Next Article in Journal
Encapsulation and Biological Activity of Hesperetin Derivatives with HP-β-CD
Previous Article in Journal
Solvent-Induced Lag Phase during the Formation of Lysozyme Amyloid Fibrils Triggered by Sodium Dodecyl Sulfate: Biophysical Experimental and In Silico Study of Solvent Effects
Previous Article in Special Issue
Enhancing Efficiency of Natural Product Structure Revision: Leveraging CASE and DFT over Total Synthesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Four New Diterpenoids from the South China Sea Soft Coral Sinularia nanolobata and DFT-Based Structure Elucidation

1
College of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
2
State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
3
School of Medicine, Shanghai University, Shanghai 200444, China
4
Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(19), 6892; https://doi.org/10.3390/molecules28196892
Submission received: 2 September 2023 / Revised: 26 September 2023 / Accepted: 28 September 2023 / Published: 30 September 2023
(This article belongs to the Special Issue Advances in Computer Assisted Structure Elucidation (CASE))

Abstract

:
Three new cembranoids (13) and a new casbanoid (4), along with three known analogues (57), have been isolated from the soft coral Sinularia nanolobata collected off Ximao Island. The structures, including the absolute configurations of new compounds, were established using extensive spectroscopic data analysis, time-dependent density functional theory/electronic circular dichroism (TDDFT-ECD) calculations, and the comparison with spectroscopic data of known compounds. In the in vitro bioassay, compounds 1 and 5 exhibited moderate cytotoxic activities against human erythroleukemia (HEL) cell lines, with IC50 values of 37.1 and 42.4 μM, respectively.

Graphical Abstract

1. Introduction

Soft corals of the genus Sinularia (subclass Octocorallia, order Alcyonacea, family Alcyoniidae) have been well studied by organic chemists for a long time [1,2]. To date, hundreds of secondary metabolites have been discovered in approximately 50 species of this genus [2]. Chemically, the structures of these metabolites can be classified into three main types, namely terpenes, steroids, and prostaglandins, which are responsible for a diverse range of significant bioactivities, especially cytotoxic and anti-inflammatory potentials [1,2,3].
The first chemical and biological study on the soft coral S. nanolobata was performed in 1997, resulting in the isolation of four cytotoxic amphilectane-type diterpenoids [4]. In the following decades, a variety of diterpenoids, nor-diterpenoids, sesquiterpenoids, nor-sesquiterpenoids, steroids, seco-steroids, and steroidal glycosides, which exhibited interesting biological activities, such as anti-inflammatory, cytotoxic, and neuroprotective activities, were isolated from the titled animals [5,6,7,8,9,10,11,12,13]. Their unique structures and excellent bioactivities attracted our ongoing interest to search for more bioactive secondary metabolites from the South China Sea soft corals.
Recently, the soft coral S. nanolobata was collected off Ximao Island, Hainan Province, China, in May 2019. Our previous chemical study on the titled animal resulted in the isolation and characterization of a series of polyoxygenated diterpenoids [14]. Further chemical investigation of the acetone extract of the same sample led to the isolation and characterization of three previously undescribed cembrane-type diterpenoids (13) and a new casbane-type diterpenoid (4) (Figure 1). Herein, we report the isolation, structure elucidation, and cytotoxic activity of these newly isolated compounds.

2. Results

The acetone extract of S. nanolobata was portioned between Et2O and H2O to afford the Et2O-soluble fraction, which was subjected to silica gel column chromatography to yield 12 subfractions. The subfractions were further purified using repeated silica gel, Sephadex LH-20, and reversed-phase HPLC to afford compounds 17. The known compounds were rapidly characterized as grandilobatin D (5) [6], 11,12-epoxy-1E,3E,7E-cembratrien-15-ol (6) [15], and casbene (7) [16] by comparing the observed and reported spectroscopic data.
Compound 1 was obtained as an optically active {[α ] D 20 −13.5 (c 0.25, CHCl3)} colorless oil. Its molecular formula, C21H34O2, was determined using the HR-EIMS ion peak at m/z 318.2554 [M]+ (calcd. for C21H34O2, Figure S1g), implying 5 degrees of unsaturation. The IR spectrum showed the presence of a carbonyl group (1708 cm−1, Figure S1h). The 1H NMR data (Table 1) of 1 displayed two vinyl methyls at δH 1.75 (3H, s) and 1.70 (3H, s), two tertiary methyls at δH 1.29 (3H, s) and 1.30 (3H, s), a bimodal methyl at δH 0.98 (3H, d, J = 6.9 Hz), an oxymethyl at δH 3.02 (3H, s), and three olefinic protons at δH 6.17 (1H, d, J = 10.7 Hz), 5.89 (1H, d, J = 10.7 Hz), and 5.24 (1H, m), respectively. The 13C NMR data together with DEPT and HSQC spectra indicated the presence of 21 carbon signals, which were classified as 6 methyls, 6 methylenes, 4 methines, and 5 quaternary carbons. The aforementioned data revealed that 1 was a cembrane-type diterpenoid and closely resembled that of co-occurring known compound, grandilobatin D (5) [6], with the only difference being the methoxyl group at C-15 in 1 instead of the C-15 hydroxyl group in 5, which is in agreement with the mass data. This replacement caused the 13C NMR resonance of C-15 to be shifted downfield from δC 73.9 to 78.2 in 1. The position of the methoxyl group at C-15 was further confirmed by the HMBC correlation from −OMe (δH 3.02) to C-15 (δC 78.2) (Figure 2). The geometries of the double bonds at Δ1,2, Δ3,4, and Δ7,8 were both assigned to be E by the observed chemical shifts (<20 ppm) of the two vinyl methyls resonance at δC 17.4 (C-18) and δC 17.6 (C-19), along with the NOESY correlations of H-2 (δH 6.17)/Me-18 (δH 1.75), H-3 (δH 5.89)/H2-14 (δH 2.23 and 1.97), and H-7 (δH 5.24)/H2-9 (δH 3.03).
The TDDFT-ECD calculation was carried out to deduce the absolute configuration of 1, which has been proven to be a reliable structure elucidation method for the determination of the absolute configuration of natural products. In detail, torsional sampling conformational searches using MMFFs (Merck Molecular Force Field) were carried out by means of the conformational search module in the Macromodel by applying an energy window of 21 kJ/mol, which afforded 145 conformers for (12S)-1. The Boltzmann populations of the conformers were obtained based on the potential energy provided by the MMFFs, which afforded five conformers for re-optimization. The re-optimization and the following TDDFT calculations of the re-optimized geometries were all performed using Gaussian 09 at the B3LYP/6-311G(d,p) level with IEFPCM (Polarizable Continuum Model using the Integral Equation Formalism variant) solvent model for acetonitrile. Frequency analysis was performed as well to confirm that the re-optimized geometries were at the energy minima. Finally, the SpecDis 1.62 software was used to obtain the Boltzmann-averaged ECD spectra and visualize the results. Detailed comparison of the experimental ECD spectrum with those of calculated ones revealed that the Boltzmann-averaged ECD spectrum of (12S)-1 displayed an identical curve compared to the experimental one (Figure 3). Consequently, the absolute configuration of 1 was determined as 12S.
Compound 2, which was isolated as a colorless oil, gave the molecular formula C21H34O2, the same as that of 1, on the basis of HR-EIMS ion peak at m/z 318.2558 [M]+ (calcd. for C21H34O2, 318.2553). The 1H and 13C NMR data (Table 1) of 2 were virtually identical to those of the known co-isolated compound, 11,12-epoxy-1E,3E,7E-cembratrien-15-ol (6), with the exception of a methoxyl group at C-15 in 2 instead of the C-15 hydroxyl group in 6. The planar structure of 2 was further elucidated via 1H–1H COSY and HMBC experiments (Figure 2). The E geometries of the double bonds Δ3,4 and Δ7,8 in 2 were determined using the chemical shifts (<20 ppm) of the C-18 (δC 18.3) and C-19 (δC 15.1) methyl groups, which were further confirmed by the NOESY cross-peaks of H-2 (δH 6.14)/Me-18 (δH 1.74), and H-7 (δH 5.29)/H2-9 (δH 2.26) (Figure 2). Moreover, the NOESY correlations of H-2/Me-16 (δH 1.30) and H-3 (δH 5.82)/H2-14 (δH 2.12 and 2.03) assigned the E geometry of the double bond Δ1,2. The relative configuration of C-11 and C-12 of 2 were suggested to be the same 11R*, 12R* as those of 6 due to the similar NMR data and the diagnostic NOESY relationships of H-11 (δH 2.90)/H-13β (δH 1.33) and Me-20 (δH 1.25)/H-10β (δH 1.45) (Figure 2). The absolute configuration of 2 was established by the application of the TDDFT-ECD calculation method. In this case, conformational search afforded 171 conformers for (11R, 12R)-2 and 5 conformers for re-optimization and the following TDDFT-ECD calculation. As shown in Figure 3, the Boltzmann-averaged ECD spectrum of (11R, 12R)-2 was matched to the experimental ECD spectrum of 2. Accordingly, the structure of 2 was elucidated as depicted in Figure 1.
Compound 3 was also obtained as a colorless oil with the molecular formula of C21H34O2 on the basis of HR-EIMS ion peak at m/z 318.2566 [M]+ (calcd. for C21H34O2, 318.2553). Analysis of the 1H and 13C NMR data of 3 (Table 2) revealed similarities to 2, except for the location of the methoxyl group from the C-15 in 2 transferred to C-4 in 3, and accompanied by the isomerization of olefins from Δ1,2 and Δ3,4 to Δ1,15 and Δ2,3, respectively. These observations were supported by the HMBC correlations from the methyl protons Me-18 (δH 1.31) to C-3 (δC 130.5), C-4 (δC 77.3), and C-5 (δC 41.7); −OMe (δH 3.07) to C-4 (δC 77.3); Me-16 (δH 1.81) to C-1 (δC 129.5) and C-15 (δC 131.7); and from the olefinic proton H-2 (δH 5.71) to C-15 (Figure 2). The large coupling constant (J2,3 = 16.3 Hz) and the 13C chemical shift of the methyl group Me-19 (δC 14.8) established the E geometries of the double bonds Δ2,3 and Δ7,8. Its relative configuration at C-11 and C-12 was proven to be the same 11R* and 12R* as those of 2 on the basis of the NOESY experiment (Figure 2). The whole relative configuration of the remaining chiral center C-4 and the distant stereochemical domain C-11/C-12 were defined using the QM-NMR calculation and DP4+ analysis [17,18]. These calculation methods utilize Bayes’s theorem to estimate the probability of the selected candidate being correct. The common stages included the generation of plausible isomers and conformational search for each isomer in the gas phase using the MMFFs as applied in the Macromodel software Schrodinger2015-2. Finally, the NMR parameters on the two possible candidate isomers (Figure S5a, 3a: 4R*, 11R*, and 12R*; 3b: 4S*, 11R*, and 12R*) were calculated by the means of gauge including atomic orbitals (GIAO) method at the mPW1PW91/6-31+G(d) level of theory following the DP4+ protocols. As a result, the experimentally observed NMR data of 3 gave the best match of over 99% to the 3b isomer (Figure S5b).
With the relative configuration assigned, the following task was the determination of the absolute configuration of 3. Similarly, TDDFT-ECD calculation method was again applied in this case to determine the absolute configuration of 3. The conformational search of isomer (4S, 11R, 12R)-3 afforded 125 conformers and obtained 5 conformers with Boltzmann populations of more than 1% for the following re-optimization and TDDFT-ECD calculations. As shown in Figure 4, the Boltzmann-averaged ECD spectrum of (4S, 11R, and 12R)-3 highly matched the experimental ECD curve of 3. In light of these evidence, the structure of compound 3 was established as depicted in Figure 1.
Compound 4 was isolated as a colorless oil, possessing the molecular formula of C20H30 by the HR-EIMS ion peak at m/z 270.2342 [M]+ (calcd. for C20H30, 270.2342), suggesting that 4 possessed 6 degrees of unsaturation. The 1H and 13C NMR data (Table 2) of 4 resembled those of the known co-isolated compound, casbene (7), with the exception of a conjugated terminal double bond in 4 instead of the vinyl methyl in 7. This replacement caused the presence of another three olefinic protons and the absence of a methyl signal in the 1H NMR of 4. The planar structure of 4 was further confirmed by the analysis of its 1H–1H COSY and HMBC correlations (Figure 2). The geometries of the double bonds Δ3,4 and Δ7,8 were assigned to be both E by the shielded carbon resonances of the two vinyl methyls at δC 15.8 (C-18) and 18.0 (C-19), along with the obvious NOESY correlations of Me-18 (δH 1.65)/H-2 (δH 1.32) and Me-19 (δH 1.64)/H2-6 (δH 2.16) (Figure 2). Moreover, the large coupling constants (J10,11 = 16.2 Hz) between H-10 and H-11 established the E geometry of the double bond Δ10,11. The 1,2-cis-configuration of C-1 and C-2 was determined by the NOE relationships of H-1/H-2/Me-16 (Figure 2) and the large ∆δC value (13.1 ppm) between the gem-dimethyls C-16 (δC 29.2) and C-17 (δC 16.1). Moreover, the TDDFT-ECD calculation method was also applied to determine the absolute configuration of 4. As a result, the Boltzmann-averaged ECD spectrum of (1S, 2R)-4 highly matched to the experimental one, while the ECD profile of enantiomer (1R, 2S)-4 showed completely opposite curve (Figure 4). Consequently, the absolute configuration of 4 was determined to be 1S, 2R.
In the in vitro bioassay, cembrane-type diterpenoids have been well documented to display the growth inhibitory activities against various cancer cell lines [19]. Accordingly, the cytotoxic activities of all the isolated compounds 17 were evaluated in vitro against HEL (human erythroleukemia cells), H1975 (human lung adenocarcinoma cells), A549 (human non-small cell lung cancer cells), H1299 (human non-small cell lung cancer cells), and MDA-MB-231 (human breast cancer cells) by using the CCK8 and MTT methods. The dose-dependent assay was performed for the determination of IC50 values for the active compounds, and only compounds 1 and 5 exhibited medium cytotoxic activities against HEL cells with IC50 values of 37.09 and 42.37 μM, respectively, compared to that of the positive control doxorubicin (IC50 = 0.05 μM for HEL). In light of the above data, the primary structure–activity relationships of 17 were summarized, and the moderate potency of 1, 5 and the inactivity of 24, 6, 7 suggested that the carbonyl group at C-10 seemed to have a significant impact on the cytotoxic activity against the tested cell lines. Furthermore, the structural comparison for the pair of 1 and 5 revealed that the substitutes at C-15 also contributed to the activity.

3. Discussion

Although this is not the first chemical investigation that we conducted on the soft coral S. nanolobata from the South China Sea, we still obtained some new structures from it in this study. Structurally, all the new compounds 14 shared the same cembrane or casbane-type carbon skeleton with known analogues 57, and these molecules differed from each other mainly in different substituents or double bond positions, which suggested that they underwent a common biosynthesis pathway. In the bioassay, ketone carbonyl compounds 1 and 5 showed potential cytotoxic activities against HEL cells compared to that of inactive compounds, which provided a possible lead scaffold for further structural modifications to design novel anti-tumor drug. Further research should be conducted on the ecological roles of these bioactive secondary metabolites formed during the biosynthesis process of the soft coral.

4. Materials and Methods

4.1. The General Experimental Procedures

Optical rotations were measured on a Perkin-Elmer 241MC polarimeter (PerkinElmer, Fremont, CA, USA). IR spectra were recorded using a Nicolet 6700 spectrometer (Thermo Scientific, Waltham, MA, USA); peaks were reported in cm–1. The NMR spectra were measured at 300 K on Bruker DRX 400 and Avance 600 MHz NMR spectrometers (Bruker Biospin AG, Fallanden, Germany); chemical shifts were reported in parts per million (δ) in CDCl3 (δH reported referred to CHCl3 at 7.26 ppm; δC reported referred to CDCl3 at 77.16 ppm) and coupling constants (J) in Hz; assignments were supported by 1H–1H COSY, HSQC, HMBC, and NOESY experiments. EIMS and HR-EIMS spectra were recorded using a Finnigan-MAT-95 mass spectrometer (ThermoFisher Scientific, Waltham, USA). Semi-preparative HPLC was performed on an Agilent-1260 system equipped with a DAD G1315D detector using ODS-HG-5 (250 mm × 9.4 mm, 5 µm) by eluting with CH3OH–H2O or CH3CN–H2O system at 3 mL/min. Commercial silica gel (200−300 and 400−500 mesh; Qingdao, China) was used for column chromatography. Precoated SiO2 plates (HSGF-254; Yantai, China) were used for analytical TLC. Spots were detected using TLC under UV light or by heating after spraying with an anisaldehyde H2SO4 reagent. All solvents used for extraction and isolation were of analytical grade.

4.2. Biological Material

Specimens of titled animals were collected along the coast of Ximao Island, Hainan province, China, in May 2019, at a depth of −20 m, and were frozen immediately after collection. The high-definition photos and biological samples of the titled animals were sent to Hainan University, and the specimens were accordingly identified as S. nanolobata by Prof. Xiu-Bao Li. The voucher sample is available for inspection at the Shanghai Institute of Materia Medica, SIBS-CAS (No. 19-XD-12).

4.3. Extraction and Isolation

The frozen soft coral (856 g, dry weight after extraction) was extracted exhaustively with acetone at room temperature (3 × 5.0 L). The acetone extract (40 g) was then partitioned between Et2O (3 × 1.0 L) and H2O (3 × 1.0 L), and the Et2O-soluble fraction was concentrated under reduced pressure to obtain a brown residue (16.5 g). Subsequently, the residue was separated into 12 fractions (A-L) via gradient silica gel column chromatography. Fraction A (264 mg) was partially purified using semi-preparative RP-HPLC (CH3CN–H2O, 97:3, 3.0 mL/min) to yield compounds 4 (0.6 mg, tR = 24.4 min) and 7 (4.0 mg, tR = 29.8 min). Fraction G (583 mg) was initially chromatographed using a Sephadex LH-20 column and eluted with PE/DCM/MeOH (2:1:1), affording four subfractions (G1–G4). Purification of subfraction G3 using semi-preparative RP-HPLC (CH3CN–H2O, 60:40) yielded compounds 1 (2.5 mg, tR = 19.4 min), 2 (39.7 mg, tR = 20.8 min), and 3 (4.9 mg, tR = 21.7 min). Fraction H (356 mg) was further chromatographed using a Sephadex LH-20 column and eluted with PE/DCM/MeOH (2:1:1), affording five subfractions (H1–H5). Subfraction H3 was subsequently separated via silica gel column chromatography (300–400 mesh) and eluted with PE–DCM (1:1) to give compounds 5 (21.8 mg) and 6 (3.9 mg).

4.3.1. 12α-methyl-1E,3E,7E-cembratrien-10-one (1)

Colorless oil; [α ] D 20 −13.5 (c 0.25 CHCl3); IR (KBr) νmax = 2928, 2871, 1708, 1456, 1376, 1154, 1072 cm−1; UV (MeCN) λmax 249.0 nm (log ε 4.65); 1H and 13C NMR data, see Table 1; HR-EIMS m/z 318.2554 [M]+ (calcd. for C21H34O2, 318.2553).

4.3.2. 15-methoxyl-11,12-epoxy-1E,3E,7E-cembratrien (2)

Colorless oil; [α ] D 20 −2.8 (c 0.53 CHCl3); IR (KBr) νmax = 2977, 2937, 1144, 1071 cm−1; UV (MeCN) λmax 192.0 nm (log ε 4.29); 1H and 13C NMR data, see Table 1; HR-EIMS m/z 318.2558 [M]+ (calcd. for C21H34O2, 318.2553).

4.3.3. 4α-methoxyl-11,12-epoxy-1,2E,7E-cembratrien (3)

Colorless oil; [α ] D 20 +9.9 (c 0.28 CHCl3); IR (KBr) νmax = 2974, 2934, 1374, 1075 cm−1; UV (MeCN) λmax 243.5 nm (log ε 3.80); 1H and 13C NMR data, see Table 2; HR-EIMS m/z 318.2566 [M]+ (calcd. for C21H34O2, 318.2553).

4.3.4. 2E,7E,10E,12-casbatetraen (4)

Colorless oil; [α ] D 20 −96.7 (c 0.04 CHCl3); IR (KBr) νmax = 2923, 2853, 1456 cm−1; CD (MeCN) λ (∆ε) 207.5 (−3.94), 240.5 (+0.99); UV (MeCN) λmax 203.0 nm (log ε 4.05); 1H and 13C NMR data, see Table 2; HR-EIMS m/z 270.2342 [M]+ (calcd. for C20H30, 270.2342).

4.4. Computational Methods

Conformational searches were carried out using the torsional sampling (MCMM) method and the MMFFs force field. Conformers above 1% of the population were re-optimized at the B3LYP/6-311G(d,p) level using the IEFPCM solvent model for acetonitrile. Subsequently, NMR calculations were performed at the PCM/mPW1PW91/6-31G(d) level, as recommended for DP4+. NMR shielding constants were calculated by using the GIAO method. Finally, the shielding constants were averaged over the Boltzmann distribution obtained for each stereoisomer and correlated with the experimental NMR data. For the resulting geometries, ECD spectra were obtained via TDDFT calculations performed with Gaussian 09 using the same functional, basis set, and solvent model as the energy optimization. At last, the Boltzmann-averaged ECD spectra were obtained using SpecDis 1.62 software.

4.5. Bioactivity Assays

The cytotoxicity of compounds 17 was evaluated by using the CCK8 (HEL) and MTT (H1975, MDA MB-231, A549, and H1299) methods, with doxorubicin (DOX) as the positive control. The growth inhibition of compounds on cancer cells from different tissue sources was tested using five concentration gradients. The maximum concentration of the compounds was 50 μM, diluted five times, and the cancer cells were treated with five concentration gradients for 72 h. Compounds with the highest concentration of 50 μM and an inhibition rate greater than 60% were re-screened, and the half-maximal inhibition (IC50) values were calculated.

5. Conclusions

In summary, three new cembrane-type and one new casbane-type diterpenoids were isolated and characterized from the soft coral S. nanolobata collected off Ximao Island, Hainan Province, China. The structures of the new compounds were established by a combination of extensive spectroscopic analysis, comparison with literature data, and DFT-based quantum chemical calculation-aided configuration analysis. In particular, the relative configuration of 3 was defined using the QM-NMR calculation and DP4+ analysis, and the absolute configurations of 14 were determined using TDDFT ECD calculations. In the in vitro bioassay, compounds 1 and 5 exhibited moderate cytotoxic activities against HEL cells, with IC50 values of 37.09 and 42.37 μM, respectively. The discovery of these new bioactive secondary metabolites once again proved the chemical diversity of the soft coral S. nanolobata.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28196892/s1, Figures S1–S4: HREIMS, 1D NMR, 2D NMR, IR, ECD and UV spectra of compounds 14; Figure S5: QM-NMR calculation of compound 3; Figure S6: Experimental ECD spectra and TDDFT-ECD calculated ECD curves of compounds 14.

Author Contributions

Conceptualization, H.L. and Y.-W.G.; methodology, Y.-W.G.; software, S.-W.L.; validation, S.-W.L. and M.-Z.S.; formal analysis, J.L.; investigation, D.-D.Y. and L.-M.K.; resources, L.-G.Y.; data curation, S.-W.L.; writing—original draft preparation, D.-D.Y. and L.-M.K.; writing—review and editing, S.-W.L.; visualization, J.L.; supervision, M.-Z.S.; project administration, H.L. and Y.-W.G.; funding acquisition, Y.-W.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Key Research and Development Program of China, grant number 2022YFC2804100, and the National Science Foundation of China, grant number 81991521. J. Liu is thankful for the financial support of the Syngenta-SIMM-PhD Studentship Project.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available in Electronic Supporting Information (ESI).

Acknowledgments

The authors would like to thank X.-B. Li from Hainan University for the taxonomic identification of the soft coral material.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are available from the authors.

References

  1. Chen, W.-T.; Li, Y.; Guo, Y.-W. Terpenoids of Sinularia soft corals: Chemistry and bioactivity. Acta Pharm. Sin. B 2012, 2, 227–237. [Google Scholar] [CrossRef]
  2. Yan, X.; Liu, J.; Leng, X.; Ouyang, H. Chemical diversity and biological activity of secondary metabolites from soft coral genus Sinularia since 2013. Mar. Drugs 2021, 19, 335. [Google Scholar] [CrossRef] [PubMed]
  3. Kamel, H.N.; Slattery, M. Terpenoids of Sinularia: Chemistry and biomedical applications. Pharm. Biol. 2008, 43, 253–269. [Google Scholar] [CrossRef]
  4. Yamada, K.; Ujiie, T.; Yoshida, K.; Miyamoto, T.; Higuchi, R. Sinulobatins A-D, New amphilectane-type diterpenoids from the Japanese soft coral Sinularia nanolobata. Tetrahedron 1997, 53, 4569–4578. [Google Scholar] [CrossRef]
  5. Ahmed, A.F.; Su, J.-H.; Shiue, R.-T.; Pan, X.-J.; Dai, C.-F.; Kuo, Y.-H.; Sheu, J.-H. New β-caryophyllene-derived terpenoids from the soft coral Sinularia nanolobata. J. Nat. Prod. 2004, 67, 592–597. [Google Scholar] [CrossRef] [PubMed]
  6. Ahmed, A.F.; Tai, S.-H.; Wen, Z.-H.; Su, J.-H.; Wu, Y.-C.; Hu, W.-P.; Sheu, J.-H. A C-3 methylated isocembranoid and 10-oxocembranoids from a Formosan soft coral, Sinularia grandilobata. J. Nat. Prod. 2008, 71, 946–951. [Google Scholar] [CrossRef] [PubMed]
  7. Tseng, Y.-J.; Wen, Z.-H.; Dai, C.-F.; Chiang, M.Y.; Sheu, J.-H. Nanolobatolide, a new C18 metabolite from the Formosan soft coral Sinularia nanolobata. Org. Lett. 2009, 11, 5030–5032. [Google Scholar] [CrossRef] [PubMed]
  8. Tseng, Y.-J.; Wang, S.-K.; Duh, C.-Y. Secosteroids and norcembranoids from the soft coral Sinularia nanolobata. Mar. Drugs 2013, 11, 3288–3296. [Google Scholar] [CrossRef] [PubMed]
  9. Chao, C.-H.; Huang, T.-Z.; Wu, C.-Y.; Chen, B.-W.; Huang, C.-Y.; Hwang, T.-L.; Dai, C.-F.; Sheu, J.-H. Steroidal and α-tocopherylhydroquinone glycosides from two soft corals Cladiella hirsute and Sinularia nanolobata. RSC Adv. 2015, 5, 74256–74262. [Google Scholar] [CrossRef]
  10. Chao, C.-H.; Wu, C.-Y.; Huang, C.-Y.; Wang, H.-C.; Dai, C.-F.; Wu, Y.-C.; Sheu, J.-H. Cubitanoids and cembranoids from the soft coral Sinularia nanolobata. Mar. Drugs 2016, 14, 150. [Google Scholar] [CrossRef] [PubMed]
  11. Ngoc, N.-T.; Huong, P.T.M.; Thanh, N.V.; Cuong, N.X.; Nam, N.H.; Thung, D.C.; Kiem, P.V.; Minh, C.V. Steroid constituents from the soft coral Sinularia nanolobata. Chem. Pharm. Bull. 2016, 64, 1417–1419. [Google Scholar] [CrossRef] [PubMed]
  12. Ngoc, N.-T.; Huong, P.T.M.; Thanh, N.V.; Cuong, N.X.; Nam, N.H.; Thung, D.C.; Kiem, P.V.; Minh, C.V. Sesquiterpene constituents from the soft coral Sinularia nanolobata. Nat. Prod. Res. 2017, 31, 1799–1804. [Google Scholar] [CrossRef] [PubMed]
  13. Hsu, F.-Y.; Wang, S.-K.; Duh, C.-Y. Xeniaphyllane-derived terpenoids from soft coral Sinularia nanolobata. Mar. Drugs 2018, 14, 40. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, J.; Li, S.-W.; Zhao, Q.-M.; Zhang, Z.-Y.; Yao, L.-G.; Gu, Y.-C.; Lan, L.-F.; Guo, Y.-W. Nanolobatone A, unprecedented diterpenoid and related casbanoids from the Hainan soft coral Sinularia nanolobata. Chem.-Eur. J. 2023, 29, e202300055. [Google Scholar] [CrossRef] [PubMed]
  15. Duh, C.-Y.; Hou, R.-S. Cytotoxic cembranoids from the soft corals Sinularia gibberosa and Sarcophyton trocheliophorum. J. Nat. Prod. 1996, 59, 595–598. [Google Scholar] [CrossRef]
  16. Sitton, D.; West, C.A. Casbene: An anti-fungal diterpene produced in cell-free extracts of Ricznus communis seedings. Phytochemistry 1975, 14, 1921–1925. [Google Scholar] [CrossRef]
  17. Grimblat, N.; Zanardi, M.M.; Sarotti, A.M. Beyond DP4: An improved probability for the stereochemical assignment of isomeric compounds using quantum chemical calculations of NMR shifts. J. Org. Chem. 2015, 80, 12526–12534. [Google Scholar] [CrossRef] [PubMed]
  18. Grimblat, N.; Gavin, J.A.; Daranas, A.H.; Sarotti, A.M. Combining the power of J coupling and DP4 analysis on stereochemical assignments: The J-DP4 methods. Org. Lett. 2019, 21, 4003–4007. [Google Scholar] [CrossRef] [PubMed]
  19. Rodrigues, I.G.; Miguel, M.G.; Mnif, W. A brief review on new naturally occurring cembranoid diterpene derivatives from the soft corals of the genera Sarcophyton, Sinularia, and Lobophytum since 2016. Molecules 2019, 24, 781. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The chemical structures of compounds 17.
Figure 1. The chemical structures of compounds 17.
Molecules 28 06892 g001
Figure 2. The 1H–1H COSY, key HMBC, and NOESY correlations of compounds 14.
Figure 2. The 1H–1H COSY, key HMBC, and NOESY correlations of compounds 14.
Molecules 28 06892 g002
Figure 3. Experimental and calculated ECD spectra of 1 and 2.
Figure 3. Experimental and calculated ECD spectra of 1 and 2.
Molecules 28 06892 g003
Figure 4. Experimental and calculated ECD spectra of 3 and 4.
Figure 4. Experimental and calculated ECD spectra of 3 and 4.
Molecules 28 06892 g004
Table 1. The 1H and 13C NMR data of 1 and 2 in CDCl3 a.
Table 1. The 1H and 13C NMR data of 1 and 2 in CDCl3 a.
No.12
δH, Mult (J in Hz)δC, MultδH, Mult (J in Hz)δC, Mult
1-143.2, s-143.0, s
26.17, d (10.7)122.1, d6.14, d (10.0)122.3, d
35.82, d (10.7)121.8, d5.82, d (10.0)120.3, d
4-137.3, s-138.8, s
52.18, m39.0, t2.17, m38.2, t
2.18, m 2.17, m
62.23, m25.5, t2.27, m25.1, t
2.23, m 2.17, m
75.24, m128.9, d5.28, m127.2, d
8-129.4, s-133.6, s
93.03, m53.1, t2.26, m37.0, t
3.03, m 2.11, m
10-209.9, s2.01, m24.5, t
1.45, m
112.55, dd (14.6, 8.7)50.9, t2.90, dd (9.2, 3.5)61.3, t
2.19, m
122.06, m30.2, d-61.4, d
131.43, m37.4, t2.08, m38.8, t
1.27, m 1.33, m
142.23, m24.6, t2.12, m23.1, t
1.97, m 2.03, m
15-78.2, s-78.0, s
161.29, s27.0, q1.29, s26.4, q
171.30, s25.2, q1.29, s25.8, q
181.75, s17.4, q1.74, s18.3, q
191.70, s17.6, q1.66, s15.1, q
200.98, d (6.9)20.4, q1.25, s17.4, q
−OMe3.02, s50.4, q3.02, s50.4, q
a 1H NMR at 600 MHz, values are reported in ppm referenced to CHCl3 (δH 7.26). 13C NMR at 150 MHz, values are reported in ppm referenced to CDCl3 (δC 77.16). Assignments were aided by HSQC and HMBC experiments.
Table 2. The 1H and 13C NMR data of 3 and 4 in CDCl3 a.
Table 2. The 1H and 13C NMR data of 3 and 4 in CDCl3 a.
No.34
δH, Mult (J in Hz)δC, MultδH, Mult (J in Hz)δC, Mult
1-129.5, s0.62, dt (8.2, 2.6)29.5, d
26.48, d (16.3)127.4, d1.32, m26.0, d
35.71, d (16.3)130.5, d4.83, d (8.2)122.9, d
4-77.3, s-134.8, s
51.92, m41.7, t2.22, dd (12.1, 4.2)39.6, t
1.60, m 2.06, dd (12.1, 4.9)
62.64, m23.0, t2.16, m23.8, t
1.95, m 2.16, m
75.34, br d (7.7)128.6, d5.14, t (5.7)124.2, d
8-132.5, s-134.2, s
92.33, d (13.0)36.9, t2.73, dd (16.4, 5.0)40.8, t
2.10, dd (13.0, 3.1) 2.62, dd (16.4, 9.1)
102.18, dt (12.9, 3.0)24.4, t5.67, ddd (16.2, 9.1, 5.0)130.5, d
1.32, m
112.79, dd (10.8, 2.6)62.6, d5.94, d (16.2)130.8, d
12-61.6, s-147.5, s
132.01, m37.6, t2.31, m34.3, t
1.02, m 2.31, m
142.46, m26.4, t1.49, m25.5, t
2.04, m 1.38, m
15-131.7, s-20.1, s
161.81, s21.5, q1.07, s29.2, q
171.81, s20.4, q0.93, s16.1, q
181.31, s23.3, q1.65, s15.8, q
191.70, s14.8, q1.64, s18.0, q
201.30, s16.3, q4.87, s113.0, t
4.82, s
−OMe3.07, s50.3, q
a 1H NMR at 600 MHz, values are reported in ppm referenced to CHCl3 (δH 7.26). 13C NMR at 150 MHz, values are reported in ppm referenced to CDCl3 (δC 77.16). Assignments were aided by HSQC and HMBC experiments.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yu, D.-D.; Ke, L.-M.; Liu, J.; Li, S.-W.; Su, M.-Z.; Yao, L.-G.; Luo, H.; Guo, Y.-W. Four New Diterpenoids from the South China Sea Soft Coral Sinularia nanolobata and DFT-Based Structure Elucidation. Molecules 2023, 28, 6892. https://doi.org/10.3390/molecules28196892

AMA Style

Yu D-D, Ke L-M, Liu J, Li S-W, Su M-Z, Yao L-G, Luo H, Guo Y-W. Four New Diterpenoids from the South China Sea Soft Coral Sinularia nanolobata and DFT-Based Structure Elucidation. Molecules. 2023; 28(19):6892. https://doi.org/10.3390/molecules28196892

Chicago/Turabian Style

Yu, Dan-Dan, Lin-Mao Ke, Jiao Liu, Song-Wei Li, Ming-Zhi Su, Li-Gong Yao, Hui Luo, and Yue-Wei Guo. 2023. "Four New Diterpenoids from the South China Sea Soft Coral Sinularia nanolobata and DFT-Based Structure Elucidation" Molecules 28, no. 19: 6892. https://doi.org/10.3390/molecules28196892

Article Metrics

Back to TopTop