Next Article in Journal
Effect of Fiber–Matrix Interface Friction on Compressive Strength of High-Modulus Carbon Composites
Next Article in Special Issue
The Isolation and Structure Elucidation of Spirotetronate Lobophorins A, B, and H8 from Streptomyces sp. CB09030 and Their Biosynthetic Gene Cluster
Previous Article in Journal
Phytotoxicity of Bisphenol A to Allium cepa Root Cells Is Mediated through Growth Hormone Gibberellic Acid and Reactive Oxygen Species
Previous Article in Special Issue
Berbamine Hydrochloride Inhibits African Swine Fever Virus Infection In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Flavonoids from the Roots of Sophora flavescens and Their Potential Anti-Inflammatory and Antiproliferative Activities

Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 19 January 2023 / Revised: 21 February 2023 / Accepted: 21 February 2023 / Published: 22 February 2023

Abstract

:
The phytochemical investigation of the roots of the traditional Chinese medicinal plant Sophora flavescens led to the isolation of two novel prenylflavonoids with an unusual cyclohexyl substituent instead of the common aromatic ring B, named 4′,4′-dimethoxy-sophvein (17) and sophvein-4′-one (18), and 34 known compounds (1–16, 19–36). The structures of these chemical compounds were determined by spectroscopic techniques, including 1D-, 2D-NMR, and HRESIMS data. Furthermore, evaluations of nitric oxide (NO) production inhibitory activity against lipopolysaccharide (LPS)-treated RAW264.7 cells indicated that some compounds exhibited obvious inhibition effects, with IC50 ranged from 4.6 ± 1.1 to 14.4 ± 0.4 μM. Moreover, additional research demonstrated that some compounds inhibited the growth of HepG2 cells, with an IC50 ranging from 0.46 ± 0.1 to 48.6 ± 0.8 μM. These results suggest that flavonoid derivatives from the roots of S. flavescens can be used as a latent source of antiproliferative or anti-inflammatory agents.

1. Introduction

The roots of Sophora flavescens are ordinarily served in the traditional Chinese medicine (TCM), “Ku Shen”, for the curing of skin diseases, cancer, dysentery, hematochezia, jaundice, pruritus vulvae, eczema, and hepatitis [1]. Modern pharmacological research shows that it exhibits outstanding activities toward tumors, inflammation, diabetes, and microbial infections [2,3,4,5,6,7]. Phytochemical studies demonstrated that alkaloids and flavonoids are the major chemical classes of S. flavescens compounds [8,9,10]. Matrine, as a reflective component of the alkaloids of S. flavescens, has been used as an antitumor drug (Compound matrine injection) in China. With more in-depth research on the alkaloids of S. flavescens, the anticancer effects of some alkaloids have been shown to be more potent than those of flavonoids of S. flavescens in vitro and in vivo [11].
In order to discover new compounds with antiproliferative and anti-inflammatory activities from S. flavescens, we isolated 36 flavonoids from this medicinal plant (Figure 1), including flavanones, isoflavones, flavonols, flavanonols, and chalcones. Among them, compounds 17 and 18 were two new dihydroflavones with an unusual cyclohexyl substituent instead of the common aromatic ring B. Many studies have shown an interesting link between chronic inflammation and cancers [12]. Thus, all isolated components were evaluated for their anti-inflammatory activity by acting on LPS-stimulated macrophage (RAW 264.7) cell lines in vitro. In addition, all components were assessed for their antiproliferative activities against HepG2 cell lines.
Here, we characterize the isolation, chemical structure elucidation, antiproliferative activity against HepG2 cells, and NO production inhibitory activity of all isolates.

2. Results and Discussion

The comprehensive use of separation materials and chromatographic methods such as normal silica gel, hydroxypropyl dextran gel (Sephadex LH-20), MPLC, TLC, MCI reversed-phase column, and HPLC, with the help of MS and NMR and other spectroscopy methods, was carried out to isolate and identify 36 compounds, including 2 new ones: 4′,4′-dimethoxy-sophvein (17) and sophvein-4′-one (18).

2.1. Structural Elucidation

Compound 17 was acquired as a white amorphous powder. The HR-ESI-MS demonstrated the molecular ion peak at m/z 429.1894 [M+Na]+ (calcd. for 429.1884), corresponding to the molecular formula C22H30O7, which indicated eight degrees of unsaturation. The 1H spectroscopic data of 17 (Table 1) demonstrated the existence of one aromatic proton signal (δH 5.84 (1H, s)), one isopentenyl side chain signal (δH 1.68 (3H, s); 1.59 (3H, s); 3.14 (2H, d, J = 6.9 Hz); 5.11 (1H, br t, J = 6.9 Hz)), and two methoxy groups signals (δH 3.15 (3H, s) and 3.10 (3H, s)). By interpreting the DEPT and 13C NMR spectra, 22 carbon signals (Table 1) were observed, including two methoxy groups (δC 47.9 and 48.1), two methyls (δC18.1 (C-5″) and 25.9 (C-4″)), three methylidynes (δC 84.6 (C-2); 96.4 (C-6); 124.4 (C-2″)), six methylenes (δC 22.4 (C-1″); 28.4 (C-3′); 28.5 (C-5′); 29.9 (C-2′); 31.3 (C-6′); 37.0 (C-3)), three oxygenated aromatic carbons (δC161.3 (C-9); 163.0 (C-5); 166.0 (C-7)), and six quaternary carbons (δC 72.2(C-1′); 101.1(C-4′); 103.2 (C-10); 109.0 (C-8); 131.6 (C-3″); 198.8 (C-4)). According to the above NMR data, it is speculated that this compound may be an isopentenyl substituted flavanone. However, from the 1H NMR spectra of compound 17, the proton signal of the typical aromatic flavonoid B-ring in the flavonoid compound completely disappeared, and a complex signal of eight aliphatic protons emerged instead.
These data suggest the presence of a hydrogenated B-ring unit in the compound, and this inference is also confirmed by the HMQC and HMBC spectra (Figure 2). Comparing the 13C NMR data, except for the isopentenyl signal, the remaining 17 signals are very similar to (2S)-4′,4′-dimethoxy-ongokein [13]. The main difference between the two compounds is that the H-8 (δH 5.98 (d, J = 2.0 Hz)) signal in (2S)-4′,4′-dimethoxy-ongokein disappears, and the chemical shift of C-8 also decreases to the lower field. These data indicate that the isopentenyl group is attached at the C-8 position. This inference was identified via the HMBC correlation between H-1″ (δH 3.14, 2H, d, J = 6.9 Hz) and C-9 (δC 161.3), C-8 (δC 109.0), and C-7 (δC 166.0). On the other hand, the attachment of the two methoxy groups to the C-4′-position was shown by the HMBC correlation peaks between two methoxy signals (δH 3.15 and 3.10) and C-4′ (δC 101.1).
By comparison, the coupling constants of H-2 (δH 4.04 (d, J = 13.6 Hz)), H-3a (δH 2.81 (dd, J = 17.0, 13.6 Hz)), and H-3b (δH 2.51 (d, J = 17.0 Hz)) were found to be very close to (2S)-4′,4′-dimethoxy-ongokein [13], so it is inferred that the C-2 of compound 17 is in the S configuration. Additionally, the configuration of the B-ring of cyclohexane was defined to be the 1′C4′ configuration via the NOE effect in the ROESY experiment (Figure 2). According to the above analysis, the structure of compound 17 was confirmed and named 4′,4′-dimethoxy-sophvein.
Compound 18 was acquired as a white amorphous powder. The HR-ESI-MS displayed the molecular ion peak at m/z 361.1650 [M+H]+ (calcd for 361.1646), suggesting the molecular formula C20H24O6 with 9° of unsaturation. The 1H and 13C NMR spectroscopic data of 18 are similar to 17 (Table 1), with the main difference being the disappearance of the two methoxyl signals in 18. Furthermore, the chemical shift of C-4′ exhibited a downfield shift (from δC 101.1 to 210.3). Combined with HR-ESI-MS, it was found that the molecular weight of compound 18 is 46 less than that of 17. The above data indicate that the C-4′ of the B-ring of compound 18 has a carbonyl group. This inference was further defined by the HMBC correlation of the δH 2.03 (H-3′b), 2.21 (H-2′), and 2.10 (H-6′) proton signals with C-4′ (δC 210.3). Finally, by comparing the NOE effects in the ROESY spectra of the two compounds, it was found that 18 and 17 have the same configuration. The optical rotation values of the two compounds [ α D 24 = + 49.88 (c 0.25, MeOH) for 17 and α D 24 = +38.69 (c 0.29, MeOH)] for 18 also support this inference. Based on the above data analysis, the structure of 18 was confirmed and named sophvein-4′-one. See Supplementary Materials for HR-ESI-MS, 1H and 13C NMR, HMQC, HMBC, 1H-1H COSY, and ROESY of compounds 17 and 18.
The known compounds (116 and 1936) were determined based on a comparison with published NMR data in the references to be sophorafiavanone B (1) [14], isoxanthohumol (2) [15], kenusanonoe I (3) [16], kushenol S (4) [17], leachianone G (5) [18], 5-methoxy-7,2′,4′-trihydroxy-8-[3,3-dimethyl-allyl]flavanone (6) [19], kushenol W (7) [17], 8-(3-Hydroxymethyl-2-butenyl)-5,7,2′,4′-tetra-hydroxyflavanone (8) [20], kushenol V (9) [17], alopecurone G (10) [21], sophoraflavone G (11) [22], leachianone A (12) [23], kurarinone (13) [24], (2S)-2′-methoxykurarinone (14) [25], kushenol E (15) [26], kushenol B (16) [26], noranhyoicaritin (19) [27], sophoflavescenol (20) [28], 8-(3,3-dimethylallyl)-tamarixetin (21) [29], 8-lavandulylkaempferol (22) [30], kushenol Z (23) [31], kushenol C (24) [32], 5-O-methylkushenol C (25) [33], (2R)-3β,7,4′-trihydrox-y-5-methoxy-8-prenyl- flavanone (26) [20], kushenol X (27) [23], kushenol N (28) [34], kushenol I (29) [35], 5,7,4′-trihydroxyisoflavone (30) [36], 7,3′-dihydroxy-4′-methoxyisoflavanone (31) [37], calycosin (32) [36], xanthohumol (33) [38], xanthogalenol (34) [39], kuraridin (35) [34], and kushenol D (36) [40].

2.2. Biological Studies

The potential cytotoxicity of compounds on RAW 264.7 macrophages cells was determined before executing further studies. Macrophage cells were treated with compounds, and a mitochondria colorimetric (MTT) assay was used to test cell survival. Cell viability (%) = [(O.DDrug − O.DBlank)/(O.DControl − O.DBlank)] ×100%, and SPSS version 21.0 (probit analysis) was used for calculating CC50 extract. Table 2 shows that some compounds exhibited obvious cytotoxicity on macrophages. Among them, compound 22 was the most toxic composition with a CC50 value of 13.8 ± 0.6 μM.
NO is one of the immune effectors used by macrophages to defend our bodies against intracellular pathogens. To determine if compounds 136 can modulate the NO production by macrophages, the Griess reagent was used to analyze the NO levels. The cells were incubated with compounds 136 (3.12, 6.25, 12.5, 25, and 50 μM) and then treated with LPS for 24 h. When activated by LPS, NO was produced from macrophages after inducing iNOS genes and subsequent protein expression. NO production inhibition (%) = [(O.DLPS − O.DDrug)/(O.DLPS − O.DBlank)] × 100%. The determination of IC50 was calculated using probit analysis in SPSS version 21.0. The research results show that some compounds exhibited obvious NO production inhibitory activity, with IC50 ranging from 4.6 ± 1.1 to 14.4 ± 0.4 μM, as shown in Table 2. Among them, compound 35 displayed the most significant inhibition of NO production, with an IC50 value of 4.6 ± 1.1 μM.
The inhibitory rate of HepG2 cells growth of compounds 136 was measured by the MTT assay and compared with a negative control and positive control (cisplatin, IC50 = 24.5 ± 0.8 μM). Inhibitory rate (%) = [(O.DDrug − O.DBlank)/(O.DControl − O.DBlank)] ×100%. The IC50 values were calculated with probit analysis using software SPSS version 21.0. The results show that eight compounds inhibited the growth of HepG2 cells with an IC50 ranging from 0.46 ± 0.1 to 48.6 ± 0.8 μM (Table 3), while others were inactive (IC50 > 50 μM). Compound 22 exhibited the best antiproliferative effects on HepG2 cell lines, with an IC50 value of 0.46 ± 0.1 μM. The present work revealed that many secondary metabolites of S. flavescens have antiproliferative and NO production inhibitory activities, indicating its application in traditional Chinese medicine.

3. Materials and Methods

3.1. General Experimental Procedure

The optical rotation value was recorded with a Jasco DIP-370 polarimeter (JASCO Corporation, Tokyo, Japan). The ultraviolet (UV) spectrum was recorded by a UV2700 spectrophotometer (Shimadzu, Kyoto, Japan). The infrared (IR) spectrum was obtained by an FT-IR spectrophotometer (PerkinElmer, Waltham, MA, USA) using KBr pellets. High-resolution electrospray ionization mass spectroscopy (HRESIMS) was obtained with an Agilent 6500 LC/Q-TOF mass spectrometer (Agilent, Waldbronn, Germany). The 1H NMR, 13C NMR, HMBC, HSQC, 1H-1H COSY, and ROESY spectra were measured by a Bruker Advance III HD using tetramethylsilane (TMS) as an internal standard (600 MHz, Bruker BioSpin, Zürich, Switzerland). Chemical shifts are demonstrated in δ (ppm) and relative to the residual solvent signals. Silica gel (100–200 and 200–300 mesh, Qingdao Marine Chemical Industry Co., Qingdao, China), polyamide (60–90 mesh, Changfeng Chemical Factory Co., Gulou, Nanjing, China), sephadex LH-20 (Sigma-Aldrich Corp., St. Louis, MO, USA), and RP-18 reverse-phase silica gel (20–45 µm, Fuji Silysia Chemical Ltd., Kasugai-shi, Japan) have been used for column chromatography (CC). The reverse-phase medium-pressure liquid chromatography (RP-MPLC) system comprises a Büchi pump (Büchi Labortechnik AG, Meierseggstrasse 40 Postfach CH-9230, Flawil, Switzerland), column and precolumn (310 × 36 mm and 110 × 36 mm, Soochow high tech chromatography CO., Ltd., Suzhou, China), and MCI-gel CHP-20P (75–150 µm, Mitsubishi Chemical Co., Tokyo, Japan). Thin-layer chromatography (TLC) was performed using precoated silica gel plates (GF254, Qingdao Marine Chemical Factory, Qingdao, China), and the spots were visualized by spraying with 10% sulfuric acid ethanolic solution or α-naphthol-sulfuric acid solution and heating at 110 °C for 3–5 min. Murine macrophage (RAW 264.7) cells were acquired from Kunming Institute of Zoology (KIZ), Chinese Academy of Sciences (CAS). Macrophage cells were placed in a constant temperature incubator and cultured at a temperature of 37 °C and a concentration of 5% CO2. Human hepatoma HepG2 cell lines were also obtained from the Kunming Institute of Zoology (KIZ), Chinese Academy of Sciences (CAS). HepG2 cells were cultured at 37 °C under 5% CO2. Additionally, an inverted phase contrast microscope was used to observe cell morphology.

3.2. Plant Material

The roots of S. flavescens were collected in Honghe, Yunnan Province, People’s Republic of China, in June 2019. The sample was identified by one of the authors (Xuan-Qin Chen). A voucher specimen (number: KUMST20190628) was conserved at the Key Laboratory of Phytochemistry, Kunming University of Science and Technology, China.

3.3. Extraction and Isolation

The air-dried roots of S. flavescens (20 Kg) were crushed and extracted with 95% aq. ethanol (24 h × 3 times). The ethanol extracts were percolated and evaporated in vacuo to obtain a residue. This residue was suspended in water and then extracted with ethyl acetate (3 times) and concentrated to produce an ethyl acetate phase (493 g). The ethyl acetate phase was segmented and enriched by macroporous resin to obtain 308 g of total flavonoids. The total flavonoid extract (300 g) was segmented using a normal phase silica gel column chromatography (CC) and stepwise gradient elution with a gradient of petroleum ether-ethyl acetate (4:1–1:1) to obtain 7 subfractions (Fr.1–7).
Fr.1 (3.4 g) was further segmented by a silica gel chromatography column (CC) stepwise gradient elution with petroleum ether-dichloromethane (1:2) to yield two subfractions (F1-1~F1-2). F1-1 (1.9 g) was further segmented via Sephadex LH-20 (methanol) to obtain 8 subfractions (F1-1-1~F1-1-8). F1-1-5 (120.6 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (150:1) to obtain compound 3 (14.0 mg). F1-2 (567.0 mg) was further segmented via Sephadex LH-20 (methanol) to obtain 8 subfractions (F1-2-1~F1-2-8). F1-2-5 (59.0 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-ethyl acetate (9:1–2:1) to obtain compounds 1 (141.4 mg) and 4 (33.2 mg). F1-2-8 (80.0 mg) was segmented by CC (silica gel) and stepwise gradient elution with petroleum ether-ethyl acetate (10:1–5:1) to yield compound 22 (30.0 mg).
Fr.2 (1.5 g) was further segmented via Sephadex LH-20 (methanol) to obtain 6 subfractions (F2-1~F2-6). F2-4 (366.0 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (150:1) to obtain 5 subfractions (F2-4-1~F2-4-5). F2-4-4 (28.0 mg) was further segmented via Sephadex LH-20 (methanol) to obtain compounds 15 (8.3 mg) and 16 (7.5 mg). F2-4-5 (15.0 mg) was further segmented via Sephadex LH-20 (methanol) to obtain compound 11 (6.0 mg). F2-6 (246.0 mg) was further segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (100:1–50:1) to obtain 5 subfractions (F2-6-1~F2-6-5). F2-6-3 (32.5 mg) was further segmented via Sephadex LH-20 (methanol) to yield compound 19 (24.3 mg). F2-6-5 (508.6 mg) was further segmented via Sephadex LH-20 (methanol) to obtain compound 21 (468.8 mg).
Fr.3 (30.0 g) was further segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (50:1–10:1) to obtain 2 subfractions (F3-1~F3-2). F3-1 (4.0 g) was further segmented via Sephadex LH-20 (methanol) to yield 7 subfractions (F3-1-1~F3-1-7). F3-1-3 (501.6 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (50:1–20:1) to obtain 4 subfractions (F3-1-3-1~F3-1-3-4). F3-1-3-2 (61.2 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-ethyl acetate (30:1–10:1) to obtain compound 10 (5.6 mg). F3-1-4 (1.1 g) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (50:1–10:1) to obtain 6 subfractions (F3-1-4-1~F3-1-4-6). F3-1-4-5 (642.5 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-ethyl acetate (30:1–10:1) to obtain 3 subfractions (F3-1-4-5-1~F3-1-4-5-3). F3-1-5 (2.3 g) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-ethyl acetate (50:1–30:1) to obtain 6 subfractions (F3-1-5-1~F3-1-5-6). F3-1-5-1 (400.4 mg) was further segmented via Sephadex LH-20 (methanol) to obtain 2 subfractions (F3-1-5-1-1~F3-1-5-1-2). F3-1-5-1-1 (119.2 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-acetone (4:1–1:1) to obtain compound 34 (27.8 mg). F3-1-5-1-2 (70.3 mg), followed by semipreparative high-performance liquid chromatography (HPLC) (70%, MeOH-H2O, 3 mL/min), was used to obtain compound 31 (4.1 mg). F3-2 (25.0 g) was first subjected to reverse-phase RP-18 chromatography with 40–100% methanol-water as the eluent, and the same fractions were combined by thin-layer chromatography to obtain 8 subfractions (F3-2-1~F3-2-8). F3-2-5 (276.5 mg) was further segmented via Sephadex LH-20 (methanol) to yield compound 30 (140.2 mg).
Fr.4 (11.0 g) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (60:1–10:1) to obtain 6 subfractions(F4-1~F4-6). F4-3 (900.7 mg) was further segmented via Sephadex LH-20 (methanol) to obtain 5 subfractions (F4-3-1-F4-3-5). F4-3-4 (163.3 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-ethyl acetate (20:1–5:1) to obtain compound 36 (68.2 mg). F4-3-5 (78.5 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-ethyl acetate (20:1–5:1) to obtain compound 33 (30.1 mg). F4-4 (900.6 mg) was further segmented via Sephadex LH-20 (methanol) to obtain 3 subfractions (F4-4-1~F4-4-3). F4-4-1 (20.3 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-ethyl acetate (3:1) to obtain compound 17 (9.0 mg). F4-4-2 (102.3 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-ethyl acetate (3:1) to obtain compound 18 (12.2 mg). F4-4-3 (500.1 mg) was segmented by CC (silica gel) stepwise gradient elution with chloroform-methanol (80:1–20:1) to obtain 4 subfractions (F4-4-3-1~F4-4-3-4). F4-5 (1.1 g) was further segmented via Sephadex LH-20 (methanol) to obtain 5 subfractions (F4-5-1~F4-5-5). F4-5-5 (350.4 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-ethyl acetate (4:1–1:1) to obtain 4 subfractions (F4-5-5-1~F4-5-5-4). F4-5-5-1 (60.3 mg) was segmented by CC (silica gel) stepwise gradient elution with trichloromethane-methanol (60:1–10:1) to obtain compound 7 (27.5 mg). F4-5-5-3 (200.3 mg) was further segmented via Sephadex LH-20 (methanol) to obtain compound 9 (123.9 mg). F4-6-4 (2.1 g) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-ethyl acetate (4:1–1:1) to obtain 2 subfractions (F4-6-4-1~F4-6-4-2). F4-6-4-1 (1.2 g) was further segmented via Sephadex LH-20 (methanol) to obtain compound 27 (20.7 mg). F4-6-4-2-2 (1.0 g) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-ethyl acetate (3:1–1:1), and then was further segmented via Sephadex LH-20 (methanol) to obtain compounds 5 (60.6 mg) and 35 (40.2 mg).
Fr.5 (92.0 g) was passed through a normal-phase silica gel column, firstly subjected to reversed-phase RP-18 silica gel eluting with methanol-water (60–100%) to obtain 11 subfractions (F5-1~F5-11). F5-1 (1.1 g) was further segmented via Sephadex LH-20 (methanol) to obtain 4 subfractions (F5-1-1~F5-1-4). F5-2 (2.0 g) was further segmented via Sephadex LH-20 (methanol) to obtain 2 subfractions (F5-2-1~F5-2-2). F5-2-1 (90.8 mg) was repeatedly recrystallized to produce compound 32 (10.3 mg). F5-2-2 (1.3 g) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-acetone (2:1–1:1) to obtain 5 subfractions (F5-2-2-1~F5-2-2-5). F5-2-2-3 (201.4 mg) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (25:1–10:1) and then was further segmented via Sephadex LH-20 (methanol) to obtain compound 8 (5.0 mg). F5-3 (10.0 g) was segmented by CC (silica gel) stepwise gradient elution with trichloromethane-methanol (80:1–30:1) to obtain 6 subfractions (F5-3-1~F5-3-6). F5-3-3-7 (91.5 mg), followed by semipreparative HPLC (55%, MeOH-H2O, 3 mL/min), was used to obtain compound 2 (4.0 mg). F5-3-6 (5.0 g) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-acetone (3:1–1:1) to obtain 4 subfractions (F5-3-6-1~F5-3-6-4). F5-3-6-3, (76.1 mg) followed by semipreparative HPLC (50%, MeOH-H2O, 3 mL/min), was used to obtain compound 26 (4.2 mg). F5-3-6-4 (47.6 mg), followed by semipreparative HPLC (55%, MeOH-H2O, 3 mL/min), was used to obtain compound 28 (4.2 mg) and 29 (4.0 mg). F5-4 (2.0 g) was further segmented via Sephadex LH-20 (methanol) to obtain 9 subfractions (F5-4-1~F5-4-9). F5-4-1 (614.5 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-acetone (3:1–1:1) to obtain compound 13 (500.1 mg). F5-9 (2.0 g) was further segmented via Sephadex LH-20 (methanol) to obtain 10 subfractions (F5-9-1~F5-9-10). F5-9-1 (1.0 g) was segmented by CC (silica gel) stepwise gradient elution with trichloromethane-methanol (50:1–10:1) to obtain compound 14 (806.2 mg). F5-10 (5.5 g) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-acetone (2:1–1:1) to obtain compound 23 (20.1 mg).
Fr.6 (54.0 g) was firstly subjected to reversed-phase RP-18 silica gel eluting with methanol-water (60–100%) to obtain 7 subfractions (F6-1~F6-7). F6-5 (25 g) was segmented by CC (silica gel) stepwise gradient elution with trichloromethane-methanol (100:1–50:1) to obtain compound 13 (10.0 g) and compound 20 (801.5 mg). F6-5-5 (511.6 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-ethyl acetate (3:1–1:1) to obtain 2 subfractions (F6-5-5-1~F6-5-5-2). F6-5-5-2 (105.3 mg) was further segmented via Sephadex LH-20 (methanol) to obtain compound 24 (89.5 mg).
Fr.7 (43.0 g) was segmented by CC (silica gel) stepwise gradient elution with dichloromethane-methanol (15:1–1:1) to obtain 7 subfractions (F7-1~F7-7). F7-5 (4.0 g) was further segmented via Sephadex LH-20 (methanol) to obtain 7 subfractions (F7-5-1~F7-5-7). F7-5-3 (203.5 mg) was further segmented via Sephadex LH-20 (methanol) to obtain 3 subfractions (F7-5-3-1~F9-5-3-3). F7-5-3-3 (30.6 mg) was segmented by CC (silica gel) stepwise gradient elution with petroleum ether-acetone (2:1–1:1) to obtain compound 6 (5.2 mg). F7-5-7 (40.9 mg) was segmented by CC (silica gel) stepwise gradient elution with trichloromethane-acetone (15:1–1:1) to obtain compound 25 (30.7 mg).
4′,4′-Dimethoxy-sophvein (17): white amorphous powder; α D 24 = + 49.88 (c 0.25, MeOH); UV λmax (MeOH) nm (log ε): 214 (4.74), 229 (4.64), 285 (4.58); IR (KBr) νmax: 3456, 2950, 1638, 1400, 1098 cm−1; 1H NMR (Methanol-d4, 600 MHz) and 13C NMR (Methanol-d4, 150 MHz), see Table 1; HRESIMS at m/z 429.1894 [M+Na]+ (calcd for C22H30O7Na+, 429.1884).
Sophvein-4′-one (18): white amorphous powder; α D 24 = +38.69 (c 0.29, MeOH); UV λmax (MeOH) nm (log ε): 214 (4.48), 231 (4.06), 286 (3.97); IR (KBr) νmax: 3446, 2952, 1635, 1406, 1086 cm−1; 1H NMR (Acetone-d6, 600 MHz) and 13C NMR (Acetone-d6, 150 MHz), see Table 1; HRESIMS at m/z 361.1650 [M+Na]+ (calcd for C20H24O6 Na+, 361.1646).

3.4. Cell Culture Conditions

Murine macrophages (RAW 264.7) were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with the addition of 10% fetal bovine serum (FBS) and 1% streptomycin (10,000 μg/mL)-penicillin (10,000 U/mL) at 37 °C in a humidified incubator under 5% CO2. HepG2 cells were cultured at 37 °C under 5% CO2. The optimal fermentation culture medium comprised DMEM, 1% mixed penicillin (10,000 U mL−1), 1% HEPES (BioFroxx, Hesse, Einhausen), and 10% fetal bovine serum (FBS), as well as streptomycin (SM) (10,000 μg/mL−1) fluid. In all experiments, cells were left to acclimate for 24 h before any treatments.

3.5. Cell Viability Examination

To measure cell viability, an MTT assay was performed [41]. Macrophages (4 × 104 per well) were cultured in 96-well plates for 24 h; then, they were treated with several concentrations of compounds 136 (3.12, 6.25, 12.5, 25, and 50 μM) for 1 h, and then they were stimulated with LPS (1 μg/mL) for another 24 h. After washing twice with a PBS buffer, 20 μL of the MTT solution was added to each well, and incubation continued for 4 h. Optical density (O.D) was measured at 490 nm by a microplate reader (Thermo Fisher Scientific, Waltham, MA, USA). The relative cell viability was calculated in contrast to the normal control group.

3.6. NO Production Measurement

Activated macrophage cells could express inducible nitric oxide synthase (iNOS), catalyzing the production of NO from L-arginine. To determine the effect of compounds 136 on treatments on NO production, the accumulation of nitrites (NO2−) in the culture medium was recorded as an indicator of NO production [42,43]. Macrophages (8 × 104 per well) were seeded onto 96-well plates and pretreated by compounds 136 (3.12, 6.25, 12.5, 25, and 50 μM) 1 h prior to treatment by LPS (1 μg/mL). Afterward, costimulation for 24 h at 37 °C was carried out in an incubator under 5% CO2. Then, Griess reagents I and II (100 μL) were mixed with cell culture medium (70 μL). Prior to measuring the optical density, plates were incubated at room temperature for 10 min, and the absorbance at 540 nm was measured using a Thermo Fisher Scientific microplate reader. N(G)-monomethyl-L-arginine, monoacetate salt (L-NMMA), and the medium were used as positive and negative controls, respectively.

3.7. Antiproliferative Assay

Briefly, the cells were incubated in 96-well microplates (1 × 105 per well) and allowed to adhere for 24 h before drug administration. Then, the cell lines were treated with test compounds 136 at five concentrations (3.12, 6.25, 12.5, 25, and 50 μM). At 24 and 48 h of incubation, cells were treated with MTT (200 µL, 5.0 mg/mL) and dissolved in the culture medium, for 1 h under at 37 °C under a 5% CO2 humidified atmosphere. The MTT was then removed carefully and resolved with DMSO (150 μL/well). Optical density was recorded using a Thermo Fisher Scientific microplate reader at 490 nm. Cisplatin (CP) and medium were used as positive and negative controls, respectively.

3.8. Statistical Analysis

The data were analyzed using Statistical Package for Social Sciences (SPSS Version 21.0) software and are presented as the mean ± S.D. values of three different experiments. The results were analyzed via one-way analysis of variance (ANOVA), and statistical significance was defined as p < 0.05.

4. Conclusions

In summary, the systematic phytochemical study on the roots of S. flavescens led to the isolation of 36 flavonoids (136), including 18 dihydroflavonoids (118), 7 flavonols (1925), 4 dihydroflavonols (2629), 3 isoflavones (30–32), and 4 chalcones (3336), identified by spectroscopic methods (1H, 13C NMR, HSQC, 1H-1H COSY, HMBC, HRESIMS), and chemical and physical methods. Bioactivity assays indicated that nine compounds exhibited the most significant NO inhibitory activity, better than that of the L-NMMA positive control (IC50 = 21.8 ± 0.9 μM). Furthermore, the antiproliferative activities against the HepG2 hepatoma cell lines of all isolates were assessed using the MTT method. The results demonstrated that compound 22 exhibited significant cytotoxic activity, with an IC50 value of 0.46 ± 0.1 μM (positive control cisplatin: IC50 = 24.5 ± 0.8 μM). This study lays the foundation for studying the potential therapeutic applications of flavonoid derivatives from S. flavescens for inflammatory diseases and hepatoma.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/molecules28052048/s1, Figures S1–S14 HRESIMS, 1H and 13C NMR, HMQC, HMBC, 1H-1H COSY, and ROESY of compounds 17 and 18.

Author Contributions

Conceptualization, R.-T.L. and Z.-J.Z.; methodology, G.-X.L.; formal analysis, Y.-F.Y. and T.-T.L.; investigation, Y.-F.Y., T.-T.L., G.-X.L., X.-Q.C. and Z.-J.Z.; data curation, G.-X.L.; writing—original draft preparation, T.-T.L.; writing—review and editing, Y.-F.Y. and T.-T.L.; supervision, Z.-J.Z.; funding acquisition, R.-T.L. and Z.-J.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported financially by the National Natural Science Foundation of China (No. 31960092 and 82204243), the Innovative Team of Yunnan Province (No. 2019HC018), and Key Laboratory of Colleges and Universities in Yunnan Province.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the Supplementary Materials.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

Abbreviations

CC, chromatograph column; COSY, correlation spectroscopy; DEPT, distortionless enhancement by polarization transfer; HPLC, high-performance liquid chromatography; HR-ESI-MS, high-resolution electrospray ionization mass spectroscopy; HMBC, heteronuclear multiple bond correlation; HSQC, heteronuclear single quantum coherence; L-NMMA, N(G)-monomethyl-L-arginine; LPS, lipopolysaccharide; MPLC, medium-pressure liquid chromatography; NMR, nuclear magnetic resonance; MS, mass spectrometry; NO, nitric oxide; NOE, nuclear overhauser effect; ROESY, rotating-frame overhauser effect spectroscopy; TCM, traditional Chinese medicine; TLC, thin-layer chromatography; TMS, tetramethylsilane.

References

  1. Lu, K.-Z.; Feng, Z.-M.; Yuan, X.; Yang, Y.-N.; Jiang, J.-S.; Zhang, X.; Zhang, P.-C. Five novel pterocarpan derivatives from Sophora flavescens. Chin. J. Chem. 2021, 39, 2763–2768. [Google Scholar] [CrossRef]
  2. He, X.-R.; Fang, J.-C.; Huang, L.-H.; Wang, J.-H.; Huang, X.-Q. Sophora flavescens Ait.: Traditional usage, phytochemistry and pharmacology of an important traditional Chinese medicine. J. Ethnopharmacol. 2015, 172, 10–29. [Google Scholar] [CrossRef]
  3. Zhou, Y.-J.; Gou, Y.-J.; Yang, X.-L.; Ou, Z.-L. Anti-cervical cancer role of matrine, oxymatrine and Sophora flavescens alkaloid gels and its mechanism. J. Cancer 2018, 9, 1357–1364. [Google Scholar] [CrossRef] [Green Version]
  4. Shen, C.-C.; Lin, T.-W.; Huang, Y.-L.; Wan, S.-T.; Shien, B.-J.; Chen, C.-C. Phenolic constituents of the roots of Sophora flavescens. J. Nat. Prod. 2006, 69, 1237–1240. [Google Scholar] [CrossRef]
  5. Kwon, M.; Ko, S.K.; Jang, M.; Kim, G.H.; Ryoo, I.J.; Son, S.; Ryu, H.W.; Oh, S.R.; Lee, W.K.; Kim, B.Y.; et al. Inhibitory effects of flavonoids isolated from Sophora flavescens on indoleamine 2,3-dioxygenase 1 activity. J. Enzyme Inhib. Med. Chem. 2019, 34, 1481–1488. [Google Scholar] [CrossRef] [Green Version]
  6. Zhou, H.-P.; Lutterodt, H.M.; Cheng, Z.-H.; Yu, L.-L. Anti-inflammatory and antiproliferative activities of trifolirhizin, a flavonoid from Sophora flavescens roots. J. Agric. Food. Chem. 2009, 57, 4580–4585. [Google Scholar] [CrossRef] [Green Version]
  7. Kim, J.H.; Cho, C.W.; Kim, H.Y.; Kim, K.T.; Choi, G.S.; Kim, H.H.; Cho, I.S.; Kwon, S.J.; Choi, S.K.; Yoon, J.Y.; et al. α-Glucosidase inhibition by prenylated and lavandulyl compounds from Sophora flavescens roots and in silico analysis. Int. J. Biol. Macromol. 2017, 102, 960–969. [Google Scholar] [CrossRef]
  8. Zhang, S.-Y.; Li, W.; Nie, H.; Liao, M.; Qiu, B.; Yang, Y.-L.; Chen, Y.-F. Five new alkaloids from the roots of Sophora flavescens. Chem. Biodivers. 2018, 15, e1700577. [Google Scholar] [CrossRef]
  9. Li, J.-J.; Zhang, X.; Shen, X.-C.; Long, Q.-D.; Xu, C.-Y.; Tian, C.-J.; Lin, Y. Phytochemistry and biological properties of isoprenoid flavonoids from Sophora flavescens Ait. Fitoterapia 2020, 143, 104556–104562. [Google Scholar] [CrossRef]
  10. Zhu, H.; Yang, Y.-N.; Feng, Z.-M.; Jiang, J.-S.; Zhang, P.-C. Sophoflavanones A and B, two novel prenylated flavanones from the roots of Sophora flavescens. Bioorg. Chem. 2018, 79, 122–125. [Google Scholar] [CrossRef]
  11. Yang, Y.-N.; Zhu, H.; Yuan, X.; Zhang, X.; Feng, Z.-M.; Jiang, J.-S.; Zhang, P.-C. Seven new prenylated flavanones from the roots of Sophora flavescens and their anti-proliferative activities. Bioorg. Chem. 2021, 109, 104716. [Google Scholar] [CrossRef]
  12. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  13. Gerold, J.; Reiner, W.; Hans, A. Cyclohexanoid protoflavanones from the stem-bark and roots of Ongokea gore. Phytochemistry 2005, 66, 1698–1706. [Google Scholar]
  14. Milligan, S.; Kalita, J.; Heyerick, A.; Rong, H.; Cooman, L.D.; Keukeleire, D.D. Identification of a potent phytoestrogen in hops (Humulus lupulus L.) and beer. J. Clin. Endocr. Metab. 1999, 84, 2249. [Google Scholar] [CrossRef]
  15. Jeong, T.S.; Ryu, Y.B.; Kim, H.Y.; Curtis-Long, M.J.; An, S.J.; Lee, J.H.; Lee, W.S.; Park, K.H. Low Density lipoprotein (LDL)-antioxidant flavonoids from roots of Sophora flavescens. Biol. Pharm. Bull. 2008, 31, 2097–2102. [Google Scholar] [CrossRef] [Green Version]
  16. Wollenweber, E.; Roitman, J.N. New reports on surface flavonoids from Chamaebatiaria (Rosaceae), Dodonaea (Sapindaceae), Elsholtzia (Lamiaceae), and Silphium (Asteraceae). Nat. Prod. Commun. 2007, 2, 385–389. [Google Scholar] [CrossRef]
  17. Kuroyanagi, M.; Arakava, T.; Hirayama, Y.; Hayashi, T. Antibacterial and antiandrogen flavonoids from Sophora flavescens. J. Nat. Prod. 1999, 62, 1595–1599. [Google Scholar] [CrossRef]
  18. Iinuma, M.; Ohyama, M.; Tanaka, T.; Lang, F.A. Three New Phenolic Compounds from the Roots of Sophora leachiana. J. Nat. Prod. 1993, 56, 2212. [Google Scholar] [CrossRef]
  19. Yang, X.; Yang, J.; Xu, C.; Huang, M.; Zhou, Q.; Lv, J.; Ma, X.; Ke, C.; Ye, Y.; Shu, G. Antidiabetic effects of flavonoids from Sophora flavescens EtOAc extract in type 2 diabetic KK-ay mice. J. Ethnopharmacol. 2015, 170, 161–170. [Google Scholar] [CrossRef]
  20. Huang, R.; Liu, Y.; Zhao, L.L.; Chen, X.X.; Wang, F.; Cai, W.; Chen, L. A new flavonoid from Sophora flavescens Ait. Nat. Prod. Res. 2017, 31, 2228–2232. [Google Scholar] [CrossRef]
  21. Iinuma, M.; Ohyama, M.; Tanaka, T. Six flavonostilbenes and a flavanone in roots of Sophora alopecuroides. Phytochemistry 1995, 38, 519–525. [Google Scholar] [CrossRef]
  22. Shirataki, Y.; Yokoe, I.; Noguchi, M.; Tomimoi, T.; Komatsu, M. Studies on the Constituents of Sophora Species. XXII.: Constituents of the root of Sophora moorcroftiang BENTH. ex BAKER. (1). Chem. Pharm. Bull. 1988, 36, 2220–2225. [Google Scholar] [CrossRef] [Green Version]
  23. Ibieta, H.P.; Michael, W. Binding of flavonoids from Sophora flavescens to the rat uterine estrogen receptor. Planta Med. 2005, 71, 1065–1068. [Google Scholar]
  24. Hatayama, K.; Komatsu, M. Studies on the constituents of Sophora species. V. Constituents of the root of Sophora angustifolia Sieb. Et Zucc.(2). Chem. Pharm. Bull. 1971, 19, 2126–2131. [Google Scholar] [CrossRef]
  25. Kang, T.H.; Jeong, S.J.; Ko, W.G.; Kim, N.Y.; Lee, B.H.; Inagaki, M.; Miyamoto, T.; Higuchi, R.; Kim, Y.C. Cytotoxic lavandulyl flavanones from Sophora flavescens. J. Nat. Prod. 2000, 63, 680–681. [Google Scholar] [CrossRef]
  26. Shi, Y.R.; Lee, H.S.; Kim, Y.K.; Kim, S.H. Determination of isoprenyl and lavandulyl positions of flavonoids from Sophora flavescens by NMR experiment. Arch. Pharm. Res. 1997, 20, 491–495. [Google Scholar]
  27. Wang, Z.Q.; Lou, Y.J. Proliferation-stimulating effects of icaritin and desmethylicaritin in MCF-7 cells. Eur. J. Pharmacol. 2004, 504, 147–153. [Google Scholar] [CrossRef]
  28. Jung, M.J.; Kang, S.S.; Jung, H.A.; Kim, G.J.; Choi, J.S. Isolation of flavonoids and a cerebroside from the stem bark of Albizzia julibrissin. Arch. Pharm. Res. 2004, 27, 593–599. [Google Scholar] [CrossRef]
  29. Kawamura, T.; Hayashi, M.; Mukai, R.; Terao, J.; Nemoto, H. An Efficient Method for C8-Prenylation of Flavonols and Flavanones. Synthesis 2012, 44, 1308–1314. [Google Scholar]
  30. Jung, J.; Kang, S.S.; Woo, J.J.; Choi, J.S. A new lavandulylated flavonoid with free radical and onoo-scavenging activities from Sophora flavescens. Arch. Pharm. Res. 2005, 28, 1333–1336. [Google Scholar] [CrossRef]
  31. Chen, H.; Yang, J.; Hao, J.; Lv, Y.; Chen, L.; Lin, Q.; Yuan, J.; Yang, X. A Novel Flavonoid kushenol Z from Sophora flavescens mediates mTOR pathway by inhibiting phosphodiesterase and Akt activity to induce apoptosis in non-small-cell lung cancer cells. Molecules 2019, 24, 4425. [Google Scholar] [CrossRef] [Green Version]
  32. Ohmoto, T.; Aikawa, R.; Nikaido, T.; Sankawa, U.; Wu, L.J.; Ueno, A.; Hukushima, S. Inhibition of adenosine 3′, 5′-cyclic monophosphate phosphodiesterase by components of Sophora flavescens Aiton. Chem. Pharm. Bull. 1986, 34, 2094–2099. [Google Scholar] [CrossRef] [Green Version]
  33. Yagi, A.; Fukunaga, M.; Okuzako, N.; Mifuchi, I.; Kawamoto, F. Antifungal substances from Sophora flavescens. Jpn. J. Pharmacogn. 1989, 43, 343–347. [Google Scholar]
  34. Sato, S.; Takeo, J.; Aoyama, C.; Kawahara, H. Na+-glucose cotransporter (SGLT) inhibitory flavonoids from the roots of Sophora flavescens. Bioorgan. Med. Chem. 2007, 15, 3445–3449. [Google Scholar] [CrossRef]
  35. Wu, L.; Miyase, T.; Ueno, A.; Kuroyanagi, M.; Noro, T.; Fukushima, S. Studies on the Constituents of Sophora flavescens Aiton. III. Yakugaku Zasshi 1985, 105, 736–741. [Google Scholar] [CrossRef] [Green Version]
  36. Selepe, M.A.; Drewes, S.E.; Heerden, F.R. Total synthesis of the pyranoisoflavone kraussianone I and related isoflavones. J. Nat. Prod. 2010, 73, 1680–1685. [Google Scholar] [CrossRef]
  37. Umehara, K.; Nemoto, K.; Matsushita, A.; Terada, E.; Monthakantirat, O.; De-Eknamkul, W.; Miyase, T.; Warashina, T.; Degawa, M.; Noguchi, H. Flavonoids from the heartwood of the Thai medicinal plant Dalbergia parviflora and their effects on estrogenic-responsive human breast cancer cells. J. Nat. Prod. 2009, 72, 2163–2168. [Google Scholar] [CrossRef]
  38. Bartmańska, A.; Tronina, T.; Popłoński, J.; Milczarek, M.; Filip-Psurska, B.; Wietrzyk, J. Highly cancer selective antiproliferative activity of natural prenylated flavonoids. Molecules 2018, 23, 2922. [Google Scholar] [CrossRef] [Green Version]
  39. Stevens, J.F.; Taylor, A.W.; Nickerson, G.B.; Ivancic, M.; Deinzer, M.L. Prenylflavonoid variation in Humulus lupulus: Distribution and taxonomic significance of xanthogalenol and 4′-O-methylxanthohumol. Phytochemistry 2000, 53, 759–775. [Google Scholar] [CrossRef]
  40. Wu, L.; Miyase, T.; Ueno, A.; Kuroyanagi, M.; Noro, T.; Fukushima, S. Studies on the constituents of Sophora flavescens Aiton. II. Chem. Pharm. Bull. 1985, 33, 3231–3236. [Google Scholar] [CrossRef] [Green Version]
  41. El-Readi, M.Z.; Eid, S.; Ashour, M.L.; Tahrani, A.; Wink, M. Modulation of multidrug resistance in cancer cells by chelidonine and Chelidonium majus alkaloids. Phytomedicine 2013, 20, 282–294. [Google Scholar] [CrossRef]
  42. Li, J.-C.; Dai, W.-F.; Liu, D.; Jiang, M.-Y.; Zhang, Z.-J.; Chen, X.-Q.; Chen, C.-H.; Li, R.-T.; Li, H.-M. Bioactive ent-isopimarane diterpenoids from Euphorbia neriifolia. Phytochemistry 2020, 175, 112373. [Google Scholar] [CrossRef]
  43. Cao, L.; Li, R.; Chen, X.; Xue, Y.; Liu, D. Neougonin A inhibits lipopolysaccharide -induced inflammatory responses via downregulation of the NF-kB signaling pathway in RAW 264.7 macrophages. Inflammation 2016, 39, 1939–1948. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of isolated compounds.
Figure 1. Chemical structures of isolated compounds.
Molecules 28 02048 g001
Figure 2. The structure and the key 2D NMR correlations of 17.
Figure 2. The structure and the key 2D NMR correlations of 17.
Molecules 28 02048 g002
Table 1. 1H and 13C NMR data of compounds 17 and 18 a.
Table 1. 1H and 13C NMR data of compounds 17 and 18 a.
Position17 b18 c
δH (J in Hz)δCδH (J in Hz)δC
24.04 (d, 13.6)84.64.40 (dd, 13.6, 2.7)83.7
3a2.81 (dd, 17.0,13.6)37.03.02 (dd, 17.0, 2.7)36.9
3b2.51 (d, 17.0) 2.68 (dd,17.0, 2.7)
4198.8198.0
5163.0162.8
65.84 (s)96.46.01 (s)96.3
7166.0164.8
8109.0108.1
9161.3160.7
10103.2103.0
1′72.271.3
2′a1.75 (m)29.92.21 (m)36.7
2′b1.67 (m) 2.21 (m)
3′a1.86 (m)28.42.39 (m)33.0
3′b1.86 (m) 2.03 (m)
4′101.1210.3
5′a1.82 (m)28.52.74 (m)36.8
5′b1.65 (m) 2.74 (m)
6′a1.72 (d,11.7)31.32.10 (m)34.0
6′b1.45 (d,11.7) 2.10 (m)
1a″3.14 (d, 6.9)22.43.24 (d, 6.9)22.1
1b″3.14 (d, 6.9) 3.24 (d, 6.9)
2″5.11 (br t, 6.9)124.45.16 (br t, 6.9)124.1
3″131.6131.0
4″1.59 (s)25.91.62 (s)25.8
5″1.68 (s)18.11.71 (s)17.9
4′-OMe3.15 (s)47.9
4′-OMe3.10 (s)48.1
a Assignments were supported with HSQC, HMBC, and 1H-1H COSY experiments. b Measured in methanol-d4 at 600 MHz for 1H NMR and at 150 MHz for 13C NMR. c Measured in acetone-d6 at 600 MHz for 1H NMR and at 150 MHz for 13C NMR.
Table 2. Inhibitory effects of compounds 136 on LPS-induced NO production in macrophages (IC50 values less than 20 μM are listed).
Table 2. Inhibitory effects of compounds 136 on LPS-induced NO production in macrophages (IC50 values less than 20 μM are listed).
CompdsIC50 b (μM)CC50 c (μM)CompdsIC50 b (μM)CC50 c (μM)
28.4 ± 0.7>50225.7 ± 0.413.8 ± 0.6
1211.2 ± 1.125.7 ± 1.1297.2 ± 0.620.6 ± 1.1
1314.4 ± 0.433.8 ± 1.0354.6 ± 1.121.9 ± 0.4
159.3 ± 0.718.8 ± 0.3366.7 ± 0.616.2 ± 0.9
1912.4 ± 0.9>50L-NMMA a21.8 ± 0.9>50
a L-NMMA was used as positive control. b IC50: 50% inhibitory concentration. (Mean ± SD of 3 tests.) c CC50: 50% cytotoxic concentration. (Mean ± SD of 3 tests.)
Table 3. Antiproliferative effects of compounds 136 on HepG2 cell lines (IC50 values less than 50 μM are listed).
Table 3. Antiproliferative effects of compounds 136 on HepG2 cell lines (IC50 values less than 50 μM are listed).
CompoundsIC50 b (μM)CompdsIC50 b (μM)
134.9 ± 0.32126.4 ± 0.3
537.6 ± 0.7220.46 ± 0.1
1630.8 ± 1.13448.6 ± 0.8
1941.4 ± 0.63546.2 ± 0.7
Cisplatin a24.5 ± 0.8--
a Cisplatin was used as positive control. b IC50: 50% inhibitory concentration. (Mean ± SD of 3 tests.).
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

Yang, Y.-F.; Liu, T.-T.; Li, G.-X.; Chen, X.-Q.; Li, R.-T.; Zhang, Z.-J. Flavonoids from the Roots of Sophora flavescens and Their Potential Anti-Inflammatory and Antiproliferative Activities. Molecules 2023, 28, 2048. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28052048

AMA Style

Yang Y-F, Liu T-T, Li G-X, Chen X-Q, Li R-T, Zhang Z-J. Flavonoids from the Roots of Sophora flavescens and Their Potential Anti-Inflammatory and Antiproliferative Activities. Molecules. 2023; 28(5):2048. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28052048

Chicago/Turabian Style

Yang, Yan-Fei, Ting-Ting Liu, Guo-Xian Li, Xuan-Qin Chen, Rong-Tao Li, and Zhi-Jun Zhang. 2023. "Flavonoids from the Roots of Sophora flavescens and Their Potential Anti-Inflammatory and Antiproliferative Activities" Molecules 28, no. 5: 2048. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules28052048

Article Metrics

Back to TopTop