Next Article in Journal
Chemical Fingerprint Analysis and Quantitative Analysis of Rosa rugosa by UPLC-DAD
Previous Article in Journal
Polyphenol Composition, Antioxidant Activity and Cytotoxicity of Seeds from Two Underexploited Wild Licania Species: L. rigida and L. tomentosa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Iridoid Glucosides from Caryopteris incana (Thunb.) Miq. and Their α-Glucosidase Inhibitory Activities

1
The MOE Key Laboratory of Standardization of Chinese Medicines, and SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China
2
Shanghai R&D Center for Standardization of Chinese Medicines, Shanghai 201203, China
*
Author to whom correspondence should be addressed.
Submission received: 10 October 2016 / Revised: 1 December 2016 / Accepted: 9 December 2016 / Published: 21 December 2016
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
In our continued investigations of the plant Caryopteris incana, five new iridoid glucosides 15, including two cis-trans-isomers, 3 and 4, along with six known compounds 611, were isolated from the n-butyl alcohol (n-BuOH) soluble fraction of whole dried material of Caryopteris incana. Their structures were established by a combination of spectroscopic techniques, including 1D and 2D NMR and high resolution electrospray ionization mass spectroscopy (HR-ESI-MS). Furthermore, all isolates were evaluated for their yeast α-glucosidase inhibitory effects. Among these compounds, 48 and 10 exhibited potent inhibition of α-glucosidase.

Graphical Abstract

1. Introduction

The incidence of obesity and diabetes in humans is high and continues to increase. In particular, type-II diabetes mellitus (T2DM) is one of the most common chronic diseases in nearly all countries, and it continues to be an increasing international health burden and hidden killer [1]. Since the early 1990s, comparatively effective glucosidase inhibitors have been generally available to type-II diabetics in order to depress postprandial hyperglycemia induced by the digestion of carbohydrates in the intestines [2]; however, they are accompanied by side effects.
Traditional Chinese medicines are known to be a potential rich source, and chemical pool, for finding effective treatment agents with mild or no side effects. Caryopteris incana (Thunb.) Miq., known in China as “Lanxiangcao”, is a Chinese folk medicine for the relief of colds, pertussis, rheumatic pains, bronchitis, eczema, gastroenteritis, itchy skin, and venomous snake bites [3]. Up to now, there have been many reports regarding its chemical constituents, such as diterpenes [4], iridoids [5], flavonoids [6], and phenolic glycosides [7].
Recently, we reported 12 new diterpenes caryopincaolide A–L, including four, new, rearranged abietanes, caryopincaolide A–D, with unprecedented skeletons, and twenty-eight known diterpenes, isolated from the petroleum ether and ethyl acetate soluble fractions of whole plant of C. incana; additionally, some compounds exhibited moderate inhibitory effects against dipeptidyl peptidase IV (DPP-IV) [8]. Furthermore, in the previous literature, some iridoid glycosides and phenylpropanoid glycosides displayed potential α-glucosidase inhibitory activities [9,10]. Thus, the aim of the present work was to continue phytochemical investigations on the n-butyl alcohol soluble fraction of this plant in order to discover effective hypoglycaemic agents to remedy serious T2DM.
In this paper, the isolation and structural identification of five new iridoid glycosides are described: Caryocanoside B (1), 5-hydroxy-2′′′-O-caffeoylcaryocanoside B (2), 2′′′-O-(E)-p-coumaroyl caryocanoside B (3), 2′′′-O-(Z)-p-coumaroyl caryocanoside B (4), and 2′-O-(E)-p-coumaroyl asystasioside A (5) (Figure 1). Six known compounds were isolated and determined by comparison of the physical data with those reported in the literature: Two iridoid glycosides, 8-O-acetylharpagide (6) [11] and 8-acetyl-6′-O-(p-coumaroyl) harpagide (7) [12], two phenethyl alcohol glycoside, galactosylmartynoside (8) [13] and 6‴-O-feruloylincanoside D (9) [14], an abietatriene-type diterpene glycoside, ajugaside A (10) [13], and a flavonoid luteolin 7-O-glucoside (11) [15], respectively. All compounds were tested for in vitro α-glucosidase inhibitory activity; compounds 4 and 10 possess α-glucosidase inhibitory effects with IC50 values of 0.377 mM and 0.328 mM, respectively.

2. Results

Compound 1, was obtained as a yellow powder, [ α ] D 20 = −0.098 (c 0.1, MeOH), its molecular formula, C33H48O18, was established by its high resolution electrospray ionization mass spectroscopy (HR-ESI-MS) ion peak (m/z 731.2802 [M − H]), which corresponded to 10 units of unsaturation. The absorptions in the infrared (IR) spectrum at 3357 and 1633 cm−1 showed the existence of hydroxy and carbonyl groups. The 1H NMR spectrum (Table 1) of CD3OD revealed characteristic of bis-iridoid glucoside signals attributed to the disubstituted olefinic protons at δH 6.26 (1H, dd, J = 2.4, 6.0 Hz, H-3), δH 4.77 (1H, brd, J = 6.0 Hz, H-4), a trisubstituted olefinic proton at δH 7.42 (1H, s, H-3′), two hemiacetal protons at δH 5.87 (1H, s, H-1), δH 5.47 (1H, d, J = 4.4, H-1′) together with two β-anomeric protons at δH 4.68 (1H, d, J = 8.0 Hz, H-1″), δH 4.64 (1H, d, J = 8.0 Hz, H-1‴). The 13C NMR spectrum (Table 1) showed the presence of two β-glucopyranosyl moieties and two carbonyl groups, suggesting that 1 was an ester dimmer of iridoid glycoside. By comparison of the 13C-NMR data with those known iridoid glycosides, one unit (A) (Figure 1) of 1 was determined to be ajugoside [16,17], and another (B) (Figure 1) to be 7-deoxy-8-epi-loganic acid [18]. The main difference between unit A and ajugoside was that the C-6 in unit A moved to the downfield region (from δC 76.9 to 79.4), and unit B, between 7-deoxyloganic acid, was an upfield shift of C-11′ in unit B (from δC 172.2 to 168.4), indicating that units A and B were connected via esterification between the hydroxyl group at C-6 of ajugoside (unit A) and the carboxylic acid group of 7-deoxyloganic acid (unit B), which was confirmed by heteronuclear multiple bond correlation (HMBC) (Figure 2) correlation between δH 4.97 (H-6) and δC 168.4 (C-11′). HMBC of 1 showed the following correlations: H-1/C-5, C-8; H-3/C-1, C-5; H-4/C-6, C-9; H-5/C-4, C-6, C-8; H-7/C-5, C-9, C-10; H-10/C-8 and H-1′/C-5′, C-8’; H-3′/C-1′, C-5′, C-11′; H-7′/C-5′, C-9′, C-10′; H-10′/C-8′, which further proves the existence of unit A and B. The Nuclear Overhauser Effect Spectroscopy (NOESY) (Figure 3) experiment established the stereochemistry of 1; for unit A, the correlations between δH 5.87 (1H, s, H-1) and 1.58 (3H, s, H-10), δH 4.97 (1H, d, H-6) and H-10; and between δH 2.98 (1H, brd, H-5) with 2.81 (1H, brd, H-9) revealed that H-1, H-6, and H-10 were on the same face, an α-orientation, whereas H-5 and H-9 were on the opposite face, a β-position, showing that the relative configuration of unit A was identical to ajugoside. For unit B, obvious Nuclear Overhauser Effect (NOE) cross-peaks of δH 5.47 (1H, d, H-1′) with 1.08 (3H, d, H-10′), H-1′ with δH 1.38 (1H, m, Hα-7); and δH 2.91 (1H, brdd, H-5′) with 2.07 (1H, m, Hβ-6′), H-5′ with δH 2.27 (1H, m, H-9′) were observed, demonstrating H-5′and H-9′ to be of a β-orientation, and H-1′ and H-10′ to be in the α-position. The relative configuration of unit B was also the same as 7-deoxy-8-epi-loganic acid. Thus, compound 1 was an ester dimer connected by an ester bond and the bridging ester bond was located at the 6-position of ajugoside, and was named caryocanoside B (shown in Figure 1).
Compound 2, a yellow powder, [ α ] D 20 = −0.266 (c 0.1, MeOH), with a molecular formula of C42H54O22, was established from HR-ESI-MS data ([M − H], m/z 909.3038), requiring 15 degrees of unsaturation. IR spectra indicated the presence a hydroxyl group at 3357 cm−1, a carbonyl group at 1704 cm−1 and an olefinic group at 1669 cm−1 for 2. The NMR spectra of 2 (Table 1) had a close structural similarity to those of compound 1, except for the addition of a hydroxyl at C-5 (δC 72.7), and a caffeoyl group approved by a set of ABX-type aromatic protons at δH 7.06 (1H, d, J = 2.0 Hz, H-2′′′′), 6.97 (1H, dd, J = 8.0, 2.0 Hz, H-6′′′′) and 6.79 (1H, d, J = 8.0 Hz, H-5′′′′); δC 168.3 (C-9′′′′), δC 114.9 (C-8′′′′), and δC 147.1 (C-7′′′′). The connectivity of these fragments was supported by a careful analysis of 2D-NMR (heteronuclear single quantum correlation (HSQC), 1H-1H correlation spectroscopy (1H-1H COSY), and HMBC) spectra, which corroborated that the caffeoyl group was located at C (2′′′), based on the HMBC correlation of δH 4.79 (1H, d, H-2′′′) with δC 168.3 (C-9′′′′). The relative configuration of compound 2 was established by its NOESY, and is the same as 1 (All spectroscopic data are available from Supplementary Material). Therefore, structure 2 was elucidated, named 5-hydroxy-2′′′-O-caffeoylcaryocanoside B, and is shown in Figure 1.
Compound 3 was isolated as a white amorphous powder, [ α ] D 20 = −0.168 (c 0.1, MeOH), and its molecular formula of C42H54O20 was determined from HR-ESI-MS data (m/z 877.3147 [M − H]). The similarity of the IR and NMR data (Table 1) of 3 and 1, suggested that 3 was also a derivative of 1. The main difference was that 3 presented a p-coumaroyl group, with signals at δH 6.26 (1H, d, J = 15.6 Hz, H-8′′′′), δH 7.56 (1H, d, J = 15.6 Hz, H-7′′′′), δH 7.47 (2H, d, J = 8.4 Hz, H-2′′′′ and H-6′′′′), δH 6.83 (2H, d, J = 8.4 Hz, H-3″′ and H-5′′′′); δC 168.1 (C-9′′′′), 115.0 (C-8′′′′), 146.5 (C-7′′′′), 127.2 (C-1′′′′), 131.4 (C-2′′′′ and C-6′′′′), 116.9 (C-3′′′′ and C-5′′′′), and 161.2 (C-4′′′′). Additionally, the coumaroyl group was attached at C-2′′′, as confirmed by HMBC correlation of δH 4.79 (1H, t, H-2‴) with δC 168.1 (C-9′′′′). The relative configuration of 3 was identical to that of 1, which was determined from its NOESY spectrum. Thus, structure 3 was elucidated as 2′′′-O-(E)-p-coumaroyl caryocanoside B and is shown in Figure 1.
Compound 4 was obtained as a white amorphous powder; [ α ] D 20 = −0.09 (c 0.1, MeOH). The HR-ESI-MS data (m/z 901.3092 [M + Na]+) and 13C NMR data (Table 1) of 4 indicated that 4 has the same molecular formula as that of compound 3. The only significant difference was the replacement of the (E)-p-coumaroyl group in 3 by the (Z)-p-coumaroyl group in 4. This was supported by the coupling constant (J = 12.8 Hz) of H-7′′′′ and H-8′′′′. Therefore, compound 4 was determined to be a cis-isomer of 3, and was named as 2′′′-O-(Z)-p-coumaroyl caryocanoside B (shown in Figure 1).
Compound 5 was obtained as a pale yellow powder; [ α ] D 20 = −0.058 (c 0.1, MeOH). It was shown to have a molecular formula of C31H40O16 from the [M − H] ion at m/z 667.2257, determined by HR-ESI-MS. The IR spectrum indicated the presence of a hydroxy at 3357 cm−1, carbonyl groups at 1698 cm−1, an olefinic group at 1632 cm−1, and a phenyl at 1603 cm−1. The similarity in the NMR spectroscopic data of 5 (Table 2) and those of asystasioside A [19] suggested that 5 is a derivative of asystasioside A. Interpretation of the NMR spectra of 5 allowed us to find an additional (E)-p-coumaroyl group in its structure; key signals at δH 6.29 (1H, d, J = 15.6 Hz, H-8′′′), δH 7.60 (1H, d, J = 15.6 Hz, H-7′′′), δH 7.52 (2H, d, J = 8.4 Hz, H-2′′′ and H-6′′′), and δH 6.83 (2H, d, J = 8.4 Hz, H-3′′′ and H-5′′′). The correlation between δH 4.81 (1H, overlap, H-2′) with δC 168.5 (C-9′′′) in the HMBC data (Figure 4) of 5 showed that the coumaroyl group was located at C-2′. In addition, the HMBC correlations from δH 4.83 (1H, overlap, H-1′) to δC 95.6 (C-1), and from δH 5.08 (1H, d, J = 8.4 Hz, H-1″) to δC 167 (C-11), supported that two β-glucopyranosyl moieties were at positions C-1 and C-11. The stereochemistry of 5 was determined from its NOESY spectrum (Figure 5), which displayed NOE cross-peaks of δH 5.50 (1H, d, H-1) with 1.24 (1H, m, Hα-7), H-1 with δH 1.03 (3H, d, H-10); δH 2.82 (1H, m, H-5) with 1.92 (1H, m, Hβ-6), H-5 with δH 2.27 (1H, m, H-9), indicated that 5 had the same relative configuration as asystasioside A. Thus, on the basis of the above-mentioned data, the structure of 5 was established as 2′-O-(E)-p-coumaroyl asystasioside A.
Compounds 611 (Figure 6) were identified by comparison with the literature. They were identified as 8-O-acetylharpagide (6), 8-acetyl-6′-O-(p-coumaroyl) harpagide (7), galactosylmartynoside (8), 6′′′-O-feruloylincanoside D (9), ajugaside A (10), and luteolin 7-O-glucoside (11).
The isolated compounds were evaluated for their inhibitory effects against α-glucosidase. Compounds 4 and 10 exhibited stronger inhibitory effects against α-glucosidase than the positive control, acarbose, with IC50 values of 0.377 mM and 0.328 mM, respectively (Table 3).

3. Experimental Section

3.1. General

IR spectra were measured on a PerkinElmer FT-IR spectrometer (PerkinElmer, Shanghai, China). Optical rotations were measured on an Autopol VI (Rudolph Research Analytical, Hackettstown, NJ, USA). A Bruker AV-400 spectrometer and Bruker AV-600 spectrometer (Bruker Co., Rheinstetten, Germany) were used for NMR spectroscopy, with tetramethylsilane (TMS) (Sigma-Aldrich Co., St. Louise, MO, USA) as the internal standard. HR-ESI-MS spectra were obtained on a Waters UPLC Premior QTOF spectrometer (Waters Co., Milford, MA, USA). Column chromatography (CC) was performed with silica gel (200–300 mesh; Qingdao Marine Chemical Factory, Qingdao, China), Sephadex LH-20 (GE Healthcare Bio-Sciences AB, Uppsala, Sweden), and YMC gel ODS-A-HG (50 μm, YMC Co., Ltd., Kyoto, Japan). Thin-layer chromatography (TLC) was conducted on silica gel GF254 plates (Qingdao Marine Chemical Factory, Qingdao, China). Preparative HPLC was performed on an Agilent 1260 series instrument (Agilent Co., Santa Clara, CA, USA) cooperated with a Shiseido Capcellpak Prep C18 ODS column (250 × 20 mm, 5 μm, Shiseido Co., Tokyo, Japan) with a flow rate of 16 mL∙min−1.

3.2. Plant Materials

Plant materials of Caryopteris incana (Thunb.) Miq. were harvested in Hexian County of Anhui Province, the People’s Republic of China, in September 2012. The fresh material was air-dried and ground to coarse powder. The identification of plant materials was verified by Prof. Qingshan Yang of Anhui University of Traditional Chinese Medicine. A voucher specimen (no. 20120915-1) was deposited in the Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, 201203, China.

3.3. Extraction and Isolation

The dried and crushed plants of Caryopteris incana (Thunb.) Miq. (19.5 kg) were completely extracted with EtOH at room temperature; the ethanolic extract was concentrated under reduced pressure, the residue was dissolved in hot water (10 L, 60 °C), and partitioned successively with petroleum ether (PE), ethyl acetate (EtOAc), and butyl alcohol (n-BuOH) in the same volume, 3–4 times in order to get the PE-(234 g), EtOAc-(355 g) and n-BuOH soluble (200 g) fractions. In this part, we performed continuous research on the n-BuOH soluble fraction of C. incana.
The n-BuOH soluble fraction (200 g) was subjected to column chromatography (CC) over silica gel (100–200 mesh), eluted with a gradient mixture of EtOAc/MeOH/H2O (15:2:1, 10:2:1, 8:2:1), to afford ten fractions 1–10. Fractions 2–5 were subjected to octadecyl silane (ODS) CC eluting with a gradient of MeOH/H2O (5:95–60:40, v/v) to give four sub-fractions (Fr. I–Fr. IV), Fr. I, Fr. II, and Fr. IV were further chromatographed on a Sephadex LH-20 column using MeOH/H2O (80:20, v/v) as eluent to afford compounds 6 (40 mg), 2 (3 mg) and 8 (15 mg), respectively; Fr. III was separated by preparative TLC (developer: EtOAc/MeOH/H2O 10:2:1; eluent: Acetone), followed by a Sephadex LH-20 column and eluted with (MeOH/H2O 80:20, v/v) to yield compound 1 (10 mg). Fr. 6–Fr. 8 were subjected to ODS CC eluting with a gradient of MeOH/H2O (5:95–50:50, v/v) to give three sub-fractions (Fr. V–Fr. VII); Fr. V was chromatographed on a Sephadex LH-20 column using MeOH/H2O (80:20, v/v) as eluent to acquire compound 11 (9 mg), followed by prep-HPLC on a Shiseido Capcellpak Prep C18 column, using acetonitrile/H2O (30:1, 16 mL∙min−1, detection at 254 nm) to afford compound 5 (5 mg); retention time was 15 min. Fr. VI was chromatographed on a Sephadex LH-20 column using MeOH/H2O (80:20, v/v) as eluent to get a mixture of compounds 3 and 4, followed by prep-HPLC on a Shiseido Capcellpak Prep C18 column using MeOH/H2O (29:1, 16 mL∙min−1, detection at 254 nm) to afford compound 3 (11 mg) and compound 4 (4 mg); the retention time of compounds 3 and 4 were 11 and 13 min, respectively. Fr. VII was subjected to the same method as Fr. VI, and produced compounds 7 (6 mg), 9 (13 mg) and 10 (7 mg); the retention times of compounds 7, 9 and 10 were 10, 16, and 13 min, respectively. (prep-HPLC detected at 254 nm, acetonitrile/H2O, 27:1).

3.4. Spectroscopic Data

Caryocanoside B (1) was obtained as a yellow amorphous powder; [ α ] D 20 = −0.098 (c 0.1, MeOH); IR νmax: 3357, 2921, 2851, 1704, 1633, 1422, 1369, 1274, 1194, 1071, 1010, 901, and 859 cm−1; HR-ESI-MS m/z 731.2802 [M − H], (calcd. for 731.2762); 1H- and 13C-NMR data, see Table 1.
5-Hydroxy-2′′′-O-caffeoylcaryocanoside B (2) was obtained as a yellow amorphous powder; [ α ] D 20 = –0.266 (c 0.1, MeOH); IR νmax: 3357, 1703, 1669, 1599, 1515, 1373, 1233, 1186, 1067, 1011, 901, 851, 810, and 773 cm−1; HR-ESI-MS m/z 909.3038 [M − H], (calcd. for 909.3040); 1H- and 13C-NMR data, see Table 1.
2′′′-O-(E)-p-Coumaroyl caryocanoside B (3) was obtained as a white amorphous powder; [ α ] D 20 = −0.168 (c 0.1, MeOH); IR νmax: 3313, 2949, 2162, 1979, 1702, 1646, 1603, 1587, 1515, 1444, 1365, 1326, 1271, 1171, 1009, 973, 897, and 836 cm−1; HR-ESI-MS m/z 877.3147 [M − H], (calcd. for 877.3130); 1H- and 13C-NMR data, see Table 1.
2′′′-O-(Z)-p-coumaroyl caryocanoside B (4) was obtained as a white amorphous powder; [ α ] D 20 = −0.09 (c 0.1, MeOH); IR νmax: 3358, 2921, 2851, 1704, 1633, 1604, 1514, 1424, 1370, 1261, 1190, 1067, 1008, 901, 854, 800, and 766 cm−1; HR-ESI-MS m/z 901.3092 [M + Na]+, (calcd. for 901.3106); 1H- and 13C-NMR data, see Table 1.
2′-O-(E)-p-coumaroyl asystasioside A (5) was obtained as a yellow amorphous powder; [ α ] D 20 = −0.058 (c 0.1, MeOH); IR νmax: 3357, 2922, 2851, 2161, 1698, 1632, 1603, 1514, 1425, 1263, 1169, 1061, 1024, 901, 834, 764 and 700 cm−1; HR-ESI-MS m/z 667.2257 [M − H], (calcd. for 667.2238); 1H- and 13C-NMR data, see Table 2.

3.5. Alpha-Glucosidase Inhibitory Assay

The α-glucosidase inhibition assay was performed according to a slightly modified, previously reported method [20]; α-Glucosidase (0.1 U/mL) was dissolved in potassium phosphate buffer (pH 6.8) as the enzyme solution. The tested compounds (50 μL, 5 mM) dissolved in potassium phosphate buffer containing 3% DMSO were mixed with 50 μL of enzyme solution. After incubation at 37 °C for 10 min, a p-nitrophenyl-α-glucopyranoside (pNPG) solution (100 μL) (5.0 mM pNPG in 0.1 M potassium phosphate buffer (pH 6.8)) was added. The enzymatic reaction proceeded at 37 °C for 20 min and was terminated by the addition of 100 μL of 0.2 M Na2CO3. 4-nitrophenol absorption was immediately measured at 405 nm by using a microplate reader. The experiments were performed in triplicate. The percent inhibition of α-glucosidase was calculated as inhibition rate (%) = 100 × [1 − (Asample − As-blank)/(Acontrol − Ablank)]. The α-glucosidase from Saccharomyces cerevisiae, pNPG, and acarbose were purchased from Sigma-Aldrich.

4. Conclusions

Five new iridoid glucosides, 15, including two (cis/trans) isomers, together with six known compounds were isolated from the whole plant of C. incana. The discovery of these compounds further expands our knowledge of the structural diversity of the glycosides produced by the plant. Furthermore, compounds 48 and 10 exhibited a significant α-glucosidase inhibitory activity, with IC50 values from 3.35 to 0.33 mM. 2′′′-O-(Z)-p-coumaroyl caryocanoside B (4) and ajugaside A (10) manifested the highest activities with IC50 values at 0.38 and 0.33 mM. Two isomers (3 and 4) showed distinct results to inhibition of α-glucosidase; only cis-isomer (4) had a potent effect. This mechanism needs to be further investigated. Combined with the research, which was completed by our predecessors, the potential hypoglycaemic activity of C. incana should be substantiated, and this plant could be regarded as a suitable herb-derived drug for the prevention and treatment of T2DM.

Supplementary Materials

Supplementary materials can be accessed at: https://0-www-mdpi-com.brum.beds.ac.uk/1420-3049/21/12/1749/s1.

Acknowledgments

This work was financially supported by grant from the National Nature Science Foundation of China (No. 21672152).

Author Contributions

Isolation and purification of Caryopteris incana and evaluation of α-glucosidase inhibitory activities of the isolated compounds was carried by Xu-Dong Mao. Collection of the plant was carried by Sen-Miao Zhao. Cheng-Gang Zhang performed the research and recorded the spectra. Gui-Xin Chou designed the research.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Shaw, J.E.; Sicree, R.A.; Zimmet, P.Z. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res. Clin. Pract. 2010, 87, 4–14. [Google Scholar] [CrossRef] [PubMed]
  2. Zhao, D.G.; Zhou, A.Y.; Du, Z.; Zhang, Y.; Zhang, K.; Ma, Y.Y. Coumarins with α-glucosidase and α-amylase inhibitory activities from the flower of Edgeworthia gardneri. Fitoterapia 2015, 107, 122–127. [Google Scholar] [CrossRef] [PubMed]
  3. Jiangsu New Medical College. The Chinese Medicine Dictionary; Shanghai People’s Publishing House: Shanghai, China, 1977; p. 774. [Google Scholar]
  4. Gao, J.; Han, G. Cytotoxic abietane diterpenoids from Caryopteris incana. Phytochemistry 1997, 44, 759–761. [Google Scholar]
  5. Yoshikawa, K.; Harada, A.; Iseki, K.; Hashimoto, T. Constituents of Caryopteris incana and their antibacterial activity. J. Nat. Med. 2014, 68, 231–235. [Google Scholar] [CrossRef] [PubMed]
  6. Yoshizaki, M.; Fujino, H.; Masuyama, M.; Arisawa, M.; Morita, N. A chemotaxonomic study of flavonoids in the leaves of six Trichosanthes species. Phytochemistry 1987, 26, 2557–2558. [Google Scholar] [CrossRef]
  7. Gao, J.; Igarashi, K.; Nukina, M. Three new phenylethanoid glycosides from Caryopteris incana and their antioxidative activity. Chem. Pharm. Bull. 2000, 48, 1075–1078. [Google Scholar] [CrossRef] [PubMed]
  8. Zhao, S.M.; Chou, G.X.; Yang, Q.S.; Wang, W.; Zhou, J.L. Abietane diterpenoids from Caryopteris incana (Thunb.) Miq. Org. Biomol. Chem. 2016, 14, 3510–3520. [Google Scholar] [CrossRef] [PubMed]
  9. Hua, J.; Qi, J.; Yu, B.Y. Iridoid and phenylpropanoid glycosides from Scrophularia ningpoensis Hemsl. and their α-Glucosidase inhibitory activities. Fitoterapia 2014, 93, 67–73. [Google Scholar] [CrossRef] [PubMed]
  10. Liu, Z.X.; Liu, C.T.; Liu, Q.B.; Ren, J.; Lia, L.Z.; Huang, X.X.; Wang, Z.Z.; Song, S.J. Iridoid glycosides from the flower buds of Lonicera japonica and their nitric oxide production and α-glucosidase inhibitory activities. J. Funct. Foods 2015, 18, 512–519. [Google Scholar] [CrossRef]
  11. Li, G.Z.; Mishig, D.; Pu, X.; Yi, J.H.; Zhang, G.L.; Luo, Y.G. Chemical Components of Aerial Parts of Lagochilus ilicifolius. Chin. J. Appl. Environ. Biol. 2012, 18, 924–927. [Google Scholar] [CrossRef]
  12. Zhang, Y.H.; Cheng, D.L. Two new iridoid glycosides from Caryopteris mongholica. Chin. Chem. Lett. 2000, 11, 319–322. [Google Scholar]
  13. Takasaki, M.; Yamauchi, I.; Haruna, M.; Konoshima, T. New glycosides from Ajuga decumbens. J. Nat. Prod. 1998, 61, 1105–1109. [Google Scholar] [CrossRef] [PubMed]
  14. Park, S.; Son, M.J.; Yook, C.S.; Jin, C.; Lee, Y.S.; Kim, H.J. Chemical constituents from aerial parts of Caryopteris incana and cytoprotective effects in human HepG2 cells. Phytochemistry 2014, 101, 83–90. [Google Scholar] [CrossRef] [PubMed]
  15. Markham, K.R.; Ternai, B.; Stanley, R.; Geiger, H.; Mabry, T.J. Carbon-13 NMR studies of flavonoids—III: Naturally occurring flavonoid glycosides and their acylated derivatives. Tetrahedron 1978, 34, 1389–1397. [Google Scholar] [CrossRef]
  16. Wang, J.H.; Cong, Y.; Li, X. Isolation and identification of the chemical constituents of Leonurus japonicus Houtt. Chin. J. Med. Chem. 2002, 12, 146–148. [Google Scholar]
  17. Morvai, M.; Nagy, T.; Kocsis, A.; Szabó, L.F.; Podanyi, B. Effect of oxygen substituents on two- and three-bond carbon-proton spin-spin coupling constants. Magn. Reson. Chem. 2000, 38, 343–359. [Google Scholar] [CrossRef]
  18. Boros, C.A.; Stermitz, F.R. Iridoids. An Updated Review. Part I. J. Nat. Prod. 1990, 53, 1055–1147. [Google Scholar] [CrossRef]
  19. Demuth, H.; Jensen, S.R.; Nielsen, B.J. Iridoid glucosides from Asystasia bella. Phytochemistry 1989, 28, 3361–3364. [Google Scholar] [CrossRef]
  20. Liu, Q.; Hu, H.J.; Li, P.F.; Yang, Y.B.; Wu, L.H.; Chou, G.X.; Wang, Z.T. Diterpenoids and phenylethanoid glycosides from the roots of Clerodendrum bungei and their inhibitory effects against angiotensin converting enzyme and α-glucosidase. Phytochemistry 2014, 103, 196–202. [Google Scholar] [CrossRef] [PubMed]
  • Sample Availability: Samples of the compounds 25 are available from the authors.
Figure 1. Chemical structures of compounds 15.
Figure 1. Chemical structures of compounds 15.
Molecules 21 01749 g001
Figure 2. Selected heteronuclear multiple bond correlations (HMBC) for compound 1.
Figure 2. Selected heteronuclear multiple bond correlations (HMBC) for compound 1.
Molecules 21 01749 g002
Figure 3. Selected Nuclear Overhauser Effect Spectroscopy (NOESY) correlations for compound 1.
Figure 3. Selected Nuclear Overhauser Effect Spectroscopy (NOESY) correlations for compound 1.
Molecules 21 01749 g003
Figure 4. Selected HMBC correlations for compound 5.
Figure 4. Selected HMBC correlations for compound 5.
Molecules 21 01749 g004
Figure 5. Selected NOESY correlations for compound 5.
Figure 5. Selected NOESY correlations for compound 5.
Molecules 21 01749 g005
Figure 6. Chemical structures of isolated compounds 611.
Figure 6. Chemical structures of isolated compounds 611.
Molecules 21 01749 g006
Table 1. 1H- and 13C-NMR data for 14.
Table 1. 1H- and 13C-NMR data for 14.
Position1 a2 a3 b4 a
δH (J)δCδH (J)δCδH (J)δCδH (J)δC
15.87 (s)94.06.03 (s)94.25.80 (s)94.05.8 (s)94.1
36.26 (dd, 2.4, 6.0)142.26.35 (d, 6.4)144.06.17 (d, 6.6)141.96.25 (dd, 6.4, 2.4)142.3
44.77 (brd, 6.0)103.04.81 (brd, 8.0)106.24.36 (d, 6.0)103.04.70 (d, 5.2)103.0
52.98 (d, 8.0)39.7 72.72.66, overlap39.22.91 (d, 8.0)39.7
64.97 (d, 4.4)79.44.34 (d, 4.0)79.24.21 (d, 4.8)79.34.71, overlap79.4
72.35 (brd, 15.2)46.01.96 (m)44.32.07 (d, 15.6)46.22.12 (d, 15.6)46.1
2.19 (brd, 15.2)1.67 (m)1.80 (dd, 4.8, 15.6)2.0 (m)
8 89.9 88.2 89.6 89.8
92.81 (d, 8.4)50.12.79 (s)55.82.66, overlap49.93.35, overlap49.8
101.58 (s)22.51.38 (s)22.11.45 (s)22.51.49 (s)22.4
COOCH3 172.9 172.9 172.9 173.0
COOCH31.99 (s)22.21.96 (s)22.21.93 (s)22.31.94 (s)22.2
1′5.47 (d, 4.4)96.35.53 (d, 2.4)95.35.52 (brd, 1.8)95.55.48 (d, 2.8)96.4
3′7.42 (s)153.07.48 (s)152.47.30 (s)152.07.32 (s)152.4
4′ 113.6 114.7 114.7 114.5
5’2.91 (brdd, 14.4, 8.0)34.42.84 (m)33.02.81 (td, 8.4, 3.6)32.62.83 (td, 8.4, 4.4)32.9
6′1.56, overlap32.51.86 (m)32.21.87 (m)32.21.92 (m)32.4
2.07 (m)1.79 (m)1.61 (m)1.60 (m)
7′1.38 (m)33.21.23 (m)34.31.21 (m)34.31.26 (m)34.0
1.79 (m)1.72 (m)1.72 (m)1.73 (m)
8′2.28 (m)37.62.24 (m)36.42.23 (m)36.22.25 (m)36.7
9′2.27 (m)44.42.34 (td, 8.8, 2.0)44.22.36 (td, 9.6, 1.8)44.12.34 (td, 8.8, 2.8)44.3
10′1.08 (d, 6.8)16.61.00 (d, 7.2)16.50.98 (d, 7.2)16.51.01 (d, 6.8)16.4
11′ 168.4 168.1 167.9 167.9
1″4.68 (d, 8.0)99.84.57 (d, 8.0)99.94.63 (d, 7.8)99.84.65 (d, 8.0)99.9
2″3.21 (m)74.73.20 (t, 8.4)74.43.20 (t, 8.4)74.73.20 (t, 8.0)74.7
3″3.37 (m)78.03.37 (m)77.63.38, overlap77.93.37, overlap78.0
4″3.31, overlap71.63.30, overlap71.53.31, overlap71.63.27, overlap71.6
5″3.31, overlap78.23.30, overlap78.13.39, overlap78.13.35, overlap78.2
6″3.85 (m)62.93.91 (dd, 12.0)62.73.88 (d, 12.0)62.83.89 (dd)62.9
3.65 (m)3.69 (dd, 5.2, 11.6)3.69 (m)3.70 (d, 5.2)
1′′′4.64 (d, 8.0)99.84.86, overlap96.74.85, overlap97.04.81 (d, 8.0)97.9
2′′′3.19 (m)74.84.79 (d, 8.4)74.64.79 (t, 8.4)74.64.77 (t, 8.4)74.3
3′′′3.37 (m)78.03.60 (t, 8.8)75.93.62 (t, 9.0)75.83.55 (t, 8.8)75.9
4′′′3.26, overlap71.73.37 (m)71.83.38, overlap71.73.36, overlap71.8
5′′′3.31, overlap78.43.37 (m)78.53.38, overlap78.43.36, overlap78.5
6′′′3.89 (m)62.93.91 (dd, 12.0)62.83.94 (d,10.8)62.73.94 (d)62.7
3.69 (m)3.69 (dd, 5.2, 11.6)3.69 (m)3.67 (d, 5.6)
1′′′′ 127.8 127.2 127.5
2′′′′ 7.06 (d, 2.0)115.87.47 (d, 8.4)131.47.70 (d, 8.4)134.2
3′′′′ 146.66.83 (d, 8.4)116.96.74 (d, 8.4)115.7
4′′′′ 149.5 161.2 160.1
5′′′′ 6.79 (d, 8.0)116.46.83 (d, 8.4)116.96.74 (d, 8.4)115.7
6′′′′ 6.97 (dd, 1.6, 8.0)123.37.47 (d, 8.4)131.47.70 (d, 8.4)134.2
7′′′′ 7.49 (d, 16.0)147.17.56 (d, 15.6)146.56.87 (d, 12.8)145.9
8′′′′ 6.20 (d, 16.0)114.96.26 (d, 15.6)115.05.70 (d, 12.8)116.2
9′′′′ 168.3 168.1 166.8
a Compound 1, 2, 4: Measured at 1H (400 MHz) and 13C (100 MHz), CD3OD; b Compound 3: Measured at 1H (600 MHz), and 13C (150 MHz), CD3OD. brd: broad doublet; brdd: broad doublet of doublets; d: doublet; dd: doublet of doublets; m: multiplet; s: singlet; t: triplet; td: triplet of doublets.
Table 2. 1H- and 13C-NMR data for 5, measured at 1H (600 MHz), and 13C (150 MHz), CD3OD.
Table 2. 1H- and 13C-NMR data for 5, measured at 1H (600 MHz), and 13C (150 MHz), CD3OD.
PositionCompound 5
δH (J)δC
15.50 (d, 3.0)95.6
37.53 (s)153.7
4 113.3
52.82 (m)33.7
61.92 (m)31.7
1.60 (m)
71.24 (m)33.7
1.74 (m)
82.26 (m)36.5
92.27 (m)44.2
101.03 (d, 6.6)16.6
11 167.0
1′4.83, overlap97.2
2′4.81, overlap74.7
3′3.59 (t, 9.0)75.9
4′3.37 (m)71.8
5′3.37 (m)78.6
6′3.76 (dd,1.8,12.0)62.2
3.63 (dd,4.8,12.0)
1″5.08 (d, 8.4)95.5
2″3.32, overlap73.9
3″3.36, overlap77.9
4′′′3.34, overlap70.9
5″3.15 (m)78.5
6″3.93 (d, 11.4)62.8
3.69 (d, 12.0)
1′′′ 127.4
2′′′7.52 (d, 8.4)131.7
3′′′6.83 (d, 8.4)116.7
4′′′ 161.1
5′′′6.83 (d, 8.4)116.7
6′′′7.52 (d, 8.4)131.7
7′′′7.60 (d, 15.6)147.2
8′′′6.29 (d, 15.6)114.9
9′′′ 168.5
Table 3. The IC50 (mM) values of α-glucosidase inhibitory activity of the isolated compounds, 111, and acarbose as the control. Each value is shown as a mean ± standard derivation of three replicates.
Table 3. The IC50 (mM) values of α-glucosidase inhibitory activity of the isolated compounds, 111, and acarbose as the control. Each value is shown as a mean ± standard derivation of three replicates.
CompoundsIC50CompoundsIC50
1>5.071.38 ± 0.27
2>5.080.82 ± 0.15
3>5.09>5.0
40.38 ± 0.015100.33 ± 0.06
53.35 ± 0.1211>5.0
61.89 ± 0.7acarbose3.49 ± 0.15

Share and Cite

MDPI and ACS Style

Mao, X.-D.; Chou, G.-X.; Zhao, S.-M.; Zhang, C.-G. New Iridoid Glucosides from Caryopteris incana (Thunb.) Miq. and Their α-Glucosidase Inhibitory Activities. Molecules 2016, 21, 1749. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules21121749

AMA Style

Mao X-D, Chou G-X, Zhao S-M, Zhang C-G. New Iridoid Glucosides from Caryopteris incana (Thunb.) Miq. and Their α-Glucosidase Inhibitory Activities. Molecules. 2016; 21(12):1749. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules21121749

Chicago/Turabian Style

Mao, Xu-Dong, Gui-Xin Chou, Sen-Miao Zhao, and Cheng-Gang Zhang. 2016. "New Iridoid Glucosides from Caryopteris incana (Thunb.) Miq. and Their α-Glucosidase Inhibitory Activities" Molecules 21, no. 12: 1749. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules21121749

Article Metrics

Back to TopTop