DOI QR코드

DOI QR Code

Dammarane-type triterpene oligoglycosides from the leaves and stems of Panax notoginseng and their antiinflammatory activities

  • Li, Juan (Department of Pharmacognosy, China Pharmaceutical University) ;
  • Wang, Ru-Feng (The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM, Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine) ;
  • Zhou, Yue (The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM, Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine) ;
  • Hu, Hai-Jun (The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM, Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine) ;
  • Yang, Ying-Bo (The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM, Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine) ;
  • Yang, Li (The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM, Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine) ;
  • Wang, Zheng-Tao (Department of Pharmacognosy, China Pharmaceutical University)
  • Received : 2017.06.10
  • Accepted : 2017.11.16
  • Published : 2019.07.15

Abstract

Background: Inflammation is widespread in the clinical pathology and closely associated to the progress of many diseases. Triterpenoid saponins as a key group of active ingredients in Panax notoginseng (Burk.) F.H. Chen were demonstrated to show antiinflammatory effects. However, the chemical structures of saponins in the leaves and stems of Panax notoginseng (PNLS) are still not fully clear. Herein, the isolation, purification and further evaluation of the antiinflammatory activity of dammarane-type triterpenoid saponins from PNLS were conducted. Methods: Silica gel and reversed-phase C8 column chromatography were used. Furthermore, preparative HPLC was used as a final purification technique to obtain minor saponins with high purities. MS, NMR experiments, and chemical methods were used in the structural identifications. The antiinflammatory activities of the isolated saponins were assessed by measuring the nitric oxide production in RAW 264.7 cells stimulated by lipopolysaccharides. Real-time reverse transcription polymerase chain reaction was used to measure the gene expressions of inflammation-related gene. Results: Eight new minor dammarane-type triterpene oligoglycosides, namely notoginsenosides LK1-LK8 (1-8) were obtained from PNLS, along with seven known ones. Among the isolated saponins, gypenoside IX significantly suppressed the nitric oxide production and inflammatory cytokines including tumor necrosis $factor-{\alpha}$, interleukin 10, interferon-inducible protein 10 and $interleukin-1{\beta}$. Conclusion: The eight saponins may enrich and expand the chemical library of saponins in Panax genus. Moreover, it is reported for the first time that gypenoside IX showed moderate antiinflammatory activity.

Keywords

1. Introduction

Panax notoginseng, Panax ginseng Meyer, and Panax quinquefolium L. belong to Panax genus under Araliaceae family. As is known to all, triterpenoid saponins have made great contributions to the biological activities of these three medicinal herbs mentioned above, from which more than 200 saponins have already been isolated. According to the structures of the aglycone, these known saponins were classified into five types, while the protopanaxadiol (PPD) and protopanaxatriol type accounted for the majority [1]. Depending on the numbers of sugar units, they can also be divided into monoglycoside, diglycoside, and oligoglycoside. The major components of these three medicinal herbs were reported to be PPDand protopanaxatriol-type oligoglycosides, such as ginsenoside Rb1, Rc, Rb3, and Re [2].

Inflammation occurs widely in the process of clinical pathology and plays a significant role in the progress of many diseases [3], and therefore the compounds holding antiinflammatory effects will be used for the treatment of various inflammation-related diseases. Previous reports have shown that ginsenoside Rc and Rd belonging to PPD-type oligoglycosides exerted antiinflammatory activity by suppressing the gene expressions of tumor necrosis factor-α (Tnf-α) and nuclear factor kappa B, respectively [4,5]. The leaves and stems of P. notoginseng (PNLS) were rich in PPD-type oligoglycosides and could be an alternative source of bioactive saponins and a possible replacement of the roots, which required long growth period for harvest. So far, approximately 30 saponins have been purified and characterized from PNLS, such as notoginsenoside Fc and ginsenoside Rb3 and Rc [6e8]. However, chemical structures of a few saponins are still uncertain, especially pairs of isomeric ones undistinguished by MS spectrum [9]. Hence, isolation from PNLS was conducted to enhance chemical diversity of Panax plants and provide materials for activity screening to find out potential agents with antiinflammatory effects.

In this report, fifteen dammarane-type triterpene oligoglycosides were obtained from PNLS. Among them, gypenoside IX was demonstrated to exhibit the highest inflammatory activities by suppressing the nitric oxide (NO) production and inflammatory cytokines including Tnf-α, interleukin 10 (Il-10), interferoninducible protein 10 (Cxcl10) and interleukin-1β (Il-1β) in RAW 264.7 cells stimulated by lipopolysaccharides (LPSs).

2. Materials and methods

2.1. General experimental procedures

The total saponins of PNLS were purchased from Qidan Co. Ltd. (Wenshan, Yunnan, China). Optical rotations were measured on a Perkin-Elmer 341B polarimeter. IR spectra were obtained from a Nicolet-380 spectrometer. 1H-NMR (600 MHz) and 13C-NMR (150 MHz) data were acquired on a Bruker AVANCE-III spectrometer in deuterated pyridine. The spectra of high resolution electrospray ionization mass spectroscopy (HRESIMS) were obtained from a Q-TOF mass spectrometer (Waters, UK) in the negative ion mode. Configuration determination of sugar components was performed on HPLC-UV (Agilent 1100) with a Shim-Pack HRC-ODS column (250 × 4.6 mm, 5 mm, Shimadzu, Japan). Silica gel (200e300 mesh, Qingdao Haiyang Chemical Group Co., Ltd., China) and reversedphase C8 gel (40e60 mm, YMC, Japan) were used in the open column chromatography (CC). Preparative HPLC was performed on a preparative HPLC system (Tong Heng Innovation Technology Co., Ltd., China) equipped with a T-Sharp C18 column (150 × 30 mm, 5 mm, Xuanmei Co. Ltd., Shanghai, China) and semipreparative HPLC with a Zorbax SB C18 column (250 × 9.4 mm, 5 mm, Agilent Technologies, USA). TLC was carried on the HSGF254 plates (Qingdao Jiangyou Group Co., Ltd., China) with developing solvents CHCl3/ MeOH/H2O (7.5:2.5:0.3, 7:3:0.5, 6.5:3.5:0.5, v/v). Authentic samples used for determination of configurations of the sugar moiety were obtained from Sigma-Aldrich (Steinheim, Germany). L-cysteine methyl ester and o-tolylisothiocyanate as derivatization reagents were bought from Tokyo Chemical Industry (Tokyo, Japan).

2.2. Extraction and isolation

The total saponins (440 g) from PNLS were divided into six fractions (AeF) by silica gel CC, eluted with CH2Cl2/MeOH/H2O (8:2:0.2, 7.5:2.5:0.3, 7:3:0.5, 6.5:3.5:0.5, 1:1:0). F (200 g) was chromatographed over silica gel again to yield four subfractions (F1eF4). F4 (120 g) was fractionated using Rp-C8 gel, eluted with MeOH/H2O (4:6 → 9:1) to give four subfractions (F4.1: 1.5 g, F4.2: 6 g, F4.3: 40 g, F4.4: 34 g). Then F4.2 (1g ×6) was further purified by preparative HPLC on T-Sharp C18 column with CH3CN/H2O (28:72 / 32:68) as mobile phase, 40 ml/min, 6 times repeated. Every 20 ml of eluent were collected and followed by a detection of TLC. Eluent in each tube with similar Rf value was combined to give compound 1 (8 mg), 2 (12 mg), 3 (5 mg), and four subfractions (F4.2.1-F4.2.4). After further purification of F4.2.3 (150 mg) by semipreparative HPLC on an Agilent Zorbox SB-C18 column (CH3CN/H2O, 30:70, UV detection at 203 nm), compound 5 (3 mg) and 9 (85 mg) were obtained. In a similar way, compound 4 (7 mg), 6 (32 mg), and 7 (18 mg) were obtained from F4.2.4 (100 mg). Purification of F4.1 (1.5 g) was performed in the same condition as F4.2 to give compound 8 (12 mg). Purification of 1 g of F4.3 was carried out on the silica gel CC (CHCl3/MeOH/H2O, 7.5:2.5:0.5 → 7:3:0.5) and prep-HPLC (CH3CN/H2O, 26:74 → 30:70) to give 10 (350 mg), 11 (10 mg), 12 (5 mg), and 13 (230 mg). Then 1 g of F4.4 was further chromatographed with silica gel (CHCl3/MeOH/H2O, 8:2:0.2 → 7.5:2.5:0.5) to yield 14 (300 mg). Finally, 100 mg of F3 was further separated by semipreparative HPLC (CH3CN/H2O, 32:68 → 36:64) and 15 (20 mg) was obtained.

2.3. Spectroscopic data

Notoginsenoside LK1 (1): white, amorphous powder; \([\alpha]^{25}_D\) + 2.2 (c 0.09, MeOH); IR νmax 3,352, 2,920, 1,662, 1,367, 1,164, 1,077, 1,027,893 cm-1. 1H-NMR and 13C-NMR data are shown in Table 1; m/z 1,091.5648 [M-H]-(calcd for C53H88O23) from HRESIMS.

Notoginsenoside LK2 (2): white, amorphous powder; \([\alpha]^{25}_D\) + 5.6 (c 0.10, MeOH); IR νmax 3,354, 2,926, 1,645, 1,367, 1,163, 1,072, 1,038, 893cm-1. 1H-NMR and 13C-NMR data are shown in Table 1; m/z 1,207.6134 [M-H]-(calcd for C58H96O26) from HRESIMS.

Notoginsenoside LK3 (3): white, amorphous powder; \([\alpha]^{25}_D\) +10.6 (c 0.07, MeOH); IR νmax 3,368, 2,943, 1,650, 1,368, 1,164,1,073, 1,027, 894 cm-1.1H-NMR and 13C-NMR data are shown inTable 1; m/z 1,237.6258 [M-H]-(calcd for C59H98O27) from HRESIMS.

Notoginsenoside LK4 (4): white, amorphous powder; \([\alpha]^{25}_D\) -1.1 (c 0.10, MeOH); IR νmax 3,355, 2,928, 1,667, 1,367, 1,163, 1,073, 1,028,894 cm-1. 1H-NMR and 13C-NMR data are shown in Table 1; m/z 1.223.6122 [M-H]-(calcd for C58H96O27) from HRESIMS.

Notoginsenoside LK5 (5): white, amorphous powder; \([\alpha]^{25}_D\) +2.5 (c 0.08, MeOH); IR νmax 3,344, 2,943, 1,647, 1,366, 1,162, 1,073, 1,027, 897 cm-1. 1H-NMR and 13C-NMR data are shown in Table 2; m/z 1,223.6112 [M-H]-(calcd for C58H96O27) from HRESIMS.

Notoginsenoside LK6 (6): white, amorphous powder; \([\alpha]^{25}_D\) +2.2 (c 0.09, MeOH); IR νmax 3,342, 2,881, 1,650, 1,367, 1,163, 1,075, 1,027, 896 cm-1. 1H-NMR and 13C-NMR data are shown in Table 2; m/z 1,093.5806 [M-H]-(calcd for C53H90O23) from HRESIMS.

Notoginsenoside LK7 (7): white, amorphous powder; \([\alpha]^{25}_D\) -11.1 (c 0.09, MeOH); IR νmax 3,353, 2,925, 1,666, 1,367, 1,167, 1,078, 1,027, 894 cm-1. 1H-NMR and 13C-NMR data are shown in Table 2; m/z 1,093.5853 [M-H]-(calcd for C53H90O23) from HRESIMS.

Notoginsenoside LK8 (8): white, amorphous powder; \([\alpha]^{25}_D\) +2.2 (c 0.10, MeOH); IR νmax 3,339, 2,885, 1,650, 1,368, 1,170, 1,077, 1,027,895cm-1.1H-NMR and 13C-NMR data are shown in Table 2; m/z 1.931.5287 [M-H] (calcd for C47H80O18) from HRESIMS.

2.4. Acid hydrolysis of compounds 1-8

Compounds 1-8 (1 mg each) were hydrolyzed with trifluoroacetic acid (TFA) (2 mol/L) at 120°Cfor 2 h. After cooling, the remaining acid was taken away with repeat evaporation of MeOH. And then the residue was dissolved in water and removed off impurities with ether. After concentration of the water layer, the residue and authentic samples were analyzed for a comparison through TLC.

2.5. Absolute configuration of sugars

The sugar mixtures were obtained as described above. Further derivatizations were conducted according to the method reported [10]. In short, the residue dissolved in anhydrous pyridine was reacted at 60°Cfor 1 h with L-cysteine methyl ester hydrochloride followed by addition of o-tolylisothiocyanate and heat treatment of another 1 h. HPLC-UV was used to analyze the reaction mixture we got.

Table 1 13C NMR (150 MHz) and 1H NMR (600 MHz) data of compounds 1-4

2.6. Determination of NO production

Compounds 1-15 (purity >95%) were dissolved in dimethyl sulfoxide (DMSO), and the final concentration of DMSO was 0.1%. A pretreatment of each compound for 30 min was conducted in RAW 246.7 cells and then followed by a stimulation of LPS (100 ng/ml) for 24 h. DMSO was used as negative control and 20(S)-ginsenoside Rg3 (100 μM) as positive group [11]. All compounds were tested in nontoxic concentration. The Griess reaction was carried out to measure the NO production [12]. Briefly, 80 ml of cell culture supernatant was added into Griess reagent in the same volume and reacted for 10 min. Then the absorbance at 540 nm was obtained from a microplate reader.

Table 2 13C NMR (150 MHz) and 1H NMR (600 MHz) data of compounds 5-8

2.7. Measurement of inflammation-related mRNA expressions of gypenoside IX treated cells

RAW 246.7 cells were pretreated with gypenoside IX (100 μM) or ginsenoside Rg3 (100 μM) for 30 min and then stimulated with LPS (100 ng/ml) for 4 h. Total RNA was extracted using RNA fast 200 (Fastagen, Shanghai, China) and reverse transcribed into cDNA with PrimeScript RT Master Mix (TaKaRa-Bio, Otsu, Japan), according to the manufacturer instructions. The cDNA was amplified quantitatively using SYBR Premix Ex Taq (TaKaRa-Bio, Otsu, Japan). The the forward and reverse primers were 5'-CTCTTCTCATTCCTGCTTGT-3'and 5'-GTGGTTTGTGAGTGTGAGG-3'for mouse Tnf-ɑ, 5'-CCTGGTAGAAGTGATGCC-3'and 5'-ACTGCCTT-GCTCTTATTTTC-3'for Il-10, 5'-CCTATGGCCCTCATTCTCAC-3'and 5'-CGTCATTTTCTGCCTCATCCT-3'for Cxcl10, and 5'-TTAGTCCTCGGC-CAAGACAG-3'and 5'-GGCAAGGAGGAAAACACAGG-3' for Il-1β. Real-time reverse transcription polymerase chain reaction was performed using an ABI ViiA 7 Real time PCR system (Applied Biosystems, CA, USA). The data was normalized to β-Actin mRNA.

2.8. Statistical analysis

Data from three independent experiments were presented as the mean ± standard deviation. Student’s t test and Prism Graphpad program were used in the data analysis and calculation, respectively.

3. Results

3.1. Structure elucidation of new compounds

Compound 1 had a molecular formula C53H88O23 deduced from m/z 1091.5648 (M-H) - in the HRESIMS. The D-glucose and Dxylose were suggested from the acid hydrolysis of 1, and their relative configurations were determined as β because of signals at δ 4.94 (d, 7.2 Hz), 4.93 (d, 7.5 Hz), 5.12 (d, 8.0 Hz), and 5.40 (d, 7.5 Hz). Seven aglycone methyls were suggested from corresponding singlets at δ 0.8, 0.9, 1.0, 1.1, 1.3, 1.6, and 1.8 and olefinic protons from two singlets at δ 6.4 and 5.7 in the 1H-NMR spectrum. In addition, a carbonyl group (δ 203.0) and a terminal double bond (δ 144.7 and 125.8) in the 13C-NMR spectrum were also observed. (cid:3) According to the above characteristic peaks, compound 1 was indicated to show the same aglycone as notoginsenoside B, especially the α, β-unsaturated ketone in the side chain [13]. Apart from 30 signals due to the skeleton, the remaining signals of compound 1 were assigned to sugar chain and identical to those of ginsenoside Rb3, especially four anomeric carbon signals at δ 106.4, 106.4, 105.5, and 98.3 [14]. The positions of the functional groups in the side chain were further determined by observation of the correlations between H3-27 (δ 1.84) and C-24, C-25, and C-26 (δ 203.0, 144.7 and 125.8) and H2-26 (δ 6.38 and 5.72) and C-24, C-25 and C-27 (δ 18.2) in the heteronuclear multiple bond correlation (HMBC) spectrum. The glycosylations were indicated to happen at C-3 and C-20 from the downfield signals at the corresponding position. Besides, the correlation between C-3 and H-1 from the glc, and C-20 and H-1 from the glc'' further corroborated the finding. Sugar sequences were indicated to be a 1/2 linkage type attaching to C-3 and 1/6 to C-20 from the correlations between signals at δ 83.8 and δ 4.93 and δ 70.5 and δ 5.40.

Accordingly, compound 1 was identified as 3-O-[β-D-glucopyranosyl(1→2)-β-D-glucopyranosyl]-20-O-[β-D-xylopyranosyl (1→6)-β-D-glucopyranosyl]3β,12β,20(S)-trihydroxydammar-25-en-24-one (Fig. 1).

Fig. 1. The structures of compounds 1-15. Ara(f), a-L-arabinofuranosyl; Ara(p), α-L-arabinopyranosyl; Glc, β-D-glucopyranosyl; Xyl, β-D-xylopyranosyl

Compound 2 was corresponded to a molecular formula of C58H96O26 determined from m/z 1207.6134 (M-H)-in the HRESIMS. The absolute configurations of the glucose and xylose were proved to be D and arabinose to be L by acid hydrolysis of 2. The distinctive carbon signals at δ 143.0, 136.0, 127.8, and 115.3 suggested a conjugated double bond; the position of which was deduced from the correlations between H3-27 (δ 1.95) and C-24, C-25, and C-26 (δ 136.0, 143.0 and 115.3) and H2-26 (δ 5.04, 4.95) and C-24, C-25, and C-27 (δ 19.3) in the HMBC experiment. Furthermore, signals attributed to the aglycone particularly the above characteristic peaks were shown to be identical to those of quinquenoside L1 [15]. Sugar chains were deduced to consist of β-D-glucopyranosyl, β-D-xylopyranosyl, and a-L-arabinofuransyl unit from the signals for five anomeric carbons along with the coupling constants of the corresponding protons. The trisaccharide moiety was suggested to attach at C-3 and the disaccharide moiety at C-20 of the aglycone from the correlations observed between signals at δ 4.94 and δ 89.3 and δ 5.18 and δ 83.7. Then we found that the remaining 28 signals for the sugar parts showed consistence with those of ginsenoside FP2 (11) [16]. Finally, the structure of compound 2 was determined as 3-O-[β-D-xylopyranosyl(1→2)-β-D-glucopyranosy (1→2)-β-D-glucopyranosyl]-20-O-[α-L-arabinofuransyl(1→6)-β-D-glucopyranosyl]3β,12β,20(S)-trihydroxydammar-23,25-diene.

Compound 3 possessed a molecular formulaC59H98O27 established from m/z 1,237.6258 (M-H)-in the HRESIMS. The absolute configurations of glucose and xylose were all determined as D according to the HPLC chromatogram of the acid hydrolysate. NMR spectra of 3 showed high similarities with those of notoginsenoside LK2 (2). The only difference laid at signals due to the sugar chain, which were consistent with those of notoginsenoside Fa [17]. Based on this elucidation along with two-dimensional spectra, such as heteronuclear single quantum correlation (HSQC) and HMBC, compound 3 was identified as 3-O-[β-D-xylopyranosyl(1→2)-β-D-glucopyranosy(1→2)-β-D-glucopyranosyl]-20-O-[β-D-glucopyranosy(1→6)-β-D-glucopyranosyl]3β,12β,20(S)-trihydroxydammar-23, 25-diene.

Compound 4 was corresponded to a molecular formula C58H96O27 deduced from m/z 1,223.6122 (M-H)-in the HRESIMS. The absolute configurations of glucose and xylose were shown to be D. The carbon and proton signals of 4 agreed mostly with those of notoginsenoside LK1 (1), especially the distinctive signals at δ 144.7 and δ 125.8 due to olefinic carbons and the signals at δ 203.0 assigned to a carbonyl group. The only differences were five more carbon signals due to a xylopyranosyl unit in 4 than 1. Furthermore, the sugar sequences of 4 were shown the same as those of notoginsenoside Fc (10) [17]. Finally, based on this clarification above and two-dimensional spectra, compound 4 was deduced as 3-O-[β-D-xylopyranosyl(1→2)-β-D-glucopyranosyl(1→2)-β-D-glucopyranosyl]-20-O-[β-D-xylopyranosyl(1→6)β-D-glucopyranosyl]3β,12β,20(S)-trihydroxydammar-25-en-24-one.

Compound 5 possessed a molecular formula C58H96O27 deter-(cid:3) . The absolute configurations of mined from m/z 1,223.6112 (M-H)- the glucose, xylose, and arabinose from acid hydrolysate of compound 5 were proved to be D, D, and L, respectively. The carbon and proton signals of 5 agreed well with those of notoginsenoside LK4 (4). The only difference was the replacement of the terminal-β-D-xylopyranosyl moiety at C-20 in 4 by an α-L-arabinopyranosyl group in notoginsenoside LK5. At last, in view of the illuminations above and detailed displays of two-dimensional spectra, compound 5 was identifiedas 3-O-[β-D-xylopyranosyl(1→2)-β-D-glucopyranosyl(1→2)-β-D-glucopyranosyl]-20-O-[α-L-arabinopyranosyl(1→6)-β-D-glucopyranosyl]3β,12β,20(S)-trihydroxydammar--25-en-24-one (Fig. 1).

Compound 6 had a molecular formula C53H90O23 deduced from (cid:3) . The absolute configuration of glucose and m/z 1,093.5806 (M-H)- xylose were all proved to be D by HPLC analysis. The unique signals at δ 110.7 and 149.4 at low field were assigned to olefinic carbons and signal at δ 76.4 was due to oxygenated carbon, which indicated a terminal double bond and a hydroxyl group. The 13C-NMR spectrum of 6 matched well with those of gypenoside LXXI (9) [18], except for slight difference of the signals at C-24 and C-26 between 6 and 9. Specifically, signals went to high field by movement of δ 0.1 for C-24 and δ 0.5 for C-26 from 6 to 9 (6: δ 76.4 at C-24, δ 110.7 at C-26; 9: δ76.3 at C-24, δ 110.3 at C-26), which suggested 6 and 9 were obtained as a pair of C-24 epimers. Furthermore, the configuration at C-24 in the 6 was identified to be R after a comparison of signals between 6 and reported C-24 epimers, such as vina-ginsenoside R9 and majoroside F1 [19]. Accordingly, compound 6 was deduced as 3-O-[β-D-glucopyranosyl(1→2)-β-D-glucopyranosyl]-20-O-[β-D-xylopyranosyl(1→6)-β-D-glucopyranosyl]3β,12β,20(S)-trihydrox-ydammar-24(R)-hydroxyl-25-ene.

Compound 7 was assigned to a molecular formula C53H90O23(cid:3) . The 13C-NMR and 1H-NMR established by m/z 1,093.5853 (M-H)- spectrum of 7 resembled to those of notoginsenoside LK6. The only difference was an α-L-arabinofuranosyl group in notoginsenoside LK7 by substitution of a terminal-β-D-xylopyranosyl moiety at C-20 position in 6. Accordingly, compound 7 was identified as 3-O-[β-D-glucopyranosyl(1→2)-β-D-glucopyranosyl]-20-O-[α-L-arabinofuranosyl (1→6)-β-D-glucopyranosyl]3β,12β,20(S)-trihydroxydammar-24(R)-hydr oxyl-25-ene.

Compound 8 was assigned to a molecular formula C47H80O18 deduced from HRESIMS. The 13C-NMR spectra of 8 and notoginsenoside Ft3 showed high similarities but a slight difference in signals for C-24 and C-26 [20]. However, the structure of notoginsenoside Ft3 was not fully determined owing to the unconfirmed hydroxyl group at C-24 position. In our study, the configuration at C-24 of compound 8 was suggested to be S from the higher-field shift at δ 76.3 (C-24) and δ 110.2 (C-26). Accordingly, compound 8 was identified as 3-O-[β-D-xylopyranosyl(1→2)-β-D-glucopyranosyl (1→2)-β-D-glucopyranosyl]3β,12β,20(S)-trihydroxydammar-24(S)-hy droxyl-25-ene.

3.2. Structure identification of known compounds

The known compounds 9-15 were identified as gypenoside LXXI (9) [18], notoginsenoside Fc (10) [17], notoginsenoside FP2 (11) [16], notoginsenoside Fz (12) [8], ginsenoside Rc (13) [14], ginsenoside Rb3 (14) [14] and gypenoside IX (15) [21] compared to the NMR and MS data reported.

3.3. Antiinflammatory activity

Previous reports have revealed that saponins exerted antiinflammatory activity, and therefore all the isolated compounds were screened for antiinflammatory activity by measuring NO production of LPS-stimulated Raw 264.7 cells. As the results indicate (Fig. 2), only pretreatment of gypenoside IX at noncytotoxic concentration (100 mM) significantly decreased the NO production to 72.0% compared with the control group (100%), which indicated the antiinflammatory activity of gypenoside IX. Furthermore, the expressions of inflammation-related genes Tnf-α, Il-10, Cxcl10, and Il-1β were suppressed by gypenoside IX (Fig. 3). Taken together, gypenoside IX has the potential to be used as an antiinflammatory agent.

Fig. 2. Effect of compounds 1e15 on nitric oxide (NO) production in LPS-stimulated RAW 264.7 cells. The data were presented as ratio (%) compared to the group of DMSO-treated cell (control group). 20(S)-Rg3 was used as positive control. Compounds, which showed less than 80% of NO production, can be regarded as the potential antiinflammatory inhibitors. Data from three independent experiments were expressed as the mean ± SD. **p < 0.01 and *p < 0.05 versus the control group. DMSO, dimethyl sulfoxide; LPS, lipopolysaccharide; Rg3, 20(S) ginsenoside Rg3; SD, standard deviation.

Fig. 3. Effect of gypenoside IX. (A) On Tnf-α expression in the LPS-stimulated RAW 264.7 cells. (B) Il-10 expression in the LPS-stimulated RAW 264.7 cells. (C) Cxcl10 expression in the LPS-stimulated RAW 264.7 cells. (D) Il-1β expression in the LPS-stimulated RAW 264.7 cells. Data from six independent experiments were expressed as the mean ± SD. **p < 0.01 and *p < 0.05 versus the Model group. LPS (100 ng/ml) were used to build the model. Cxcl10, interferon-inducible protein 10; Il-10, interleukin 10; Il-1b, interleukin-1β; IX, gypenoside IX; LPS, lipopolysaccharide; Rg3, 20(S) ginsenoside Rg3; SD, standard deviation; Tnf-α, tumor necrosis factor-α.

4. Discussion

PNLS were rich in dammarane-type saponins and could be considered as an alternative source of bioactive saponins. In the present study, eight new notoginsenosides bearing dehydrogenated or oxidized side chains along with seven known PPD-type saponins with common side chains have been isolated and identified from PNLS. Among them, compounds1, 4, and 5 showed the same α. β-unsaturated ketone as previously isolated saponins, such as notoginsenoside B from the dried root of P. notoginseng, ginsenoside III from the flower buds of P. ginseng, and vina-ginsenoside R20 from the roots of Panax vietnamensis Ha et Grushv. [13,22,23]. It has been indicated that this kind of side chain existed widely in genus Panax. Before our study, only quinquenaside L1 from the leaves and stems of P. quinquefolium was reported to bear a side chain with conjugated double bonds [15]. The discovery of 2 and 3 has greatly enhanced the chemical diversity of saponins of this type. In addition, this is the first report on a pair of C-24 epimers [notoginsenoside LK6 (6) and gypenoside LXXI (9)] from P. notoginseng, although this kind of C-24 epimers (vina-ginsenoside R9 and majoroside F1) has been previously isolated from P. vietnamensis [19]. In the previous study, 7 and 8 with undefined configurations at C-24 were proposed as floranotoginsenoside D and notoginsenoside Ft3, respectively [20,24]. Besides, only 1D NMR and HRESIMS without 2D NMR were listed for the identification of floranotoginsenoside D, and acid treatment was used to obtain notoginsenoside Ft3 from leaves of P. notoginseng. In this study, the configurations of 7 and 8 were determined by comparing their chemical shifts to those of the C-24 epimers (6 and 9). Moreover, the structure of 7 was completely characterized by chemical and spectroscopic methods, and 8 was formed naturally.

Gypenoside IX was firstly isolated from Gynostemma pentaphyllum Makino and found in P. notoginseng and P. ginseng [25,26]. Although many reports demonstrated that G. pentaphyllum and gypenosides from this plant displayed antiinflammatory properties [27e29], there have been very few reports of gypenoside IX about antiinflammatory activity. Our data showed the moderate antiinflammatory effect of gypenoside IX, which could be served as an important pharmalogical active saponin in the genus of Panax or Gynostemma.

According to our data (Fig. 2), isolated saponins with four or five sugar units such as ginsenoside Rb3 (14) and 1-7 showed no effect

on our experiment model. However, Ma et al [30] reported that ginsenoside Rb3 protected cardiomyocytes against ischemiareperfusion injury by suppressing the nuclear factor kappa B pathway. Ginsenoside Rb1 with similar structure to ginsenoside Rb3 was reported to be metabolized after oral administration to compound K and then to present bioactivities on treating allergic inflammation [31]. Hence, it may be suggested that 1-7 might show effects by metabolizing to less polar saponins through orally administration. Thus, further pharmacological studies of these new compounds in vivo models should be processed.

In this study, we have isolated fifteen dammarane-type triterpene oligoglycosides especially eight new ones from the PNLS, which may enrich and expand the chemical library of saponins in Panax plants. Morever, gypenoside IX as a PPD-type oligoglycoside was demonstrated to show antiinflammatory effect by suppressing the NO production and inflammation-related genes in LPS-stimulated RAW 264.7 cells. Further investigation is expected to increase insight into mechanisms of antiinflammation of gypenoside IX.

Conflicts of interest

All authors declare no conflicts of interest.

Acknowledgments

This work was financially supported by the National Natural Science Foundation of China (81530096, 81573581, 81603229, 81603156) and China Postdoctoral Science Foundation (2016T90382) as well as Young Eastern Scholar Program (QD2016038) and Chenguang Program (16CG49) supported by Shanghai Education Devel- opment Foundation and Shanghai Municipal Education Commission.

References

  1. Yang WZ, Hu Y, Wu WY, Ye M, Guo DA. Saponins in the genus Panax L. (Araliaceae): a systematic review of their chemical diversity. Phytochemistry 2014;106:7-24. https://doi.org/10.1016/j.phytochem.2014.07.012
  2. Kim DH. Chemical diversity of Panax ginseng, Panax quinquifolium, and Panax notoginseng. J Ginseng Res 2012;36:1-15. https://doi.org/10.5142/jgr.2012.36.1.1
  3. Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell 2005;3:211-7. https://doi.org/10.1016/j.ccr.2005.02.013
  4. Yu T, Yang YY, Kwak YS, Song GG, Kim MY, Rhee MH, Cho JY. Ginsenoside Rc from Panax ginseng exerts anti-inflammatory activity by targeting TANKbinding kinase 1/interferon regulatory factor-3 and p38/ATF-2. J Ginseng Res 2017;41.
  5. Kim DH, Chung JH, Yoon JS, Ha YM, Bae SJ, Lee EK, Jung KJ, Kim MS, Kim YJ, Kim MK, et al. Ginsenoside Rd inhibits the expressions of iNOS and COX-2 by suppressing $NF-{\kappa}B$ in LPS-stimulated RAW264. 7 cells and mouse liver. J Ginseng Res 2013;37:54-63. https://doi.org/10.5142/jgr.2013.37.54
  6. Guo XJ, Zhang XL, Feng JT, Guo ZM, Xiao YS, Liang XM. Purification of saponins from leaves of Panax notoginseng using preparative two-dimensional reversed-phase liquid chromatography/hydrophilic interaction chromatography. Anal Bioanal Chem 2013;405:3413-21. https://doi.org/10.1007/s00216-013-6721-8
  7. Guo XJ, Zhang XL, Guo ZM, Liu YF, Shen AJ, Jin GW, Liang XM. Hydrophilic interaction chromatography for selective separation of isomeric saponins. J Chromatogr A 2014;1325:121-8. https://doi.org/10.1016/j.chroma.2013.12.006
  8. Li DW, Cao JQ, Bi XL, Xia XC, Li W, Zhao YQ. New dammarane-type triterpenoids from the leaves of Panax notoginseng and their protein tyrosine phosphatase 1B inhibitory activity. J Ginseng Res 2014;38:28-33. https://doi.org/10.1016/j.jgr.2013.11.013
  9. Mao Q, Yang J, Cui XM, Li JJ, Qi YT, Zhang PH, Wang Q. Target separation of a new anti-tumor saponin and metabolic profiling of leaves of Panax notoginseng by liquid chromatography with eletrospray ionization quadrupole time-of-flight mass spectrometry. J Pharmaceut Biomed 2012;59:67-77. https://doi.org/10.1016/j.jpba.2011.10.004
  10. Tanaka T, Nakashima T, Ueda T, ToMii K, KouNo I. Facile discrimination of aldose enantiomers by reversed-phase HPLC. Chem Pharm Bull 2007;55:899-901. https://doi.org/10.1248/cpb.55.899
  11. Shin YM, Jung HJ, Choi WY, Lim CJ. Antioxidative, anti-inflammatory, and matrix metalloproteinase inhibitory activities of 20(S)-ginsenoside Rg3 in cultured mammalian cell lines. Mol Biol Rep 2013;40:269-79. https://doi.org/10.1007/s11033-012-2058-1
  12. Kim HK, Cheon BS, Kim YH, Kim SY, Kim HP. Effects of naturally occurring flavonoids on NO production in the macrophage cell line RAW 264.7 and their structureeactivity relationships. Biochem Pharmacol 1999;58:759-65. https://doi.org/10.1016/S0006-2952(99)00160-4
  13. Yoshikawa M, Murakami T, Ueno T, Yashiro K, Hirokawa N, Murakami N, Yamahara J, Matsuda H, Saijoh R, Tanaka O. Bioactive saponins and glycosides. VIII. Notoginseng (1): new dammarane-type triterpene oligoglycosides, notoginsenosides-A, -B, -C, and -D, from the dried root of Panax notoginseng (Burk.) F.H. Chen. Chem Pharm Bull 1997;45:1039-45. https://doi.org/10.1248/cpb.45.1039
  14. Liu C, Han JY, Duan YQ, Huang X, Wang H. Purification and quantification of ginsenoside Rb3 and Rc from crude extracts of caudexes and leaves of Panax notoginseng. Sep Purif Technol 2007;54:198-203. https://doi.org/10.1016/j.seppur.2006.09.004
  15. Wang JH, Lia W, Sha Y, Tezuka Y, Kadota S, Li X. Triterpenoid saponins from leaves and stems of Panax quinquefolium L. J Asian Nat Prod Res 2001;3:123-30. https://doi.org/10.1080/10286020108041379
  16. Wang XY, Wang D, Ma XX, Zhang YJ, Yang CR. Two new Dammarane-Type Bisdesmosides from the fruit pedicels of Panax notoginseng. Helv Chim Acta 2008;91:60-6. https://doi.org/10.1002/hlca.200890013
  17. Yang TR, Kasai R, Zhou J, Tanaka O. Dammarane saponins of leaves and seeds of Panax notoginseng. Phytochemistry 1983;22:1473-8. https://doi.org/10.1016/S0031-9422(00)84039-X
  18. Yoshikawa K, Takemoto T, Arihara S. Studies on the constituents of cucurbitaceae plants. XVI. On the saponin constituents of Gynostemma pentaphyllum Makino. (11). Yakugaku Zasshi 1987;107:262-7. https://doi.org/10.1248/yakushi1947.107.4_262
  19. Duc NM, Kasai R, Ohtani K, Ito A, Nham NT, Yamasaki K, Tanaka O. Saponins from Vietnamese ginseng, Panax vietnamensis Ha et Grushv. Collected in central Vietnam. II. Chem Pharm Bull 1994;42:115-22. https://doi.org/10.1248/cpb.42.115
  20. Chen JT, Li HZ, Wang D, Zhang YJ, Yang CR. New Dammarane Monodesmosides from the acidic deglycosylation of Notoginseng-Leaf Saponins. Helv Chim Acta 2006;89(7):1442-8. https://doi.org/10.1002/hlca.200690144
  21. Takemoto T, Arihara S, Nakajima T, Okuhira M. Studies on the constituents of Gynostemma pentaphyllum Makino. I. Structures of Gypenoside IeXIV. Yakugaku Zasshi 1983;103:173-85. https://doi.org/10.1248/yakushi1947.103.2_173
  22. Qiu F, Ma ZZ, Xu SX, Yao XS, Chen YJ, Che ZT. Studies on dammarane-type saponins in the flower-buds of Panax ginseng CA Meyer. J Asian Nat Prod Res 1998;1:119-23. https://doi.org/10.1080/10286029808039853
  23. Duc NM, Kasai R, Yamasaki K, Nham NT, Tanaka O. New dammarane saponins from Vietnamese ginseng. Stud Plan Sci 1999;6:77-82. https://doi.org/10.1016/S0928-3420(99)80010-X
  24. Wang JR, Yamasaki Y, Tanaka T, Kouno I, Jiang ZH. Dammarane-type triterpene saponins from the flowers of Panax notoginseng. Molecules 2009;14(6):2087-94. https://doi.org/10.3390/molecules14062087
  25. Kim JH, Yi YS, Kim MY, Cho JY. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res 2017;41.
  26. Yuan J, Chen Y, Liang J, Wang CZ, Liu XF, Yang ZH, Tang Y, Li JK, Yuan CS. Component analysis and target cell-based neuroactivity screening of Panax ginseng by ultra-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry. J Chromatogr B 2016;1038:1-11. https://doi.org/10.1016/j.jchromb.2016.10.014
  27. Luthje P, Lokman EF, Sandstrom C, Ostenson CG, Brauner A. Gynostemma pentaphyllum exhibits anti-inflammatory properties and modulates antimicrobial peptide expression in the urinary bladder. J Funct Foods 2015;17:283-92. https://doi.org/10.1016/j.jff.2015.03.028
  28. Xie Z, Liu W, Huang H, Slavin M, Zhao Y, Whent M, Blackford J, Lutterodt H, Zhou H, Chen P, et al. Chemical composition of five commercial Gynostemma pentaphyllum samples and their radical scavenging, antiproliferative, and anti-inflammatory properties. J Agr Food Chem 2010;58:11243-9. https://doi.org/10.1021/jf1026372
  29. Yang F, Shi HM, Zhang XW, Yang HS, Zhou Q, Yu LL. Two new saponins from tetraploid jiaogulan (Gynostemma pentaphyllum), and their anti-inflammatory and ${\alpha}$-glucosidase inhibitory activities. Food Chem 2013;141:3606-13. https://doi.org/10.1016/j.foodchem.2013.06.015
  30. Ma LJ, Liu HM, Xie ZL, Yang S, Xu W, Hou JB, Yu B. Ginsenoside Rb3 protects cardiomyocytes against ischemia-reperfusion injury via the inhibition of JNKmediated $NF-{\kappa}B$ pathway: a mouse cardiomyocyte model. PLoS One 2014;9(8), e103628. https://doi.org/10.1371/journal.pone.0103628
  31. Park EK, Shin YW, Lee HU, Kim SS, Lee YC, Lee BY, Kim DH. Inhibitory effect of ginsenoside Rb1 and compound K on NO and prostaglandin E2 biosyntheses of RAW264. 7 cells induced by lipopolysaccharide. Biol Pharm Bull 2005;28:652-6. https://doi.org/10.1248/bpb.28.652

Cited by

  1. Evaluation of Anti-inflammatory Nutraceuticals in LPS-induced Mouse Neuroinflammation Model: An Update vol.18, pp.7, 2019, https://doi.org/10.2174/1570159x18666200114125628
  2. Dammarane-type triterpenoid saponins from Salvia russellii Benth. vol.184, 2021, https://doi.org/10.1016/j.phytochem.2020.112653
  3. Identification of Bioactive Natural Product from the Stems and Stem Barks of Cornus walteri: Benzyl Salicylate Shows Potential Anti-Inflammatory Activity in Lipopolysaccharide-Stimulated RAW 264.7 Mac vol.13, pp.4, 2019, https://doi.org/10.3390/pharmaceutics13040443
  4. Antidiabetic Flavonoids from Fruits of Morus alba Promoting Insulin-Stimulated Glucose Uptake via Akt and AMP-Activated Protein Kinase Activation in 3T3-L1 Adipocytes vol.13, pp.4, 2021, https://doi.org/10.3390/pharmaceutics13040526
  5. Neuroprotective triterpene saponins from the leaves of Panax notoginseng vol.35, pp.14, 2019, https://doi.org/10.1080/14786419.2019.1677657
  6. Restoring perturbed oxylipins with Danqi Tongmai Tablet attenuates acute myocardial infarction vol.90, 2019, https://doi.org/10.1016/j.phymed.2021.153616