Next Article in Journal
The EFSA Health Claim on Olive Oil Polyphenols: Acid Hydrolysis Validation and Total Hydroxytyrosol and Tyrosol Determination in Italian Virgin Olive Oils
Previous Article in Journal
Metal-Free Air Oxidation in a Convenient Cascade Approach for the Access to Isoquinoline-1,3,4(2H)-triones
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Bioactive Dimeric Abietanoid Peroxides from the Bark of Cryptomeria japonica

1
Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
2
Research Center for Active Natural Products Development, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
3
Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
4
Department of Life Science, National Taitung University, Taitung 950, Taiwan
5
Bioresource Collection and Research Center (BCRC), Food Industry Research and Development Institute (FIRDI), Hsinchu 300, Taiwan
6
National Museum of Marine Biology and Aquarium, Pingtung 944, Taiwan
7
Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
8
Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung 404, Taiwan
9
Department of Biotechnology, Asia University, Taichung 413, Taiwan
10
Chinese Medicine Research Center, China Medical University, Taichung 404, Taiwan
*
Author to whom correspondence should be addressed.
These two authors contributed equally to this paper.
Molecules 2019, 24(11), 2178; https://doi.org/10.3390/molecules24112178
Submission received: 10 May 2019 / Revised: 6 June 2019 / Accepted: 8 June 2019 / Published: 10 June 2019
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
Three new dimeric abietane-type diterpenoids, abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7α-yl peroxide (1), abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7β-yl peroxide (2), and 12-hydroxyabieta-8,11,13-trien-7β-yl 7-oxoabieta-5,8,11,13-tetraen-12-yl peroxide (3), together with four known abietane-type diterpenoids (47) were isolated from the methanol extract of the bark of Cryptomeria japonica. Their structures were elucidated on the basis of spectroscopic analysis and comparison of NMR data with those of known analogues. At a concentration of 50 μM, compounds 1, 2, and 3 showed 26.2%, 23.6%, and 35.7% inhibition towards xanthine oxidase enzyme, respectively. In addition, compound 3 also showed 24.9% inhibition toward angiotensin-converting enzyme (ACE).

1. Introduction

The Japanese cedar, Cryptomeria japonica D. Don (Cupressaceae), belongs to the monotypic genus in the Cupressaceae [1]. It is a massive evergreen coniferous tree, growing up to 50 m in height. Due to the aromatic, reddish-pink in color, soft, lightweight but strong, and waterproof properties of its wood, it is planted in large quantities and used as building materials and wood products. This plant has been cultivated as an important coniferous tree species in Taiwan since 1906. Phytochemical investigations indicated the presence of monoterpenoids, sesquiterpenoids, and diterpenoids [2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24] in the leaves, heartwood, and barks of C. japonica. The crude extracts and secondary metabolites from this species exhibited a wide array of bioactivities including cytotoxic [23], antifungal [24], antibacterial [23], antioxidant [25], anti-inflammatory [26], and insect antifeedant [27] and repellent [18] properties. While searching for bioactive chemical ingredients of the bark of C. japonica, we have already reported the isolation of three sesquarterpenoids [28,29] and ten abietane-type diterpenoids [30,31,32]. Herein, the isolation and structure elucidation of three new dimeric abietane-type diterpenoids are described as well as their inhibitory activity towards xanthine oxidase and angiotensin-converting enzymes.

2. Results and Discussion

2.1. Isolation and Structural Elucidation

The MeOH extract of the bark of C. japonica was suspended in H2O and then partitioned with EtOAc and n-BuOH, successively. The EtOAc-soluble portion was submitted to repeated silica gel column chromatography and semipreparative normal phase-HPLC to afford compounds 17 (Figure 1).
The high resolution electron impact mass spectrum (HR-EI-MS) of 1 gave a molecular ion at m/z 584.4238, corresponding to the molecular formula of C40H56O3, with thirteen degrees of unsaturation. The electron impact mass spectrum (EI-MS) of 1 displayed the fragmental ions at m/z 300 [C20H28O2]+ and 284 [C20H28O]+ (Figure 2) and forty carbon signals were observed in the 13C-NMR spectrum, indicating that 1 was a dimeric diterpenoid (Figure 2). The UV and IR spectra showed absorption bands for hydroxyl (3409 cm−1) and aromatic (λmax 220 and 279 nm; 3049, 1593, and 1487 cm−1) groups. The 1H- and 13C-NMR data of 1 (Table 1) showed one set of dehydroabietane diterpene signals for constituent upper monomer-1 including three tertiary methyl groups (δH 0.97, 0.98, and 1.36 (each 3H, s, Me-18, Me-19, and Me-20)), an isopropyl group attached to a phenyl group (δH 0.78 (3H, d, J = 7.0 Hz, Me-16), 1.01 (3H, d, J = 7.0 Hz, Me-17), and 2.85 (1H, sept, J = 7.0 Hz, H-15)), two para aromatic protons (δH 6.34 (1H, s, H-14) and 6.69 (1H, s, H-11)), a phenolic hydroxyl proton (δH 4.33 (1H, s, exchangeable with D2O)), and a typical downshifted Hβ-1 signal of a dehydroabietane diterpene (δH 1.96 (1H, br d, J = 12.0 Hz)) [33]. A downshifted benzyl proton connected with a peroxyl group (δH 5.86 (1H, br s, H-7)), instead of a hydroxyl group [19,33,34], was assigned as H-7, suggested by the 1H-1H COSY correlations with the two methylene protons of H-6 (δH 2.28 (1H, m), δH 2.32 (1H, m)), and HMBC correlations with C-5 (δC 43.5) and C-8 (δC 145.6; Figure 3). In addition, the 1H-NMR signal of H-7 was a broad singlet peak and showed the NOESY correlation with both Hα-6 (δH 2.32) and Hβ-6 (δH 2.28), hinted that the peroxyl group was attached on C-7 in α-axial orientation [33] (Figure 3). These data proved that the structure of constituent monomer-1 was related to 7α-peroxyferruginol. The 1H- and 13C-NMR data of 1 (Table 1) also exhibited another set of dehydroabietane diterpene signals for constituent lower monomer-2 including three tertiary methyl groups (δH 0.94, 1.04, and 1.11 (each 3H, s, Me-18′, Me-19′, and Me-20′)), an isopropyl group attached to a phenyl group (δH 0.81 (3H, d, J = 7.0 Hz, Me-16′), 0.98 (3H, d, J = 7.0 Hz, Me-17′), and 2.82 (1H, sept, J = 7.0 Hz, H-15′)), two para aromatic protons (δH 6.98 (1H, s, H-11′) and 6.77 (1H, s, H-14′)), an ABX coupling system of one methine proton (δH 2.07 (1H, dd, J = 3.0, 2.5 Hz, H-5′)), and two vinyl protons (δH 5.87 (1H, dd, J = 9.5, 2.5 Hz, H-6′) and 6.45 (1H, dd, J = 9.5, 3.0 Hz, H-7′)), together with a typical downshifted Hβ-1 signal of a dehydroabietane diterpene (δH 2.22 (1H, br d, J = 13.0 Hz)) [33]. The above NMR spectroscopic data suggested constituent lower monomer-2 was related to 6,7-dehydroferruginol [35]. Thus, the gross structure of 1 is composed of 7α-hydrperoxyferruginol and 6,7-dehydroferruginol. The chemical shift of H-7 appeared at the lower field region (δH 5.86) in constituent monomer-1, comparing to that of the 7α-hydroxyferruginol analogues [34] implied that the connectivity of two monomers between C-7 and C-12′ through a peroxide functionality. The NOESY correlation between H-7 and H-11′ (δH 6.98; Figure 3) further confirmed this proposal. Interestingly, there are some electrostatic attraction between the electron-rich aryl, the phenol functionality of upper 7α-peroxyferruginol derivative, and the electron-deficient aryl, with peroxide moiety of lower 6,7-dehydroferruginol, formed the most stable conformer as shown in Figure 3. Due to the anisotropic effect from the opposite phenyl group, H-14, H-15, H-16, H-17, H-14′, H-15′, H-16′, H-17′, and phenol of 1 were posited in the shielding region and thus showed the higher field chemical shifts than that of the usual dehydroabietane diterpene. In contrast, H-5, H-7, and H-20’ of 1 were located in the deshielding region and thus exhibit a lower field chemical shifts than that of the usual dehydroabietane diterpene [33]. Complete 1H- and 13C-NMR chemical shifts were established by 1H-1H COSY, HMQC, HMBC, and NOESY spectra. Based on these above evidences, compound 1 was elucidated as abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7α-yl peroxide.
Compound 2 was an isomer of 1 with the same molecular formula C40H56O3, determined by the molecular ion of HR-EI-MS at m/z 584.4237. Its EI-MS also showed the fragmental ions at m/z 300 [C20H28O2]+ and 284 [C20H28O]+, indicating that 2 was also a dimeric diterpenoid. The absorptions for hydroxyl (3423 cm−1) and aromatic (3048, 1590, and 1493 cm−1; λmax 217 and 276 nm) groups were also found in the UV and IR spectra. Comparison of 1H and 13C-NMR data of 2 and 1 (Table 1) showed that the signals of constituent lower monomer-2 of 2 were almost the same as those of 1, indicating the structure of constituent lower monomer-2 is related to 6,7-dehydroferruginol. The 1H- and 13C-NMR data of 2 (Table 1) also showed another set of dehydroabietane diterpene signals for constituent upper monomer-1 including three tertiary methyl groups (δH 0.95, 1.02, and 1.37 (each 3H, s, Me-18, Me-19, and Me-20)), an isopropyl group attached to a phenyl group (δH 1.16 (3H, d, J = 7.0 Hz, Me-17), 1.18 (3H, d, J = 7.0 Hz, Me-16), and 3.01 (1H, sept, J = 7.0 Hz, H-15)), two para aromatic protons (δH 6.72 (1H, s, H-14) and 6.74 (1H, s, H-11)), a phenolic hydroxyl proton (δH 4.52 (1H, s)), and a typical downshifted Hβ-1 signal of a dehydroabietane diterpene (δH 1.93 (1H, br d, J = 12.0 Hz)) [33]. A downshifted benzyl proton connected with a peroxyl group (δH 5.20 (1H, d, J = 8.0 Hz, H-7)) was assigned as H-7, suggesting by the 1H-1H COSY correlations with the two methylene protons of H-6 (δH 2.24 (1H, m, Hα-6), δH 2.40 (1H, br d, J = 8.0 Hz, Hβ-6)) and HMBC correlations with C-5 (δC 47.6) and C-8 (δC 147.2; Figure 3). Since the 1H-NMR signal of H-7 was a doublet peak with a constant coupling constant, 8.0 Hz, the peroxyl group on C-7 was in β-equational orientation (Figure 2), instead of in α-axial orientation in 1 [33]. H-7 showed the NOESY correlation with Hα-6 (δH 2.24) and 1.59 (H-5, m), but the lack of NOESY correlation with Hβ-6 (δH 2.40) further confirmed this proposal (Figure 3). Thus, the structure of 2 was identified as abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7β-yl peroxide. Compound 2 did not exhibit the most stable conformer as in 1. Therefore, the chemical shifts of two isopropyl groups in 2 were not shifted to the high field region.
The HR-EI-MS of 3 showed a molecular ion at m/z 598.4018, which corresponded to the molecular formula, C40H54O4, indicating fourteen degrees of unsaturation. The EI-MS fragmental ions of 3 at m/z 300 [C20H28O2]+ and 298 [C20H26O3]+ indicated that 3 was also a dimeric diterpenoid. The absorptions for hydroxyl (3376 cm−1), benzoyl (1639 cm−1; λmax 233, 282, and 310 nm) and aromatic (3049, 1586, and 1467 cm−1) groups were also found in its UV and IR spectra. Comparison of 1H- and 13C-NMR data of 3 and 2 (Table 1) showed that the signals of constituent monomer-1 of 3 were almost the same as those of 2, indicating the structure of constituent upper monomer-1 is related to 7α-peroxyferruginol. The 1H- and 13C-NMR data of 3 (Table 1) also showed another set of dehydroabietane diterpene signals for constituent lower monomer-2 as follows: Three tertiary methyl groups (δH 0.97, 1.03, and 1.49 (each 3H, s, Me-18′, Me-19′, and Me-20)), an isopropyl group attached to a phenyl group (δH 1.20 (3H, d, J = 7.0 Hz, Me-17’), 1.23 (3H, d, J = 7.0 Hz, Me-16’) and 3.32 (1H, sept, J = 7.0 Hz, H-15′)), two para aromatic protons (δH 7.19 (1H, s, H-11′) and 8.03 (1H, s, H-14′)), one trisubstituted double bond (δH 6.48 (1H, s, H-6′); δC 124.5 (C-6′), 173.2 (C-5′)), and a typical downshifted Hβ-1 signal of a dehydroabietane diterpene (δH 2.51 (1H, br d, J = 13.5 Hz, H-1′)). The NMR spectroscopic data of constituent lower monomer-2 showed a close structural resemblance to that of 5,6-dehydrosugiol [36]. Thus, the structure of constituent lower monomer-2 was tentatively determined as 5,6-dehydrosugiol-related abietane. H-7 exhibited a doublet of a doublet signal with two coupling constants, 9.0 and 2.5 Hz and showed the NOESY correlation with Hα-6 (δH 2.24) and H-5 (δH 1.72), but the lack of NOESY correlation with Hβ-6 (δH 2.41), which confirmed the peroxyl group was attached on C-7 in β-equational orientation (Figure 2) [33]. Thus, the structure of 3 was identified as 12-hydroxyabieta-8,11,13-trien-7β-yl 7-oxoabieta-5,8,11,13-tetraen-12-yl peroxide.
Four known compounds were identified by comparison of the NMR data with those described in the literatures as sugiol (4) [37], 16-hydroxysugiol (5) [38], 12-dehydroxy-15-hydroxysugiol (6) [39], and 12-O-acetylsugiol (7) [40].

2.2. Inhibitory Activities Toward Xanthine Oxidase and Angiotensin-Converting Enzyme

Xanthine oxidase is a key enzyme in purine metabolic pathway, catalyzing oxypurines (hypoxanthine and xanthine) to uric acid and plays an important role in causing gout [41]. Additionally, the angiotensin-converting enzyme (ACE) plays a key physiological role in blood pressure regulation of the renin–angiotensin system due to its action in the formation of angiotensin II, a potent vasoconstrictor, and in the degradation of bradykinin, a vasodilator [42]. Compounds 13 were evaluated using the above two enzyme inhibitory activities [43,44]. At the concentration of 50 μM, compounds 13 exhibited 26.2%, 23.6%, and 35.7% xanthine oxidase inhibitory activity, respectively. Compound 3 also showed 24.9% ACE inhibitory activity, while compounds 1 and 2 were inactive. Analysis of the relationship between structure and activity in compounds 13 showed that the inhibitory activities toward the two above enzymes of compound 3 containing a 5,6-dehydrosugiol moiety at C-7 were higher than that of compounds 1 and 2 with a 6,7-dehydroferruginol moiety at C-7. Furthermore, the different orientations of the 7-substituent between compounds 1 and 2 had no significant effect on their xanthine oxidase inhibitory activity.

3. Experimental Section

3.1. Chemicals

Xanthine, Xanthine oxidase, and ACE (EC 3.4.15.1) from rabbit lungs, hippuryl-l-histidyl-l-leucine (HHL), ferulic acid (FA), sodium chloride (NaCl), and sodium hydroxide (NaOH) were purchased from Sigma Chemical Co. (St. Louis, MO, USA). Other chemicals used in this experiment were analytical grade. The water was obtained from a Milli-Q® (Millipore) water purification system (Billerica, MA, USA).

3.2. General

Optical rotations were made on a JASCO DIP-180 digital polarimeter. UV and IR spectra were recorded on a Shimadzu UV-1601PC and a Perkin-Elmer 983 G spectrophotometer, respectively. 1H- and 13C-NMR spectra were acquired on a Varian-Unity-Plus-400 spectrometer with residual solvent signals as internal reference. Chemical shifts are given in δ values and coupling constants (J) are given in hertz (Hz). EI-MS and HR-EI-MS were measured with a Jeol-JMS-HX300 mass spectrometer. Column chromatography (CC) was performed with silica gel (230–400 mesh; Merck & Co., Inc., Kenilworth, NJ, USA). TLC was performed with pre-coated silica gel plates (60 F-254; Merck & Co., Inc., Kenilworth, NJ, USA). Semi-preparative HPLC was performed using a normal phase column (Purospher STAR Si, 5 mm, 250 × 10 mm; Merck & Co., Inc., Kenilworth, NJ, USA) on a LDC Analytical-III system.

3.3. Plant Material

The bark of C. japonica D. Don was collected in Sitou, Taiwan in June, 2000. The plant material was identified by Dr. Yen-Hsueh Tseng, Department of Forestry, National Chung-Hsing University (NCHU). A voucher specimen (TCF13443) was deposited at the Herbarium of the Department of Forestry, NCHU, Taiwan.

3.4. Extraction and Isolation

The air-dried bark of C. japonica (16.0 kg) was extracted by maceration with MeOH (100 L) three times (seven days each time) at room temperature. After filtration, the combined MeOH extract was evaporated under reduced pressure to give a crude extract (480 g). The obtained extract was suspended in H2O (1 L), and successively partitioned with EtOAc (1 L) and n-BuOH (1 L) three times. The EtOAc soluble fraction (430 g) was loaded onto a silica gel (4.0 kg) column and eluted with n-hexane–EtOAc and EtOAc–MeOH mixtures to give 11 fractions, fr. 1 (2.6 g), 2 (29.4 g), 3 (47.8 g), 4 (92.4 g), 5 (21.6 g), 6 (18.1 g), 7 (22.5 g), 8 (35.8 g), 9 (19.2 g), 10 (44.2 g), and 11 (72.2 g). Fr. 3 from hexane/EtOAc (9:1) elution was further purified through a silica gel column (7 cm × 60 cm) and eluted with hexane/CH2Cl2 (1:0–0:1 v/v) to obtain nine fractions, 3A–3I. Further purification of subfraction 3E by HPLC gave 4 (15.1 mg) and 7 (1.5 mg) using hexane/EtOAc (9:1 v/v). Further purification of subfraction 3G by HPLC gave 3 (2.4 mg) using hexane/EtOAc (10:1 v/v). Further purification of subfraction 3H by HPLC gave 1 (1.6 mg) and 2 (3.3 mg) using hexane/EtOAc (9:1 v/v). Fr. 5 from n-hexane–EtOAc (7:3 v/v) elution was further purified over a silica gel column (5 cm × 45 cm), eluted with n-hexane–CH2Cl2–EtOAc (8:8:1 to 0:1:1 v/v/v) to yield fifteen fractions, 5A–5O. Further purification of subfraction 5E by HPLC gave 5 (0.5 mg) and 6 (1.9 mg) using n-hexane–EtOAc (7:3 v/v).
Abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7α-yl peroxide (1). Gum; [α ] D 25 : −21.2 (c = 0.7, CHCl3); EI-MS (70 eV) m/z (rel. int.): 584 ([M]+, 1), 300 (77), 284 (100), 269 (18), 227 (15), 213 (39), 202 (530), 189 (91); HR-EI-MS m/z: 584.4238 [M]+ (calculated for C40H56O3 584.4232); UV (MeOH) λmax (log ε): 220 (4.74), 279 (4.41) nm; IR (KBr) νmax: 3409, 3049, 1593, 1487, 1460, 1407, 1255, 1169, 1043, 1009, 890 cm−l; 1H-NMR and 13C-NMR (400/100 MHz, in CDCl3): See Table 1; chemical spectra is in the Supplementary Materials.
Abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7β-yl peroxide (2). Gum; [α ] D 25 : −54.6 (c = 1.0, CHCl3); EI-MS (70 eV) m/z (rel. int.): 584 ([M]+, 1), 582 (3), 473 (7), 313 (5), 300 (45), 284 (100), 269 (9), 213 (15), 202 (30), 189 (25), 59 (14); HR-EI-MS: m/z: 584.4237 [M]+ (calculated for C40H56O3 584.4232); UV (MeOH) λmax (log ε): 217 (4.80), 276 (4.45) nm; IR (KBr) νmax 3423, 3048, 1590, 1493, 1460, 1407, 1261, 1169, 1062, 1036, 1003, 963, 890, 738 cm−1; 1H-NMR and 13C-NMR (400/100 MHz, in CDCl3): See Table 1; chemical spectra is in the Supplementary Materials.
12-Hydroxyabieta-8,11,13-trien-7β-yl 7-oxoabieta-5,8,11,13-tetraen-12-yl peroxide (3). Gum; [α ] D 25 : −39.7 (c = 0.8, CHCl3); EI-MS m/z 598 ([M]+, 1), 300 (50), 298 (26), 285 (63), 229 (47), 213 (41), 203 (37), 189 (100), 69 (37), 55 (44); HR-EI-MS m/z [M]+: 598.4018 [M]+ (calculated for C40H54O4 598.4024); UV (MeOH) λmax (log ε): 233 (4.34), 282 (4.08), 310 (3.88) nm; IR (KBr) νmax 3376, 3049, 1639, 1586, 1467, 1407, 1275, 1182, 1036, 990, 897 cm−1; 1H-NMR and 13C-NMR (400/100 MHz, in CDCl3): See Table 1; chemical spectra is in the Supplementary Materials.

3.5. Xanthine Oxidase Inhibition Assay

The inhibitory effect on xanthine oxidase of compounds 1–3 was measured spectrophotometrically according to the method reported by Chen et al. with minor modifications [43]. The mixture assay consisted of a 35 μL of 0.1 mM phosphate buffer (pH = 7.5), 30 μL of enzyme solution (0.01 units/mL in 0.1 mM phosphate buffer, pH = 7.5), and 20 μL of the sample solution (final concentration was 50 μM). The mixture was pre-incubated at 25 °C for 15 min, and then was initiated by adding 60 μL of substrate solution (150 mM xanthine in the same buffer). The reaction mixture was incubated for further 30 min at 25 °C. The reaction was stopped by adding 50 μL of 2 N HCl, and the absorbance was measured at 290 nm using a microplate reader. The percentage activity of xanthine oxidase was calculated as following the formula: XO Inhibition (%) = (1 − B/A) × 100, where A and B are the activities of the enzyme without and with test sample. Quercetin, a known inhibitor of xanthine oxidase, was used as a positive control, whereas a negative control was performed without any inhibitor.

3.6. Angiotensin-I Converting Enzyme (ACE) Inhibition Assay

ACE assay was performed using the modified spectrophotometric method described by Cushman and Cheung with minor modifications [44]. The assay mixture contained 30 µL 2.5 mM Hippuryl-l-histidyl-l-leucine (HHL), 10 µL of testing sample at a certain concentration, and 20 µL of ACE (0.05 mU/µL) in 200 mM borate buffer containing 300 mM NaCl (adjusted to pH 8.3). The mixture was incubated at 37 °C for 60 min, and then was halted by addition of 30 μL of 2 N HCl. The substrate HHL and product hippuric acid (HA) liberated through hydrolysis of HHL were determined by HPLC equipped with a Hypersil GOLD C-18 analytical column (250 mm × 4.6 mm, 5 μm). The column was eluted with a mobile phase of 23% ACN containing 0.1% TFA at a constant flow rate of 1 mL/min for 15 min and monitored for absorbance at 228 nm. The inhibition activity was calculated using the following formula: ACE Inhibition (%) = [1 − (∆AInhibitor/∆ABlank)] × 100, where ∆AInhibitor and ∆ABlank were the peak areas of HA in testing and blank samples, respectively. Captopril, a known inhibitor of ACE, was used as a positive control, whereas a negative control was performed without any inhibitor.

4. Conclusions

In this study, three new dimeric abietanoid peroxides, abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7α-yl peroxide (1), abieta-6,8,11,13-tetraen-12-yl 12-hydroxyabieta-8,11,13-trien-7β-yl peroxide (2), and 12-hydroxyabieta-8,11,13-trien-7β-yl 7-oxoabieta-5,8,11,13-tetraen-12-yl peroxide (3), together with four known abietane-type diterpenoids (47) were isolated and characterized from the bark of C. japonica. At a concentration of 50 μM, the three new compounds exhibited the xanthine oxidase inhibitory activity. In addition, compounds 3 also showed ACE inhibitory activity.

Supplementary Materials

The following are available online. 1H-NMR, 13C-NMR, HMQC, HMBC, 1H-1H COSY, NOESY, EIMS, infrared, and ultraviolet-visible spectra of compounds 13. 1H-NMR spectra of compounds 47.

Author Contributions

The study was designed by Y.-H.K. and was performed the experiments, analyzed the data, and wrote the manuscript by C.-I.C., C.-C.C. and C.-R.C. C.-I.C., C.-C.C. and M.-D.W. analyzed the data, and contributed to discussions. Y.-H.K., M.-J.C. and P.-J.S. contributed to discussions and reviewed the manuscripts. All authors read and approved the final version of the manuscript.

Funding

This work was financially supported by the “Chinese Medicine Research Center, China Medical University” from The Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan (CMRC-CHM-4) and Taiwan Ministry of Health and Welfare Clinical Trial Center (MOHW108-TDU-B-212-133004).

Acknowledgments

We thank Shu-Yun Sun and Lih-Mei Sheu for the EI-MS and HR-EI-MS measurement in the Instrumentation Center of the College of Science, National Taiwan University and National Chung Hsing University. We are also grateful to the National Center for High-performance Computing for computer time and facilities.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gan, W.S. Manual of Medicine Plants in Taiwan; National Research Institute of Chinese Medicine: Taipei, Taiwan, 1958; Volume 1, p. 54. [Google Scholar]
  2. Shieh, M.; Iizuka, Y.; Matsubara, Y. Monoterpenoid and sesquiterpenoid constituents of the essential oil of sugi (Cryptomeria japonica D. Don.). Agric. Biol. Chem. 1981, 45, 1493–1495. [Google Scholar] [CrossRef]
  3. Nagahama, S.; Tazaki, M. Terpenoids of wood oil of sugi (Cryptomeria japonica). Peculiarities of Obisugi variety. Mokuzai Gakkaishi 1993, 39, 1077–1083. [Google Scholar]
  4. Nagahama, S.; Tazaki, M.; Sanetika, T.; Nishimura, K.; Tajima, M. Terpenoids of the wood oil of sugi (Cryptomeria japonica). III. Components of Yakusugi. Mokuzai Gakkaishi 1996, 42, 1121–1126. [Google Scholar]
  5. Nagahama, S.; Tazaki, M.; Nomura, H.; Nishimura, K.; Tajima, M.; Iwasita, Y. Terpenoids of the wood oil of sugi (Cryptomeria japonica) IV. Components of form Yabukuguri. Mokuzai Gakkaishi 1996, 42, 1127–1133. [Google Scholar]
  6. Nagahama, S.; Tazaki, M.; Sanetika, T.; Nishimura, K.; Tajima, M. Terpenoids of the wood oil of sugi (Cryptomeria japonica) V. Components of from Ayasugi. Mokuzai Gakkaishi 1998, 44, 282–286. [Google Scholar]
  7. Morita, S.; Yatagai, M.; Fujita, S. Distributions of the extracts and sesquiterpenes in the trunk of Yakusugi (Cryptomeria japonica). Mokuzai Gakkaishi 1995, 41, 938–944. [Google Scholar]
  8. Narita, H.; Yatagai, M.; Ohira, T. Chemical composition of the essential oils from bogwood of Cryptomeria japonica D. Don. J. Essent. Oil Res. 2006, 18, 68–70. [Google Scholar] [CrossRef]
  9. Shimizu, M.; Tsuji, H.; Shogawa, H.; Fukumura, H.; Tanaami, S.; Hayashi, T.; Arisawa, M.; Morita, N. Anti-inflammatory constituents of topically applied crude drugs. II. Constituents and anti-inflammatory effect of Cryptomeria japonica D. Don. Chem. Pharm. Bull. 1988, 36, 3967–3973. [Google Scholar] [CrossRef] [PubMed]
  10. Nagahama, S.; Tazaki, M.; Kobayashi, H.; Sumimoto, M. Sesquiterpene alcohols from Cryptomeria japonica and C. Fortunei leaf oil. Phytochemistry 1993, 33, 879–882. [Google Scholar] [CrossRef]
  11. Su, W.C.; Fang, J.M.; Cheng, Y.S. Hexacarbocyclic triterpenes from leaves of Cryptomeria japonica. Phytochemistry 1993, 34, 779–782. [Google Scholar] [CrossRef]
  12. Su, W.C.; Fang, J.M.; Cheng, Y.S. Abietanes and kauranes from leaves of Cryptomeria japonica. Phytochemistry 1994, 35, 1279–1284. [Google Scholar] [CrossRef]
  13. Su, W.C.; Fang, J.M.; Cheng, Y.S. Labdanes from leaves of Cryptomeria japonica. Phytochemistry 1994, 37, 1109–1114. [Google Scholar]
  14. Su, W.C.; Fang, J.M.; Cheng, Y.S. Sesquiterpenes from leaves of Cryptomeria japonica. Phytochemistry 1995, 39, 603–607. [Google Scholar]
  15. Su, W.C.; Fang, J.M.; Cheng, Y.S. Diterpenoids from leaves of Cryptomeria japonica. Phytochemistry 1996, 41, 255–261. [Google Scholar]
  16. Su, W.C.; Fang, J.M.; Cheng, Y.S. Synthesis and structure determination of cryptomanhydride, an uncommon natural terpenic anhydride. Tetrahedron Lett. 1995, 36, 5367–5370. [Google Scholar] [CrossRef]
  17. Chen, X.H.; Kim, C.S.; Kashiwagi, T.; Tebayashi, S.I.; Horiike, M. Antifeedants against Acusta despesta from the Japanese cedar, Cryptomeria japonica II. Biosci. Biotechnol. Biochem. 2001, 65, 1434–1437. [Google Scholar] [CrossRef] [PubMed]
  18. Morisawa, J.; Kim, C.S.; Kashiwagi, T.; Tebayashi, S.I.; Horiike, M. Repellents in the Japanese cedar, Cryptomeria japonica, against the pill-bug, Armadillidium vulgare. Biosci. Biotechnol. Biochem. 2002, 66, 2424–2428. [Google Scholar] [CrossRef] [PubMed]
  19. Arihara, S.; Umeyama, A.; Bando, S.; Imoto, S.; Ono, M.; Tani, M.; Yoshikawa, K. A new abietane and two dimeric abietane diterpenes from the black heartwood of Cryptomeria japonica. Chem. Pharm. Bull. 2004, 52, 354–358. [Google Scholar] [CrossRef]
  20. Shibuya, T. Cryptoquinonemethides D and E, C 30-terpene quinone methides, from Cryptomeria japonica. Phytochemistry 1992, 31, 4289–4294. [Google Scholar] [CrossRef]
  21. Yoshikawa, K.; Tanaka, T.; Umeyama, A.; Arihara, S. Three abietane diterpenes and two diterpenes incorporated sesquiterpenes from the bark of Cryptomeria japonica. Chem. Pharm. Bull. 2006, 54, 315–319. [Google Scholar] [CrossRef]
  22. Yoshikawa, K.; Suzuki, K.; Umeyama, A.; Arihara, S. Abietane diterpenoids from the barks of Cryptomeria japonica. Chem. Pharm. Bull. 2006, 54, 574–578. [Google Scholar] [CrossRef] [PubMed]
  23. Kofujita, H.; Ota, M.; Takahashi, K.; Kawai, Y.; Hayashi, Y. A diterpene quinone from the bark of Cryptomeria japonica. Phytochemistry 2002, 61, 895–898. [Google Scholar] [CrossRef]
  24. Moiteiro, C.; Esteves, T.; Ramalho, L.; Rojas, R.; Alvarez, S.; Zacchino, S.; Bragança, H. Essential oil characterization of two Azorean Cryptomeria japonica populations and their biological evaluations. Nat. Prod. Commun. 2013, 8, 1785–1790. [Google Scholar] [CrossRef] [PubMed]
  25. Horiba, H.; Nakagawa, T.; Zhu, Q.; Ashour, A.; Watanabe, A.; Shimizu, K. Biological activities of extracts from different parts of Cryptomeria japonica. Nat. Prod. Commun. 2016, 11, 1337–1342. [Google Scholar] [CrossRef] [PubMed]
  26. Shyur, L.F.; Huang, C.C.; Lo, C.P.; Chiu, C.Y.; Chen, Y.P.; Wang, S.Y.; Chang, S.T. Hepatoprotective phytocompounds from Cryptomeria japonica are potent modulators of inflammatory mediators. Phytochemistry 2008, 69, 1348–1358. [Google Scholar] [CrossRef] [PubMed]
  27. Wu, B.; Kashiwagi, T.; Kuroda, I.; Chen, X.H.; Tebayashi, S.I.; Kim, C.S. Antifeedants against Locusta migratoria from the Japanese Cedar, Cryptomeria japonica II. Biosci. Biotechnol. Biochem. 2008, 72, 611–614. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, C.C.; Wu, J.H.; Yang, N.S.; Chang, J.Y.; Kuo, C.C.; Wang, S.Y.; Kuo, Y.H. Cytotoxic C35 terpenoid cryptotrione from the bark of Cryptomeria japonica. Org. Lett. 2010, 12, 2786–2789. [Google Scholar] [CrossRef] [PubMed]
  29. Chang, C.I.; Wang, S.S.; Wu, M.D.; Cheng, M.J.; Ko, H.H.; Chang, H.S.; Chen, J.J.; Chen, C.C.; Kuo, Y.H. Two new sesquarterpenoids from the bark of Cryptomeria japonica. Phytochem. Lett. 2017, 22, 56–60. [Google Scholar] [CrossRef]
  30. Chang, C.I.; Chen, C.C.; Wang, S.Y.; Chao, C.Y.; Chao, L.K.; Chen, J.J.; Ko, H.H.; Chen, C.C.; Kuo, Y.H. Three new abietane-type diterpenes from the bark of Cryptomeria japonica. Helv. Chim. Acta 2016, 99, 710–715. [Google Scholar] [CrossRef]
  31. Chang, C.I.; Chen, C.C.; Wang, S.Y.; Chang, H.S.; Chao, L.K.; Chen, J.J.; Chen, C.C.; Kuo, Y.H. Three new abietane-type diterpenes from the bark of Cryptomeria japonica. Phytochem. Lett. 2017, 19, 46–49. [Google Scholar] [CrossRef]
  32. Chang, C.I.; Chen, C.C.; Chao, C.Y.; Ko, H.H.; Chang, H.S.; Wang, S.Y.; Chen, J.J.; Chen, C.C.; Kuo, Y.H. Two new abietane-type diterpenes from the bark of Cryptomeria japonica. Nat. Prod. Commun. 2017, 12, 1553–1555. [Google Scholar] [CrossRef]
  33. Kuo, Y.H.; Chen, C.H.; Wein, Y.S. New dimeric monoterpenes and dimeric diterpenes from the heartwood of Chamaecyparis obtusa var. formosana. Helv. Chim. Acta 2002, 85, 2657–2663. [Google Scholar] [CrossRef]
  34. Dellar, J.E.; Cole, M.D.; Waterman, P.G. Antimicrobial abietane diterpenoids from Plectranthus elegans. Phytochemistry 1996, 41, 735–738. [Google Scholar] [CrossRef]
  35. Ernst, H.; Guenter, M. Synthese von (+)-Taxodion. Justus Liebigs Ann. Chem. 1989, 7, 677–686. [Google Scholar]
  36. Yang, J.; Peng, C.Z.; Jiang, J.; Huang, G.; Liu, Y. Chemical Constituents of Podocarpus wallichiana. Chem. Nat. Compd. 2016, 52, 142–143. [Google Scholar] [CrossRef]
  37. Zhao, H.; Li, H.; Huang, G.; Chen, Y. A new abietane mono-norditerpenoid from Podocarpus nagi. Nat. Prod. Res. 2017, 31, 844–848. [Google Scholar] [CrossRef]
  38. Miron-Lopez, G.; Bazzocchi, I.L.; Jimenez-Diaz, I.A.; Moujir, L.M.; Quijano-Quinones, R.; Quijano, L.; Mena-Rejon, G.J. Cytotoxic diterpenes from roots of Crossopetalum gaumeri, a Celastraceae species from Yucatan Peninsula. Bioorganic. Med. Chem. Lett. 2014, 24, 2105–2109. [Google Scholar] [CrossRef] [PubMed]
  39. Pereda-Miranda, R.; Delgado, G.; Vivar, A.R.D. An abietane diterpenoid from Salvia sapinae. Phytochemistry 1986, 25, 1931–1933. [Google Scholar] [CrossRef]
  40. Kuo, Y.H.; Wu, T.R.; Cheng, M.C.; Wang, Y. Five new compounds from the heartwood of Juniperus formosana HAYATA. Chem. Pharm. Bull. 1990, 38, 3195–3201. [Google Scholar] [CrossRef]
  41. Cos, P.; Ying, L.; Calomme, M.; Hu, J.P.; Cimanga, K.; Van, P.B.; Pieters, L.; Vlietinck, A.J.; Vanden, B.D. Structure-activity relationship and classification of flavonoids as inhibitors of xanthine oxidase and superoxide scavengers. J. Nat. Prod. 1998, 61, 71–76. [Google Scholar] [CrossRef]
  42. Soffer, R.L. Angiotensin-converting enzyme and the regulation of vasoactive peptides. Annu. Rev. Biochem. 1976, 45, 73–94. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, C.H.; Chan, H.C.; Chu, Y.T.; Ho, H.Y.; Chen, P.Y.; Lee, T.H.; Lee, C.K. Antioxidant activity of some plant extracts towards xanthine oxidase, lipoxygenase and tyrosinase. Molecules 2009, 14, 2947–2958. [Google Scholar] [CrossRef] [PubMed]
  44. Cushman, D.W.; Cheung, H.S. Spectrophotometric assay and properties of the angiotensin-converting enzyme of rabbit lung. Biochem. Pharm. 1971, 20, 1637–1648. [Google Scholar] [CrossRef]
Sample Availability: Samples of the compounds are not available from the authors.
Figure 1. The chemical structures of compounds 17 isolated from C. japonica.
Figure 1. The chemical structures of compounds 17 isolated from C. japonica.
Molecules 24 02178 g001
Figure 2. Some key electron impact (EI)-Mass fragmentations of compounds 1 and 3.
Figure 2. Some key electron impact (EI)-Mass fragmentations of compounds 1 and 3.
Molecules 24 02178 g002
Figure 3. Significant HMBC (one-headed arrows) and NOESY (two-headed arrows) correlations of compounds 13.
Figure 3. Significant HMBC (one-headed arrows) and NOESY (two-headed arrows) correlations of compounds 13.
Molecules 24 02178 g003
Table 1. NMR (nuclear magnetic resonance) data (CDCl3) of compound 13. δ in ppm, J in Hz.
Table 1. NMR (nuclear magnetic resonance) data (CDCl3) of compound 13. δ in ppm, J in Hz.
No.123
δC aδH bδCδHδCδH
141.41.78 m, 1.96 br d (12.0)40.81.80 m, 1.93 br d (12.0)41.01.78 m, 1.93 br d (12.1)
218.81.59 m18.61.60 m18.61.59 m, 1.77 m
342.81.30 td (13.0, 3.5), 1.50 m42.61.25 m, 1.51m42.41.25 m, 1.51 m
434.5 34.8 34.7
543.51.96 m47.61.59 m47.31.72 m
634.02.28 m, 2.32 m34.82.24 br d (14.0), 2.40 m34.22.24 br d (14.3), 2.41 m
797.05.86 br s104.15.20 d (8.0)102.75.38 dd (9.0, 2.5)
8145.6 147.2 146.0
9142.2 143.1 142.5
1040.6 40.7 41.5
11113.36.69 s113.56.74 s114.06.79 s
12148.4 148.9 149.5
13131.9 132.8 133.1
14121.36.34 s120.96.72 s120.76.68 s
1526.32.85 sept (7.0)27.33.01 sept (7.0)27.33.02 sept (7.0)
1622.30.78 d (7.0)22.61.18 d (7.0)22.51.16 d (7.0)
1722.21.01 d (7.0)21.71.16 d (7.0)21.71.11 d (7.0)
1833.50.97 s33.80.95 s33.70.97 s
1923.20.98 s23.11.02 s23.11.01 s
2021.91.36 s21.41.37 s21.71.36 s
12-OH 4.33 s 4.52 s 4.52
1′36.21.55 m, 2.22 br d (13.0)36.31.65 m, 2.13 m38.01.72 m, 2.51 br d (13.5)
2′19.01.75 m, 1.65 m18.81.69 m18.72.00 br d (13.5), 1.70 m
3′41.01.23 m, 1.51m40.91.23 m, 1.51m40.31.45 m, 1.70 m
4′32.8 32.8 37.5
5′51.12.07 dd (3.0, 2.5)51.12.13 dd (3.0, 2.0)173.2
6′127.65.87 dd (9.5, 2.5)128.05.90 dd (9.5,3.0)124.56.48 s
7′127.36.45 dd (9.5, 3.0)127.46.51 dd (9.5,2.0)185.4
8′126.5 127.3 124.5
9′146.6 146.9 153.5
10′38.1 37.9 41.4
11′107.76.98 s109.16.95 s109.67.19 s
12′152.9 153.6 158.2
13′133.6 135.4 136.9
14′124.46.77 s124.56.92 s124.58.03 s
15′27.02.82 sept (7.0)26.53.34 sept (7.0)26.93.32 sept (7.0)
16′21.90.81 d (7.0)22.91.21 d (7.0)22.61.23 d (7.0)
17′22.20.98 d (7.0)22.81.19 d (7.0)22.51.20 d (7.0)
18′32.60.94 s32.60.97 s32.60.97 s
19′22.51.04 s22.51.03 s29.01.03 s
20′20.51.11 s23.01.00 s32.31.49 s
Recorded at a 100 MHz (13C); and b 400 MHz (1H).

Share and Cite

MDPI and ACS Style

Chang, C.-I.; Chen, C.-C.; Chen, C.-R.; Wu, M.-D.; Cheng, M.-J.; Sung, P.-J.; Kuo, Y.-H. Bioactive Dimeric Abietanoid Peroxides from the Bark of Cryptomeria japonica. Molecules 2019, 24, 2178. https://doi.org/10.3390/molecules24112178

AMA Style

Chang C-I, Chen C-C, Chen C-R, Wu M-D, Cheng M-J, Sung P-J, Kuo Y-H. Bioactive Dimeric Abietanoid Peroxides from the Bark of Cryptomeria japonica. Molecules. 2019; 24(11):2178. https://doi.org/10.3390/molecules24112178

Chicago/Turabian Style

Chang, Chi-I, Cheng-Chi Chen, Chiy-Rong Chen, Ming-Der Wu, Ming-Jen Cheng, Ping-Jyun Sung, and Yueh-Hsiung Kuo. 2019. "Bioactive Dimeric Abietanoid Peroxides from the Bark of Cryptomeria japonica" Molecules 24, no. 11: 2178. https://doi.org/10.3390/molecules24112178

Article Metrics

Back to TopTop