table 4. The best results for the docking of LC–MS/MS polyphenolic ligands with AChE target.
Table 4. The best results for the docking of LC–MS/MS polyphenolic ligands with AChE target.
Binding Energy (Kcal/mol)Hydrogen InteractionsHydrophobic InteractionsVan der Waals
Interactions
Galantamine−10.3Ser203, Glu202, Tyr124, Asp74, His447Tyr337, Gly121, Phe338, Phe295, Phe297, His447, Trp86Gly122, Ser125, Tyr341, Gly120, Tyr133, Gly448
Luteolin−10.8Asn87, Tyr133, Gly448, Ser125Trp86Pro88, Gln71, Gly121, Gly120, Tyr199, Glu202, Ile451, His447, Tyr337, Tyr124, Asp74, Tyr72, Val73
Quercetin−10.6Ser125, Asp74, Tyr72, Asn87Trp86, Tyr337Ile451, His447, Tyr449, Gly448, Glu202, Ser203, Tyr133, Gly120, Gly121, Tyr124, Gly126, Val73, Pro88, Gln71
Naringenin−10.2Ser125, Asn87, Glu202Trp86, Tyr337Val73, Tyr124, Tyr449, His447, Gly448, Ser203, Ile451, Gly120, Tyr133, Gly121, Pro88, Gly121
Rosmarinic acid−10.2Tyr124, Gly121, Gly122, His447, Ser203, Trp86, Tyr133, Val294Trp86, Trp286Ser293, Phe295, Phe297, Tyr337, Tyr119, Ala127, Gly126, Leu130, Ser125, Gly120, ALA204, Phe338, Tyr341
Kaempferol−10.0Glu202, His447, Gly121Trp86, Tyr337Tyr124, Tyr72, Val73, Asn87, Pro88, Gly126, Gly120, Tyr133, Ile451, Ser203, Gly448, Tyr449
Table 5. Drug-like properties of the top active ligands of LC–MS/MS.
Table 5. Drug-like properties of the top active ligands of LC–MS/MS.
MW g/molLogPLogSHBAHBDTPSA (Å2)AMRnRBLipinskiVeber
Galantamine287.351.91−2.931441.9384.051YesYes
Luteolin286.241.73−3.7164111.1376.011YesYes
Quercetin302.241.23−3.1675131.3678.031YesYes
Naringenin272.251.84−3.495386.9971.571YesYes
Rosmarinic acid360.311.52−3.4485144.5291.407YesNo
Kaempferol286.241.58−3.3164111.1376.011YesYes
HBA, Num. H-bond acceptors; HBD, Num. H-bond donors; nRB, Num. rotatable bonds; AMR, atom molar refractivity.
Table 6. Drug-like properties of the top active ligands of LC–MS/MS.
Table 6. Drug-like properties of the top active ligands of LC–MS/MS.
CriteriaGalantamineLuteolinQuercetinNaringeninChlorogenic AcidKaempferol
Absorption-DistributionBBB penetrationYesNoNoNoNoNo
Caco2HighHighLowHighLowHigh
HIAHighHighHighHighLowHigh
MetabolismCYP1A2 inhibitorNoyesYesNoNoYes
CYP2C19 inhibitorNoNoNoNoNoNo
CYP2C9 inhibitorNoNoNoNoNoNo
CYP2D6 inhibitorYesYesYesYesNoYes
CYP3A4 inhibitorNoYesYesYesNoYes
ExcretionClLowModerateModerateHighLowModerate
ToxicityhERG BlockersNoNoNoNoNoNo
AMES ToxicityNoYesYesNoNoNo
CarcinogenicityNoNoYesNoNoNo
CytotoxicityNoNoNoYesNoNo
ImmunotoxicityYesNoNoNoYesNo
H-HTNoNoNoNoNoNo
NR-ARNoNoNoNoNoNo
NR-ERNoYesYesYesNoYes
SR-p53NoNoNoYesNoNo
BBB, Blood–brain barrier; HIA, human intestinal absorption; Caco2, permeability assay; hERG, human ether-a-go-go-related gene potassium channel; H-HT, human hepatotoxicity; NR-AR, androgen receptor disruptor; NR-ER, estrogen receptor disruptor; SR-p53, tumor suppressor protein p53 activator; Cl, clearance of the molecule; No, inactive; Yes, active.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lekmine, S.; Bendjedid, S.; Benslama, O.; Martín-García, A.I.; Boussekine, S.; Kadi, K.; Akkal, S.; Nieto, G.; Sami, R.; Al-Mushhin, A.A.M.; et al. Ultrasound-Assisted Extraction, LC–MS/MS Analysis, Anticholinesterase, and Antioxidant Activities of Valuable Natural Metabolites from Astragalus armatus Willd.: In Silico Molecular Docking and In Vitro Enzymatic Studies. Antioxidants 2022, 11, 2000. https://doi.org/10.3390/antiox11102000

AMA Style

Lekmine S, Bendjedid S, Benslama O, Martín-García AI, Boussekine S, Kadi K, Akkal S, Nieto G, Sami R, Al-Mushhin AAM, et al. Ultrasound-Assisted Extraction, LC–MS/MS Analysis, Anticholinesterase, and Antioxidant Activities of Valuable Natural Metabolites from Astragalus armatus Willd.: In Silico Molecular Docking and In Vitro Enzymatic Studies. Antioxidants. 2022; 11(10):2000. https://doi.org/10.3390/antiox11102000

Chicago/Turabian Style

Lekmine, Sabrina, Samira Bendjedid, Ouided Benslama, Antonio Ignacio Martín-García, Samira Boussekine, Kenza Kadi, Salah Akkal, Gema Nieto, Rokayya Sami, Amina A. M. Al-Mushhin, and et al. 2022. "Ultrasound-Assisted Extraction, LC–MS/MS Analysis, Anticholinesterase, and Antioxidant Activities of Valuable Natural Metabolites from Astragalus armatus Willd.: In Silico Molecular Docking and In Vitro Enzymatic Studies" Antioxidants 11, no. 10: 2000. https://doi.org/10.3390/antiox11102000

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