Next Article in Journal
Antiulcer Potential of Psidium guajava Seed Extract Supported by Metabolic Profiling and Molecular Docking
Next Article in Special Issue
Impact of Gastrointestinal Digestion on the Anti-Inflammatory Properties of Phlorotannins from Himanthalia elongata
Previous Article in Journal
Chemical Characterization of Taif Rose (Rosa damascena) Methanolic Extract and Its Physiological Effect on Liver Functions, Blood Indices, Antioxidant Capacity, and Heart Vitality against Cadmium Chloride Toxicity
Previous Article in Special Issue
Microwave-Assisted Dendropanax morbifera Extract for Cosmetic Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Bioactive Components from Ampelopsis japonica with Antioxidant, Anti-α-Glucosidase, and Antiacetylcholinesterase Activities

1
Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
2
Department of Chemistry, College of Science, National Kaohsiung Normal University, Kaohsiung 82444, Taiwan
3
Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
4
Ministry of Health and Welfare, National Research Institute of Chinese Medicine, Taipei 112026, Taiwan
5
Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan
6
Department of Physical Medicine and Rehabilitation, Kaohsiung Veterans General Hospital Tainan Branch, Tainan City 710011, Taiwan
7
Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan City 717302, Taiwan
8
Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404332, Taiwan
*
Authors to whom correspondence should be addressed.
Antioxidants 2022, 11(7), 1228; https://doi.org/10.3390/antiox11071228
Submission received: 19 May 2022 / Revised: 8 June 2022 / Accepted: 20 June 2022 / Published: 23 June 2022

Abstract

:
The dried root of Ampelopsis japonica (Thunb.) Makino (A. japonica.) is a traditional medicine used to treat fever, pain, and wound healing. It exhibits anti-inflammatory, antitumor, antityrosinase, and antimelanogenic activities. In this paper, we used different solvent extracts from the root of A. japonica to determine their antioxidant activity. Acetone extract showed relatively strong antioxidant properties by 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), 2,2-diphenyl-1-(2,4,6-trinitrophenyl)hydrazyl (DPPH), superoxide radical scavenging activity, and ferric reducing antioxidant power (FRAP) assays. In addition, these extracts also showed significant α-glucosidase and acetylcholinesterase (AChE) inhibitory activities. Acetone extract significantly inhibited α-glucosidase with an IC50 value of 8.30 ± 0.78 μg/mL, and ethanol extract remarkably inhibited AChE with an IC50 value of 37.08 ± 7.67 μg/mL. Using HPLC analysis and comparison with the chemical composition of various solvent extracts, we isolated seven active compounds and assessed their antioxidant, anti-α-glucosidase, and anti-AChE activities. Catechin (1), gallic acid (2), kaempferol (3), quercetin (4), resveratrol (6), and epicatechin (7) were the main antioxidant components in the root of A. japonica. According to the results of DPPH, ABTS, and superoxide radical scavenging assays, these isolates showed stronger antioxidant capacity than butylated hydroxytoluene (BHT). Moreover, 1, 3, 4, euscaphic acid (5), 6, and 7 also expressed stronger anti-α-glucosidase activity than the positive control acarbose, and all the isolated compounds had a good inhibitory effect on AChE. Molecular docking models and hydrophilic interactive modes for AChE assays suggest that 1 and 5 exhibit unique anti-AChE potency. This study indicates that A. japonica and its active extracts and components may be a promising source of natural antioxidants, α-glucosidase, and AChE inhibitors.

1. Introduction

Oxidation is a natural process that forms free radicals called reactive oxygen species (ROS) and reactive nitrogen species (RNS) through a series of intermediate byproducts [1]. Free radicals are very active atoms or molecules. Because they have unpaired electrons, they can easily snatch electrons from other molecules and cause damage. These free radicals cause oxidative damage to biological macromolecules and damage to physiological macromolecules such as DNA, proteins, and lipids [2]. Cardiovascular disease, diabetes, cancer, neurodegenerative diseases, Alzheimer’s disease, and inflammatory diseases may occur in the body when antioxidants are lacking to eliminate excess reactive free radicals [3]. In order to reduce the oxidative damage of reactive free radicals, many synthetic antioxidants such as butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT) have strong antioxidant activities and have been widely used in commercial foods in the form of additives. In recent years, the use of synthetic antioxidants has been limited due to their possible carcinogenic and toxic effects [4]. Therefore, natural antioxidants present in food and other biological materials are of interest due to their safety and potential nutritional value [5]. Antioxidants in Chinese herbal medicine (CHM) are popular for their low toxicity and desirable pharmacological activity [6,7]. Among the secondary metabolites of CHM, polyphenols have important inhibitory activities on enzymes such as acetylcholinesterase (AChE) and α-glucosidase [8].
Diabetes is a metabolic disorder caused by high blood sugar. One of the goals of reducing hyperglycemia is to reduce the activity of α-glucosidase, which is responsible for the hydrolysis of carbohydrates. α-Glucosidase inhibitors delay the intestinal absorption of glucose, thereby limiting the fluctuation of postprandial blood glucose [9]. There are several antidiabetic medications, such as acarbose, voglibose, and miglitol, which reduce high blood sugar levels by inhibiting the activity of α-glucosidase. However, continued use of synthetic drugs often results in adverse side effects such as liver toxicity, abdominal cramps, diarrhea, and vomiting [10,11]. On the other hand, acetylcholinesterase inhibitors are the most suitable modern and primary treatments against neurodegenerative diseases [12]. It is also a key enzyme in one of the treatment strategies for Alzheimer’s disease (AD). Acetylcholinesterase inhibitors block the action of AChE, thereby increasing the level of acetylcholine in the brain, which would otherwise be hydrolyzed to acetic acid and choline [13]. Currently, AChE inhibitors such as galantamine, donepezil, and rivastigmine have been used for the treatment of AD [14]. However, the efficacy of these drugs is hindered by their side effects, e.g., hepatotoxicity, gastrointestinal disturbances, and hypotension [15]. Therefore, natural antioxidants may be favorable candidates for the treatment of related diseases such as neurodegenerative diseases and diabetes.
The dried root of Ampelopsis japonica (Thunb.) Makino is a traditional remedy for fever, pain, and wound healing. Numerous pharmacological properties of this material have been reported, such as anti-inflammatory, antitumor, antityrosinase, and antimelanogenic activities [16]. In this study, we investigated the effects of various solvent extracts and major bioactive compounds from the root of A. japonica on antioxidant, anti-α-glucosidase, and AChE inhibitory activities.

2. Materials and Methods

2.1. Chemicals and General Procedures

Gallic acid, Folin–Ciocalteu reagent, hydrogen peroxide solution, α-glucosidase, acetylthiocholine iodide, 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid), acetylcholinesterase, 5,5′-dithio-bis(2-nitrobenzoic) acid, 2,4,6-tripyridyl-s-triazine, and Trolox were bought from Sigma-Aldrich. Nitroblue tetrazolium, 2,2-diphenyl-1-(2,4,6-trinitrophenyl)hydrazyl, loroglucinol, and phenazine methosulphate were supplied by Tokyo Chemical Industry Co., Ltd. (Tokyo, Japan). Aluminum chloride, ferric chloride, and p-nitrophenyl-α-D-glucopyranoside were bought from Alfa Aesar. Potassium dihydrogenphosphate, sodium carbonate, potassium peroxodisulfate, disodium hydrogenphosphate, and sodium carbonate were bought from the Showa Chemical Industry Co., Ltd. (Tokyo, Japan). Sodium acetate, butyl hydroxytoluene, potassium acetate, and nicotinamide adenine dinucleotide were supplied by Acros Organics. Galanthamine hydrobromide was bought from MedchemExpress.

2.2. Preparation of A. japonica Extracts

Preparation of A. japonica extracts was carried out as previously described [6]. Samples were collected, air-dried, and ground to powder. 200 mL of different solvents (n-hexane, dichloromethane, chloroform, acetone, ethyl acetate, ethanol, methanol, and water) were added to 20 g of powder and incubated with shaking by orbital shakers for 24 h at room temperature. The extracts were filtered and condensed under reduced pressure at 38 °C.

2.3. Preparation of Active Compounds

The root (100 g) of A. japonica was pulverized and extracted 3 times with MeOH (3 × 500 mL, 3 d each). The MeOH extract was concentrated under reduced pressure at 37 °C to give a residue (Fraction M, 5.64 g). Fraction M (5.46 g) was separated by column chromatography (215 g of silica gel, 70–230 mesh; n-hexane/ethyl acetate gradient) to obtain 11 fractions: M1–M11. Part (120.6 mg) of Fraction M2 was further by semi-preparative HPLC (reversed-phase C18 silica gel; 0.2% acetic acid in water (v/v)/acetonitrile acetate gradient) to obtain 5 (5.3 mg) and 6 (10.2 mg). Part (160.4 mg) of Fraction M3 was further by semi-preparative HPLC (reversed-phase C18 silica gel; 0.2% acetic acid in water (v/v)/methanol acetate gradient) to afford 7 (8.45 mg). Part (220 mg) of Fraction M4 was further purified by semi-preparative HPLC (normal-phase silica gel; n-hexane/acetone acetate gradient) to afford 3 (9.24 mg), and 4 (10.3 mg). Part (182 mg) of Fraction M5 was further by semi-preparative HPLC (reversed-phase C18 silica gel; 0.2% acetic acid in water (v/v)/methanol acetate gradient) to obtain 2 (26.3 mg). Part (86.4 mg) of Fraction M7 was further by semi-preparative HPLC (reversed-phase C18 silica gel; 0.2% acetic acid in water (v/v)/acetonitrile acetate gradient) to obtain 1 (15.4 mg). The structures of 1-7 were identified by NMR spectra (Figures S1–S7).

2.4. Reversed-Phase HPLC

The reversed-phase HPLC assay to measure the seven components was performed as previously described with minor modifications [17,18]. Reversed-phase separations were executed using a LiChrospher® 100 RP-18 Endcapped (5 μm; column of dimensions 4.6 × 250 mm). Gradient separation using 0.2% acetic acid in water (v/v) (solvent A) and methanol (solvent B) as mobile phase was as follows: 0-8 min, isocratic elution with 3% B; 8–10 min, gradient elution from 3 to 5% B; 10–25 min, isocratic elution with 5% B; 25–30 min, gradient elution from 5 to 20% B; 30–35 min, isocratic elution with 20% B; 35–150 min, gradient elution from 20 to 100% B; 150–155 min, back to initial conditions at 3% B; and 155–160 min, isocratic elution with 3% B. The flow rate was 1.0 mL/min, and the injection volume was 50 μL at room temperature. Peaks were detected at 280 nm. Different compounds were identified by retention time.

2.5. Determination of Total Phenolic Content (TPC)

TPC of different solvent extracts was determined in accordance with the method previously reported [19].

2.6. Determination of Total Flavonoid Content (TFC)

TFC of various solvent extracts was measured by the reference method [20].

2.7. DPPH Radical Scavenging Activity

This assay was determined using the procedure previously reported [21].

2.8. ABTS Radical Scavenging Assay

This assay was assessed using the procedure previously described [22].

2.9. Superoxide Radical Scavenging Assay

This assay was determined by the method previously described [23].

2.10. Ferric Reducing Antioxidant Power (FRAP) Assay

The FRAP assay was measured using the procedure previously described [24].

2.11. α-Glucosidase Inhibitory Activity Assay

α-Glucosidase inhibitory activity was measured according to the method previously described [25].

2.12. Acetylcholinesterase (AChE) Inhibitory Assay

AChE inhibitory assay was determined using the reference method, with minor changes [26]. Briefly, 140 μL of 0.1 M sodium phosphate buffer (pH 8.0), 10 µL of DTNB, 20 µL of sample, and 15 µL of AChE solution were added in a 96-well microplate and incubated for 10 min. The reaction was then initiated with the addition of 10 µL of acetylthiocholine iodide, followed by an additional 10 min incubation. The absorbance was evaluated at 405 nm using a spectrophotometer.

2.13. Molecular Modeling Docking Study

All calculations were performed by Discovery Studio 2019 (San Diego, CA, USA) software. This study was calculated using the method previously described [17,18].

2.14. Statistical Analysis

The data are displayed as mean ± SEM. Statistical analysis was performed using Student’s t-test. A probability of 0.05 or less was considered statistically significant.

3. Results

3.1. Determination of TPC, TFC, and Yields in Each Solvent Extract

Various solvent extracts from A. japonica were assessed for TPC, TFC, and yields (Table 1). The yields of A. japonica different solvent extracts ranged from 0.28 ± 0.08% (n-hexane extract) to 16.46 ± 0.31 % (water extract). There were significant differences in TPC of various solvent extracts, among which the acetone extract of A. japonica had the highest TPC content (142.89 ± 4.07 mg/g), in succeeding order of methanol (95.98 ± 6.68 mg/g) > ethanol (95.69 ± 7.80 mg/g) > ethyl acetate (79.09 ± 8.45 mg/g) > water (47.38 ± 2.18 mg/g) > chloroform (20.92 ± 1.47 mg/g) > dichloromethane (20.19 ± 1.99 mg/g) > n-hexane (15.01 ± 0.44 mg/g). There were obvious differences in TFC content of different solvent extracts, among which the chloroform extract had the highest TFC content (94.22 ± 1.34 mg/g), followed by ethyl acetate (80.05 ± 7.82 mg/g), acetone (71.72 ± 2.66 mg/g), dichloromethane (62.60 ± 3.51 mg/g), n-hexane (56.36 ± 3.86 mg/g), ethanol (22.75 ± 1.24 mg/g), methanol (5.72 ± 1.13 mg/g), and water (4.45 ± 1.21 mg/g).

3.2. DPPH Radical Scavenging Activity

The acetone extract (SC50 = 54.88 ± 4.04 μg/mL) showed strong DPPH radical scavenging activity, followed by methanol (SC50 = 84.73 ± 7.82 μg/mL), ethanol (SC50 = 87.12 ± 6.45 μg/mL), ethyl acetate (SC50 = 92.14 ± 8.12 μg/mL), and water (SC50 = 98.54 ± 7.09 μg/mL), as shown in Table 2. In addition, the extracts of n-hexane, chloroform, and dichloromethane had no significant effect (SC50 > 200 μg/mL).

3.3. ABTS Free Radical Scavenging Effect

The acetone extract (SC50 = 33.88 ± 2.13 μg/mL) displayed the strongest ABTS radical scavenging activity, in succeeding order of methanol (SC50 = 53.77 ± 4.65 μg/mL), ethyl acetate (SC50 = 57.45 ± 4.74 μg/mL), ethanol (SC50 = 64.56 ± 4.80 μg/mL), and water (SC50 = 99.30 ± 7.02 μg/mL), as shown in Table 2. In addition, dichloromethane, chloroform, and n-hexane extracts had no significant effect (SC50 > 200 μg/mL).

3.4. Superoxide Radical Scavenging Effect

From the result in Table 2, all extracts of A. japonica were tested for their superoxide radical scavenging activity. Notably, the results exhibited that, except for methanol (SC50 = 290.83 ± 15.23 μg/mL), ethanol (SC50 = 307.20 ± 22.39 μg/mL), and water (SC50 = 313.84 ± 20.24 μg/mL), other extracts had no significant effect (SC50 > 400 μg/mL).

3.5. Ferric Reducing Antioxidant Power (FRAP) Effect

The acetone extract displayed the highest FRAP value (1001.00 ± 46.17 TE mM/g), followed by ethanol (736.95 ± 14.40 TE mM/g), methanol (712.56 ± 18.32 TE mM/g), ethyl acetate (587.11 ± 20.61 TE mM/g), and water (413.34 ± 21.08 TE mM/g) in Table 2. The solvent extracts with low polarities, such as dichloromethane, chloroform, and n-hexane, showed relatively weak FRAP values.

3.6. Anti-α-Glucosidase Effect

The anti-α-glucosidase activity of the acetone extract of A. japonica was the strongest (IC50 = 8.30 ± 0.78 μg/mL), followed by ethanol (IC50 = 11.06 ± 2.07 μg/mL), ethyl acetate (IC50 = 12.51 ± 2.42 μg/mL), methanol (IC50 = 19.27 ± 1.12 μg/mL), dichloromethane (IC50 = 28.00 ± 0.14 μg/mL), n-hexane (IC50 = 28.43 ± 3.78 μg/mL), and chloroform (IC50 = 34.16 ± 3.88 μg/mL), as shown in Table 3. Most various solvent extracts showed more potent anti-α-glucosidase activity than acarbose (IC50 = 335.50 ± 2.14 μg/mL). These results indicated that various solvent extracts of A. japonica had strong α-glucosidase inhibitory effects, except the water extract.

3.7. AChE Inhibition Activity

A previous study showed that chlorogenic acid has an AChE inhibitory effect [27]. Thus, chlorogenic acid was employed as a positive control. The ethanolic extract of A. japonica exhibited the highest inhibitory activity against AChE with an IC50 value of 37.08 ± 7.67 μg/mL, followed by acetone, methanol, n-hexane, water, dichloromethane, chloroform, and ethyl acetate, with IC50 values of 61.95 ± 5.54, 77.99 ± 5.08, 83.97 ± 8.90, 85.82 ± 8.74, 91.47 ± 26.03, 91.64 ± 8.77, and 103.30 ± 2.15 μg/mL, respectively (Table 3). All extracts showed good AChE inhibitory activity.

3.8. Quantitation of Active Components in Different Solvent Extracts

The HPLC methods using reversed-phase columns for the quantification of seven components isolated from A. japonica were verified regarding linearity, LOD, and LOQ. The linearity was validated by the data from six different concentrations (1.0, 5.0, 10.0, 25.0, 50.0, and 100.0 μg/mL) of the standard solutions. The linear regression parameters of calibration curves, correlation coefficient, LOD, and LOQ are shown in Table S1. Figure S8 shows the HPLC chromatogram of seven active ingredients.
Figures S9–S16 shows the HPLC analyses of seven active ingredients [catechin (1), gallic acid (2), kaempferol (3), quercetin (4), euscaphic acid (5), resveratrol (6), and epicatechin (7)] (Figure 1) from different solvent extracts of A. japonica quantification (Table 4). Of all the extracts, the acetone extract had the highest content of the seven active ingredients. Of the seven active ingredients in all solvent extracts, compound 2 has the highest content.

3.9. Antioxidant Activities of Isolated Components

Seven isolated compounds were tested for antioxidant activity. The results are shown in Table 5, compound 2 (SC50 = 2.60 ± 0.67 μg/mL) showed the strongest DPPH radical scavenging activity, followed by 7 (SC50 = 2.78 ± 0.25 μg/mL), 4 (SC50 = 3.36 ± 0.58 μg/mL), 1 (SC50 = 10.08 ± 3.09 μg/mL), 3 (SC50 = 12.48 ± 3.01 μg/mL), and 6 (SC50 = 13.19 ± 4.78 μg/mL). There are six components showed stronger ABTS radical scavenging activities than BHT, in succeeding order of 2 (SC50 = 1.45 ± 0.14 μg/mL) > 1 (SC50 = 2.23 ± 0.22 μg/mL) > 6 (SC50 = 2.81 ± 0.12 μg/ mL) > 4 (SC50 = 3.15 ± 0.49 μg/mL) > 7 (SC50 = 3.78 ± 0.03 μg/mL) > 3 (SC50 = 5.24 ± 0.45 μg/mL). Compound 4 (SC50 = 31.89 ± 2.03 μg/mL) exhibited stronger superoxide radical scavenging activity than all isolated compounds. Furthermore, compound 2 (28,512.82 ± 43.27 mM TE/g), 4 (16,038.26 ± 86.89 mM TE/g), 7 (13,122.77 ± 182.42 mM TE/g), 1 (8729.33 ± 424.55 mM TE/g), 3 (7912.47 ± 220.08 mM TE/g), and 6 (7453.94 ± 60.09 mM TE/g) had higher antioxidant activity than BHT via FRAP assay.

3.10. Anti-α-Glucosidase Activities of Isolated Components

To further evaluate the inhibitory activity of α-glucosidase, the main components isolated from A. japonica were investigated. The results are shown in Table 6, compound 3 (IC50 = 5.81 ± 2.70 μg/mL), 4 (IC50 = 14.39 ± 5.93 μg/mL), 5 (IC50 = 20.38 ± 2.13 μg/mL), 6 (IC50 = 28.81 ± 5.65 μg/mL), 1 (IC50 = 81.78 ± 11.58 μg/mL), and 7 (IC50 = 88.73 ± 10.94 μg/mL) showed stronger α-glucosidase inhibitory activity than the positive control acarbose (IC50 = 334.53 ± 2.22 μg/mL).

3.11. AChE Inhibition Assay of Isolated Components

The inhibitory activity of seven main components from A. japonica against AChE is shown in Table 6. The results indicated that 5 (IC50 = 11.64 ± 2.69 μg/mL), 1 (IC50 = 26.35 ± 9.55 μg/mL), 2 (IC50 = 41.59 ± 7.57 μg/mL), 7 (IC50 = 53.38 ± 7.30 μg/mL), and 3 (IC50 = 55.04 ± 8.57 μg/mL) displayed stronger anti-AChE activity than chlorogenic acid (positive control) (IC50 = 64.42 ± 0.16 μg/mL).

3.12. Molecular Modeling Docking

The 3D crystal structure of the acetylcholinesterase complexed with acetylcholine (PDB: 2ACE) from Torpedo californica exhibits that the substrate binding site of acetylcholinesterase is formed by 14α-helices, 14β-sheets, and numerous loops in a gorge shape [28]. Once acetylcholine enters the substrate binding pocket by leaning its acetyl group toward the catalytic site (esteric site) and resides its trimethylamine group to the anionic site, it is surrounded by hydrophilic and hydrophobic residues including Trp 84, Gly 119, Glu 199, Ser 200, Phe 288, Phe 299, Glu 327, Phe 331, and His 440. Among these residues, Ser 200, Glu 327, and His 440 are viewed as the key residues at the esteric site to perform the hydrolytic reaction for acetylcholine when Ser 200 acts as the nucleophile. Furthermore, Trp 84 is considered as the essential residue at the anionic site. For the interactive binding mode of acetylcholine in the substrate binding pocket, the carbonyl group of acetylcholine is reduced to the hydroxyl group, which further interacts with the backbone of Gly 119 by acting as the H-bond acceptor. Apart from this H-bond interaction, there is no significant hydrophilic interaction between acetylcholine and the substrate binding pocket, especially the trimethylamine group of acetylcholine, which has been hypothesized to possibly make essential ionic interaction with the residues at the anionic site.
For the binding mode of galanthamine (Figure 2a) in the substrate binding pocket of acetylcholinesterase, the crystal structure of the acetylcholinesterase complexed with (−)-galanthamine (PDB: 1W6R) from Torpedo californica has been disclosed [29]. Galanthamine binds to the substrate binding pocket by leaning its A, B, and C rings at a similar position as the acetyl group of acetylcholine, and locates its D ring at the anionic site. Since the A, B, and C rings of galanthamine are nearby Ser 200 and His 440, their three hydrophilic groups can form hydrophilic interactions with the esteric site, including (1) the 9-methoxy group on the A ring interacts with Ser 200 by performing as the H-bond acceptor, (2) the oxygen atom on the B ring also serves as the H-bond acceptor to interact with Ser 200, and (3) the 14-hydroxyl group on the C ring contacts with Glu 199 by serving as the H-bond donor. Additionally, the D ring of galanthamine, containing a tertiary amino group, which does not exhibit obvious ionic interaction, employs the methyl substituent on the amino group to make the nonclassical H-bond interaction with Asp 72. More importantly, galanthamine makes several essential π–π interactions in the substrate binding pocket, including (1) the A ring of galanthamine interacts with Phe 331, (2) the C ring of galanthamine contacts with Trp 84, and (3) the D ring of galanthamine interacts with Trp 84 and Phe 330.
To further study the interaction between catechin (1) (Figure 2b) (or gallic acid (2)) and acetylcholinesterase, and try to interpret how 1 (or 2) might exert its antagonistic effect, The crystal structure of the acetylcholinesterase from Electrophorus electricus (PDB: 1C2B) was also used in this study [30]. The crystal structures of acetylcholinesterase from Electrophorus electricus and Torpedo californica share high sequence homology and have very similar conformation. In the substrate binding site for both structures, the key residues, including Ser 203 (Ser 200 in Torpedo californica), His 447 (His 440 in Torpedo californica), Glu 334 (Glu 327 in Torpedo californica), Trp 86 (Trp 84 in Torpedo californica), Tyr 133 (Tyr 130 in Torpedo californica), Phe 338 (Phe 331 in Torpedo californica), and Tyr 449 (Tyr 442 in Torpedo californica), are almost identical. For the binding model of 2, 2 resided in the substrate binding pocket by leaning its three hydroxyl substituents toward the esteric site and locating its benzoic acid moiety at the anionic site. Compound 2 made two H-bond interactions comprising (1) the 3-hydroxyl group docked with the backbone of Gly 120 by serving as the H-bond donor, and (2) the carboxylate group made H-bond contact with Tyr 124 and Ser 125 by performing as the H-bond acceptor. Additionally, 2 made the essential π–π interaction with Trp 86 at the anionic site.
The docking model of catechin (1) exhibited that 1 resided at a similar position as galanthamine, as shown in Figure 3. In the substrate binding site, the B ring of 1 as the B ring of galanthamine leaned toward Ser 203 at the terminus of the α-helice shown by the purple color in Figure 3, and the A ring of 1 as the C ring of galanthamine located nearby Trp 86. However, the C ring of 1 resided at a different position than the D ring of galanthamine. The C ring of 1 stayed between Tyr 124 and Phe 338, but the D ring of galanthamine was located between Trp 84 (Trp 86 in Electrophorus electricus) and Phe 331 (Phe 338 in Electrophorus electricus). Once compound 1 entered the substrate binding pocket, it made significant hydrophilic and hydrophobic interactions, including (1) the 3-hydroxyl group acted as the H-bond donator to make contact with Ser 203 and also served as the H-bond acceptor to interact with the backbone of Gly122; (2) the A ring of 1 contacted with Trp 86 and Tyr 337 by π–π interaction; and (3) the C ring of 1 interacted with Tyr 124, Phe 338, and Tyr 341 by π–π interaction.
The substrate binding pocket of acetylcholinesterase mainly includes two important sites, esteric and anionic sites. The esteric site contains some significant hydrophilic residues such as Glu 202 (Glu 199 in Torpedo californica), Ser 203 (Ser 200 in Torpedo californica), His 447 (His 440 in Torpedo californica), and Glu 334 (Glu 327 in Torpedo californica), and the potential acetylcholinesterase antagonist should have the structural moiety to make contact with these residues for good inhibition activity. For example, in galanthamine derivatives, whose 9-methoxy group is replaced by a phenoxy or 14-hydroxyl group is replaced by a carbonyl group, their antagonistic effect is largely decreased [31]. For catechin (1), galanthamine, and gallic acid (2), 1 and galanthamine make similar H-bond interactions, but 2 only makes one H-bond interaction with the backbone of the unimportant residue at the esteric site. Based on these results, the antagonistic effect of 2 should be lower than that of 1 or galanthamine. On the contrary, the anionic site of acetylcholinesterase mainly contains hydrophobic residues such as Trp 86 (Trp 84 in Torpedo californica), Tyr 133 (Tyr 130 in Torpedo californica), Tyr 341 (Tyr 334 in Torpedo californica), Phe 338 (Phe 331 in Torpedo californica) and Tyr 449 (Tyr 442 in Torpedo californica), so the potential acetylcholinesterase antagonist should have structural moieties to interact with these residues for good inhibition activity. Additionally, the tertiary amino moiety of the potential acetylcholinesterase antagonist located at the anionic site also plays an important role in the antiacetylcholinesterase activity, although it does not exhibit obvious ionic or hydrophilic interaction in the substrate binding pocket. The QSAR result shows that the galanthamine derivative containing a C(3) = N(4) double bond exhibits a better antiacetylcholinesterase effect than that having a C(3)–N(4) single bond. Since the anionic site of acetylcholinesterase contains key aromatic residues, the tertiary amino moiety might interact with the anionic site by the aromatic hydrophobic interaction rather than the ionic interaction. In particular, Trp 86 (Trp 84 in Torpedo californica) is the key residue at the anionic site, and it is frequently shown to exhibit a π–π interaction with the acetylcholinesterase antagonist. For example, the crystal structure of the acetylcholinesterase complexed with huperzine A (PDB: 1VOT) from Torpedo californica indicates that huperzine A does not make an H-bond interaction with Ser 200 or Glu 199 at the esteric site, and the pyridine moiety of huperzine A makes a strong π–π interaction with Trp 84 [28]. Furthermore, for the binding modes of 1 and galanthamine at the anionic site, the key residue Trp 86 (Trp 84 in Torpedo californica), only contacts with the A ring of 1, but Trp 84 makes three π–π interactions with the C and D rings of galanthamine. According to the evidence mentioned above, it is highly exhibited that the difference in the binding modes of 2, 1, and galanthamine at the anionic site might lead to their distinct antiacetylcholinesterase potency.
In addition, the interaction between euscaphic acid (5) (Figure 4) and acetylcholinesterase was also evaluated. The crystal structure of the acetylcholinesterase from Electrophorus electricus (PDB: 1C2B) was employed in this study. For the binding model of 5 (Figure 5), the A ring of 5 stayed between Tyr 124 and Phe 338, a similar position to the C ring of 1. However, the A ring of 5 resided at a different position from the D ring of galanthamine. The D ring of galanthamine was located between Trp 84 (Trp 86 in Electrophorus electricus) and Phe 331 (Phe 338 in Electrophorus electricus). 5 made two H-bond interactions comprising (1) the 2-hydroxyl group on the A ring of 5 interacts with Phe 295 by performing as the H-bond acceptor, and (2) the 19-hydroxyl group on the E ring of 5 interacts with Trp 286 by performing as the H-bond acceptor. Additionally, the A ring of 5 interacted with Tyr 124, Tyr 341, and Phe 297 by π–alkyl interaction.
The lowest binding energy of each ligand was regarded as the optimal conformation. In this study, chlorogenic acid and galanthamine were employed as positive control. The binding energies of compounds 5, 1, 2, 3, 7, 4, and 6 were −8.7, −8.5, −8.3, −8.1, −8.1, −7.8, and −7.6 kcal/mol, respectively (Table 7). Compared with chlorogenic acid, the binding energies of compounds 13, 5, and 7 were lower than −8.0 kcal/mol. This shows that 13, 5, and 7 can dock into the pocket of the crystal structure of the acetylcholinesterase from Electrophorus electricus more effectively than that of chlorogenic acid.
To further study the interaction between compounds 35 and α-glucosidase. The crystal structure of the isomaltase from Saccharomyces cerevisiae (PDB: 3A4A) was also employed in this study. Compounds 35 showed potent α-glucosidase inhibitory activity. Therefore, the interaction between 35 and α-glucosidase was evaluated by molecular model docking.
In this research, acarbose was employed as a positive control. The binding energies of compounds 3, 4, 5, 6, 1, 7, and 2 were −8.5, −8.0, −7.8, −7.8, −7.6, −7.5, and −5.1 kcal/mol, respectively (Table 8). Compared with the positive control, the binding energies of compounds 1 and 37 were less than −5.3 kcal/mol. This shows that compounds 1 and 37 could dock into the pocket of the crystal structure of isomaltase from Saccharomyces cerevisiae more effectively than that of acarbose.
As shown in Figure 6, compound 3 was bound with Gln 353, Gln 277, and Asp 69 through conventional hydrogen bonds, while other interactions (π–π T-shaped, π–alkyl, and π–anion) were also observed with Phe 303, Tyr 72, Val 216, Asp 352, Glu 277, and Glu 411. These permitted 3 and the enzyme to create a stable complex.
For compound 4 (Figure 7), binding to Glu 277 was via conventional hydrogen bonds, while other interactions (π–π T-shaped, π–alkyl, and π–cation) were also observed with Tyr 158, Arg315, and Arg 442.
Finally, compound 5 was bound with Arg 315 and Leu 313 via conventional hydrogen bonds, while other interactions (alkyl and π–alkyl) were also perceived with Arg 442, Phe 303, and Lys 156 (Figure 8).
As a positive control, acarbose binds to Arg 442, Asp 69, Asp 215, Asp 352, Asn 415, and Gln 353 via conventional hydrogen bonds. Glu 277 forms a carbon–hydrogen bond with acarbose. Furthermore, two hydroxyl groups provide unfavorable donor–donor interactions with Arg 213 and Gln 279 residues (Figure 9).
On the basis of our data, the docking binding energies of compounds 1 and 35 are lower than that of acarbose, suggesting that they have better binding capability. In our research, the active ingredients 1 and 35 not alone exhibited α-glucosidase inhibitory activity but, likewise, had stronger binding potentiality with the active sites of α-glucosidase from Saccharomyces cerevisiae. This suggests that these components can be worthy of further research as natural anti-α-glucosidase agents.

4. Discussion

Different methods have been used to extract natural products for use as alternatives to modern medicines. Organic solvents play a crucial role in natural product chemistry and are applied to obtain extract products, comprising all kinds of metabolites, based on the property and polarity of the component of interest [32]. Variations in solvent polarity result in dramatic dissimilarities in phytochemical compositions and biological activities. Thus, we used solvents of different polarities to obtain and evaluate these various metabolites from A. japonica. In this study, we discovered all kinds of metabolites with different biological activities owing to different solvent polarities.
In this study, among all solvent extracts evaluated by antioxidant assays such as DPPH, ABTS, and FRAP, acetone extract of A. japonica displayed higher antioxidant activity, which may be related to TPC in the extracts. The dissimilarity in antioxidant capacity of various extracts may be due to the different TPC or antioxidant components in each extract. This study is the first report on the comparative evaluation of antioxidant, TFC, and TPC analyses of various solvent extracts from the root of A. japonica. This could give a direction for the choice of a suitable solvent for TPC, TFC, and antioxidant extraction methods.
α-Glucosidase has been recognized as a therapeutic target for regulating postprandial hyperglycemia. Inhibition of intestinal α-glucosidase delays carbohydrate digestion and absorption, thereby suppressing postprandial hyperglycemia [33,34]. In the anti-α-glucosidase assay, compounds 1 and 35 exhibited more potent α-glucosidase inhibitory activities than the positive control acarbose. In this study, it was displayed that 1 was approximately 4-fold stronger than acarbose against α-glucosidase. This study first evaluated the molecular docking study of 5 with Saccharomyces cerevisiae α-glucosidase. In addition, the binding energy of 5 to α-glucosidase from Saccharomyces cerevisiae was calculated for the first time in our study.
The primary role of acetylcholinesterase is to rapidly hydrolyze acetylcholine at cholinergic synapses, terminating the transmission of nerve impulses. The use of acetylcholinesterase inhibitors to enhance cholinergic function in the brain is a major strategy in the treatment of Alzheimer’s disease (AD) [35]. In previous studies, 1 was used as a pleiotropic drug for the treatment of AD through its cholinesterase inhibitory activity and metal chelating activity. Compound 1 was considered a suitable candidate for the development of neuroprotective agents [36]. In our study, 1, 2, and 5 also exhibited anti-acetylcholinesterase activities, which deserves further study. The molecular docking study for 5 with acetylcholinesterase from Electrophorus electricus is first assessed in this study. In addition, the binding energy of euscaphic acid to Electrophorus electricus acetylcholinesterase was calculated for the first time in our study.

5. Conclusions

Different solvent extracts of A. japonica were studied by anti-acetylcholinesterase, anti-α-glucosidase, and antioxidant assays. The contents of TPC in methanol and ethanol extracts were about 5 to 6 times those of TPC in chloroform, dichloromethane and n-hexane extracts, which proved that the suitable relative polarity range of TPC in A. japonica extraction solvents was 0.355 to 0.762. The low-polarity solvent extracts, like chloroform and ethyl acetate, had higher amount of TFC than high-polarity solvent extracts. The TFC in the chloroform extract was almost 20-fold that of the water extract. In our study, acetone, ethanol, and methanol extracts displayed relatively strong ABTS, DPPH, superoxide radical scavenging, and FRAP activities, which may be consistent with TPC in the extracts. The acetone extract had the highest antioxidant and anti-α-glucosidase activities. For the AChE inhibitory activities, the ethanol, methanol and acetone extract of A. japonica showed higher inhibitory effects than other solvent extracts. Biological activity assays showed that compounds 14 and 67 displayed antioxidant activities, and compounds 1 and 37 had strong anti-α-glucosidase effects. Compound 15 and 7 all showed good AChE inhibitory activity. As the result of molecular docking, 1 and 5 at the anionic site might lead to its distinct anti-acetylcholinesterase potency. On the other hand, compounds 35 had better binding capacity to the active site of α-glucosidase from Saccharomyces cerevisiae.
In summary, this study revealed that the extraction solvent of A. japonica affected the extraction yield, antioxidant effect and other biological activities. The acetone, methanol, and ethanol extracts showed relatively high TPC levels and antioxidant activities. The acetone extract contains the most polyphenols, and the chloroform extract contains the most flavonoids. In addition, the ethanol, acetone and methanol extracts showed higher inhibitory effects against α-glucosidase and AChE. According to our antioxidant results, the active antioxidant components of A. japonica were compounds 14 and 67. The above bioactive components could be applied as herbal antioxidants against oxidative damage. It is also worth noting that they also act as natural α-glucosidase and acetylcholinesterase inhibitors.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antiox11071228/s1. Table S1: Retention time, LODs, LOQs, and regression analysis for seven components in Ampelopsis japonica in reverse phase. Figure S1: 1H-NMR spectrum (acetone-d6, 400 MHz) of catechin (1). Figure S2: 1H-NMR spectrum (acetone-d6, 400 MHz) of gallic acid (2). Figure S3: 1H-NMR spectrum (acetone-d6, 400 MHz) of kaempferol (3). Figure S4: 1H-NMR spectrum (acetone-d6, 400 MHz) of quercetin (4). Figure S5: 1H-NMR spectrum (methanol-d4, 500 MHz) of euscaphic acid (5). Figure S6: 1H-NMR spectrum (acetone-d6, 400 MHz) of resveratrol (6). Figure S7: 1H-NMR spectrum (methanol-d4, 400 MHz) of epicatechin (7). Figure S8: Reversed-phase HPLC chromatogram of isolated pure compounds. Figure S9: Reversed-phase HPLC chromatogram of water extract. Figure S10: Reversed-phase HPLC chromatogram of methanol extract. Figure S11: Reversed-phase HPLC chromatogram of ethanol extract. Figure S12: Reversed-phase HPLC chromatogram of acetone extract. Figure S13: Reversed-phase HPLC chromatogram of ethyl acetate extract. Figure S14: Reversed-phase HPLC chromatogram of dichloromethane extract. Figure S15: Reversed-phase HPLC chromatogram of chloroform extract. Figure S16: Reversed-phase HPLC chromatogram of n-hexane extract. Figure S17: Reversed-phase HPLC chromatogram of all solvent extracts with the same scale.

Author Contributions

J.-H.L. performed the isolation, structure elucidation of the constituents, bioassay, and manuscript writing. H.-R.L. conducted the molecular docking. C.-S.Y. analyzed quantification of components and manuscript writing. C.-C.L. carried out NMR experiments. I.-C.W. analyzed bioassay data. J.-J.C. and I.-C.W. planned, designed, and organized all the research of this study and the preparation of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a grant from the Ministry of Science and Technology (MOST), Taiwan (No. MOST 109-2320-B-010-029-MY3), awarded to J.-J. Chen. This work was also supported by the grants from Kaohsiung Veterans General Hospital Tainan Branch (VHYK 110-07 and VHYK 111-02).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the main text and the supplementary materials of this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Meena, D.K.; Sahoo, A.K.; Srivastava, P.P.; Sahu, N.P.; Jadhav, M.; Gandhi, M.; Swain, H.S.; Borah, S.; Das, B.K. On valorization of solvent extracts of Terminalia arjuna (arjuna) upon DNA scission and free radical scavenging improves coupling responses and cognitive functions under in vitro conditions. Sci. Rep. 2021, 11, 10656. [Google Scholar] [CrossRef] [PubMed]
  2. Yew, P.N.; Lee, L.W.L.; Lim, Y.Y. Antioxidant and intracellular reactive oxygen species/reactive nitrogen species scavenging activities of three porcupine bezoars from Hystrix brachyura. Phcog. Res. 2017, 9, 366–372. [Google Scholar] [PubMed]
  3. Ndhlala, A.R.; Moyo, M.; Van Staden, J. Natural antioxidants: Fascinating or mythical biomolecules? Molecules 2010, 15, 6905–6930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Tanvir, E.M.; Hossen, M.S.; Hossain, M.F.; Afroz, R.; Gan, S.H.; Khalil, M.I.; Karim, N. Antioxidant properties of popular turmeric (Curcuma longa) varieties from Bangladesh. J. Food Qual. 2017, 2017, 8471785. [Google Scholar] [CrossRef] [Green Version]
  5. Kaur, C.; Kapoor, H.C. Antioxidants in fruits and vegetables—The millennium’s health. Int. J. Food Sci. Technol. 2001, 36, 703–725. [Google Scholar] [CrossRef]
  6. Li, C.W.; Chu, Y.C.; Huang, C.Y.; Fu, S.L.; Chen, J.J. Evaluation of antioxidant and anti-α-glucosidase activities of various solvent extracts and major bioactive components from the seeds of Myristica fragrans. Molecules 2020, 25, 5198. [Google Scholar] [CrossRef]
  7. Guo, J.M.; Weng, X.C.; Wu, H.; Li, Q.H.; Bi, K.S. Antioxidants from a Chinese medicinal herb—Psoralea corylifolia L. Food Chem. 2005, 91, 287–292. [Google Scholar]
  8. Siebert, D.A.; Campos, J.S.; Alberton, M.D.; Vitali, L.; Micke, G.A. Dual electrophoretically-mediated microanalysis in multiple injection mode for the simultaneous determination of acetylcholinesterase and α-glucosidase activity applied to selected polyphenols. Talanta 2021, 224, 121773. [Google Scholar] [CrossRef]
  9. Zaidi, H.; Ouchemoukh, S.; Amessis-Ouchemoukh, N.; Debbache, N.; Pacheco, R.; Serralheiro, M.L.; Araujo, M.E. Biological properties of phenolic compound extracts in selected Algerian honeys—The inhibition of acetylcholinesterase and α-glucosidase activities. Eur. J. Integr. Med. 2019, 25, 77–84. [Google Scholar] [CrossRef]
  10. Etxeberria, U.; de la Garza, A.L.; Campión, J.; Martínez, J.A.; Milagro, F.I. Antidiabetic effects of natural plant extracts via inhibition of carbohydrate hydrolysis enzymes with emphasis on pancreatic alpha amylase. Expert Opin. Ther. Targets 2012, 16, 269–297. [Google Scholar] [CrossRef] [Green Version]
  11. Van De Laar, F.A. Alpha-glucosidase inhibitors in the early treatment of type 2 diabetes. Vasc. Health Risk Manag. 2008, 4, 1189–1195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Orhan, I.; Kartal, M.; Tosun, F.; Şener, B. Screening of various phenolic acids and flavonoid derivatives for their anticholinesterase potential. Z. Naturforsch. C 2014, 62, 829–832. [Google Scholar] [CrossRef] [PubMed]
  13. Mihaylova, D.; Desseva, I.; Popova, A.; Dincheva, I.; Vrancheva, R.; Lante, A.; Krastanov, A. GC-MS metabolic profile and α-glucosidase-, α-amylase-, lipase-, and acetylcholinesterase-inhibitory activities of eight peach varieties. Molecules 2021, 26, 4183. [Google Scholar] [CrossRef] [PubMed]
  14. Abbas-Mohammadi, M.; Farimani, M.M.; Salehi, P.; Ebrahimi, S.N.; Sonboli, A.; Kelso, C.; Skropeta, D. Acetylcholinesterase-inhibitory activity of Iranian plants: Combined HPLC/bioassay-guided fractionation, molecular networking and docking strategies for the dereplication of active compounds. J. Pharm. Biomed. Anal. 2018, 158, 471–479. [Google Scholar] [CrossRef] [PubMed]
  15. Son, M.; Park, C.; Rampogu, S.; Zeb, A.; Lee, K.W. Discovery of novel acetylcholinesterase inhibitors as potential candidates for the treatment of Alzheimer’s disease. Int. J. Mol. Sci. 2019, 20, 1000. [Google Scholar] [CrossRef] [Green Version]
  16. Lee, C.L.; Jhan, Y.L.; Chiang, H.M.; Chen, Y.H.; Chen, C.J.; Chang, Y.S. Characterization of chemical constituents with their antioxidant and anti-melanogenesis activities from the roots of Ampelopsis japonica. Nat. Prod. Res. 2021, 36, 1–5. [Google Scholar] [CrossRef]
  17. Chu, Y.C.; Yang, C.S.; Cheng, M.J.; Fu, S.L.; Chen, J.J. Comparison of various solvent extracts and major bioactive components from unsalt-fried and salt-fried rhizomes of Anemarrhena asphodeloides for antioxidant, anti-α-glucosidase, and anti-acetylcholinesterase activities. Antioxidants 2022, 11, 385. [Google Scholar] [CrossRef]
  18. Lin, Y.T.; Lin, H.R.; Yang, C.S.; Liaw, C.C.; Sung, P.J.; Kuo, Y.H.; Cheng, M.J.; Chen, J.J. Antioxidant and Anti-α-glucosidase activities of various solvent extracts and major bioactive components from the fruits of Crataegus pinnatifida. Antioxidants 2022, 11, 320. [Google Scholar] [CrossRef]
  19. Koivikko, R.; Loponen, J.; Honkanen, T.; Jormalainen, V. Contents of soluble, cell-wall-bound and exuded phlorotannins in the brown alga Fucus vesiculosus, with implications on their ecological functions. J. Chem. Ecol. 2005, 31, 195–212. [Google Scholar] [CrossRef] [Green Version]
  20. Do, Q.D.; Angkawijaya, A.E.; Tran-Nguyen, P.L.; Huynh, L.H.; Soetaredjo, F.E.; Ismadji, S.; Ju, Y.H. Effect of extraction solvent on total phenol content, total flavonoid content, and antioxidant activity of Limnophila aromatica. J. Food Drug Anal. 2014, 22, 296–302. [Google Scholar] [CrossRef] [Green Version]
  21. Takao, T.; Kitatani, F.; Watanabe, N.; Yagi, A.; Sakata, K. A simple screening method for antioxidants and isolation of several antioxidants produced by marine bacteria from fish and shellfish. Biosci. Biotechnol. Biochem. 1994, 58, 1780–1783. [Google Scholar] [CrossRef] [Green Version]
  22. Re, R.; Pellegrini, N.; Proteggente, A.; Pannala, A.; Yang, M.; Rice-Evans, C. Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free. Radic. Biol. Med. 1999, 26, 1231–1237. [Google Scholar] [CrossRef]
  23. Zhang, Q.F.; Zhang, Z.R.; Cheung, H.Y. Antioxidant activity of Rhizoma Smilacis Glabrae extracts and its key constituent-astilbin. Food Chem. 2009, 115, 297–303. [Google Scholar] [CrossRef]
  24. Alvarez-Jubete, L.; Wijngaard, H.; Arendt, E.K.; Gallagher, E. Polyphenol composition and in vitro antioxidant activity of amaranth, quinoa buckwheat and wheat as affected by sprouting and baking. Food Chem. 2010, 119, 770–778. [Google Scholar] [CrossRef]
  25. Pujirahayu, N.; Bhattacharjya, D.K.; Suzuki, T.; Katayama, T. α-Glucosidase inhibitory activity of cycloartane-type triterpenes isolated from Indonesian stingless bee Propolis and their structure–activity relationship. Pharmaceuticals 2019, 12, 102. [Google Scholar] [CrossRef] [Green Version]
  26. Ellman, G.L.; Courtney, K.D.; Andres, V.; Featherstone, R.M. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  27. Omena, C.M.B.; Valentim, I.B.; Guedes, G.D.; Rabelo, L.A.; Mano, C.M.; Bechara, E.J.H.; Sawaya, A.C.H.F.; Trevisan, M.T.S.; da Costa, J.G.; Ferreira, R.C.S.; et al. Antioxidant, anti-acetylcholinesterase and cytotoxic activities of ethanol extracts of peel, pulp and seeds of exotic Brazilian fruits. Food Res. Int. 2012, 49, 334–344. [Google Scholar] [CrossRef] [Green Version]
  28. Raves, M.L.; Harel, M.; Pang, Y.P.; Silman, I.; Kozikowski, A.P.; Sussman, J.L. Structure of acetylcholinesterase complexed with the nootropic alkaloid, (−)-Huperzine A. Nat. Struct. Biol. 1997, 4, 57–63. [Google Scholar] [CrossRef]
  29. Greenblatt, H.M.; Guillou, C.; Guénard, D.; Argaman, A.; Botti, S.; Badet, B.; Thal, C.; Silman, I.; Sussman, J.L. The complex of a bivalent derivative of galanthamine with Torpedo acetylcholinesterase displays drastic deformation of the active-site gorge: Implications for structure-based drug design. J. Am. Chem. Soc. 2004, 126, 15405–15411. [Google Scholar] [CrossRef]
  30. Bourne, Y.; Grassi, J.; Bougis, P.E.; Marchot, P. Conformational flexibility of the acetylcholinesterase tetramer suggested by X-ray crystallography. J. Biol. Chem. 1999, 274, 30370–30376. [Google Scholar] [CrossRef] [Green Version]
  31. Bartolucci, C.; Perola, E.; Pilger, C.; Fels, G.; Lamba, D. Three-dimensional structure of a complex of galanthamine (Nivalin®) with acetylcholinesterase from Torpedo californica: Implications for the design of new anti-Alzheimer drugs. Proteins 2001, 42, 182–191. [Google Scholar] [CrossRef]
  32. Yamamoto, K.; Miyake, H.; Kusunoki, M.; Osaki, S. Crystal structures of isomaltase from Saccharomyces cerevisiae and in complex with its competitive inhibitor maltose. FEBS J. 2010, 277, 4205–4214. [Google Scholar] [CrossRef] [PubMed]
  33. Yao, Y.; Cheng, X.; Wang, L.; Wang, S.; Ren, G. A determination of potential α-glucosidase inhibitors from azuki beans (Vigna angularis). Int. J. Mol. Sci. 2011, 12, 6445–6451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Liu, Y.; Ma, L.; Chen, W.H.; Park, H.; Ke, Z.; Wang, B. Binding mechanism and synergetic effects of xanthone derivatives as noncompetitive α-glucosidase inhibitors: A theoretical and experimental study. J. Phys. Chem. B 2013, 117, 13464–13471. [Google Scholar] [CrossRef]
  35. Gholamhoseinian, A.; Moradi, M.N.; Sharifi-Far, F. Screening the methanol extracts of some Iranian plants for acetylcholinesterase inhibitory activity. Res. Pharm. Sci. 2009, 4, 105–112. [Google Scholar]
  36. Suganthy, N.; Pandima Devi, K. In vitro antioxidant and anti-cholinesterase activities of Rhizophora mucronata. Pharm. Biol. 2016, 54, 118–129. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The chemical structures of catechin (1), gallic acid (2), kaempferol (3), quercetin (4), euscaphic acid (5), resveratrol (6), and epicatechin (7) from Ampelopsis japonica.
Figure 1. The chemical structures of catechin (1), gallic acid (2), kaempferol (3), quercetin (4), euscaphic acid (5), resveratrol (6), and epicatechin (7) from Ampelopsis japonica.
Antioxidants 11 01228 g001
Figure 2. The chemical structures of galanthamine (a) and catechin (1) (b).
Figure 2. The chemical structures of galanthamine (a) and catechin (1) (b).
Antioxidants 11 01228 g002
Figure 3. Interaction of catechin (1) with the active sites of acetylcholinesterase from Electrophorus electricus. The binding mode (a) and the hydrophilic interactive model (b) of catechin (1) in the substrate binding pocket of the crystal structure (PDB: 1C2B).
Figure 3. Interaction of catechin (1) with the active sites of acetylcholinesterase from Electrophorus electricus. The binding mode (a) and the hydrophilic interactive model (b) of catechin (1) in the substrate binding pocket of the crystal structure (PDB: 1C2B).
Antioxidants 11 01228 g003
Figure 4. The chemical structure of euscaphic acid (5).
Figure 4. The chemical structure of euscaphic acid (5).
Antioxidants 11 01228 g004
Figure 5. Interactions of euscaphic acid (5) with active sites of acetylcholinesterase from Electrophorus electricus.
Figure 5. Interactions of euscaphic acid (5) with active sites of acetylcholinesterase from Electrophorus electricus.
Antioxidants 11 01228 g005
Figure 6. Interactions of kaempferol (3) with active sites of S. cerevisiae α-glucosidase.
Figure 6. Interactions of kaempferol (3) with active sites of S. cerevisiae α-glucosidase.
Antioxidants 11 01228 g006
Figure 7. Interactions of quercetin (4) with active sites of S. cerevisiae α-glucosidase.
Figure 7. Interactions of quercetin (4) with active sites of S. cerevisiae α-glucosidase.
Antioxidants 11 01228 g007
Figure 8. Interactions of euscaphic acid (5) with active sites of S. cerevisiae α-glucosidase.
Figure 8. Interactions of euscaphic acid (5) with active sites of S. cerevisiae α-glucosidase.
Antioxidants 11 01228 g008
Figure 9. Interactions of acarbose with active sites of S. cerevisiae α-glucosidase.
Figure 9. Interactions of acarbose with active sites of S. cerevisiae α-glucosidase.
Antioxidants 11 01228 g009
Table 1. Each extraction solvent from Ampelopsis japonica of TPC, TFC, and extraction yields.
Table 1. Each extraction solvent from Ampelopsis japonica of TPC, TFC, and extraction yields.
Extracting SolventsRelative PolarityTPC (mg/g) a
(GAE)
TFC (mg/g) b
(QE)
Yields (%) c
n-Hexane0.00915.01 ± 0.44 ***56.36 ± 3.86 **0.28 ± 0.08
Chloroform0.25920.92 ± 1.47 **94.22 ± 1.34 ***0.50 ± 0.08
Dichloromethane0.26920.19 ± 1.99 **62.60 ± 3.51 ***0.45 ± 0.04
Ethyl acetate0.22879.09 ± 8.45 **80.05 ± 7.82 **0.66 ± 0.10
Acetone0.355142.89 ± 4.07 ***71.72 ± 2.66 ***1.03 ± 0.27
Methanol0.76295.69 ± 7.80 **5.72 ± 1.13 **1.90 ± 0.50
Ethanol0.65495.98 ± 6.68 **22.75 ± 1.24 ***4.87 ± 0.03
Water1.00047.38 ± 2.18 ***4.45 ± 1.21 *16.46 ± 0.31
a TPC was shown as milligrams of gallic acid equivalents (GAE) per gram of extract. b TFC was shown as milligrams of quercetin equivalents (QE) per gram of extract; c Yield was counted as % yield = (weight of extract/initial weight of dry Ampelopsis japonica) × 100; * p < 0.05; ** p < 0.01; *** p < 0.001 compared with the control.
Table 2. Antioxidant effects of various solvent extracts from Ampelopsis japonica determined by DPPH, ABTS, superoxide radical scavenging, and FRAP assays.
Table 2. Antioxidant effects of various solvent extracts from Ampelopsis japonica determined by DPPH, ABTS, superoxide radical scavenging, and FRAP assays.
Extracting SolventsSC50 (μg/mL) aTE (mM/g) d
DPPHABTSSuperoxideFRAP
n-Hexane>200>200>40026.03 ± 1.90 **
Chloroform>200>200>40094.07 ± 7.35 ***
Dichloromethane>200>200>400104.20 ± 9.18 ***
Ethyl acetate92.14 ± 8.12 *57.45 ± 4.74 *>400587.11 ± 20.61 ***
Acetone54.88 ± 4.40 *33.88 ± 2.31 **>4001001.00 ± 46.17 ***
Methanol84.73 ± 7.82 *53.77 ± 4.65 **290.83 ± 15.23 *712.56 ± 18.32 ***
Ethanol87.12 ± 6.45 * 64.56 ± 4.80 **307.20 ± 22.39 *736.95 ± 14.40 ***
Water98.54 ± 7.09 *99.30 ± 7.02 *313.84 ± 20.24 *413.34 ± 21.08 ***
BHT b33.04 ± 2.12 **14.09 ± 0.24 **N.A. c4257.97 ± 145.90 ***
a The SC50 value was defined as the concentration of the samples causing 50% free radical scavenging, and was displayed as mean ± SD (n = 3); b BHT was employed as positive control; c N.A. means unavailable (poor solubility); d FRAP was expressed in millimolar of Trolox equivalents (TE) per gram of extract; * p < 0.05, ** p < 0.01, and *** p < 0.001 compared with the control.
Table 3. Different solvent extracts from Ampelopsis japonica of α-glucosidase and AChE inhibitory activities.
Table 3. Different solvent extracts from Ampelopsis japonica of α-glucosidase and AChE inhibitory activities.
Extracting SolventsIC50 (μg/mL) a
α-GlucosidaseAChE
n-Hexane28.43 ± 3.78 *83.97 ± 8.90 *
Chloroform34.16 ± 3.88 *91.64 ± 8.77 *
Dichloromethane28.00 ± 0.14 *91.47 ± 26.03 *
Ethyl acetate12.51 ± 2.42 *103.30 ± 2.15 *
Acetone8.30 ± 0.78 **61.95 ± 5.54 **
Methanol19.27 ± 1.12 *77.99 ± 5.08 *
Ethanol11.06 ± 2.07 *37.08 ± 7.67 *
Water>40085.82 ± 8.74 *
Acarbose b335.50 ± 2.14 *
Chlorogenic acid b66.69 ± 0.16 *
a The IC50 value was defined as half-maximal inhibitory concentration of each free radical scavenging activity; BHT was employed as positive control; b Acarbose and chlorogenic acid were employed as positive controls; * p < 0.05 and ** p < 0.01 compared with the control.
Table 4. Quantification and identification of the main active compounds from Ampelopsis japonica in various extracts.
Table 4. Quantification and identification of the main active compounds from Ampelopsis japonica in various extracts.
Extracting Solventsmg/kg
1234567Total Amount
Water6.27 ± 0.588.43 ± 0.734.64 ± 0.2222.30 ± 1.620.68 ± 0.03N.D.N.D.42.32 ± 3.18
Methanol10.27 ± 1.0812.73 ± 1.427.24 ± 0.649.30 ± 0.911.26 ± 0.083.01 ± 0.280.67 ± 0.0344.48 ± 4.44
Ethanol7.85 ± 0.5216.43 ± 1.642.94 ± 0.033.71 ± 0.280.86 ± 0.0711.22 ± 1.020.58 ± 0.0443.59 ± 3.60
Acetone8.43 ± 0.6312.43 ± 1.723.21 ± 0.066.64 ± 0.741.21 ± 0.0212.42 ± 1.443.41 ± 0.1847.75 ± 4.79
Ethyl acetate3.79 ± 0.168.86 ± 0.631.54 ± 0.083.62 ± 0.211.01 ± 0.082.60 ± 0.110.52 ± 0.0921.94 ± 1.36
Chloroform4.12 ± 0.342.63 ± 0.087.83 ± 0.554.02 ± 0.430.91 ± 0.060.70 ± 0.080.84 ± 0.0721.05 ± 1.61
Dichloromethane2.74 ± 0.161.63 ± 0.064.62 ± 0.432.66 ± 0.120.84 ± 0.081.83 ± 0.060.71 ± 0.0715.03 ± 0.98
n-Hexane1.43 ± 0.083.46 ± 0.261.12 ± 0.072.63 ± 0.220.69 ± 0.076.23 ± 0.52N.D.15.56 ± 1.22
Results are expressed as milligrams of each compound in kilograms of extract. N.D. means no detectable.
Table 5. Antioxidant effects of isolated compounds from Ampelopsis japonica measured with DPPH, ABTS, superoxide radical scavenging, and FRAP assays.
Table 5. Antioxidant effects of isolated compounds from Ampelopsis japonica measured with DPPH, ABTS, superoxide radical scavenging, and FRAP assays.
CompoundsSC50 (μg/mL) aTE (mM/g)
DPPHABTSSuperoxideFRAP
110.08 ± 3.09 **2.23 ± 0.22 **64.43 ± 7.73 *8729.33 ± 424.55 ***
22.60 ± 0.67 *1.45 ± 0.14 **47.40 ± 3.01 *28,512.82 ± 43.27 ***
312.48 ± 3.01 **5.24 ± 0.45 *N.A. −b7912.47 ± 220.08 ***
43.36 ± 0.58 **3.15 ± 0.49 *31.89 ± 2.03 **16,038.26 ± 86.89 ***
5>400>400>4008.67 ± 3.93 *
613.19 ± 4.78 *2.81 ± 0.12 **66.16 ± 5.23 *7453.94 ± 60.09 ***
72.78 ± 0.25 *3.78 ± 0.03 **41.76 ± 4.20 *13,122.77 ± 182.42 ***
BHT b36.99 ± 4.54 *17.36 ± 3.14 *N.A. c3997.23 ± 144.35 ***
a The SC50 value was defined as the concentration of the samples causing 50% free radical scavenging, and was displayed as mean ± SD (n = 3); b BHT was employed as positive control; c N.A. means unavailable (poor solubility); * p < 0.05, ** p < 0.01, and *** p < 0.001 compared with the control.
Table 6. Main isolated components from Ampelopsis japonica of α-glucosidase and AChE inhibitory activities.
Table 6. Main isolated components from Ampelopsis japonica of α-glucosidase and AChE inhibitory activities.
CompoundsIC50 (μg/mL)
α-GlucosidaseAChE
181.78 ± 11.58 **26.35 ± 9.55 **
2>40041.59 ± 7.57 *
35.81 ± 2.70 **55.04 ± 8.57 **
414.39 ± 5.93 **66.34 ± 5.09 **
520.38 ± 2.13 *11.64 ± 2.69 **
628.81 ± 5.65 *80.75 ± 9.21 **
788.73 ± 10.94 *53.38 ± 7.30 *
Acarbose a334.53 ± 2.22 *
Chlorogenic acid a64.42 ± 0.16 *
Galanthamine hydrobromide a0.57 ± 0.09 *
a Acarbose, chlorogenic acid and galanthamine hydrobromide were employed as positive controls; * p < 0.05 and ** p < 0.01 compared with the control.
Table 7. Binding energies of active components with acetylcholinesterase from Electrophorus electricus calculated in silico.
Table 7. Binding energies of active components with acetylcholinesterase from Electrophorus electricus calculated in silico.
CompoundsAffinity (kcal/mol)
1−8.5
2−8.3
3−8.1
4−7.8
5−8.7
6−7.6
7−8.1
Chlorogenic acid a−8.0
Galanthamine a−9.4
a Chlorogenic acid and galanthamine employed as positive controls.
Table 8. Binding energies of active components and acarbose with α-glucosidase from Saccharomyces cerevisiae calculated in silico.
Table 8. Binding energies of active components and acarbose with α-glucosidase from Saccharomyces cerevisiae calculated in silico.
CompoundsAffinity (kcal/mol)
1−7.6
2−5.1
3−8.5
4−8.0
5−7.8
6−7.8
7−7.5
Acarbose a−5.3
a Acarbose employed as a positive control.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liang, J.-H.; Lin, H.-R.; Yang, C.-S.; Liaw, C.-C.; Wang, I.-C.; Chen, J.-J. Bioactive Components from Ampelopsis japonica with Antioxidant, Anti-α-Glucosidase, and Antiacetylcholinesterase Activities. Antioxidants 2022, 11, 1228. https://doi.org/10.3390/antiox11071228

AMA Style

Liang J-H, Lin H-R, Yang C-S, Liaw C-C, Wang I-C, Chen J-J. Bioactive Components from Ampelopsis japonica with Antioxidant, Anti-α-Glucosidase, and Antiacetylcholinesterase Activities. Antioxidants. 2022; 11(7):1228. https://doi.org/10.3390/antiox11071228

Chicago/Turabian Style

Liang, Jia-Hua, Hsiang-Ru Lin, Chang-Syun Yang, Chia-Ching Liaw, I-Chou Wang, and Jih-Jung Chen. 2022. "Bioactive Components from Ampelopsis japonica with Antioxidant, Anti-α-Glucosidase, and Antiacetylcholinesterase Activities" Antioxidants 11, no. 7: 1228. https://doi.org/10.3390/antiox11071228

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