Next Article in Journal
Application of Cyclocarya paliurus–Kiwifruit Composite Fermented to Enhance Antioxidant Capacity, Flavor, and Sensory Characteristics of Kiwi Wine
Next Article in Special Issue
In Vitro and In Silico of Cholinesterases Inhibition and In Vitro and In Vivo Anti-Melanoma Activity Investigations of Extracts Obtained from Selected Berberis Species
Previous Article in Journal
The Impact of Furfural on the Quality of Meads
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Isoprenylated Flavonoids and 2-Arylbenzofurans from the Root Bark of Morus alba L. and Their Cytotoxic Activity against HGC27 Cancer Cells

1
Natural Products Research Center, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
2
Chengdu Institute of Biology, University of Chinese Academy of Sciences, Beijing 100049, China
3
Pharmaceutical Department, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming 650500, China
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(1), 30; https://doi.org/10.3390/molecules29010030
Submission received: 14 November 2023 / Revised: 13 December 2023 / Accepted: 16 December 2023 / Published: 20 December 2023

Abstract

:
Three new compounds (1, 11, and 12), together with 32 known ones, were isolated from the root bark of Morus alba L. using various chromatographic methods. The structures of the undescribed compounds were elucidated based on 1D, 2D NMR, and HRESIMS dataanalysis, while the known ones were identified by comparison of their spectroscopic data with those reported in the literature. All the isolates were evaluated for their cytotoxic activities against human gastric cancer HGC27 cells by CCK-8 assay. Among them, compounds 5, 8, 10, and 30 exhibited cytotoxic activities on HGC27 cells with IC50 values of 33.76 ± 2.64 μM, 28.94 ± 0.72 μM, 6.08 ± 0.34 μM, and 10.24 ± 0.89 μM, respectively. Furthermore, compound 10 was confirmed to reduce proliferation ability, increase apoptosis rate, and inhibit cell migration pathway by annexin V/PI double staining experiment, transwell experiment, and Western blot analysis.

Graphical Abstract

1. Introduction

Morus alba L., the root bark of Morus mulberry, is a well-known traditional Chinese medicine (TCM) documented in Chinese Pharmacopoeia [1]. The root bark of M. alba has been widely used in Chinese herbal medicinal prescriptions for the treatment of pneumonia, hypertension, diabetes, and arthritis [2,3]. Previous phytochemical studies revealed that M. alba contained a variety of polyphenolic constituents such as prenylated flavonoids, 2-arylbenzofurans, stilbene, coumarin, and mulberry Diels–Alder-type adducts. In addition, pharmacological studies exhibited that some polyphenols from M. alba had a broad spectrum of biological activities, such as hepatoprotective activity, whitening ability, and antiplatelet effects [4,5,6,7,8,9]. Previous studies also showed that diverse individual compounds or extracts from different parts of various mulberry plants (M. alba, M. australis, M. wittiorum, and M. yunanensis) exhibited anti-tumor activity against different tumor cell lines, such as lung cancer, liver cancer, colon cancer, breast cancer, rectal adenocarcinoma, and renal adenocarcinoma [10,11,12,13,14]. Our preliminary pharmacological experiment showed that the 75% ethanol extract of M. alba exhibited certain anti-tumor activity against gastric cancer cells, which had rarely been reported. Thus, the present paper focuses on detailed phytochemical investigation of the 75% ethanol extract of M. alba and evaluation of anti-tumor activity against gastric cancer cells of the isolated individual compounds. As a result, thirty-five compounds including three new ones were obtained. Among them, compound 10 (Albanol B) shows the most significant cytotoxicity with an IC50 value of 6.08 ± 0.34 μM. Moreover, the potential mechanism was preliminarily explored.

2. Results and Discussion

2.1. Isolation and Structure Elucidation

Phytochemical investigation on the 75% ethanol extract of the root bark of M. alba led to the isolation and identification of 3 new compounds (1, 11, and 12) and 32 known compounds (210, 1335) as shown in Figure 1. The known compounds were identified by comparing their NMR and ESIMS data with those reported in the literature as mulberrofuran D (2) [15], mulberofuran B (3) [16], mulberofuran A (4) [17], albafuran A (5) [18], mulberrofuran Y (6) [19], moracin M (7) [20], mulberrofuran G (8) [21], mulberrofuran J (9) [22], abanol B (10) [23], kuwanon T (13) [21], morusin (14) [16], kuwanon A (15) [24], notabilisin L (16) [25], 14-methoxydihydromorusin (17) [26], neocyclomorusin (18) [27], cyclomulberrin (19) [28], cyclomorusin (20) [27], artoflavone (21) [29], cudraflavone A (22) [30], kuwanon C (23) [31], mornigrol H (24) [32], austraone A (25) [33], sanggenol Q (26) [34], cycloaltilisin 7 (27) [35], sanggenol O (28) [21], kuwanon E (29) [27], benzokuwanon E (30) [36], kuwanon G (31) [16], albanin G (32) [37], moracenin D (33) [38], mulberrofuran P (34) [39], and 7,4′-dihydroxyflavone (35) [40]. Among all compounds, 110 compounds are 2-arylbenzofurans and 1135 compounds are isoprenylated flavonoids.
Compound 1 was obtained as a brown amorphous powder. The molecular formula of 1 was determined as C25H28O5 according to its HR-ESI-MS at m/z 438.1831 [M + Na]+ (calcd. for C25H28O5Na, 438.1829), indicating the presence of 12 degrees of unsaturation. Absorption maxima in the UV spectrum of 1 at λmax 216 (4.21) and 316 (4.19) nm suggested that it was a 2-arylbenzofuran derivative [19]. In the 1H NMR spectrum (Table 1), three methyl groups at δH 1.62 (3H, s, H-10″), 1.89 (3H, s, H-4″), 3.86 (3H, s, -OCH3); four methylenes at δH 1.60 (2H, m, H-6″), 2.02 (2H, m, H-5″), 3.62 (2H, d, J = 7.2, H-1″), 4.79, 4.72 (2H, s, H-9″α, β); an olefinic singlet at δH 6.92 (1H, s, H-3), two aromatic doublet signals at δH 7.32 (1H, d, J = 8.4, H-4), δH 6.90 (1H, d, J = 8.4, H-5), a 1, 3, 5-trisubstituted aromatic system (ring B) at δH 6.80 (2H, d, J = 2.0, H-2′, 6′) and δH 6.26 (1H, s, H-4′). The 13C NMR (Table 1), DEPT and HSQC spectra showed 25 carbon signals, including three methyls, four methylenes, eight methines, and ten quaternary carbons. Comparison of the 1H and 13C-NMR spectral data of 1 with those of mulberofuran B (3) indicated that they shared similar chemical structures [18]. The marked difference occurred in the position of C-7″ of the geranyl group. The carbon signals for C-5″ and C-7″ were shifted downfield to δC 36.7 (−4.2 ppm) and 76.1 (−49.3 ppm), while the C-6″, C-8″, and C-9″ signals in 1 were shifted upfield dramatically to δC 34.2 (+6.5 ppm), 148.6 (+16.3 ppm), and 111.5 (+85.7 ppm) (Table 1), respectively. Signals in the upfield of the 1H NMR spectrum of 1 were deduced to be a changed geranyl group, one of whose double bonds was hydrated. Two proton signals at δH 4.72 (1H, s) and 4.79 (1H, s) and a signal at δH 3.90 (1H, t, J = 6.8 Hz) suggested that a terminal methylene group and an oxygen-bearing methine group were present in the geranyl group. According to the HSQC spectrum, δC 111.5 was connected with δH 3.90 (1H, t, J = 6.8 Hz), and δC 76.1 was connected to two protons with δH 4.79 (a, 1H, brs) and 4.72 (β, 1H, s). In the HMBC spectrum (Figure 2), long-range correlations of H-9″/C-7″, 10″ and H-7″/C-5″, 9″, 10″, demonstrated that the geranyl group was replaced by a 7″-hydroxy-3″, 8″-dimethylbut-2″, 8″dioctenyl group [41]. Furthermore, H-1″ showed long-range correlations with C-6 and C-7a, supporting that the changed geranyl group was located at C-7. According to the above NMR data analysis, compound 1 (shown in Figure 1) was elucidated as 7″-hydroxy-3″, 8″-dimethylbut-2″, 8″-dioctenyl-6-methoxy-3′, 5′-dihydroxy-2-arylbenzofuran, and was named mulberofuran Z.
Compound 11 was obtained as a yellow powder. The molecular formula of 11 was determined as C25H26O7 according to its HR-ESI-MS at m/z 439.1751 [M + H]+ (calcd. for C25H27O7, 439.1751), indicating 13 degrees of unsaturation. The IR spectrum exhibited absorption peaks attributable to hydroxy groups (3415 cm−1), a conjugated carbonyl group (1656 cm−1), and aromatic rings (1621, 1554, 1448 cm−1). In the 1H NMR spectrum (Table 2), signals due to an isopentyl group at δH 1.61 (3H, s, CH3-4″), 1.71 (3H, s, CH3-5″), 3.36 (2H, d, J = 6.0 Hz, H-1″), and 5.13 (1H, brt, J = 6.0 Hz, H-2″) were observed. The 1H NMR spectrum also showed ortho-coupled aromatic protons (ring B) at δH 7.67 (1H, d, J = 9.0 Hz, H-6′), 6.74 d (1H, d, J = 9.0 Hz, H-5′), and meta-coupled aromatic protons (ring A) at δH 6.12 (1H, brs, H-6), 6.33 (1H, brs, H-8). Two methyl groups δH 1.18 (3H, s, CH3-12), 1.29 (3H, s, CH3-13), and an AMX coupling system at δH 2.74 (1H, dd, J = 16.2, 6.0 Hz, H-9α), 3.00 (1H, dd, J = 16.2, 3.6 Hz, H-9β), and 3.99 (1H, dd, J = 6.0, 3.6 Hz, H-10) indicated the existence of an oxepin ring [42]. The 13C NMR (Table 2), DEPT, and HSQC spectra showed the presence of 25 carbons, including 4 methyls, 2 methylenes, 6 methines, and 13 quaternary carbons. Comparison of the 1H NMR and 13C NMR data of compound 11 with those of the known compound artoxanthocarpuone A [42] suggested that the difference between them was the location of the isopentyl group at C-3′ in 11 rather than at C-6 in artoxanthocarpuone A, which was further confirmed by the key long-range correlations from δH 3.36 (H-1″) to δC 159.4 (C-2′), 119.9 (C-3′), 156.4 (C-4′), and 130.3 (C-3″) (Figure 2). Based on these observations, compound 11 can be assigned as shown in Figure 1 and named artoxanthocarpuone C.
Compound 12 was obtained as a yellow powder. The molecular formula of 12 was determined as C25H26O7 according to its HR-ESI-MS at m/z 439.1749 [M + H]+ (calcd. for C25H27O7, 439.1751), indicating 13 degrees of unsaturation. The IR spectrum exhibited absorption peaks attributable to hydroxy groups (3372 cm−1), a conjugated carbonyl group (1645 cm−1). The UV spectrum showed absorbance maxima at 336 and 215 nm, typical of a flavone skeleton. The 1H NMR spectrum (Table 2) clearly exhibited the presence of an isopentyl group at δH 1.34 (3H, s, CH3-4″), 1.58 (3H, s, CH3-5″), 3.08 (2H, dd, J = 7.2, 5.4 Hz, H-1″), and 5.07 (1H, t, J = 7.2 Hz, H-2″). The 1H NMR spectrum also showed ortho-coupled aromatic protons (ring B) at δH 6.98 (1H, d, J = 8.4 Hz, H-6′), 6.42 (1H, d, J = 8.4 Hz, H-5′), and meta-coupled aromatic protons (ring A) at δH 5.91 (1H, s, H-6), 6.00 (1H, s, H-8). Two methyl groups δH 1.36 (3H, s, CH3-12), 1.28 (3H, s, CH3-13), and an AMX coupling system at δH 2.61 (1H, dd, J = 16.8, 5.4 Hz, H-9a), 2.95 (1H, dd, J = 16.8, 7.4 Hz, H-9b), and 3.80 (1H, dd, J = 16.2, 5.4 Hz, H-10) indicated the existence of an oxepin ring [42]. The 1H NMR and 13C NMR spectra (Table 2) of 12 were similar to those of the known compound kuwanon A [24]. The carbon signals at δC 70.2 (C-10) and 27.6 (C-9) in 12 instead of δC 128.9 (C-10) and 116.3 (C-9) in kuwanon A, along with the difference of their molecular weight, demonstrated the presence of a hydroxy group at C-10 in 12. This was further verified by the long-range correlations from H-9 to C-11, C-1′ and C-4′ and from H-10 to C-11 and C-3′ in the HMBC spectrum (Figure 2) [43]. The HMBC correlations of H-1″/C-2, C-4 and H-2″/C-3, C-4″, C-5″ suggested the isopentyl group was attached to C-3 position. According to the above data, compound 12 was established as depicted in Figure 1 and named kuwanon Z.

2.2. Biological Activities of Compounds against HGC27 Cancer Cells

The effect of compounds (135) on the cell viability of HCG27 cells was evaluated by CCK-8 assay. As shown in Table 3, four compounds 5, 8, 10, and 30 had a certain inhibitory effect on the viability of HCG27 cells compared with the model group (control). In contrast, other compounds had no significant inhibitory effects on the viability of HCG27 cells; therefore, compounds 5, 8, 10, and 30 were screened to evaluate the semi-inhibition concentration (IC50) values (Table 4). Among the four compounds, compound 10 has showed the best cytotoxicity against HCG27 cells, with an IC50 value of 6.08 ± 0.34 μM, and compounds 5, 8, and 30 have showed certain cytotoxicity, with IC50 values of 33.76 ± 2.64 μM, 28.94 ± 0.72 μM, and 10.24 ± 0.89 μM, respectively. Compound 10 effectively inhibited the clone formation of HGC27 cells (Figure 3a). When cells were treated with 6 μM compound 10, the number of cell clones formed in the administered group was significantly reduced and cell growth was significantly inhibited compared to the control group (Figure 3b). We used Annexin V-FITC (Annexin V-Fluorescein Isothiocyanate) double staining assay to detect apoptotic cells. As shown in Figure 3c, the apoptosis rate of HGC27 cells treated with compound 10 significantly increased compared to the control group. Quantification of apoptotic cells by flow cytometry revealed that the percentage of HGC27 apoptotic cells increased in a dose-dependent manner (Figure 3d).
Cell migration is an essential process in tumor metastasis. The effect of compound 10 on migration was examined by the transwell method. The effect of compound 10 on the migration of HGC27 tumor cells was investigated in the premise of HGC27 cell death. As shown in Figure 3e, in vitro migration studies showed that compound 10 significantly inhibited the migratory ability of HGC27 cells in a dose-dependent manner. The number of migrating HGC27 cells was reduced from 647 to 357 compared to the control group (Figure 3f). Caspase-3 is an important protein in the regulation of cell apoptosis [44]. After treating HGC27 cells with 10 μM and 20 μM of compound 10 for 48 h, Western blot analysis showed the expression of apoptosis-related protein Caspase-3 was significantly increased compared with the control (Figure 4). Thus, compound 10 had a substantial cytotoxic effect on HGC27 cells (Figure 4). We also analyzed the expression levels of proteins Bax and Bcl-2 related to cell apoptosis [45]. Bax was upregulated in HGC27 cells, whereas Bcl-2 was downregulated. These results indicated that compound 10 has the capacity to induce apoptosis (Figure 4).

3. Experimental

3.1. General Experimental Procedures

IR and UV spectra were recorded through a PerkinElmer 1725X-FT spectrometer with KBr disks and a PerkinElmer (Boston, MA, USA) Lambda 35 spectrometer, respectively, and optical rotations were determined by using a Perkin-Elmer 341 polarimeter. 1D and 2D NMR spectra were recorded using Bruker Avance 400 and 600 spectrometers. The HR-ESI-MS data were from micrOTOF-QII mass spectrometer. The precoated silica gel GF254 10–40 μm (Qingdao Marine Chemical Group, Qingdao, China) was used for TLC. Analytical HPLC was used with the Prominence LC-20AT with a model SPD-M20A detector and Ultimate ®C18 column (250 mm × 4.60 mm, 5 μm). Preparative HPLC was carried out on P3000 with a UV3000 detector (Chengdu LaiPu Science and Technology, Chengdu, China) and Ultimate ®C18 column (250 mm × 21.2 mm, 5 μm; 250 mm × 50 mm, 10 μm, respectively). Column chromatography (CC) was performed with silica gel (100–200 mesh and 200–300 mesh, Qingdao Marine Chemical Group, China) and RP-C18 (20–40 μm, Fuji, Japan) was used for column chromatography (CC). All chemical solvents were obtained from Chron Chemicals Reagent of Chengdu. The 35 compounds with more than 96% purity were prepared in our own laboratory.

3.2. Plant Material

The root bark of M. alba (Sang-Bai-Pi) was purchased from Lotus Pond Chinese Herbal Medicine market, Sichuan, China. It has been identified by Professor Weikai Bao (Chengdu Institute of Biology, Chinese Academy of Sciences). A voucher specimen (CIB-A-431) has been deposited at the Laboratory of Phytochemistry, Chengdu Institute of Biology, Chinese Academy of Sciences.

3.3. Extraction and Isolation

The air-dried and powdered roots (30.0 kg) were extracted three times with 75% ethanol (100 L × 3, each 3 days). The resulting solution was filtered, combined, and concentrated under reduced pressure to give 3.0 Kg of brown residue. The 75% ethanol extract (3.0 Kg) was partitioned between H2O and ethyl acetate. The ethyl acetate part (2.2 Kg) was separated by silica gel column (6.0 Kg, 150 × 40 cm) eluted with petroleum ether and ethyl acetate (10:1 to 1:1) to yield four fractions (Fr.1–4).
Part of Fr.1 (88.0 g) was separated by RP-HPLC (MeOH/H2O, 88:12) to afford seven fractions (Fr.1.1–Fr.1.7). Compound 13 (6.0 g) was obtained from Fr.1.2 (8.3 g) by crystallization from MeOH. Fr.1.3 (2.0 g) was fractionated with RP-HPLC (MeOH/H2O, 72:28) to give compounds 1 (22.0 mg), 26 (120.0 mg), 16 (16.0 mg), followed by Sephadex LH-20 (MeOH/H2O, 70:30 to 70:30) to yield 11 (8.0 mg), 12 (6.6 mg). Compound 15 (8.0 g) was from Fr.1.4 (12.2 g) by crystallization from MeOH, also. Compound 2 (310.0 mg) was obtained from Fr.1.5 (1.6 g) by RP-HPLC (CH3CN/H2O, 64:36). Compound 14 (22.4 g) was isolated from Fr.1.6 (28.9 g) by crystallization in MeOH. Fr.1.7 (503.0 mg) was fractionated using RP-HPLC with CH3CN/H2O (70:30) to give compounds 17 (18.0 mg), 3 (130.0 mg), 18 (30.0 mg), 19 (46.0 mg), 4 (22.0 mg), 20 (36.0 mg).
Part of Fr.2 (33.0 g) was also separated by RP-HPLC (MeOH/H2O, 86:14) to yield six subfractions (Fr.2.1–Fr.2.6). Fr.2.2 (3.8 g) and Fr.2.4 (1.3 g) were further purified by RP-HPLC eluted with CH3CN/H2O (70:30) to give compounds 28 (109.0 mg, CH3CN/H2O, 70:30) and 21 (28.0 mg MeOH/H2O, 89:11). Compounds 27 (17.0 mg) and 22 (100.0 mg) were obtained from Fr.2.5 (300.0 mg) and Fr.2.6 (653.0 mg) by RP-HPLC eluted with (MeOH-H2O, 90:10) and (MeOH-H2O, 84:16), respectively.
Part of Fr.3 (12.6 g) was subjected to preparative RP-HPLC MeOH/H2O (86:14) to obtain six fractions (Fr.3.1–Fr.3.7). Three compounds 31 (15.7 mg), 30 (8.0 mg), and 7 (33.0 mg) were separated from Fr.3.1 (817.0 mg) through RP-HPLC eluted with MeOH/H2O (73:17). Compounds 23 (86.0 mg) and 5 (42.0 mg) were purified from Fr.3.2 (310.0 mg) by RP-HPLC (CH3CN/H2O, 63:27). Fr.3.3 (120.0 mg) and Fr.3.4 (800.0 mg) were further purified by RP-HPLC eluted with CH3CN/H2O (60:40) and CH3CN/H2O (64:36) to afford compounds 24 (28.0 mg), 6 (98.0 mg). Compound 29 (37.0 mg) was obtained from Fr.3.5 (900.0 mg) through RP-HPLC (MeOH/H2O, 78:22).
Part of Fr.4 (36.0 g) was submitted to RP- HPLC (CH3CN/H2O, 64:36) to yield six subfractions (Fr.4.1–Fr.4.6). Compounds 33 (303.0 mg), 34 (23.0 mg), and 35 (6.0 mg) were separated from Fr.4.2 (13.6 g) by RP-HPLC eluted with CH3CN/H2O (63:37). Fr. 4.3 (800.0 mg) was further purified by RP-HPLC (CH3CN-H2O, 45:55) to obtain compound 9 (168.0 mg). Fr.4.5 (500.0 mg) and Fr.4.6 (200.0 mg) were separated by RP-HPLC eluted with CH3CN/H2O (52:48) and CH3CN/H2O (62:38) to yield compounds 8 (40.0 mg) and 32 (160.3 mg). Compounds 10 (128.0 mg) and 25 (36.0 mg) were isolated from Fr.4.7 (1.6 g) through RP-HPLC (MeOH/H2O, 74:26).

3.4. Spectroscopic Data

Mulberofuran Z (1), Brown amorphous powder, [α]D20 + 3.0 (c 0.22, MeOH); UV (MeOH) λmax (log ε) 216 (4.21), 316 (4.19) nm; IR (KBr) νmax 3423, 1615, 1490, 1362, 1266, 1155, 1084 cm−1; HR-ESI-MS at m/z 431.1831 [M + Na]+ (calcd. for C25H28O5Na, 431.1829); 1H and 13C NMR data, see Table 1.
Artoxanthocarpuone C (11), Yellow powder, [α] D20 + 3.7 (c 0.22, MeOH); UV (MeOH) λmax (log ε) 216 (3.21), 327 (4.36) nm; IR (KBr) νmax 3415, 1656, 1621, 1554, 1448 cm−1; HRESIMS m/z 439.1751 [M + H]+ (calcd. for C25H27O7, 439.1751); 1H and 13C NMR data, see Table 2.
Kuwanon Z (12), Yellow amorphous powder, [α] D20 + 10.6 (c 0.22, MeOH); UV (MeOH), λmax (log ε) 215 (2.61), 336 (3.18) nm; IR (KBr) νmax 3372, 1645, 1616, 1440, 1299, 1167 cm−1; HRESIMS m/z 439.1751 [M + H]+ (calcd. for C25H27O7, 439.1749); 1H and 13C NMR data, see Table 2.

3.5. Cell Culture

HGC27 cells were purchased from American Type Culture Collection (ATCC). All cells grew adherently. HGC27 cells were cultured in RPMI 1640 media, and MRC-5 cells were cultured in a minimal essential medium (Gibco) (MA, USA). All media contained 10% endotoxin-free, heat-inactivated FBS (Gibco) and were maintained at 37 °C in a humidified atmosphere of 5% CO2.

3.6. Cytotoxicity Assay

All compounds were tested for cytotoxicity against HGC27 cell lines utilizing the CCK-8 method as previously reported [46]. HGC27 cells (4 × 103/well) were seeded in 96-well plates for CCK-8 assays after treated with compound (135) 10 µM for 24 h. Then, 10 μL of CCK-8 reagent (biofrox Z6789D144) was added into each well, and incubated for 4 h at 37 °C. Finally, the optical density (OD) was then measured at 450 nm. Cells were treated with varying concentrations of compounds 5, 8, 10, and 30 (1, 2.5, 5, 10, 20, 30, 40, and 60 µM) for 24 h. As in the above method, the IC50 values were calculated.

3.7. Colony Formation Assay

Colony formation assay was performed as described previously. Briefly, 500 cells per well were seeded in 12-well plates and then incubated for 7 d. The colonies were fixed in methanol for 20 min and stained with 2% crystal violet for 20 min. Then, the stain solution was removed using tap water, and the cells were air-dried. Finally, the number of colonies was counted.

3.8. Cell Apoptosis Assay

HGC27 cells were cultured in a 6-well plate at a density of 700,000 cells/well and incubated in a cell incubator for 24 h. When HGC27 cells were in the logarithmic growth phase, HGC27 cells were treated with 10 μM or 20 μM compound 10 for 48 h. Then, cells were collected by trypsin digestion without EDTA and washed twice with cold PBS. An amount of 5 μL Annexin V/Alexa Fluor 488 solution was added to the cell suspension and incubated in the dark at room temperature for 5 min. Then, 10 μL PI and 400 μL PBS solution were added. The stained cell suspension was transferred to a flow tube, and the proportion of apoptotic cells was detected using BD Facscanto II, (Piscataway, NJ, USA), and the experimental results were statistically analyzed using Flowjo.

3.9. Transwell Migration Assay

Before the experiment, the cells were starved by culturing in a serum-free medium for 4 h. Then, cells were collected, and 30,000 cells were cells were treated with 3 μM and 6 μM Compound 10 inoculated into 500 μL serum-free medium in the upper chamber of Transwell (Corning, NY, USA), and 500 μL culture medium with 10% FBS was added into the lower chamber. After 24 h of incubation, the culture medium was discarded. Cells were fixed with 4% paraformaldehyde (Beyotime, Shanghai, China) for 15 min, then stained with 0.4% crystal violet (Beyotime, China) for 15 min. The cells were observed and photographed under a light microscope.

3.10. Western Blot Analysis

HGC27 cells which were treated with concentrations 10 µM and 20 µM compound 10 were lysed by RIPA lysis buffer (Beyotime, Shanghai) for 20 min. After centrifugation at 13,000 rpm for 25 min at 4 °C, the sediment was removed, and the supernate was mixed with 5 × Loading Buffer boiled for 5 min at 100 °C. Next, equal amounts of proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to polyvinylidene fluoride (PVDF) membranes (Millipore, Burlington, MA, USA). After blocked with 5% non-fat milk for 2 h at room temperature (RT), the membranes were incubated with the specific primary antibodies (Signalway Antibody, Nanjing, China) overnight at 4 °C. After washing three times with Tris Buffered saline Tween 20 (TBST), the membranes were then incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (ProteinTech, Wuhan, China) for 1 h at RT. The immune-blotting signals were detected using Beyo ECL Plus electrochemiluminescence reagent (Beyotime, Shanghai, China). Next, equal amounts of proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to polyvinylidene fluoride (PVDF) membranes (Millipore, USA).

4. Conclusions

Phytochemical investigation of the 75% ethanol extract of the root bark of M. alba led to the isolation and identification of three new compounds (1, 11, and 12) and 32 known ones (210, 1335). All compounds were tested for their cytotoxic activities against gastric cancer HGC27 cells. As a result, compounds 5, 8, and 30 exhibited certain cytotoxic activities with the IC50 values ranging from 33.76 ± 2.64 μM, 28.94 ± 0.72 μM, and 10.24 ± 0.89 μM, respectively. Compound 10 showed the most significant cytotoxic activity with an IC50 value of 6.08 ± 0.34 μM. Then, compound 10 was subjected to further experiments to explore its anti-tumor mechanism. It was found that compound 10 has inhibitory effects on proliferation and migration of human cancer HGC27 cells. In addition, the experimental results indicated that compound 10 was able to induce apoptosis through upregulation of Bax protein expression and downregulation of Caspase-3 and Bcl-2 protein expression. This study was the first time to preliminarily explore the mechanism of compound 10 in inhibiting gastric cancer cell HGC27 in vitro. In conclusion, the present study provides a reference for the phytochemical and biological activities of Morus alba L.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules29010030/s1. Figures S1–S8, HR-ESI-MS data and NMR data of compound 1, Figures S9–S15, HR-ESI-MS data and NMR data of compound 11, Figures S16–S25, HR-ESI-MS data and NMR data of compound 12.

Author Contributions

Conceptualization, M.W., H.P. and L.W.; isolation and structural elucidation, H.P.; biological activity assays, D.C.; statistical analysis, H.P. and D.C.; resources, M.W.; writing—original draft preparation, H.P.; writing—review and editing, H.P., L.W., X.Z. and F.L.; supervision, M.W. and L.W.; project administration, M.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Sichuan Science and Technology Program (No. 2021ZHFP0031).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and supplementary materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. State Pharmacopoeia Committee. Chinese Pharmacopoeia; China Medical Pharmaceutical Science and Technology Publishing House: Beijing, China, 2010; p. 182. [Google Scholar]
  2. Asano, N.; Yamashita, T.; Yasuda, K.; Ikeda, K.; Kizu, H.; Kameda, Y.; Kato Nash, A.R.J.; Lee, H.S.; Ryu, K.S. Polyhydroxylated alkaloids isolated from mulberry trees (Morus alba L.) and silkworms (Bombyx mori L.). J. Agric. Food. Chem. 2001, 49, 4208–4213. [Google Scholar] [CrossRef] [PubMed]
  3. Čulenová, M.; Sychrová, A.; Hassan, S.T.S.; Berchová-Bímová, K.P.; Svobodová, P.; Helclová, A.; Michnová, H.; Hošek, J. Multiple In vitro biological effects of phenolic compounds from Morus alba root bark. J. Ethnopharmacol. 2020, 248, 112296. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, Q.J.; Li, D.Z.; Chen, R.Y.; Yu, D.Q. A new benzo-furanolignan and a new flavonol derivative from the stem of Morus australis. Chin. Chem. Lett. 2008, 19, 196–198. [Google Scholar] [CrossRef]
  5. Takahashi, M.; Takara, K.; Toyozato, T.; Wada, K. A novel bioactive chalcone of Morus australis inhibits tyrosinase activity and melanin biosynthesis in B16 melanoma cells. J. Oleo Sci. 2012, 61, 585–592. [Google Scholar] [CrossRef] [PubMed]
  6. Ko, H.H.; Wang, J.J.; Lin, H.C.; Wang, J.P.; Lin, C.N. Chemistry and biological activities of constituents from Morus australis. Biochim. Biophys. Acta Gen. Subj. 1999, 1428, 293–299. [Google Scholar] [CrossRef] [PubMed]
  7. Zheng, Z.F.; Zhang, Q.J.; Chen, R.Y.; Yu, D.Q. Four new flavonoids from Morus australis. J. Asian. Nat. Prod. Res. 2012, 143, 263–269. [Google Scholar] [CrossRef] [PubMed]
  8. Zhang, Q.J.; Tang, Y.B.; Chen, R.Y.; Yu, D.Q. Three new cytotoxic Diels-Alder-type adducts from Morus australis. Chem. Biodivers. 2007, 4, 1533–1540. [Google Scholar] [CrossRef] [PubMed]
  9. Zhang, Q.J.; Zheng, Z.F.; Chen, R.Y.; Yu, D.Q. Two new dimeric stilbenes from the stem bark of Morus australis. J. Asian Nat. Prod. Res. 2009, 11, 138–141. [Google Scholar] [CrossRef]
  10. Kikuchi, T.; Nihei, M.; Nagai, H.; Fukushi, H.; Tabata, K.; Suzuki, T.; Akihisa, T. Albanol A from the root bark of Morus alba L. induces apoptotic cell death in HL60 human leukemia cell line. Chem. Pharm. Bull. 2010, 58, 568–572. [Google Scholar] [CrossRef]
  11. Cui, L.; Lee, H.S.; Oh, W.K.; Ahn, J.S. Inhibition of sangginon G isolated from Morus alba on the metastasis of cancer cells. Chin. Herb. Med. 2011, 3, 23–26. [Google Scholar]
  12. Agabeyli, R.A. Antimutagenic activities extracts from leaves of the Morus alba, Morus nigra and their mixtures. Intl. J. Biol. 2012, 4, 166–172. [Google Scholar] [CrossRef]
  13. Devi, B.; Sharma, N.; Kumar, D.; Jeet, K. Morus alba L. inn: A phytopharmacological review. Int. J. Pharm. Pharma. Sci. 2013, 5, 14–18. [Google Scholar]
  14. Hong, S.S.; Hong, S.; Lee, H.J.; Mar, W.; Lee, D.A. A new prenylated flavanone from the root bark of Morus. B. Korean. Chem. Soc. 2013, 34, 2528–2530. [Google Scholar]
  15. Nomura, T.; Fukai, T.; Shimada, T.; Chen, I.S. Mulberrofuran D, a new isoprenoid 2-arylbenzofuran from the root barks of the mulberry tree (Morus australis Poir.). Heterocycies 1982, 19, 1855–1860. [Google Scholar] [CrossRef]
  16. Chang, Y.S.; Jin, H.G.; Lee, H.; Lee, D.S.; Woo, E.R. Phytochemical constituents of the root bark from Morus alba and their Il-6 Inhibitory activity. Nat. Prod. Sci. 2019, 25, 268–274. [Google Scholar] [CrossRef]
  17. Wu, D.L.; Zhang, X.Q.; Huang, X.J.; He, X.M.; Wang, G.C.; Ye, W.C. Chenucal constituents from root barks of Morus atropurpurea. J. Chin. Med. Mater. 2010, 35, 1978–1982. [Google Scholar]
  18. Jeong, S.H.; Ryu, Y.B.; Curtis-Long, M.J.; Ryu, H.W.; Baek, Y.S.; Kang, J.E.; Lee, W.S.; Park, K.H. Tyrosinase Inhibitory polyphenols from roots of Morus ihou. J. Agric. Food Chem. 2009, 57, 1195–1203. [Google Scholar] [CrossRef] [PubMed]
  19. Shi, Y.Q.; Fukai, T.; Sakagami, H.; Chang, W.J.; Yang, F.Q.; Wang, F.P.; Nomura, T. Cytotoxic flavonoids with isoprenoid groups from Morus mongolica. J. Nat. Prod. 2001, 64, 181–188. [Google Scholar] [CrossRef]
  20. He, X.; Chao, X.; Yang, L.; Zhang, C.; Pi, R.; Zeng, H.; Li, G.; Xu, Y.; Lin, Y. The research on chemical ingredients of the heartwood of root of Morus atpropurpurea. Nat. Prod. Res. Dev. 2014, 26, 193–196. [Google Scholar]
  21. Jung, J.W.; Koo, W.M.; Park, J.H.; Seo, K.H.; Oh, E.J.; Lee, D.Y.; Lee, D.S.; Kim, Y.C.; Lim, D.W.; Han, D.; et al. Isoprenylated flavonoids from the root bark of Morus alba and their hepatoprotective and neuroprotective activities. Arch. Pharm. Res. 2015, 38, 2066–2075. [Google Scholar] [CrossRef]
  22. Cui, X.Q.; Chen, H.; Chen, R.Y. Study on Diels-Alder type adducts from stem bark of Morus yunanensis. J. Chin. Med. Mater. 2009, 34, 286–290. [Google Scholar]
  23. Cui, X.Q.; Wang, H.Q.; Liu, C.; Chen, R.Y. Study on anti-oxidant phenolic compounds from stem bark of Morus yunanensis. J. Chin. Med. Mater. 2008, 13, 1569–1572. [Google Scholar]
  24. He, X.M.; Wu, D.L.; Zou, Y.X.; Wang, G.C.; Zhang, X.Q.; Liao, S.T.; Sun, J.; Ye, W.C. Chemical constituents from root barks of Morus atropurpurea. JMFST 2014, 30, 219–228. [Google Scholar]
  25. Zhen, P.; Ni, G.; Guo, W.Q.; Shi, G.R.; Chen, R.Y.; Yu, D.Q. Isolation and identification of pharmaceutical chemical constituents from branches of Morus notabilis. Sci. Seric. 2016, 42, 0307–0312. [Google Scholar]
  26. Guo, Y.Q.; Tang, G.H.; Lou, L.L.; Li, W.; Zhang, B.; Liu, B.; Yin, S. Prenylated flavonoids as potent phosphodiesterase-4 inhibitors from Morus alba: Isolation, modification, and structure-activity relationship study. Eur. J. Med. Chem. 2018, 144, 758–766. [Google Scholar] [CrossRef] [PubMed]
  27. Kim, J.Y.; Lee, W.S.; Kim, Y.S.; Marcus, J.C.L.; Lee, B.W.; Ryu, Y.B.; Park, K.H. Isolation of cholinesterase-Inhibiting flavonoids from Morus lhou. J. Agric. Food. Chem. 2011, 59, 4589–4596. [Google Scholar] [CrossRef] [PubMed]
  28. Geng, C.A.; Yao, S.Y.; Xue, D.Q.; Zuo, A.; Zhang, X.M.; Jiang, Z.Y.; Ma, Y.B.; Chen, J.J. New isoprenylated flavonoid from Morus alba. J. Chin. Med. Mater. 2010, 35, 1560–1565. [Google Scholar]
  29. Tseng, T.H.; Chuang, S.K.; Hu, C.C.; Chang, C.F.; Huang, Y.C.; Lin, C.W.; Lee, Y.J. The synthesis of morusin as a potent antitumor agent. Tetrahedron 2010, 66, 1335–1340. [Google Scholar] [CrossRef]
  30. Fujimoto, T.; Hano, Y.; Nomura, T.; Uzawa, J. Components of root bark of cudrania tricuspidata 2. Structures of two new isoprenylated flavones, Cudraflavones A and B. Planta Med. 1984, 50, 161–163. [Google Scholar] [CrossRef]
  31. Guo, S.; Liu, L.; Zhang, S.S.; Yang, C.; Yue, W.P.; Zhao, H.A.; Ho, C.T.; Du, J.F.; Zhang, H.; Bai, N.S. Chemical characterization of the main bioactive polyphenols from the roots of Morus australis (mulberry). Food Funct. 2019, 10, 6915–6926. [Google Scholar] [CrossRef]
  32. Wang, L.; Yang, Y.; Liu, C.; Chen, R.Y. Three new compounds from Morus nigra L. J. Asian Nat. Prod. Res. 2010, 12, 431–437. [Google Scholar] [CrossRef] [PubMed]
  33. Zheng, Z.P.; Tan, H.Y.; Wang, M.F. Tyrosinase inhibition constituents from the roots of Morus australis. Fitoterapia 2012, 83, 1008–1013. [Google Scholar] [CrossRef] [PubMed]
  34. Qin, J.; Fan, M.; He, J.; Wu, X.D.; Peng, L.Y.; Su, J.; Cheng, X.; Li, Y.; Kong, L.M.; Li, R.T.; et al. New cytotoxic and anti-inflammatory compounds isolated from Morus alba L. Nat. Prod. Res. 2015, 29, 1711–1718. [Google Scholar] [CrossRef] [PubMed]
  35. Patil, A.D.; Freyer, A.J.; Killmer, L.; Offen, P.; Taylor, P.B.; Votta, B.J.; Johnson, R.K. A new dimeric dihydrochalcone and a new prenylated flavone from the bud covers of Artocarpus altilis: Potent inhibitors of cathepsin K. J. Nat. Prod. 2002, 65, 624–627. [Google Scholar] [CrossRef]
  36. Yang, D.S.; Li, Z.L.; Yang, Y.P.; Xiao, W.L.; Li, X.L. New geranylated 2-Arylbenzofuran from Morus alba. Chin. Herb. Med. 2015, 7, 191–194. [Google Scholar] [CrossRef]
  37. Zuo, G.Y.; Yang, C.X.; Ruan, Z.J.; Han, J.; Wang, G.C. Potent anti-MRSA activity and synergism with aminoglycosides by flavonoid derivatives from the root barks of Morus alba, a traditional chinese medicine. Med. Chem. Res. 2019, 28, 1547–1556. [Google Scholar] [CrossRef]
  38. Nomura, T.; Fukai, T.; Sato, E.; Fukushima, K. The formation of moracenin-D from kuwanon-G. Heterocycles 1981, 16, 983–986. [Google Scholar] [CrossRef]
  39. Hano, Y.; Yamanaka, J.; Momose, Y.; Nomura, T. Sorocenols C-F, four new isoprenylated phenols from the root bark of Sorocea bonplandii Baillon. Heterocycles 1995, 41, 2811–2821. [Google Scholar]
  40. Liu, Y.J.; Wu, J.C.; Li, H.L.; Ma, Q.; Chen, Y.G. Alkaloid and flavonoids from the seeds of Whitfordiodendron filipes. Chem. Nat. Compd. 2016, 52, 188–190. [Google Scholar] [CrossRef]
  41. Zhang, Y.L.; Luo, J.G.; Wan, C.X.; Zhou, Z.B.; Kong, L.Y. Geranylated 2-arylbenzofurans from Morus alba var. tatarica and their α-glucosidase and protein tyrosine phosphatase 1B inhibitory activities. Fitoterapia 2014, 92, 116–126. [Google Scholar] [CrossRef]
  42. Jin, Y.J.; Lin, C.C.; Lu, T.M.; Li, J.H.; Chen, I.S.; Kuo, Y.H.; Ko, H.H. Chemical constituents derived from Artocarpus xanthocarpus as inhibitors of melanin biosynthesis. Phytochemistry 2015, 117, 424–435. [Google Scholar] [CrossRef]
  43. Tuan Hiep, N.Y.; Kwon, J.Y.; Hong, S.G.; Kim, N.Y.; Guo, Y.Q.; Hwang, B.Y.; Mar, W.C.; Lee, D.H. Enantiomeric isoflavones with neuroprotective activities from the fruits of Maclura tricuspidata. Sci. Rep. 2019, 9, 1757. [Google Scholar] [CrossRef]
  44. Zhang, X.R.; Wang, S.Y.; Sun, W.; Wei, C. Isoliquiritigenin inhibits proliferation and metastasis of MKN28 gastric cancer cells by suppressing the PI3K/AKT/mTOR signaling pathway. Mol. Med. Rep. 2018, 18, 3429–3436. [Google Scholar] [CrossRef]
  45. Dai, L.; Wang, G.; Pan, W. Andrographolide inhibits proliferation and metastasis of SGC7901 gastric cancer cells. Biomed. Res. Int. 2017, 10, 6242103. [Google Scholar] [CrossRef]
  46. Gao, Z.; Deng, G.; Li, Y.; Huang, H.; Sun, X.; Shi, H.; Luo, M. Actinidia chinensis planch prevents proliferation and migration of gastric cancer associated with apoptosis, ferroptosis activation and mesenchymal phenotype suppression. Biomed. Pharmacother. 2020, 126, 110092. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of compounds 135.
Figure 1. Chemical structures of compounds 135.
Molecules 29 00030 g001
Figure 2. The 1H-1H COSY and key HMBC correlations of compounds 1, 11, and 12.
Figure 2. The 1H-1H COSY and key HMBC correlations of compounds 1, 11, and 12.
Molecules 29 00030 g002
Figure 3. Cells apoptosis and migration assay. (a) Visual observation of cell clonogenicity. (b) Statistical analysis of cell clonogenicity. Data are mean ± SD (n = 3). Differences between treatment groups and control groups are determined, * p < 0.05, ** p < 0.01 as compared with control. (c) Apoptosis caused by 10 in HGC27 cells. (d) Treatment groups cells apoptosis rates were compared with control, * p < 0.05. Data are expressed as mean ± SD (n = 3). (e) The migration of HGC27 cells. (f) Quantitative evaluations of cell migration induced by 10 in the transwell assay, treatment groups were compared with control, * p < 0.05, ** p < 0.01. Data are expressed as mean ± SD (n = 3). Note: control: blank control group.
Figure 3. Cells apoptosis and migration assay. (a) Visual observation of cell clonogenicity. (b) Statistical analysis of cell clonogenicity. Data are mean ± SD (n = 3). Differences between treatment groups and control groups are determined, * p < 0.05, ** p < 0.01 as compared with control. (c) Apoptosis caused by 10 in HGC27 cells. (d) Treatment groups cells apoptosis rates were compared with control, * p < 0.05. Data are expressed as mean ± SD (n = 3). (e) The migration of HGC27 cells. (f) Quantitative evaluations of cell migration induced by 10 in the transwell assay, treatment groups were compared with control, * p < 0.05, ** p < 0.01. Data are expressed as mean ± SD (n = 3). Note: control: blank control group.
Molecules 29 00030 g003
Figure 4. Compound 10 influences the expression of relevant proteins. Detection of Caspase-3, Bcl-2 and Bax proteins expression level in HGC27cells by Western Blot; the treatment groups were compared with control, * p < 0.05, ** p < 0.01. Data were expressed as mean ± SD (n = 3). Note: control: blank control group.
Figure 4. Compound 10 influences the expression of relevant proteins. Detection of Caspase-3, Bcl-2 and Bax proteins expression level in HGC27cells by Western Blot; the treatment groups were compared with control, * p < 0.05, ** p < 0.01. Data were expressed as mean ± SD (n = 3). Note: control: blank control group.
Molecules 29 00030 g004
Table 1. 1H NMR (400 MHz) and 13C NMR (100 MHz) data of 1 in CD3OD (δ in ppm, J in Hz).
Table 1. 1H NMR (400 MHz) and 13C NMR (100 MHz) data of 1 in CD3OD (δ in ppm, J in Hz).
No.δH (J in Hz)δCNo.δH (J in Hz)δC
2 156.71″3.62 d (7.2)23.6
36.92 s102.32″5.40 t (7.2)123.6
3a 124.23″ 135.9
47.32 d (8.4)119.14″1.89 s16.4
56.90 d (8.8)109.35″2.02 m36.7
6 156.46″1.60 m34.2
7 114.47″3.90 t (6.8)76.1
7a 155.38″ 148.6
1′ 133.89″α4.79 s111.5
2′6.80 * d (2.0)104.19″β4.72 s
3′ 160.010″1.62 s17.5
4′6.26 s103.6-OCH33.86 s57.0
5′ 160.0
6′6.80 * d (2.0)104.1
(*) refers to overlapped signals.
Table 2. 1H NMR and 13C NMR data for the compounds 11 and 12 (δ in ppm, J in Hz).
Table 2. 1H NMR and 13C NMR data for the compounds 11 and 12 (δ in ppm, J in Hz).
11 a 12 b
No.δH (J in Hz)δCδH (J in Hz)δC
2 158.6 163.1
3 115.7 122.1
4 179.7 183.1
4a 102.0 105.4
5 157.3 163.2
66.12 brs98.95.91 a s99.7 a
7 161.3 165.8
86.33 brs93.66.00 a s94.0 a
8a 157.3 157.1
2.74 dd (16.2, 6.0)23.02.61 dd (16.8, 5.4)27.5
3.00 dd (16.2, 3.6) 2.95 dd (16.8, 7.4)
103.99 dd (6.0, 3.6)91.53.80 dd (16.2, 6.0)70.2
11 70.9 78.1
121.18 s27.31.36 s25.8
131.29 s24.11.28 s20.8
1′ 114.0 113.8
2′ 159.4 154.7
3′ 119.9 110.3
4′ 156.4 157.0
5′6.74 d (9.0)110.76.42 d (8.4)109.9
6′7.67 d (9.0)126.86.98 d (8.4)129.5
1″3.36 *d (6.0)22.53.08 dd (7.2, 5.4)24.8
2″5.13 t (7.2)123.25.07 t (7.2)122.6
3′ 130.3 132.8
4″1.61 s25.51.34 s17.6
5″1.71.s17.91.58 s25.8
5-OH13.02 s
a recorded at 600 (150) MHz in DMSO. b recorded at 600 (150) MHz in CD3OD. (*) refers to overlapped signals.
Table 3. Cell viability of compounds 135 on HGC27 cells.
Table 3. Cell viability of compounds 135 on HGC27 cells.
CompoundCell Viability (%)CompoundCell Viability (%)
196.42 ± 4.571995.67 ± 3.65
297.88 ± 2.262093.72 ± 2.86
396.27 ± 2.372198.96 ± 0.92
492.08 ± 4.252296.86 ± 1.84
567.31 ± 2.562383.99 ± 3.92
689.04 ± 3.062494.57 ± 2.74
789.47 ± 2.042588.99 ± 4.62
859.92 ± 2.162695.22 ± 3.30
996.62 ± 0.542792.97 ± 2.72
1039.71 ± 3.2728102.55 ± 3.06
1196.27 ± 4.142998.20 ± 0.37
1274.89 ± 1.583046.84 ± 3.02
1377.66 ± 6.403198.51 ± 0.97
1495.05 ± 2.783283.55 ± 1.51
1597.90 ± 1.743388.55 ± 3.20
1697.01 ± 1.833498.26 ± 1.03
1798.36 ± 1.653590.74 ± 5.24
1895.05 ± 2.30a control100.00 ± 1.31
a control: blank control group. concentration: 10 µM.
Table 4. Inhibitory effects of compounds 5, 8, 10, and 30 on HGC27 cells (mean ± SD, n = 3).
Table 4. Inhibitory effects of compounds 5, 8, 10, and 30 on HGC27 cells (mean ± SD, n = 3).
CompoundIC50 (µM)
533.76 ± 2.64
828.94 ± 0.72
106.08 ± 0.34
3010.24 ± 0.89
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pu, H.; Cao, D.; Zhou, X.; Li, F.; Wang, L.; Wang, M. Isoprenylated Flavonoids and 2-Arylbenzofurans from the Root Bark of Morus alba L. and Their Cytotoxic Activity against HGC27 Cancer Cells. Molecules 2024, 29, 30. https://doi.org/10.3390/molecules29010030

AMA Style

Pu H, Cao D, Zhou X, Li F, Wang L, Wang M. Isoprenylated Flavonoids and 2-Arylbenzofurans from the Root Bark of Morus alba L. and Their Cytotoxic Activity against HGC27 Cancer Cells. Molecules. 2024; 29(1):30. https://doi.org/10.3390/molecules29010030

Chicago/Turabian Style

Pu, Hangyi, Dongyi Cao, Xue Zhou, Fu Li, Lun Wang, and Mingkui Wang. 2024. "Isoprenylated Flavonoids and 2-Arylbenzofurans from the Root Bark of Morus alba L. and Their Cytotoxic Activity against HGC27 Cancer Cells" Molecules 29, no. 1: 30. https://doi.org/10.3390/molecules29010030

Article Metrics

Back to TopTop