Next Article in Journal
Modification of Polyhedral Oligomeric Silsesquioxanes (POSS) Molecules by Ruthenium Catalyzed Cross Metathesis
Previous Article in Journal
Effects of a Ruthenium Schiff Base Complex on Glucose Homeostasis in Diet-Induced Pre-Diabetic Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Isolation and Identification of the Anti-Oxidant Constituents from Loropetalum chinense (R. Brown) Oliv. Based on UHPLC–Q-TOF-MS/MS

Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2018, 23(7), 1720; https://doi.org/10.3390/molecules23071720
Submission received: 7 June 2018 / Revised: 3 July 2018 / Accepted: 11 July 2018 / Published: 14 July 2018

Abstract

:
The aim of this study was to identify the chemical constituents of Loropetalum chinense (R. Brown) Oliv. (LCO) and determine which of these had antioxidant effects. The chemical composition of a 70% ethanol extract of LCO was analyzed systematically using UHPLC–Q-TOF-MS/MS. The chemical components of the 70% ethanol extract of LCO were then separated and purified using macroporous resin and chromatographic techniques. Antioxidant activity was evaluated using a DPPH assay. In total, 100 compounds were identified tentatively, including 42 gallic acid tannins, 49 flavones, and 9 phenolic compounds. Of these, 7 gallium gallate, 4 flavonoid and 8 quinic acid compounds were separated and purified from the 70% ethanol extract of LCO. The compounds identified for the first time in LCO and in the genus Loropetalum were 3,4,5-trimethoxyphenyl-(6′-O-galloyl)-O-β-d-glucopyranoside, protocatechuic acid, ethyl gallate, 5-O-caffeoylquinic acid, 3-O-caffeoylquinic acid, 3,5-O-diocaffeoylquinic acid, 4,5-O-diocaffeoylquinic acid and 3,4-O-diocaffeoylquinic acid. The 50% inhibitory concentration (IC50) values of compounds 1,2,3,4,6-penta-O-galloyl-β-d-glucose, gallic acid, protocatechuic acid, and ethyl gallate were 1.88, 1.05, 1.18, and 1.05 μg/mL, respectively. Compared with the control group (VC) (2.08 μg/mL), these compounds exhibited stronger anti-oxidation activity. This study offered considerable insight into the chemical composition of LCO, with preliminary identification of the antioxidant ingredients.

Graphical Abstract

1. Introduction

Reactive oxygen species (ROS) refers to oxygen-containing reactive species and includes superoxide anions (O2), hydrogen peroxide (H2O2), and hydroxyl radicals (•OH) [1]. Under normal conditions, ROS are in a constant dynamic state of production and elimination in vivo. They play an important role in physiological metabolic processes such as enhancing leukocyte phagocytosis and prostaglandin synthesis, and participating in enzymatic pathways that contribute to immunity [2]. A net excess of ROS can follow an imbalance in ROS production and elimination. This can lead to a series of peroxidation reactions, cross-linking or breakages with subsequent cellular structural damage and dysfunction. If this occurs chronically, a number of pathophysiological processes may ensue including arteriosclerosis, cardiovascular diseases, neurodegenerative diseases, cancers, and other disorders associated with aging [3,4,5,6]. Therefore, elimination of excess ROS in the body is important for maintaining physiological health. Recently, there has been an increase in interest in antioxidants as substances that protect against oxidative damage. A particular focus has been secondary plant polyphenol or phenolic metabolites such as catechins, epigallocatechin gallate, and catechin-aldehyde polycondensates [7,8,9]. These phenolic compounds also possess other bioactivities including antimicrobial and anti-inflammatory effects [10,11]. Therefore, screening for antioxidants could be an effective strategy for sourcing new drugs or functional foods.
Loropetalum, as a member of the Hamamelidaceae family, contains three species (Loropetalum lanceum Hand-Mzt, Loropetalum subcapitatum Chun ex Chang, and Loropetalum chinense (R. Brown) Oliv.) as well as one variety (Loropetalum chinense var. rubrum) in China. Among them, Loropetalum chinense (R. Brown) Oliv. (LCO), an evergreen shrub or small arbor, was first recorded officially in the 1970 edition of the Chinese pharmacopoeia. LCO has antipyretic, hemostatic, and detoxificant effects, and is a traditional medicine widely used in the treatment of bleeding disorders, burns, skin infections, dysentery, and diarrhea [12]. Modern pharmacological research has shown that LCO has bacteriostatic, anti-inflammatory, healing, and antioxidant effects [13].
The multiple biological activities of LCO are attributed to its diverse constituents. Phytochemical studies reported the presence of tannins, flavonoids, lignans, terpenoids, and steroids in LCO [14,15,16,17]. Alkanes, aldehydes, and terpenoids have been identified as the dominant constituents of LCO essential oil [13]. However, there is little information on the biological constituents that have antioxidant effects. In earlier work, we used DPPH (2,2-diphenyl-1-picrylhydrazyl) radical scavenging activity to evaluate the antioxidant effects of four fractions (water, and 10%, 70%, and 95% ethanol eluates) separated with HPD-400 macroporous resin. Our results showed that the four fractions exhibited various antioxidant effects. The 50% inhibitory concentrations (IC50) were 22.53 μg/mL for the water eluate, 11.47 μg/mL for 10% ethanol eluate, 9.73 μg/mL for 70% ethanol eluate and 39.20 μg/mL for 95% ethanol eluate [18]. However, this work only assessed the antioxidant effects of various LCO fractions. Details of the specific constituents responsible for the antioxidant activities remain unclear.
Therefore, in the present study, we used UHPLC-Q-TOF-MS/MS to systemically analyze the chemical constituents of the ethanol extract of LCO to identify the components responsible for its antioxidant effects.

2. Materials and Methods

2.1. Chemicals and Reagents

DPPH and vitamin C (VC) were purchased from Sigma Chemicals (Shanghai, China). Chemical standards of gallic acid, protocatechuic acid, quercetin, kaempferol, and chlorogenic acid were obtained from Chengdu Munster Biotechnology Co., Ltd. (Chengdu, China). LC-MS grade methanol and HPLC grade methanol for use as solvents, and acetic acid, formic acid, and acetonitrile were purchased from Fisher Scientific (Shanghai, China). HPD-400 macroporous resin was purchased from Cangzhou Bon Adsorber Technology Co., Ltd (Jiangsu, China). Various types of silica gel were obtained from Qingdao Haiyang Chemical Co., Ltd (Shandong, China). Double distilled water was used in the LC mobile phase. All other chemicals used were analytical grade.

2.2. Plant Material and Extraction

LCO was collected in May around the city of Jindezhen (29°25′ N, 117°16′ E) (Jiangxi province, China). Plants were authenticated by Prof. Shi-lin Yang, Jiangxi University of Traditional Chinese Medicine. A voucher specimen (No. 20100521) was stored in the National Pharmaceutical Engineering Center for Solid Preparation in Chinese Materia Medica, Jiangxi University of Traditional Chinese Medicine.
Extraction parameters were as described in earlier work by our group [19]. Dried branches and leaves of LCO (17 kg) were milled and reflux-extracted twice with an 8-fold weight of 60% ethanol solution for 2 h. Extracts were then combined and filtered with vacuum suction filtration. The combined filtrates were concentrated to dryness under vacuum with a rotary evaporator to yield a yellowish-brown residue (2.754 kg) that was stored at 4 °C until needed. The residue was re-dissolved with methanol to give a final concentration of 10 mg/mL and then filtered through a 0.22 μm filter membrane before UHPLC analysis.

2.3. DPPH Assay

DPPH, control, blank and sample solutions were prepared as follows. DPPH solution: 3 mg DPPH was dissolved in absolute ethanol and mixed with 0.03 g/L reaction solution protected from light. Control: 1 mL solvent and 3 mL DPPH solution were combined in a test tube, shaken to mix and left to react for 30 min in the dark at room temperature (25 °C). Absorbance (A0) was then measured at a wavelength of 517 nm with a UV visible spectrophotometer (UV-2550, Shimadzu, Kyoto, Japan). Blank: Various concentrations of sample solution (1 mL) were added to 3 mL absolute ethanol in a test tube, shaken to mix, and then stored in the dark at 25 °C. After 30 min, absorbance (Aj) was measured at 517 nm wavelength. Sample: Various concentrations of sample solution (1 mL) were mixed with 3 mL DPPH solution in a test tube, stored in the dark at 25 °C for 30 min before measuring the absorbance (Ai) at a wavelength of 517 nm [20,21,22]. The scavenging rate was calculated as follows: scavenging activity (%) = [1 − (Ai − Aj)/A0] × 100.

2.4. UHPLC–Q-TOF-MS/MS Analysis of the Crude Extract

Chromatographic analysis was carried out on a Prominence™ UHPLC system (Shimadzu, Japan) coupled with a triple TOF™ 5600+ MS/MS system (AB Sciex, Framingham, MA, USA). Separations were accomplished on a Welch C18 column (2.1 mm × 100 mm, 1.8 μm, Shanghai, China) and 2 μL injected into UHPLC. The column oven was maintained at 40 °C. A linear gradient elution of eluents A (water containing 0.1% formic acid) and B (acetonitrile) was used for separation. The elution program was: 0.1–2 min, 1–5% B; 2.01–20 min, 8–37% B; 20–22 min, 37–50% B; 22–32 min, 50–90% B; 32.01–35 min, 95–95% B, 35.01–40 min, 1% B. The flow rate was 0.25 mL/min.
The mass spectrometer was operated both in negative and positive ion modes. The following parameter settings were used: ion spray voltage 4500 V; turbo spray temperature 600 °C; curtain gas 25 psi, nebulizer gas (GS 1) 50 psi, heater gas (GS 2) 50 psi, declustering potential 100 V. TOF MS and TOF MS/MS were scanned with the mass range of m/z 50–1250 and 50–1250, respectively. In the IDA-MS/MS experiment, collision energy was set at 35 eV and collision energy spread was (±) 10 eV. Accurate mass and composition for the precursor and fragment ions were analyzed using Peakview software (Version 1.2, AB Sciex) integrated with the instrument.

2.5. Isolation of the Crude Extract

The crude extract solution was adsorbed by the HPD-400 macroporous resin and eluted with water, and 10%, 70%, and 95% ethanol-water. The 10% and 70% fractions were concentrated under vacuum to recover the organic solvent to dryness. They were then isolated using various chromatographic techniques including silica gel, Sephadex LH-20, ODS, and MCI columns, and re-crystallization methods.
The compounds were identified using UV, MS, 1H-NMR, and 13C-NMR experiments along with comparison of their spectroscopic properties from the literature. Their antioxidant activities were measured by DPPH assay.

3. Results

3.1. UHPLC–Q-TOF-MS/MS Analysis of the Crude Extract

To obtain the abundant constituents from LCO, UHPLC conditions (type of column, mobile phase system, column temperature, and flow rate) were first optimized followed by the MS conditions (capillary voltage, declustering potential, and collision energy). UHPLC-Q-TOF-MS/MS in positive (A) and negative (B) ion modes was employed to characterize the corresponding signals. The base peak chromatogram under optimized chromatographic and MS conditions are presented in Figure 1. Retention times, observed molecular weight, and fragment ions for each metabolite, and their identities are presented in Table 1. A total of 100 compounds were identified or tentatively characterized including 42 gallic acid tannins, 49 flavones, and 9 phenolic compounds. The structures of these compounds were tentatively assigned by matching the MS/MS data with a reference or public database such as PubChem (https://pubchem.ncbi.nlm.nih.gov/) or MassBank (http://www.massbank.jp/).

3.1.1. Fragmentation of Gallic Acid Tannins

The 42 tannin compounds have gallic acid and glucose as their basic structural units, which contain or lose C7H6O5 (m/z: 170), -C7H5O5 (m/z: 169), -C7H5O4 (m/z: 153), -C6H5O3 (m/z: 125) and -C6H11O5 (m/z: 163) ion fragments in the MS/MS spectrum [23,24,25,26]. For example, in the positive ion of the MS/MS spectrum, the [M + H]+ ions of peak 5 were found at m/z: 171.0274, while m/z: 153.0180 (M + H-H2O), 135.0074 (M + H-2H2O), 125.0234 (M + H-HCHO2) and 107.0128 (M + H-HCHO2-H2O) fragment ions were consistent with gallic acid (C7H6O5). Therefore, peak 5 was determined to be gallic acid (Figure 2A1). Peak 54 had [M + H]+ ions at m/z: 303.0140, and m/z: 285.0035 (-C14H5O7), 275.0189 (-C13H7O7), 257.0082 (-C13H7O6) and 229.0130 (-C12H5O5) fragment ions were observed in the MS/MS spectrum, which were formed by loss of H2O, CO2, and CO. Following comparison with the reference substance, peak 54 was determined to be ellagic acid. Peak 4 had [M + H]+ ions at m/z 345.0818 in the MS/MS spectrum (Figure 2A2). Three fragment ion at m/z: 171.0289, 153.0185, and 125.0232 showed that peak 4 contained gallic acid units. Based on these MS/MS data, peak 4 was identified as theogallin (C14H16O10). Under the negative ion mode, peak 11 had [M − H] ions at m/z 331.0667, and fragment ions at m/z 169.0148, 151.0031, and 125.0257 indicating the presence of gallic acid units. [M − H] ions produce m/z 169.0148 by losing 162 Da (-C6H10O5), which indicated the presence of hexose units and thus peak 11 peak was determined to be monogalloylglucose.

3.1.2. Fragmentation of Flavonoids Compounds

The Retro-Diels-Alder (RDA) cleavage reaction involves loss and rearrangement of flavonoid aglycone C rings in different ways. The main fragment was derived from cleavage of C-C and C-O bonds on the C ring, and neutral fragments such as CO, CO2, H2O, and C2H2O. Hexose (m/z: 162) and pentose (m/z: 132) fragments often appear in the MS/MS cracking spectrum of flavonoid glycosides [27,28,29]. In this study, flavonoid glycosides produced gallic acid (m/z: 169) fragments. For example, peak 71 was identified as quercetin (C15H10O7) through comparison with the reference substance. [M + H]+ ions at m/z: 303.0305, and RDA cleavage of C1-C2 and C3-C4 bonds in the C ring generated the fragment m/z: 153.0181. After breakage of the C2-O and C10-O bonds of the C ring, the B ring produced m/z: 137.0229 by losing two CO. [M + H]+ ion lost one H2O (18 Da) to form m/z: 285.0037, and one CO (28 Da) to form m/z: 275.0176 (-C14H10O6). Subsequently, m/z: 275.0176 (-C14H10O6) was produced m/z: 257.0436 (C14H8O5) and m/z: 229.0493 (-C13H8O4) by losing -H2O and -CO. As peaks 57, 59, 62, and 85 had the same fragments, they were presumed to be quercetin isomers.
The [M − H] ion of peak 66 at m/z: 599.1064 was in accordance with the molecular formula C28H24O15 in the negative ion mode. m/z: 599.1064 lost 152 Da to produce ion at m/z: 447.0969. The loss of 152 Da from m/z: 599.1064 and the fragment m/z: 169.0147 showed the presence of gallic acid units. Fragment m/z: 447.0969 lost 162 Da to form m/z: 285.0414, and fragment m/z: 313.0582 in MS/MS spectrum showed the presence of hexose units. Fragments m/z: 285.0414, 151.0036, and 125.0245 were consistent with quercetin cleavage. Therefore, peak 66 was identified as astragalin-O-gallate (Figure 2B1). As peak 73 had the same fragments, it was presumed to be an isomer of peak 66.
The [M − H] ion at m/z: 593.1341 of peak 89 was found in the negative ion MS/MS spectrum. Fragments m/z: 285.0399 and 151.0038 were consistent with kaempferol cleavage. Combined with m/z: 307.0835, 163.0398, and 145.0296, we speculated that this was tribuloside (C30H26O13) (Figure 2B2), and that peaks 84 and 93 were its isomers.

3.1.3. Fragmentation of Quinine Acid Compounds

Nine caffeoylquinic acid compounds, which lost CO (m/z 28), CO2 (m/z 44), and H2O (m/z 18) during MS/MS cleavage, were observed temporarily in this study [30,31,32]. For example, [M + H]+ ion at m/z: 193.0706 of peak 3 was matched with the molecular formula, C7H12O6. Compared with [M + H]+ ions, m/z:175.0608 ions were less than 18 Da and m/z: 147.0656 ions were less than 46 Da, which meant that peak 3 contained hydroxyl and carboxyl groups. After comparison with standard products, peak 3 was identified as quinic acid.
The negative MS/MS spectra of peaks 22, 28, 37, and 68 showed the presence of quinic and caffeic acids in the structure. For example, in the MS/MS spectra of peak 22, the [M − H] ion at m/z: 353.0878 lost 162 Da and acquired m/z 191.0565, and lost 180 Da and acquired m/z 173.0445, which confirmed the presence of caffeic acid. In addition, [M − H] ion lost 192 Da and acquired m/z 161.0242, and lost 174 Da and acquired m/z 179.0344, which indicated the presence of quinic acid. Therefore, peaks 22, 28, and 37 were identified as caffeoylquinic acid isomers (C16H18O9) and peak 68 was dicaffeoylquinic acid (C25H24O12).

3.2. Structural Identification of Purified Samples

The 10% ethanol elution (101.2 g) was dissolved in water and then adsorbed by MCI-gel pore resin to obtain five fractions (Fr1–Fr5) by gradient elution of methanol/water (1:0–0:1). Fr1 underwent repeated ODS column chromatography and preparative HPLC to give compound 1. Compound 2 was obtained by repeated crystallization of Fr3. Separation of Fr4 by ODS column chromatography gave compound 3.
The 70% ethanol elution (516.2 g) was dissolved in methanol, subjected to column chromatography on silica gel (300–400 mesh) under reduced pressure, and eluted with ethyl acetate:methanol (1:0–1:1) to obtain 10 fractions (Fr1–Fr10). MCI columns, silica gel, ODS, Sephadex LH-20 and re-crystallization chromatographic methods were then use to separate compounds 38 by Fr1, compounds 912 by Fr2, compounds 1316 by Fr3, and compounds 17 and 18 by Fr4.
A total of 18 purified compounds were identified using UV, MS, 1H-NMR, and 13C-NMR methods: 1,2,3,4,6-penta-O-galloyl-β-d-glucose (1), 3,4,5-trimethoxyphenyl-(6′-O-galloyl)-O-β-d-glucopyranoside (2), 6′-O-galloylsalidroside (3), gallic acid (4), protocatechuic acid (5), ethyl gallate (6), tiliroside (7), 3-O-coumaroylquinicacid (8), kaempferol-3-O-β-d-galactopyranosyl-(1→6)-β-d-glucopyranoside (9), kaempferol-3-O-β-d-galactopyranoside (10), quercetin-3-O-β-d-glucopyranoside (11), 5-O-caffeoylquinic acid (12), 3-O-coumaroylquinic acid methyl ester (13), 3-O-caffeoylquinic acid (14), 5-O-coumaroylquinic acid (15), 3,5-O-diocaffeoylquinic acid (16), 4,5-O-diocaffeoylquinic acid (17), 3,4-O-diocaffeoylquinic acid (18) (Supplementary Material Figure S1). Preliminary information on compounds 710, 13, and 1518 has been reported elsewhere [33].
Compound 1: ESI-MS m/z: 939.1109 [M − H]; 1H-NMR (600 MHz, DMSO) δ: 6.98 (2H, s), 6.92 (2H, s), 6.86 (2H, s), 6.82 (2H, s), 6.77 (2H, s), 6.38 (1H, d, J = 8.3 Hz, H-1), 5.95 (1H, t, J = 9.6 Hz, H-2), 5.43 (2H, m, H-3,4), 4.99 (1H, d, J = 9.8 Hz, H-5), 4.70 (2H, m, H2-6).13C-NMR (DMSO, 151 MHz) δ: 145.97 (C-1′ × 5), 139.58 (C-2′ × 5), 118.54 (C-3′,5′ × 5), 109.35 (C-2′,6′ × 5), 165.05 (C-7′ × 5), 92.16 (C-1), 72.39 (C-2), 72.59 (C-3), 68.23 (C-4), 71.03 (C-5) and 61.92 (C-6). By comparing experimental data with the literature [34], we determined compound 1 to be 1,2,3,4,6-penta-O-galloyl-β-d-glucose (C41H32O26).
Compound 2: ESI-MS m/z: 497.1300 [M − H]. 1H-NMR (600 MHz, DMSO) δ: 6.30 (2H, s, H-2, 6), 3.64 (6H, s, OMe-3,5), 3.56 (3H, s, OMe-4), 4.92 (1H, d, J = 7.7 Hz, H-1′), 3.74 (1H, m, H-2′), 3.30 (3H, m, H-3′,4′,5′), 4.48 (1H, dd, J = 11.9 Hz, 1.7 Hz, H-6′a), 4.27 (1H, dd, J = 12.0, 5.7 Hz, H-6′b), 6.949 (2H, s, H-2″,6″), 9.25 (2H, br.s, OH-3″,5″), 8.95 (1H, br.s, OH-4″); 13C-NMR (151 MHz, DMSO) δ: 154.20 (C-1), 94.71 (C-2,6), 153.61 (C-3,5), 133.08 (C-4), 60.57 (3,5-OMe), 56.13 (4-OMe), 101.14 (C-1′), 73.66 (C-2′), 76.68 (C-3′), 70.17 (C-4′), 74.33 (C-5′), 64.04 (C-6′), 119.87 (C-1″), 109.08 (C-2″, 6″), 146.03 (C-3″, 5″), 138.95 (C-4″) and 166.27 (C-7″). Marrying the experimental data with the literature [35], compound 2 was identified as 3,4,5-trimethoxyphenyl-(6′-O-galloyl)-O-β-d-glucopyranoside (C22H26O13).
Compound 3: ESI-MS m/z: 451.1244 [M − H].1 H-NMR (600 MHz, MeOD) δ: 6.66 (2H, d, J = 8.5 Hz, H-2,6), 6.98 (2H, d, J = 8.5 Hz, H-3,5), 3.71 (1H, m, Ha-7), 3.93 (1H, m, Hb-7), 2.80 (2H, m, H2-8), 4.33 (1H, d, J = 7.8 Hz, H-1′), 3.23 (1H, t, J = 8.4 Hz, H-2′), 3.41 (2H, m, H-3′,4′), 3.57 (1H, m, H-5′), 4.53 (1H, dd, J = 11.8, 2.1 Hz, Ha-6′), 4.44 (1H, dd, J = 11.8, 5.8 Hz, Hb-6′), 7.11 (2H, s, H-2″, 6″); 13C-NMR (151 MHz, MeOD) δ: 167.01 (COOH), 129.27 (C-1), 129.51 (C-2,6), 114.77 (C-3,5), 155.27 (C-4), 70.39 (C-7), 35.05 (C-8), 103.12 (C-1′), 73.70 (C-2′), 76.58 (C-3′), 70.88 (C-4′), 74.08 (C-5′), 63.40 (C-6′), 120.09 (C-1″), 108.84 (C-2″,6″), 145.15 (C-3″,5″) and 138.49 (C-4″). By comparing experimental data with the literature [36], compound 3 was identified as 6′-O-galloylsalidroside (C21H24O11).
Compound 4: ESI-MS m/z: 169.0145 [M − H].1H-NMR (600 MHz, DMSO) δ: 12.20 (1H, br.s, COOH), 9.15 (2H, br.s, OH-3,5), 8.80 (1H, br.s, OH-4), 6.937 ( 2H, s, H-2,6); 13C-NMR (DMSO,151 MHz) δ: 120.90 (C-1),109.19 (C-2,6), 145.87 (C-3,5), 138.45 (C-4), 170.6 (C-7). Merging experimental and literature data [37], revealed compound 4 to be gallic acid (C7H6O5).
Compound 5: ESI-MS m/z:153.0197 [M − H]. 1H-NMR (600 MHz, MeOD) δ: 12.20 (1H, br.s, COOH), 8.48 (2H, br.s, OH-3,4), 7.47 (1H, d, J = 2.0 Hz, H-2), 6.82 (1H, d, J = 8.0 Hz, H-5), 7.45 (1H, dd, J = 8.2, 2.1 Hz, H-6); 13C-NMR (151 MHz, MeOD) δ: 122.44 (C-1), 108.95 (C-2), 149.92 (C-3), 144.60 (C-4), 114.34 (C-5), 116.37 (C-6) and 169.33 (C-7). According to the combined experimental and literature data [38], it was determined that compound 5 was protocatechuic acid (C7H6O4).
Compound 6: ESI-MS m/z: 197.0453 [M − H]. 1H-NMR (600 MHz, DMSO) δ: 6.97 (2H, s, H-2,6), 4.22 (2H, q, J = 7.1 Hz, CH2), 1.28 (3H, t, J = 7.1 Hz, CH3); 13C-NMR (151 MHz, DMSO) δ: 120.80 (C-1), 108.96 (C-2,6), 146.03 (C-3,5), 138.82 (C-4), 166.30 (C-7), 60.46 (CH2) and 14.72 (CH3). Based on the above data and the literature [39], compound 6 was identified as ethyl gallate (C9H10O5).
Compound 11: ESI-MS m/z: 463.0882 [M − H]. 1H-NMR (600 MHz, DMSO) δ: 7.70 (1H, d, J = 2.4 Hz, H-2′), 7.56 (1H, dd, J = 2.4, 8.8 Hz, H-6′), 6.84 (1H, d, J = 8.6 Hz, H-5′), 6.40 (1H, d, J = 2.0 Hz, H-8), 6.20 (1H, d, J = 2.0 Hz, H-6), 5.45 (1H, d, J = 7.4 Hz, H-1″), 3.22–3.59 (6H, m, H-2″−6″); 13C-NMR (151 MHz, DMSO) δ: 156.81 (C-2), 133.80 (C-3), 177.91 (C-4), 161.72 (C-5), 99.15 (C-6), 164.68 (C-7), 93.98 (C-8), 156.64 (C-9), 104.43 (C-10), 122.07 (C-1′), 115.68 (C-2′), 121.65 (C-6′), 116.68 (C-5′), 148.94 (C-4′), 101.36 (C-1″), 74.58 (C-2″), 76.99 (C-3″), 70.42 (C-4″), 78.04 (C-5″) and 61.46 (C-6″). Based on our findings and the literature [40], compound 11 was identified as quercetin-3-O-β-d-glucopyranoside (C21H20O12).
Compound 12: ESI-MS m/z 353.0879 [M − H]. 1H-NMR (600 MHz, MeOD) δ: 2.24 (2H, m, H-2), 4.19 (1H, br.s, H-3), 3.76 (1H, m, H-4), 5.37 (1H, d, J = 4.2 Hz, H-5), 2.10 (2H, m, H-6), 7. 06 (1H, d, J = 1.9 Hz, H-2′), 6.79 (1H, d, J = 8.2 Hz, H-5′), 6.98 (1H, dd, J = 1.9, 8.2 Hz, H-6′), 7.58 (1H, d, J = 15.9 Hz, H-7′), 6.28 (1H, d, J = 15.9 Hz, H-8′); 13C-NMR (151 MHz, DMSO) δ: 74.84 (C-1), 37.47 (C-2), 69.98 (C-3), 70.61 (C-4), 72.15 (C-5), 36.84 (C-6), 175.73 (C-7), 126.43 (C-1′), 113.90 (C-2′), 145.41 (C-3′), 148.17 (C-4′), 115.10 (C-5′), 121.58 (C-6′), 145.69 (C-7′), 113.83 (C-8′) and 167.30 (C-9′). Based on the above data and the literature [41], compound 12 was identified as 5-O-caffeoylquinic acid (C16H18O9).
Compound 14: ESI-MS m/z 353.0881 [M − H]. 1H-NMR (600 MHz, DMSO) δ: 1.79 (2H, m, H-2), 5.07 (1H, m, H-3), 3.78 (1H, m, H-4), 4.75 (1H, br.s, H-5), 1.95 (2H, m, H-6), 7.04 (1H, d, J = 1. 9 Hz, H-2′), 6.77 (1H, d, J = 8.1 Hz, H-5′), 6.98 (1H, dd, J = 1.9, 8.2 Hz, H-6′), 7.42 (1H, d, J = 15.9 Hz, H-7′), 6.15 (1H, d, J = 15.9 Hz, H-8′); 13C-NMR (151 MHz, DMSO) δ: 73.95 (C-1), 37.68 (C-2), 71.34 (C-3), 70.85 (C-4), 68.54 (C-5), 36.72 (C-6), 175.38 (C-7), 126.07 (C-1′), 115.25 (C-2′), 145.40 (C-3′), 148.81 (C-4′), 116.21 (C-5′), 121.81 (C-6′), 146.03 (C-7′), 114.77 (C-8′) and 166.19 (C-9′). Based on the above data and literature findings [41], compound 14 was identified as 3-O-caffeoylquinic acid (C16H18O9).

3.3. Antioxidant Activity Analysis of Purified Compounds

The antioxidant activity of the purified compounds was tested using DPPH methods (Table 2, Figure 3). With the exception of compound 11, all test compounds showed significant antioxidant activity. The IC50 values of compounds 2, 3, 11, 12, and 14 ranged from 3.00 to 4.05 μg/mL, and all were slightly higher than in the VC control group (2.08 μg/mL). The IC50 values of compounds 1, 4, 5, and 6 were 1.88, 1.05, 1.18, and 1.05 μg/mL, respectively, and were significantly lower than those of the VC control group.

4. Conclusions

In this study, UHPLC-Q-TOF-MS/MS and a variety of chromatographic separation techniques were used to systematically analyze and separate the active antioxidant components of LCO. A total of 100 compounds were identified from the 70% ethanol extract of LCO, and these were mainly gallic acid tannins and flavonoids. Of these, 18 pure compounds were isolated, with compounds 2, 5, 6, 12, 14, and 1618 identified for the first time in LCO and the genus Loropetalum. DPPH results showed that compounds 1, 4, 5, and 6 had significant antioxidant activity.

Supplementary Materials

The following are available online. Figure S1: The structure of compound 1~18.

Author Contributions

Conceived and designed the experiments: W.Z., Y.F., S.Y.; Performed the experiments: H.C., M.L., C.Z., W.D., H.S., W.Z.; Analyzed the data: W.Z., Y.F.; Wrote the paper: H.C., W.Z. All authors read and approved the final manuscript.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (NO. 81660650, 81660670), Science and technology research project of Jiangxi Provincial Education Department (NO.GJJ170723), Open Fund Project of Collaborative Innovation Center for Modern Science and Technology and Industrial Development of Jiangxi Traditional Medicine (NO.JXXT201402008), Study on the process evaluation of innovative drugs and preparations in Jiangxi province with 5511 advantage scientific and technological innovation team (NO. 20165BCB19009), Jiangxi Innovative Drugs and Efficient Energy Saving Devices for Pharmaceutical Collaborative Innovation Center Project( NO.GJGZ(2012)89).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Robert, L.; Jia, Z.W.; Michael, A.T. Defining ROS in biology and medicine. React. Oxyg. Species (Apex) 2016, 1, 9–21. [Google Scholar]
  2. Zhu, H.; Kassim, T.; Arben, S.; Michael, A.T.; Robert, L.Y. Oxygen and oxygen toxicity: The birth of concepts. React. Oxyg. Species (Apex) 2016, 1, 1–8. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, Y.H.; Chen, Z.W; Li, H.M.; Yan, X.F.; Feng, B. AGE/RAGE-Induced EMP release via the Nox-derived ROS pathway. J. Diabetes Res. 2018. [Google Scholar] [CrossRef] [PubMed]
  4. Ana, L.S.; Sanchari, S.; Ariel, B.L. The good, the bad, and the ugly of ROS: New insights on aging and aging-related diseases from eukaryotic and prokaryotic model organisms. Oxid. Med. Cell. Longev. 2018, 2018, 1941285. [Google Scholar] [CrossRef]
  5. Gabriella, A.; Ulla, G.K. NADPH oxidases and ROS signaling in the gastrointestinal tract. Mucosal. Immunol. 2018. [Google Scholar] [CrossRef]
  6. Miura, M.; Taguchi, Y.; Handoh, T.; Hasegawa, T.; Takahashi, Y.; Morita, N.; Matsumoto, A.; Sato, H.; Shindoh, C. Regional increase in ROS within stretched region exacerbates arrhythmias in rat trabeculae with nonuniform contraction. Pflugers Arch. 2018. [Google Scholar] [CrossRef] [PubMed]
  7. Grzesik, M.; Naparło, K.; Bartosz, G.; Sadowska-Bartosz, I. Antioxidant properties of catechins: Comparison with other antioxidants. Food Chem. 2018, 241, 480–492. [Google Scholar] [CrossRef] [PubMed]
  8. Gahruie, H.H.; Niakousari, M. Antioxidant, antimicrobial, cell viability and enzymatic inhibitory of antioxidant polymers as biological macromolecules. Int. J. Biol. Macromol. 2017, 104, 606–617. [Google Scholar] [CrossRef] [PubMed]
  9. Tanase, C.; Bara, C.I.; Popa, V.I. Cytogenetical effect of some polyphenol compounds separated from industrial by-products on maize (Zea mays L.) plants. Cellul. Chem. Technol. 2015, 49, 799–805. [Google Scholar]
  10. Miles, E.A.; Zou, B.L.; Calder, P.C. Differential anti-inflammatory effects of phenolic compounds from extra virgin olive oil identified in human whole blood cultures. Nutrition 2005, 21, 389–394. [Google Scholar] [CrossRef] [PubMed]
  11. Tanase, C.; Cosarca, S.; Toma, F.; Mare, A.; Man, A.; Miklos, A.; Imre, S.; Boz, I. Antibacterial activities of beech bark (Fagus sylvatica L.) polyphenolic extract. Environ. Eng. Manag. J. 2018, 17, 877–884. [Google Scholar]
  12. Committee for the Pharmacopoeia of People’s Republic of China. Pharmacopoeia of People’s Republic of China, 1977 ed.; Chemical Industry Publishing House: Beijing, China, 1977; p. 528. [Google Scholar]
  13. Zhang, Y.; Ren, F.C.; Yang, Y.; Wei, L.; Xu, R.; Zhao, Y.M. Chemical constituents and biological activities of Loropetalum chinense and Loropetalum chinense var, rubrum: Research advances. J. Int. Pharm. Res. 2014, 41, 307–312. [Google Scholar]
  14. Takashi, Y.; Shigeharu, T.; Yanze, L.; Yuan, K.; Ji, C.R.; Takuo, O. Hydrolysable tannins from Loropetalum Chinense. Phytochemistry 1993, 32, 1287–1292. [Google Scholar]
  15. Takashi, Y.; Osamu, N.; Chen, L.; Liu, Y.Z.; Takuo, O. Ellagitannin monomers and oligomers from Euphorbia prostrate Ait. and oligomers from Loropetalum chinense oliv. Chem. Pharm. Bull. (Tokyo) 1990, 38, 3296–3302. [Google Scholar]
  16. Liu, Y.Z.; Wu, Y.Y.; Yuan, K.; Ji, C.R.; Hou, A.J.; Takashi, Y.; Takuo, O. Astragalin 2′′,6′′-Di-O-gallate from Loropetalum chinense. Phytochemistry 1997, 46, 389–391. [Google Scholar] [CrossRef]
  17. Zhang, Q.H.; Fan, D.; Xiong, B.J.; Kong, L.B.; Zhu, X.D. Isolation of new flavan-3-ol and lignin glucoside from Loropetalum chinense and their antimicrobial activities. Fitoterapia 2013, 90, 228–232. [Google Scholar] [CrossRef] [PubMed]
  18. Shao, H.H.; He, M.Z.; Zhang, W.G.; Ou, Y.H.; Feng, Y.L.; Yang, S.L. Determination of the Antioxidant Activity of Ethanol Extract and Its Difierent Polarity Fractions from Loropetalum chinense with DPPH Assay in vitro. Lishizhen Med. Mater. Med. Res. 2013, 23, 1577–1579. [Google Scholar]
  19. Xie, Y.; Shao, H.H.; Song, Y.G.; Liu, Y.T.; Zhang, W.G.; Feng, Y.L.; Yang, S.L. Extraction technology of gallic acid and total polyphenols from Loropetalum chinense. Chin. Med. Pharmacol. Clin. 2012, 18, 9–12. [Google Scholar]
  20. Bakhta, A.; Amira, M.S.; Hamadi, F.; Monique, S.J.S.; Mohamed, B. Anti-oxidant, anti-inflammatory, analgesic and antipyretic activities of grapevine leaf extract (Vitis vinifera) in mice and identification of its active constituents by LC–MS/MS analyses. Biomed. Pharmacother. 2016, 84, 1088–1098. [Google Scholar]
  21. Isabel, M.; Leandro, L.; Teresa, Q.; Sofia, K.; Helena, P. The bark of Eucalyptus sideroxylon as a source of phenolic extractswith anti-oxidant properties. Ind. Crop. Prod. 2016, 82, 81–87. [Google Scholar]
  22. Henrique, S.A.; Gustavo, A.P.; Damila, R.M.; Marcos, N.E.; Glaucia, M.P. Determination of free, esterified, glycosylated and insoluble-bound phenolics composition in the edible part of araticum fruit (Annona crassiflora Mart.) and its by-products by HPLC-ESI-MS/MS. Food. Chem. 2018, 245, 738–749. [Google Scholar]
  23. Wang, L.L.; Sang, M.M.; Liu, E.W.; Prince, O.B.; Zhang, Y.; Wang, T.; Han, L.F.; Gao, X.M. Rapid profiling and pharmacokinetic studies of major compounds incrude extract from Polygonum multiflorum by UHPLC-Q-TOF-MS and UPLC–MS/MS. J. Pharm. Biomed. 2017, 140, 45–61. [Google Scholar] [CrossRef] [PubMed]
  24. Muhammad, Y.; Bushra, S.; Matthew, A. Biological activities of phenolic compounds extracted from Amaranthaceae plants and their LC/ESI-MS/MS profiling. J. Funct. Foods 2016, 26, 645–656. [Google Scholar]
  25. Zhang, L.; Tu, Z.C.; Xie, X.; Lu, Y.; Wang, Z.X.; Wang, H.; Sha, X.M. Antihyperglycemic, antioxidant activities of two Acer palmatum cultivars, and identification of phenolics profile by UPLC-QTOF-MS/MS: New natural sources of functional constituents. Ind. Crop. Prod. 2016, 89, 522–532. [Google Scholar] [CrossRef]
  26. Sunil, K.; Awantika, S.; Brijesh, K. Identification and characterization of phenolics and terpenoids from ethanolic extracts of Phyllanthus species by HPLC-ESI-QTOF-MS/MS. Chin. J. Pharm. Anal. 2017, 7, 214–222. [Google Scholar]
  27. Qin, Y.; Gao, B.Y.; Shi, H.M.; Cao, J.; Yin, C.L.; Lu, W.Y.; Yu, L.L.; Cheng, Z.H. Characterization of flavonol mono-, di-, tri- and tetra-O-glycosides byultra-performance liquid chromatography-electrosprayionization-quadrupole time-of-flight mass spectrometry and itsapplication for identification of flavonol glycosides in Violatianschanica. J. Pharm. Biomed. 2017, 142, 113–124. [Google Scholar]
  28. Shan, L.L.; Wu, Y.Y.; Yuan, L.; Zhang, Y.N.; Xu, Y.Y.; Li, Y.B. Rapid screening of Chemical constituents in Rhizoma anemarrhenae by UPLC-Q-TOF/MS combined with data postprocessing techniques. Evid. Based Complement. Altern. Med. 2017. [Google Scholar] [CrossRef] [PubMed]
  29. Li, H.B.; Yu, Y.; Wang, Z.Z.; Geng, J.L.; Dai, Y.; Xiao, W.; Yao, X.S. Chemical profiling of Re-Du-Ning injection by ultra-performance liquid chromatography coupled with electrospray ionization tandem quadrupole time-of-flight mass spectrometry through the screening of diagnostic ions in MSE mode. PLoS ONE 2015, 10, 1–19. [Google Scholar] [CrossRef] [PubMed]
  30. Chen, H.J.; Baskaran, S.I.; Chen, B.H. Determination of phenolic acids and flavonoids in Taraxacum formosanum kitam by liquid chromatography tandem mass spectrometry coupled with a post column derivatization technique. Int. J. Mol. Sci. 2012, 13, 260–285. [Google Scholar] [CrossRef] [PubMed]
  31. Chen, F.J.; Long, X.H.; Liu, Z.Q.; Shao, H.B.; Liu, L. Analysis of phenolic acids of jerusalem artichoke (Helianthus tuberosus L.) responding to salt-stress by liquid chromatography/tandem mass spectrometry. Sci. World J. 2014, 1–8. [Google Scholar]
  32. Shao, X.; Zhao, J.; Wang, X.; Tao, Y. Rapid screening and quantitative determination of active components in qing-hua-yu-re-formula using UHPLC-Q-TOF/MS and HPLC-UV. J. Anal. methods Chem. 2018. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, W.G.; Chen, H.F.; Shao, H.H.; Feng, Y.L.; Zhong, Y.Q.; Yang, S.L. Hemostatic activity and ingredients of Loropetalum chinense. Chin. J. Exp. Tradit. Med. Formul. 2017, 23, 47–52. [Google Scholar]
  34. Mohan, C.G.; Viswanatha, G.L.; Savinay, G.; Rajendra, C.E.; Halemani, P.D. 1,2,3,4,6 Penta-O-galloyl--d-glucose, a bioactivity guided isolated compound from Mangifera indica inhibits 11β-HSD-1 and ameliorates high fat diet-induced diabetes in C57BL/6 mice. Phytomedicine 2013, 20, 417–426. [Google Scholar] [CrossRef] [PubMed]
  35. Luisella, V.; Mario, D.A.; Andrea, G.; Marco, G.; Pierre, C.; Enrica, B. In vitro antiplasmodial activity of extracts of Tristaniopsis species and identification of the active constituents: Ellagic acid and 3,4,5-Trimethoxyphenyl-(6′-O-galloyl)-O-a-d-glucopyranoside. J. Nat. Prod. 2001, 64, 603–607. [Google Scholar]
  36. Klaus, P.L.; Maki, K.; Andreas, S.; Herbert, K. An ellagitannin, n-butyl gallate, two aryltetralin lignans, and an unprecedented diterpene ester from Pelargonium reniforme. Phytochemistry 2008, 69, 820–826. [Google Scholar]
  37. Liu, M.C.; Yang, S.J.; Jin, L.H.; Hu, D.Y.; Wu, Z.B.; Yang, S. Chemical constituents of the ethyl acetate extract of Belamcanda chinensis (L.) DC roots and their antitumor activities. Molecules 2012, 17, 6156–6169. [Google Scholar] [CrossRef] [PubMed]
  38. Yasmine, C.; Samir, B.; Eric, M.; Zhao, M.J.; Paul, M.; Fadila, B. On-line screening, isolation and identification of antioxidant compounds of Helianthemum ruficomum. Molecules 2017, 22, 239. [Google Scholar]
  39. Li, K.; Lin, Y.; Li, B.; Pan, T.W.; Wang, F.; Yuan, R.Q.; Ji, J.J.; Diao, Y.P.; Wang, S.Y. Antibacterial constituents of fructus chebulae immaturus and their mechanisms of action. BMC Complement. Altern. Med. 2016, 16, 183. [Google Scholar] [CrossRef] [PubMed]
  40. Serge, L.; Isabelle, C.; André, P.; Charles, G.; Michaël, O.; Francine, N.L.; Vakhtang, M.; Jean, L. Chemical composition and anti-herpes simplex virus type 1 (HSV-1) activity of extracts from Cornus canadensis. BMC Complement. Altern. Med. 2017, 17, 123. [Google Scholar]
  41. Wan, C.P.; Li, S.S.; Liu, L.; Chen, C.Y.; Fan, S.Y. Caffeoylquinic Acids from the Aerial Parts of Chrysanthemum coronarium L. Plants (Basel) 2017, 6, E10. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds 1,2,3,4,6-penta-O-galloyl-β-d-glucose, 3,4,5-trimethoxyphenyl-(6′-O-galloyl)-O-β-d-glucopyranoside, 6′-O-galloylsalidroside, gallic acid, protocatechuic acid, ethyl gallate, tiliroside, 3-O-coumaroylquinicacid, kaempferol-3-O-β-d-galactopyranosyl-(1→6)-β-d-glucopyranoside, kaempferol-3-O-β-d-galactopyranoside, quercetin-3-O-β-d-glucopyranoside, 5-O-caffeoylquinic acid, 3-O-coumaroylquinic acid methyl ester, 3-O-caffeoylquinic acid, 5-O-coumaroylquinic acid, 3,5-O-diocaffeoylquinic acid, 4,5-O-diocaffeoylquinic acid, 3,4-O-diocaffeoylquinic acid are available from the authors.
Figure 1. BPI chromatograms of Loropetalum chinense (R. Brown) Oliv. (LCO) in negative ion mode (A) and in positive ion mode (B).
Figure 1. BPI chromatograms of Loropetalum chinense (R. Brown) Oliv. (LCO) in negative ion mode (A) and in positive ion mode (B).
Molecules 23 01720 g001
Figure 2. The MS/MS spectrum information of different compounds in LCO. (A1) Peak 5; (A2) Peak 4; (B1) Peak 66; (B2) Peak 89.
Figure 2. The MS/MS spectrum information of different compounds in LCO. (A1) Peak 5; (A2) Peak 4; (B1) Peak 66; (B2) Peak 89.
Molecules 23 01720 g002
Figure 3. The IC50 values of purified compounds in LCO on antioxidant activity.
Figure 3. The IC50 values of purified compounds in LCO on antioxidant activity.
Molecules 23 01720 g003
Table 1. Identification of the chemical constituents in Loropetalum chinense (R. Brown) Oliv. (LCO) by HPLC–ESI-Q-TOF-MS/MS.
Table 1. Identification of the chemical constituents in Loropetalum chinense (R. Brown) Oliv. (LCO) by HPLC–ESI-Q-TOF-MS/MS.
No.tR/MinFormulaError/ppmAdductFound at Mass/DaMS2 IonsIndentification
11.21C14H16O10−0.1[M + H]+345.0815171.0275, 153.0179, 125.0235Theogallin
21.32C13H16O100.2[M − H]331.0671331.0671, 169.0144, 125.0243Monogalloyl glucose
31.32C7H12O6−0.8[M + H]+193.0706175.0608, 147.0656, 129.0551Quinic acid
41.66C14H16O100.5[M + H]+345.0818327.0726, 171.0289, 153.0185, 125.0232Theogallin
51.75C7H6O5−2.4[M + H]+171.0284153.0180, 135.0074, 125.0234, 109.0286, 107.0128, 97.0283Gallic acid
61.97C13H16O100.7[M − H]331.0673331.0673, 169.0130, 125.0241Monogalloyl glucose
72.35C13H16O10−0.1[M − H]331.0672331.0672, 169.0144, 125.0242Monogalloyl glucose
82.86C13H16O10−0.1[M − H]331.0671331.0671, 169.0142, 125.0249Monogalloyl glucose
93.63C7H6O4−1.7[M + H]+155.0336137.0230, 109.0288, 107.0112, 93.0343, 81.0343Protocatechuic acid
103.63C27H22O182.6[M − H]633.0750633.0750, 481.0649, 300.0992, 169.0153, 125.0232Corilagin
113.64C13H16O100.9[M − H]331.0674331.0685, 169.0148, 151.0031, 125.0257Monogalloyl glucose
124.06C20H20O141[M − H]483.0785483.0786, 331.0673, 313.0568, 169.0145, 151.0038, 125.0249Digalloylglucose
134.07C19H26O150.3[M − H]493.1201493.1201, 331.0648, 169.0142, 125.0248Gallic acid diglucoside
144.26C13H16O10−0.1[M − H]331.0670331.0670, 169.0141, 125.0250Monogalloyl glucose
154.49C19H26O151[M − H]493.1204493.1204, 331.0688, 313.0565, 169.0149, 125.0243Gallic acid diglucoside
164.52C27H22O183.4[M − H]633.0752633.0752, 481.0608, 463.0505, 300.0992, 169.0138, 125.0255Corilagin
174.77C8H8O5−3.8[M + H]+185.0437125.0305, 107.0121, 81.0340Methyl gallate
184.79C20H20O140.8[M − H]483.0784483.0784, 331.0676, 313.0566, 169.0150, 151.0041, 125.0254Digalloylglucose
195.31C27H22O183.5[M − H]633.0755633.0755, 463.0597, 300.0980, 169.0152Corilagin
205.33C20H20O141.2[M − H]483.0786483.0786, 331.0677, 313.0571, 169.0143, 151.0035, 125.0249Digalloylglucose
215.81C27H22O182.8[M − H]633.0751633.0751, 481.0757, 463.0505, 300.0989, 169.0143, 125.0246Corilagin
225.89C16H18O90.7[M − H]353.0889191.0565, 179.0347, 173.0445, 161.0249, 135.0447Caffeoylquinic acid
235.99C27H24O18−3[M − H]635.0955635.0955, 483.0799, 465.0670, 331.0694, 313.0574, 169.0143, 125.0253Trigalloylglucopyranose
246.02C14H10O9−0.8[M + H]+323.0395153.0179, 125.0233, 79.0185Digallic acid
256.84C8H8O5−1.1[M + H]+185.0442153.0183, 135.0078, 125.0236, 107.0103, 97.0282Methyl gallate
266.88C27H24O182.9[M − H]635.0908635.0908, 483.0788, 465.0700, 331.0683, 313.0579, 169.0149, 125.0252Trigalloylglucopyranose
276.92C27H22O183.5[M − H]633.0749633.0749, 300.0981, 169.0150, 125.0239Corilagin
287.09C16H18O90.3[M − H]353.0878191.0554, 179.0344, 173.0446, 161.0242, 135.0447Chlorogenic acid
297.43C27H30O16−1.1[M + H]+611.1784611.1784, 449.1066, 287.0559Panasenoside
307.50C27H24O183.3[M − H]635.0910635.0913, 483.0789, 465.0687, 331.0698, 313.0567, 169.0145, 125.0241Trigalloylglucopyranose
317.51C27H22O183.2[M − H]633.0753633.0753, 481.0729, 300.0991, 169.0145, 125.0255Corilagin
327.63C9H10O5−2[M + H]+199.0597181.0502, 153.0190, 140.0470, 125.0232, 107.0121, 97.0288Ethyl gallate
337.87C15H10O50.2[M + H]+271.0599271.0599, 215.0699, 177.0559, 169.0641, 153.0569, 149.0246, 119.0509Apigenin
347.87C22H18O11−1.4[M + H]+459.0915459.0915, 307.0381, 289.0714, 163.0391, 153.0178, 151.0391, 139.0389Epigallocatechin Gallate
357.97C27H30O16−2.2[M + H]+611.1593449.1059, 303.0484, 287.0534, 267.0019, 145.0489, 85.0278Rutin
368.14C16H18O80.3[M + H]+339.1073165.0545, 147.0441, 119.0489, 91.0548, 65.0396Coumaroylquinic acid
378.18C16H18O90.4[M − H]353.0878191.0558, 179.0328, 173.0328, 161.0243, 135.0456Caffeoylquinic acid
388.18C27H24O183.3[M − H]635.0911635.0911, 483.0789, 465.0690, 313.0567, 169.0143, 125.0247Trigalloylglucopyranose
398.97C21H24O11−1.1[M + H]+453.1386453.1386, 406.9998, 315.0727, 297.0602, 255.0507, 171.0283, 153.0178, 127.0393Galloylsalidroside
409.08C17H20O9−0.8[M + H]+369.1177195.0656, 177.0547, 149.0588, 145.0283, 134.0349, 117.0344, 89.0394Feruloylquinic acid
419.10C27H24O181.7[M − H]635.0901635.0901, 483.0800, 465.0679, 331.0694, 313.0566, 169.0140, 125.0253Trigalloylglucopyranose
429.20C34H28O222.8[M − H]787.1021787.1021, 635.0919, 617.0798, 483.0752, 465.0682, 313.0566, 169.0142, 125.0249Tetrakisgalloylglucopyranose
439.35C16H18O80.3[M + H]+339.1073147.0440, 119.0492, 91.0548, 65.0395Coumaroylquinic acid
449.85C41H32O26−6.3[M − H]939.1050939.1050, 787.0948, 769.0879, 635.0942, 617.0738, 313.0576, 169.0147Pentagalloylglucopyranose
4510.00C34H28O223.4[M − H]787.1026787.1026, 635.0921, 617.0811, 483.0785, 465.0679, 313.0555, 169.0141, 125.0245Tetrakisgalloylglucopyranose
4610.21C22H26O13−0.9[M + H]+499.1442315.0719, 297.0616, 275.0925, 255.0505, 185.0814, 171.0294, 153.0189, 127.03891-O-3′,4′,5′-trimethoxyphenyl-(6-O-galloyl)-β-d-glucopyranoside
4710.22C28H24O16−0.9[M + H]+617.1131617.1131, 447.0910, 303.0503, 297.0610, 153.0184Galloylhyperin
4810.29C34H28O223.5[M − H]787.1027787.1027, 635.0934, 617.0827, 483.0827, 465.0697, 313.0571, 169.0152, 125.0255Tetrakisgalloylglucopyranose
4910.51C28H24O16−1.9[M + H]+617.1129617.1147, 447.0920, 237.0385, 153.0189Galloylhyperin
5010.51C22H18O10−0.9[M + H]+443.0969273.0757, 165.0549, 153.0179, 151.0386, 147.0431, 139.0385, 123.0438Catechin gallate
5110.52C15H12O5−0.3[M + H]+273.0757273.0760, 163.0402, 153.0192, 147.0424, 135.0436, 123.0440, 105.0326Naringenin
5210.63C21H18O13−1.2[M + H]+479.0814317.0294, 285.0024, 257.0061Shikimic acid-O-digallate
5310.63C34H28O223.6[M − H]787.1028787.1028, 635.0925, 617.0823, 483.0829, 465.0656, 313.0557, 169.0143, 125.0246Tetrakisgalloylglucopyranose
5410.76C14H6O80.3[M + H]+303.0140303.0140, 285.0035, 275.0189, 257.0082, 229.0130, 201.0179, 173.0232, 145.0284Ellagic acid
5510.78C15H10O80.1[M + H]+319.0449319.0450, 301.0346, 290.0422, 273.0393, 245.0444, 217.0503, 165.0176, 153.0182Myricetin
5610.94C21H20O12−0.6[M + H]+465.1028319.0453, 303.0507, 285.0388, 257.0455, 229.0497, 145.0498, 127.0387, 97.0290Myricitrin
5710.96C15H10O7−0.7[M + H]+303.0497303.0497, 285.0037, 275.0176, 257.0436, 229.0493, 153.0181 137.0229Isomer of Quercetin
5811.21C21H20O12−0.7[M + H]+465.1025303.0508, 257.0446, 229.0492, 165.0176, 145.0491, 127.0389, 97.0289, 85.0289Hyperoside
5911.22C15H10O70.2[M + H]+303.0503303.0503, 285.0389, 257.0457, 229.0498, 153.0181 137.0232Isomer of Quercetin
6011.40C21H20O11−1.1[M + H]+449.1073287.0545, 153.0178Luteoloside
6111.66C41H32O262.4[M − H]939.1131939.1131, 787.0943, 769.0877, 635.0796, 617.0789, 313.0644, 169.0141, 125.0259Pentagalloylglucopyranose
6211.73C15H10O7−0.7[M + H]+303.0497303.0497, 285.0367, 257.0453, 229.0478, 153.0186, 137.0591Isomer of Quercetin
6311.96C20H18O11−1[M + H]+435.092303.0500, 285.0415, 257.0448, 229.0493, 153.0179, 137.0217Isomer of guaijaverin
6412.06C15H10O60.2[M + H]+287.0542287.0542, 258.0511, 241.0482, 213.0550, 165.0167, 153.0178, 137.0218, 121.0282Isomer of Kaempferol
6512.07C21H20O11−0.3[M + H]+449.1075287.0560, 165.0177, 153.0182Isomer of luteoloside
6612.16C28H24O156.6[M − H]599.1088599.1088, 447.0969, 313.0582, 285.0414, 169.0147, 151.0036, 125.0245Astragalin-O-gallate
6712.22C41H32O262.6[M − H]939.1133939.1133, 787.1020, 769.0979, 635.0901, 617.0780, 313.0514, 169.0142, 125.0262Pentagalloylglucopyranose
6812.54C25H24O120.6[M − H]515.1195515.1195, 353.0883, 191.0556, 179.0343, 135.0449Dicaffeoylquinic acids
6912.55C15H10O60.3[M + H]+287.0552287.0552, 258.0525, 241.0483, 213.0533, 165.0188, 157.0469, 153.0183, 121.0285Isomer of Kaempferol
7012.63C21H20O11−0.7[M + H]+449.1075303.0511, 287.0562, 165.0178, 145.0496, 129.0548, 127.0387Quercitrin
7112.67C15H10O70.6[M + H]+303.0505303.0505, 285.0395, 257.0445, 229.0494, 153.0182, 137.0231Isomer of Quercetin
7212.97C16H12O71.8[M + H]+317.0653317.0653, 302.0437, 285.0415, 274.0482, 246.0531, 229.0483, 153.0182Isomer of isorhamnetin
7312.99C28H24O157.6[M − H]599.1064599.1064, 447.0947, 313.0577, 285.0408, 169.0136, 151.0035, 125.0238Astragalin-O-gallate
7413.02C15H10O60.4[M + H]+287.0552287.0552, 258.0533, 231.0647, 213.0545, 165.0171, 153.0175, 137.0236, 121.0286Isomer of Kaempferol
7513.04C35H28O19−1.8[M + H]+753.1284467.0822.449.0706, 315.0705, 287.0552, 153.0181, 125.0236Astragalin-O-digallate
7613.30C15H10O60.4[M + H]+287.0561287.0561, 258.0566, 213.0567, 165.0193, 153.0175, 147.0429, 137.0240Isomer of Kaempferol
7713.68C23H24O12−1.6[M + H]+493.1332493.1332, 331.0819, 315.0505, 270.0515Tricin-O-glucopyranoside
7813.72C35H28O19−2[M + H]+753.1283753.1261, 601.1206, 467.0812, 449.0707, 287.0547, 237.0393, 153.0185Astragalin-O-digallate
7914.27C15H10O60.5[M + H]+287.0546287.0546, 258.0527, 241.0491, 213.0551, 165.0180, 153.0181, 137.0233, 121.0285Isomer of Kaempferol
8014.27C21H20O10−1[M + H]+433.1125287.0553, 165.0183, 129.0542, 85.0285, 71.0498Kaempferol-O-rhamnoside
8114.32C23H24O12−1.5[M + H]+493.3333331.0814, 315.0496, 270.0519Isomer of tricin-O-glucopyranoside
8214.32C35H28O19−2.5[M + H]+753.1279753.1366, 601.1066, 467.0804, 449.0691, 287.0543, 237.0403, 153.0183Isomer of astragalin-di-O-gallate
8314.64C16H12O71.3[M + H]+317.0649317.0649, 302.0442, 285.0392, 246.0505, 175.9679, 153.0188, 139.0399Isomer of isorhamnetin
8414.69C30H26O139.4[M − H]593.1360593.1360, 447.0949, 307.0825, 285.0413, 163.0403, 151.0030, 145.0290, 119.0508Isomer of Tribuloside
8516.27C15H10O70.4[M + H]+303.0503303.0503, 285.0399, 257.0445, 229.0491, 201.0552, 153.0182, 137.0230Quercetin
8616.36C15H10O6−0.6[M + H]+287.0553287.0553, 161.0234, 153.0183, 135.0434Isomer of Kaempferol
8716.72C16H12O7−0.7[M + H]+317.0569317.0569, 302.0420, 274.0469, 228.0420, 153.0170, 147.0435Isorhamnetin
8816.88C15H10O60.8[M + H]+287.0547287.0547, 258.0507, 241.0461, 213.0539, 165.0182, 153.0179, 121.0281Isomer of Kaempferol
8916.89C30H26O136.8[M − H]593.1341593.1341, 447.0955, 307.0835, 285.0399, 163.0398, 151.0038, 145.0296, 119.0506Tribuloside
9016.89C22H22O10−1.3[M + H]+447.128301.0705, 286.0479, 258.0536, 153.0179Methylluteolin-O-rhamnopyranosid
9117.34C17H14O7−0.2[M + H]+331.0812331.0820, 315.0498, 286.0462, 270.0522, 258.0520Quercetin-dimethyl ether
9217.39C15H10O60.8[M + H]+287.055287.0550, 258.0539, 241.0463, 213.0522, 165.0175, 153.0190, 121.0279Isomer of Kaempferol
9317.39C30H26O135.8[M − H]593.1354593.1354, 447.0938, 307.0828, 285.0403, 163.0396, 151.0031, 145.0290, 119.0505Isomer of Tribuloside
9417.48C22H22O10−0.9[M + H]+447.1281301.0710, 286.0490, 258.0527Methylluteolin-O-rhamnopyranosid
9518.11C15H12O5−0.8[M + H]+273.0755273.0736, 164.8737, 153.0183, 147.0432, 121.0273, 91.0557Isomer of naringenin
9619.05C15H10O60.9[M + H]+287.0557287.0557, 258.0527, 241.0494, 231.0652, 213.0550, 165.0185, 153.0184, 121.0286Kaempferol
9719.52C17H14O70.7[M + H]+331.0183331.0819, 315.0505, 286.0477, 270.0529, 258.0529, 242.0583Quercetin-dimethyl ether
9819.76C16H12O7−0.9[M + H]+317.0657317.0657, 302.0424, 274.0464, 153.0185Isomer of isorhamnetin
9919.97C20H18O11−1.1[M + H]+435.0924435.0924, 237.0398, 153.0175, 127.0406Guaijaverin
10020.71C17H14O7−0.3[M + H]+331.0809331.0807, 315.0495, 286.0538, 270.0528, 242.0549, 168.0608Quercetin-dimethyl ether
Table 2. The antioxidant activity of purified compounds.
Table 2. The antioxidant activity of purified compounds.
VcConcentration (µg/mL)0.731.161.452.182.90
Inhibition rate (%)18.71 ± 0.1724.26 ± 0.3634.79 ± 0.4746.19 ± 0.2869.15 ± 0.45
1Concentration (µg/mL)0.881.752.632.983.50
Inhibition rate (%)20.03 ± 0.1537.09 ± 0.2263.64 ± 0.2669.15 ± 0.4085.93 ± 0.11
2Concentration (µg/mL)2.312.783.243.704.63
Inhibition rate (%)29.08 ± 0.2045.43 ± 0.3152.63 ± 0.1162.32 ± 0.3587.81 ± 0.13
3Concentration (µg/mL)2.333.724.655.586.98
Inhibition rate (%)25.99 ± 0.1446.53 ± 0.1559.59 ± 0.3171.43 ± 0.2487.76 ± 0.16
4Concentration (µg/mL)0.520.871.211.562.08
Inhibition rate (%)22.33 ± 0.2538.75 ± 0.2752.23 ± 0.3667.66 ± 0.4082.14 ± 0.43
5Concentration (µg/mL)0.761.011.681.892.10
Inhibition rate (%)26.66 ± 0.3738.02 ± 0.3367.52 ± 0.2878.21 ± 0.1981.00 ± 0.40
6Concentration (µg/mL)0.611.011.411.822.02
Inhibition rate (%)26.25 ± 0.2742.90 ± 0.3259.54 ± 0.3376.50 ± 0.2786.47 ± 0.33
10Concentration (µg/mL)10.6321.2525.531.8842.50
Inhibition rate (%)27.55 ± 0.2950.94 ± 0.1961.29 ± 0.2678.36 ± 0.4187.37 ± 0.37
11Concentration (µg/mL)1.383.223.684.149.20
Inhibition rate (%)25.40 ± 0.2747.85 ± 0.2663.16 ± 0.2975.07 ± 0.3388.54 ± 0.33
12Concentration (µg/mL)0.992.467.399.8512.31
Inhibition rate (%)22.33 ± 0.3946.55 ± 0.4068.06 ± 0.5072.53 ± 0.3174.79 ± 0.25
14Concentration (µg/mL)2.233.564.456.688.01
Inhibition rate (%)27.06 ± 0.1939.51 ± 0.5146.28 ± 0.3766.85 ± 0.2488.77 ± 0.31

Share and Cite

MDPI and ACS Style

Chen, H.; Li, M.; Zhang, C.; Du, W.; Shao, H.; Feng, Y.; Zhang, W.; Yang, S. Isolation and Identification of the Anti-Oxidant Constituents from Loropetalum chinense (R. Brown) Oliv. Based on UHPLC–Q-TOF-MS/MS. Molecules 2018, 23, 1720. https://doi.org/10.3390/molecules23071720

AMA Style

Chen H, Li M, Zhang C, Du W, Shao H, Feng Y, Zhang W, Yang S. Isolation and Identification of the Anti-Oxidant Constituents from Loropetalum chinense (R. Brown) Oliv. Based on UHPLC–Q-TOF-MS/MS. Molecules. 2018; 23(7):1720. https://doi.org/10.3390/molecules23071720

Chicago/Turabian Style

Chen, Haifang, Mulan Li, Chen Zhang, Wendi Du, Haihua Shao, Yulin Feng, Wugang Zhang, and Shilin Yang. 2018. "Isolation and Identification of the Anti-Oxidant Constituents from Loropetalum chinense (R. Brown) Oliv. Based on UHPLC–Q-TOF-MS/MS" Molecules 23, no. 7: 1720. https://doi.org/10.3390/molecules23071720

Article Metrics

Back to TopTop