Next Article in Journal
Chitin Oligosaccharide (COS) Reduces Antibiotics Dose and Prevents Antibiotics-Caused Side Effects in Adolescent Idiopathic Scoliosis (AIS) Patients with Spinal Fusion Surgery
Previous Article in Journal
Cytotoxic Natural Products from Marine Sponge-Derived Microorganisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Metabolites and Bioactive Chlorinated Benzophenone Derivatives Produced by a Marine-Derived Fungus Pestalotiopsis heterocornis

1
Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
2
CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
3
Shanghai Institute of Pharmaceutical Industry, Shanghai 201203, China
4
Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang 110016, China
*
Authors to whom correspondence should be addressed.
Mar. Drugs 2017, 15(3), 69; https://doi.org/10.3390/md15030069
Submission received: 14 December 2016 / Revised: 19 February 2017 / Accepted: 3 March 2017 / Published: 13 March 2017

Abstract

:
Four new compounds, including two isocoumarins, pestaloisocoumarins A and B (1, 2), one sesquiterpenoid degradation, isopolisin B (4), and one furan derivative, pestalotiol A (5), together with one known isocoumarin, gamahorin (3), and three chlorinated benzophenone derivatives, pestalachloride B (6), pestalachloride E (7) and a mixture of pestalalactone atropisomers (8a/8b), were isolated from a culture of the fungus Pestalotiopsis heterocornis associated with sponge Phakellia fusca. These new chemical structures were established using NMR and MS spectroscopic data, as well as single-crystal X-ray crystallographic analysis and CD Cotton effects. All of the isolated compounds were evaluated for their antimicrobial and cytotoxic activities. Isocoumarins 13, showed antibacterial activities against Gram-positive bacteria Staphylococcus aureus and Bacillus subtilis with MIC values ranging from 25 to 100 μg/mL and weak antifungal activities. Chlorinated benzophenone derivatives 68 exhibited antibacterial activities against S. aureus and B. subtilis with MIC values ranging from 3.0 to 50 μg/mL and cytotoxicities against four human cancer cell lines with IC50 values of 6.8–87.8 μM.

1. Introduction

Fungi of the genus Pestalotiopsis are widely distributed throughout the world and have proven to be a rich source of bioactive natural products [1]. Previous studies on the genus Pestalotiopsis have resulted in the isolation of a series of new natural products, including polyketides [2,3,4,5], terpenoids [6,7,8], alkaloids [9,10] and others [11,12], and many of these compounds exhibited various biological activities [2,3,4,5,6,7,8,9,10,11,12].
In recent decades, the genus Pestalotiopsis has been isolated as an endophyte from the tropical and subtropical rainforest plants [13]. However, only a few fungi of the genus Pestalotiopsis have been reported from marine fauna [14]. Therefore, marine fungi of the genus Pestalotiopsis associated with the sponges could be expected to metabolize biologically interesting and chemically diverse compounds.
Following the above investigations, with the aim of discovering bioactive substances from marine-derived fungi, the secondary metabolites of a culture fermentation of Pestalotiopsis heterocornis, which was isolated from the sponge Phakellia fusca, were investigated. Four new compounds, including two isocoumarins, pestaloisocoumarin A (1) and pestaloisocoumarin B (2), one sesquiterpenoid degradation, isopolisin B (4), and one furan derivative, pestalotiol A (5), together with four known compounds, gamahorin (3) [15], pestalachloride B (6) [16], pestalachloride E (7) [17] and a mixture of pestalalactone atropisomers (8a/8b) [18,19], were discovered (Figure 1). The structures of the new compounds were elucidated on the basis of spectroscopic data, circular dichroism (CD) Cotton effects and single-crystal X-ray crystallographic analysis. The cytotoxicities against four human cancer cell lines, antibacterial and antifungal activities against a panel of bacteria and fungi of these isolated compounds were evaluated in the present paper.

2. Results and Discussion

Compound 1 was obtained as a colorless transparent columnar crystal and possessed a molecular formula of C12H14O5 as defined by the 13C nuclear magnetic resonance (NMR) and high-resolution electrospray ionization–mass spectrometry (HRESI–MS) data. The 1H NMR spectrum (Table 1) displayed signals for two methyls (δH 2.24, s; δH 1.36, d, J = 6.6 Hz), an oxygenated methine (δH 4.90, q, J = 6.6 Hz) and an oxygenated methylene (δH 3.81, d, J = 11.7 Hz; δH 3.55, d, J = 11.7 Hz). In addition, two ortho-coupled aromatic protons at δH 7.05 (d, J = 7.5 Hz) and δH 7.47 (d, J = 7.5 Hz) were observed. The 13C NMR spectrum (Table 1) showed 12 carbons, including six aromatic carbons, one oxygenated quaternary carbon, one oxygenated methine, one hydroxymethyl, two methyls and one ester carbonyl carbon. These observations demonstrated that compound 1 was an isocoumarin derivative. Comparing the NMR data of 1 with those of acremonone C revealed their structural similarity [20], except for the different substituent on the benzene ring. Heteronuclear multiple-bond correlation spectroscopy HMBC correlations from H-9 (δH 1.36) to C-3 (δC 78.0) and C-4 (δC 71.3), from H-10 (δH 3.81 and 3.55) to C-3 (δC 78.0), C-4 (δC 71.3) and C-4a (δC 139.2), from H-11 (δH 2.24) to C-6 (δC 136.9), C-7 (δC 125.7) and C-8 (δC 159.5) confirmed the planar structure of 1 as in Figure 1.
The relative configuration of 1 was defined on the basis of single-crystal X-ray diffraction analysis (Figure 2). The absolute configuration of 1 was determined by the electronic circular dichroism (ECD) spectra with quantum chemical calculations using the time dependent density functional theory (TDDFT) method at the B3 LYP/6-31 + G(d) level. The calculated ECD spectrum showed the same pattern as the experimental ECD spectrum of 1 (Figure 3). Thus, the absolute configuration of 1 was identified as 3R and 4S and named pestaloisocoumarin A.
The HRESI–MS analysis of 2 showed a deprotonated ion at m/z 279.0865 [M − H]. Analysis of the 1H and 13C NMR data (Table 1) indicated that 2 was very similar to gamahorin (3) [15], except that a doublet methyl in 3 was replaced by a singlet methyl in 2, and an oxygenated quaternary carbon was presented in 2 instead of a methine in 3. Furthermore, one more acetyl appeared in 2. HMBC experiments helped to determine the planar structure. HMBC correlations from H-9 (δH 1.43) to C-3 (δC 82.5) and C-4 (δC 67.9), from H-10 (δH 1.57) to C-3 (δC 82.5), C-4 (δC 67.9) and C-4a (δC 145.3), from H-11 (δH 5.17) to C-6 (δC 136.2), C-7 (δC 123.6), C-8 (δC 159.4) and C-13 (δC 171.1) confirmed that the acetyl was linked with C-11 and that C-4 was oxygenated. Nuclear overhauser effect (NOE) correlations between H-3 (δH 4.60) and H-10 (δH 1.57) suggested H-3 and the methyl at C-4 were α-orientated and the methyl at C-3 and the hydroxyl at C-4 were β-orientated. The calculated ECD spectrum of 2 showed excellent agreement with experimental results, and the 3R and 4R configurations of 2 was confirmed (Figure 3). Thus, Compound 2 was determined and named pestaloisocoumarin B.
Compound 4 was isolated as a colorless oil and gave the molecular formula C12H18O3 from HRESI–MS data. The 1H NMR and 13C NMR data (Table 2) of 4 were almost identical to those of the known compound polisin B [21], which is a 11,12,15-norbisabolane sesquiterpenoid. Analysis of the correlation spectroscopy (COSY), heteronuclear single quantum coherence (HSQC) and HMBC correlations exhibited that 4 was a isomer of polisin B. Except for carbons at C-1 (ΔδC +2.0), C-2 (ΔδC −1.2), C-4 (ΔδC +5.2), C-5 (ΔδC +1.3), C-6 (ΔδC +2.4) and C-11 (ΔδC −2.2), the carbons in the lactone ring of 4 presented the same 13C NMR data with those of polisin B, which indicated that 4 possesses the same relative configuration at C-4 and C-7 as polisin B. NOE correlations observed from H-12 (δH 1.37) to H-8α (δH 1.98), H-3α (δH 1.81) and H-6 (δH 4.00), from H-6 (δH 4.00) to H-3α (δH 1.81), from H-4 (δH 2.06) to H-8β (δH 2.24) indicated that H-4 and 6-OH were β-orientated and that H-6 and the methyl at C-7 were α-orientated (Figure 4). Thus, the structure of 4 was assigned and named isopolisin B.
Compound 5 was obtained as colorless oil with the molecular formula C11H20O4 as determined by HRESI–MS. The 1H NMR spectrum (Table 2) showed five methines, including two olefinic methines at δH 5.78, 5.60, three oxygenated methines at δH 4.13, 3.75, 3.58, three methylenes, including one oxygenated methylene at δH 3.69, 3.51 and two methyls at δH 1.75, 0.96. COSY correlations between H-1/H-2/H-3/H-4 and between H-6/H-7/H-8/H-9/H-10 gave two fragments I and II. HMBC correlations between H-11 (δH 3.69, 3.51) and C-5 (δC 81.6) deduced a Fragment III as in Figure 4. HMBC correlations between H-4 (δH 4.13) and C-5 (δC 81.6), C-8 (δC 35.9), between H-6 (δH 3.75) and C-5 (δC 81.6), between H-11 (δH 3.69, 3.51) and C-4 (δC 82.9), C-5 (δC 81.6), C-6 (δC 82.7) confirmed the linkage between Fragments I, II and III (Figure 4). NOE correlations from H-4 (δH 4.13) to H-7 (δH 3.58), H-11 (δH 3.69, 3.51), from H-6 (δH 3.75) to H-8 (δH 1.65) indicated that H-4, H-7 and the hydroxymethyl at C-5 were cis and H-6 and the propyl at C-7 were cis configurations (Figure 4). The configuration of the double bond of 5 was confirmed as E geometry based on the coupling constant values between H-2 and H-3 (JH2,3 = 14.7 Hz). Therefore, the structure of 5 was elucidated and named pestalotiol A.
Compounds 3, 68 were identified as gamahorin (3) [15], pestalachloride B (6) [16], pestalachloride E (7) [17] and a mixture of pestalalactone atropisomers (8a/8b) [18,19] by comparison of the 1H NMR, 13C NMR and mass spectroscopy MS data with those reported.
All of the isolated compounds were evaluated for their cytotoxic activities against four human cancer cell lines via the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay (MTT) assay (Table 3) and antimicrobial activities against three bacteria and three fungi using a micro broth dilution method (Table 4). Chlorinated benzophenone derivatives 6, 7 and a mixture of 8a/8b exhibited moderate cytotoxicities against four human cancer cell lines with half maximal inhibitory concentration (IC50) values 6.8–87.8 μM; while Compounds 15 did not show an obvious inhibition effect against any test cancer cell lines at 100 μM. Isocoumarins 13 and chlorinated benzophenone derivatives 68 showed antibacterial activities against Gram-positive bacteria Staphylococcus aureus and Bacillus subtilis with minimum inhibitory concentration (MIC) values ranging from 3 to 100 μg/mL. Among them, isocoumarins 13 also exhibited weak antifungal activities against three test fungi or part of them with MIC values 100 μg/mL. Compounds 68 were inactive against three test fungi at 100 μg/mL, and Compounds 4, 5 did not show antimicrobial activity against any test microorganism at 100 μg/mL.

3. Experimental Section

3.1. General Experimental Procedures

Optical rotations were determined using an AntonPaar MCP200 automatic polarimeter (Anton Paar Ltd., Graz, Austria). Ultraviolet spectra were measured with a BeckmanCoulter DU 730 nucleic acid/protein analyzer (Beckman Coulter, Inc., Brea, CA, USA). Infra-red (IR) spectra were recorded with a Bruker Tensor 27 FTIR spectrometer (film) (Bruker Optics, Ettlingen, Germany). 1D and 2D NMR spectra were collected on a Bruker AV-600 spectrometer (Bruker, Rheinstetten, Germany), δ in ppm rel. to tetramethylsilane (TMS), J in Hz. ESIMS were recorded on an Agilent 1290-6420 Triple Quadrupole LC–MS spectrometer (Agilent Technologies, Santa Clara, CA, USA). HRESI–MS were performed using a BrukerMicroTOF-Q mass spectrometer (Bruker, Daltonics, Billerica, MA, USA). Silica gel (100–200 mesh, 200–300 mesh, Qingdao Marine Chemical Ltd., Qingdao, China), Sephadex LH-20 (GE Healthcare Bio-sciences AB, Uppsala, Sweden) and YMC*GEL ODS-A (S-50 μm, 12 nm) (YMC Co., Ltd., Kyoto, Japan) were used for column chromatography. Semipreparative high performance liquid chromatography (HPLC) was performed using an Octadecylsilyl silica (ODS) column (250 × 10 mm, 5 μm, YMC-ODS-A). CD spectra were measured on a Biologic MOS-450 spectra polarimeter (Biologic Science, Claix, France). X-ray crystallographic analysis was carried out on a Bruker SMART APEX-II diffractometer (Bruker Biospin Group, Karlstuhe, Germany). MTT and antimicrobial assays were analyzed using a microplate reader (BioTek Synergy H1, BioTek Instruments, Inc., Vermont, VT, USA).

3.2. Fungal Material

The fungal strain, P. heterocornis, was isolated from the sponge P. fusca, which was collected from the Xisha Islands of China in 2012. The strain was identified by Xiuping Lin, and a voucher specimen (No. XWS03F09) was deposited in the CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.

3.3. Fermentation, Extraction and Isolation

The fungal strain P. heterocornis was cultivated in 1000 mL conical flasks containing solid rice medium (each flask contained 200 g of rice, 5 g of artificial sea salt; 200 mL of distilled water, boiled in an autoclave for 20 min at 121 °C, at 28 °C without shaking for 36 days. The total of rice culture was extracted with EtOAc three times. The combined EtOAc extract was evaporated to dryness under reduced pressure to afford 182.5 g of crude extract.
The extract was subjected to silica gel column chromatography (CC) (CH2Cl2/MeOH v/v, 50:1–0:100) to yield 6 fractions (Frs. 1–6). Fraction 3 was subjected to Sephadex LH-20 chromatography (MeOH) to produce three subfractions (Frs. 3.1–3.3). Fr. 3.1 was isolated by CC on silica gel eluted with CH2Cl2/acetone (15:1 to 0:1, v/v) to afford five subfractions (Frs. 3.1.1–3.1.5). Fr. 3.1.2 was further separated by ODS CC, eluting with MeOH/water (85%) and then purified by preparative TLC (CH2Cl2/MeOH, 20:1) to yield Compound 4 (8.5 mg). Fr. 3.1.4 was further separated by ODS CC, eluting with MeOH/water (70%) to give Compound 5 (7.5 mg). Fraction 4 was separated using silica gel column chromatography eluting with CH2Cl2/acetone (10:1) to yield five subfractions (Frs. 4.1–4.5). Fr. 4.5 was subjected to repeated column chromatography (Sephadex LH-20 and ODS) and further purified by semipreparative HPLC (65% MeOH/H2O) to give Compounds 6 (7.0 mg), 7 (5.0 mg) and a mixture of 8a/8b (2.0 mg), respectively. Fraction 6 was fractionated on Sephadex LH-20 eluted with MeOH to produce 3 fractions (Frs. 6.1–6.3). Fr. 6.2 was separated by silica gel column chromatography, eluting with CH2Cl2/acetone (4:1) to yield four subfractions (Frs. 6.2.1–6.2.4). Fr. 6.2.3 was further purified by HPLC (45% MeOH/H2O) to give Compounds 1 (4.5 mg), 2 (8.0 mg) and 3 (8.0 mg), respectively.
Pestaloisocoumarin A (1): colorless transparent columnar crystal; [ α ] D 20 +13.3 (c 0.60, MeOH); UV (MeOH) λmax (log ε) 249 (4.51), 321 (4.37) nm; IR (film) νmax 3301, 2922, 2852, 1727, 1667, 1617, 1459, 1421, 1247, 1123, 1063 cm−1; CD λmax (∆ε) 243 (−4.45), 265 (46.16), 312 (17.72); 1H NMR and 13C NMR data, see Table 1; HRESI–MS m/z 237.0786 [M − H] (calcd. for C12H13O5, 237.0763).
Pestaloisocoumarin B (2): white amorphous solid; [ α ] D 20 −16.0 (c 0.50, MeOH); UV (MeOH) λmax (log ε) 246 (4.32), 321 (4.27) nm; IR (film) νmax 3404, 2991, 2920, 2852, 1736, 1669, 1623, 1431, 1382, 1246, 1158, 1048 cm−1; CD λmax (∆ε) 249 (−12.63), 270 (4.11), 308 (4.17); 1H NMR and 13CNMR data, see Table 1; HRESI–MS m/z 279.0865 [M − H] (calcd. for C14H15O6, 279.0869).
Isopolisin B (4): colorless oil; [ α ] D 20 −19.0 (c 0.84, MeOH); IR (film) νmax 3414.6, 2922.3, 2855.4, 1763.0, 1672.4, 1605.4, 1543.5, 1451.9, 1383.0, 1288.0, 1259.1, 1203.6, 1148.3, 1035.2, 965.5 cm−1; 1H NMR and 13C NMR data, see Table 2; HRESI–MS m/z 233.1292 [M + Na]+ (calcd. for C12H18O3Na, 233.1154).
Pestalotiol A (5): colorless oil; [ α ] D 20 +28.6 (c 0.70, MeOH); IR (film) νmax 3379.5, 2958.2, 2928.2, 2872.4, 1667.0, 1604.8, 1452.8, 1408.4, 1378.4, 1089.8, 1041.6, 969.1, 851.7 cm−1; 1H NMR and 13C NMR data, see Table 2; HRESI–MS m/z 239.1394 [M + Na]+ (calcd. for C11H20O4Na, 239.1259).

3.4. X-ray Crystallography of 1

Colorless transparent columnar crystals of 1 were obtained from a 10:1 (v/v) mixture of MeOH and H2O. Crystal data of 1, C12H14O5, MW = 238.23, monoclinic, crystal size 0.10 × 0.10 × 0.08 mm, space group P21, unit cell dimensions a = 7.5403 (2) Å, b = 12.2371 (4) Å, c = 12.5115 (4) Å, α = 90°, β = 99.153 (2)°, γ = 90°, volume = 1139.75 (6) Å3, Z = 4 ρ calcd. = 1.388 g/cm3, θ range = 3.58–68.28°, Mo Kα radiation, wavelength = 1.54178 Å, temperature = 173 K, F (000) = 504, reflections collected 11,422, unique 4032 [R(int) = 0.0325], completeness to θ = 68.28°, 99.7%, the final refinement gave R1 = 0.0455 and wR2 = 0.1250 (w = 1/σ|F|2), S = 1.054, maximum transmission 0.7531, minimum transmission 0.7323, absolute structure parameter = 0.1 (2).
Bruker SMART APEX-II data collection, the structures were solved by direct methods using SHELXS-97 and refined by means of full-matrix least squares on F2.

3.5. Computational Work

The stable conformational analysis was carried out with SYBYL software (Tripos, San Francisco, CA, USA) using the MMFF94S (Merck Molecular Force Field 94S) force field with an energy cutoff of 10 kcal/mol. The stable conformers were used for geometry optimization at the B3 LYP/6-31G(d) level with a CPCM (conductor-like conductor polarizable continuum model) solvent model for methanol in the Gaussian 09 software. The ECD spectra of stable conformers were then calculated based on the TDDFT method at the B3 LYP/6-31 + G(d) level in methanol [22]. The final ECD curves were generated based on the rotatory strengths with a half-band of 0.3 eV by SpecDis [23] and calculated from the spectra of individual conformers according to their contribution to the Boltzmann weighting. The theoretical spectra have been corrected based on the UV correction.

3.6. Cytotoxicity Assay

The cytotoxicities of 18 were evaluated against four human carcinoma cell lines, including a human gastric carcinoma cell line (BGC-823), a human large-cell lung carcinoma cell line (H460), a human prostate cancer cell line (PC-3) and a human hepatocellular carcinoma cell line (SMMC-7721) in an MTT assay as previously reported [24]. The IC50 value was defined as a 50% reduction of absorbance from the control assay. Adriamycin (Sigma Inc., St Louis, MO, USA) was assayed as a positive control.

3.7. Antimicrobial Assay

A micro broth dilution assay as previously reported [25] was used to evaluate the MICs of 18 against three bacteria (Bacillus subtilis ATCC 6633, Staphylococcus aureus ATCC 25923 and Escherichia coli ATCC 25922) and three fungi (Candida albicans MYA-2867, Candida parapsilosis ATCC 22019 and Cryptococcus neoformans ATCC 208821). The MIC was defined as the lowest concentration of the antimicrobial agent that completely inhibited visual growth of an organism. Ciprofloxacin and amphotericin B (Sigma Inc.) were used as positive controls against bacteria and fungi, respectively.

4. Conclusions

In this study, eight compounds, including four new metabolites, were isolated from the marine-derived fungus P. heterocornis. The structures of the isolated compounds were elucidated by the detailed analysis of spectroscopic data, as well as single-crystal X-ray crystallographic analysis and CD Cotton effects. All compounds were evaluated for their antibacterial and cytotoxic activities. Isocoumarins 13 showed antimicrobial activities against both Gram-positive bacteria and fungi. Chlorinated benzophenone derivatives 68 showed cytotoxicities and antibacterial activities against Gram positive bacteria.

Supplementary Materials

The materials (1D and 2D NMR, HRESI–MS spectra of Compounds 12, 4, 5) are available at www.mdpi.com/1660-3397/15/3/69/s1.

Acknowledgments

This work was funded by National Natural Science Foundation of China (Nos. 81573327 and 41406187), the Basic Scientific Research Fund of Northeastern University, China (Nos. N142002001 and N152006002) and the China Ocean Mineral Resource R&D Association (DY125-22-QY-23).

Author Contributions

Hui Lei contributed to the extraction, isolation and identification. Xiuping Lin and Yonghong Liu accomplished the collection, isolation and identification the fungal strain. Li Han performed the elucidation of structures and prepared manuscript. Jian Ma and Qingjuan Ma contributed to this work by bioassay experiments. Jialiang Zhong determined the X-ray crystallographic data. Jinhui Wang and Tiemin Sun performed the ECD calculation. Xueshi Huang and Li Han revised the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Yang, X.Y.; Zhang, J.Z.; Luo, D.Q. The taxonomy, biology and chemistry of the fungal Pestalotiopsis genus. Nat. Prod. Rep. 2012, 29, 622–641. [Google Scholar] [CrossRef] [PubMed]
  2. Akone, S.H.; Amrani, M.E.; Lin, W.H.; Lai, D.W.; Proksch, P. Cytosporins F-K, new epoxyquinols from the endophytic fungus Pestalotiopsis theae. Tetrahedron Lett. 2013, 54, 6751–6754. [Google Scholar] [CrossRef]
  3. Wang, J.F.; Wei, X.Y.; Lu, X.; Xu, F.Q.; Wan, J.T.; Lin, X.P.; Zhou, X.F.; Liao, S.R.; Yang, B.; Tu, Z.C.; et al. Eight new polyketide metabolites from the fungus Pestalotiopsis vaccinii endogenous with the mangrove plant Kandelia candel (L.) Druce. Tetrahedron 2014, 70, 9695–9701. [Google Scholar] [CrossRef]
  4. Li, J.; Xie, J.; Yang, Y.H.; Li, X.L.; Zeng, Y.; Zhao, P.J. Pestalpolyols A-D, cytotoxic polyketides from Pestalotiopsis sp. cr013. Planta Med. 2015, 81, 1285–1289. [Google Scholar] [CrossRef] [PubMed]
  5. Hwang, I.H.; Swenson, D.C.; Gloer, J.B.; Wicklow, D.T. Disseminins and spiciferone analogues: Polyketide-derived metabolites from a fungicolous isolate of Pestalotiopsis disseminate. J. Nat. Prod. 2016, 79, 523–530. [Google Scholar] [CrossRef] [PubMed]
  6. Liu, L.; Li, Y.; Li, L.; Cao, Y.; Guo, L.D.; Liu, G.; Che, Y.S. Spiroketals of Pestalotiopsis fici provide evidence for a biosynthetic hypothesis involving diversified diels-alder reaction cascades. J. Org. Chem. 2013, 78, 2992–3000. [Google Scholar] [CrossRef] [PubMed]
  7. Hemberger, Y.; Xu, J.; Wray, V.; Proksch, P.; Wu, J.; Bringmann, G. Pestalotiopens A and B: Stereochemically challenging flexible sesquiterpene-cyclopaldic acid hybrids from Pestalotiopsis sp. Chem. Eur. J. 2013, 19, 15556–15564. [Google Scholar] [CrossRef] [PubMed]
  8. Wu, B.; Wu, X.D.; Sun, M.; Li, M.H. Two novel tyrosinase inhibitory sesquiterpenes induced by CuCl2 from a marine-derived fungus Pestalotiopsis sp. Z233. Mar. Drugs 2013, 11, 2713–2721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Luo, D.Q.; Chen, Y.P.; Zhang, J.; Shi, B.Z.; Yang, Z.Q.; Chen, C. A new glycine derivative and a new indole alkaloid from the fermentation broth of the plant endophytic fungus Pestalotiopsis podocarpi isolated from the Chinese podocarpaceae plant Podocarpus macrophyllus. Helv. Chim. Acta 2013, 96, 309–311. [Google Scholar] [CrossRef]
  10. Jia, Y.L.; Wei, M.Y.; Chen, H.Y.; Guan, F.F.; Wang, C.Y.; Shao, C.L. (+)- and (−)- Pestaloxazine A, a pair of antiviral enantiomeric alkaloid dimers with a symmetric spiro[oxazinane-piperazinedione] skeleton from Pestalotiopsis sp. Org. Lett. 2015, 17, 4216–4219. [Google Scholar] [CrossRef] [PubMed]
  11. Yang, X.L.; Zhang, S.; Hu, Q.B.; Luo, D.Q.; Zhan, Y. Phthalide derivatives with antifungal activities against the plant pathogens isolated from the liquid culture of Pestalotiopsis photiniae. J. Antibiot. 2011, 64, 723–727. [Google Scholar] [CrossRef] [PubMed]
  12. Li, J.; Wu, X.F.; Ding, G.; Feng, Y.; Jiang, X.J.; Guo, L.D.; Che, Y.S. α-Pyrones and pyranes from the plant pathogenic fungus Pestalotiopsis scirpina. Eur. J. Org. Chem. 2012, 12, 2445–2452. [Google Scholar] [CrossRef]
  13. Keith, L.M.; Velasquez, M.E.; Zee, F.T. Identification and characterization of Pestalotiopsis spp. causing scab disease of guava, Psidium guajava, in Hawaii. Plant Dis. 2006, 90, 16–23. [Google Scholar] [CrossRef]
  14. Wei, M.Y.; Li, D.; Shao, C.L.; Deng, D.S.; Wang, C.Y. (±)-Pestalachloride D, an antibacterial racemate of chlorinated benzophenone derivative from a soft coral-derived fungus Pestalotiopsis sp. Mar. Drugs 2013, 11, 1050–1060. [Google Scholar] [CrossRef] [PubMed]
  15. Koshino, H.; Yoshihara, T.; Okuno, M.; Sakamura, S.; Tajimi, A.; Shimanuki, T. Gamahonolides A, B, and gamahorin, novel antifungal compounds from stromata of epichloe typhina on phleum pratense. Biosci. Biotechnol. Biochem. 1992, 56, 1096–1099. [Google Scholar] [CrossRef] [PubMed]
  16. Li, E.; Jiang, L.H.; Guo, L.D.; Zhang, H.; Che, Y.S. Pestalachlorides A-C, antifungal metabolites from the plant endophytic fungus Pestalotiopsis adusta. Bioorg. Med. Chem. 2008, 16, 7894–7899. [Google Scholar] [CrossRef] [PubMed]
  17. Xing, Q.; Gan, L.S.; Mou, X.F.; Wang, W.; Wang, C.Y.; Wei, M.Y.; Shao, C.L. Isolation, resolution and biological evaluation of pestalachlorides E and F containing both point and axial chirality. RSC Adv. 2016, 6, 22653–22658. [Google Scholar] [CrossRef]
  18. Shao, C.L.; Wang, C.Y.; Xing, Q.; Wang, K.L. Manufacture of Chlorinated Benzophenone Compounds with Pestalotiopsis for Use as Marine Antifouling Agents. CN Patent 104,163,805, 26 November 2014. [Google Scholar]
  19. Slavov, N.; Cvengros, J.; Neudorfl, J.M.; Schmalz, H.G. Total synthesis of the marine antibiotic pestalone and its surprisingly facile conversion into pestalalactone and pestalachloride A. Angew. Chem. Int. Ed. 2010, 49, 7588–7591. [Google Scholar] [CrossRef] [PubMed]
  20. Rukachaisirikul, V.; Rodglin, A.; Sukpondma, Y.; Phongpaichit, S.; Buatong, J.; Sakayaroj, J. Phthalide and isocoumarin derivatives produced by an Acremonium sp. isolated from a mangrove Rhizophora apiculata. J. Nat. Prod. 2012, 75, 853–858. [Google Scholar] [PubMed]
  21. Wang, S.; Li, Z.H.; Dong, Z.J.; Liu, J.K.; Feng, T. Norbisabolane and eremophilane sesquiterpenoids from cultures of the basidiomycete Polyporus ellisii. Fitoterapia 2013, 91, 194–198. [Google Scholar] [CrossRef] [PubMed]
  22. Mazzeo, G.; Santoro, E.; Andolfi, A.; Cimmino, A.; Troselj, P.; Petrovic, A.G.; Superchi, S.; Evidente, A.; Berova, N. Absolute configurations of fungal and plant metabolites by chiroptical methods. ORD, ECD, and VCD studies on phyllostin, scytolide, and oxysporone. J. Nat. Prod. 2013, 76, 588–599. [Google Scholar] [CrossRef] [PubMed]
  23. Bruhn, T.; Schaumloeffel, A.; Hemberger, Y.; Bringmann, G. SpecDis: Quantifying the comparison of calculated and experimental electronic circular dichroism spectra. Chirality 2013, 25, 243–249. [Google Scholar] [CrossRef] [PubMed]
  24. Zheng, D.; Han, L.; Li, Y.Q.; Li, J.; Rong, H.; Leng, Q.; Jiang, Y.; Zhao, L.X.; Huang, X.S. Isostreptazolin and sannaphenol, two new metabolites from Streptomyces sannanensis. Molecules 2012, 17, 836–842. [Google Scholar] [CrossRef] [PubMed]
  25. Ding, N.; Jiang, Y.; Han, L.; Chen, X.; Ma, J.; Qu, X.; Mu, Y.; Liu, J.; Li, L.; Jiang, C.; et al. Bafilomycins and odoriferous sesquiterpenoids from Streptomyces albolongus isolated from Elephas maximus Feces. J. Nat. Prod. 2016, 79, 799–805. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structures of Compounds 18.
Figure 1. Structures of Compounds 18.
Marinedrugs 15 00069 g001
Figure 2. Single crystal X-ray structure of 1.
Figure 2. Single crystal X-ray structure of 1.
Marinedrugs 15 00069 g002
Figure 3. Electronic circular dichroism (ECD) spectra of 1 and 2.
Figure 3. Electronic circular dichroism (ECD) spectra of 1 and 2.
Marinedrugs 15 00069 g003
Figure 4. 1H-1H COSY (correlation spectroscopy) HMBC (heteronuclear multiple-bond correlation spectroscopy) correlations of 5 and NOE (nuclear overhauser effect) correlations of 4 and 5. Fragment I: -CH-CH=CH-CH3; Fragment II: -CH(OH)CHCH2CH2CH3; Fragment III: The moiety in the blue dotted line box.
Figure 4. 1H-1H COSY (correlation spectroscopy) HMBC (heteronuclear multiple-bond correlation spectroscopy) correlations of 5 and NOE (nuclear overhauser effect) correlations of 4 and 5. Fragment I: -CH-CH=CH-CH3; Fragment II: -CH(OH)CHCH2CH2CH3; Fragment III: The moiety in the blue dotted line box.
Marinedrugs 15 00069 g004
Table 1. 1H nuclear magnetic resonance (NMR) (600 MHz) and 13C NMR (150 MHz) data for Compounds 1 and 2 in CD3OD.
Table 1. 1H nuclear magnetic resonance (NMR) (600 MHz) and 13C NMR (150 MHz) data for Compounds 1 and 2 in CD3OD.
12
PositionδC, typeδH (J in Hz)δC, typeδH (J in Hz)
1168.9, C 169.0, C
378.0, CH4.90, q (6.6)82.5, CH4.60, q (6.6)
471.3, C 67.9, C
4a139.2, C 145.3, C
5115.3, CH7.05, d (7.5)114.2, CH7.16, d (7.7)
6136.9, CH7.47, d (7.5)136.2, CH7.64, d (7.7)
7125.7, C 123.6, C
8159.5, C 159.4, C
8a106.1, C 106.4, C
913.6, CH31.36, d (6.6)13.5, CH31.43, d (6.6)
1065.7, CH23.81, d (11.7)
3.55, d (11.7)
23.6, CH31.57, s
1114.0, CH32.24, s60.3, CH25.17, s
AcO 171.1, C
19.2, CH32.08, s
Table 2. 1H NMR (600 MHz) and 13C NMR (150 MHz) data for Compounds 4 and 5.
Table 2. 1H NMR (600 MHz) and 13C NMR (150 MHz) data for Compounds 4 and 5.
4 (in CD3OD)4 (in CDCl3)5 (in CD3OD)
PositionδC, typeδH (J in Hz)δC, typeδH (J in Hz)δC, typeδH (J in Hz)
1134.4, C 134.7, C 16.7, CH31.75, dd (6.4, 1.5)
2123.5, CH5.57, brd (5.0)124.2, CH5.57, brd (4.4)130.4, CH5.78, dd (14.7, 6.6)
326.1, CH22.10, m 1.81, m26.5, CH22.12, m 1.82, m126.3, CH5.60, brdd (14.7, 7.2)
437.2, CH2.06, m37.4, CH2.03, m82.9, CH4.13, d (7.9)
532.3, CH21.95, brd (13.0)
1.48, td (13.0, 4.0)
32.5, CH21.97, brd (13.2)
1.49, td (13.2, 3.7)
81.6, C
667.1, CH4.00, brs68.0, CH4.07, brs82.7, CH3.75, d (4.5)
789.0, C 88.3, C 84.7, CH3.58, td (6.6, 4.5)
830.5, CH22.24, ddd
(13.0, 10.0, 8.8)
1.98, ddd
(13.0, 10.2, 4.7)
31.2, CH22.19, ddd
(13.2, 10.1, 9.0)
1.94, ddd
(13.2, 10.0, 4.6)
35.9, CH21.65, m
928.4, CH22.72, ddd
(18.3, 10.2, 8.8)
2.58, ddd
(18.3, 10.0, 4.7)
29.0, CH22.65, ddd
(18.2, 10.0, 9.0)
2.57, ddd
(18.2, 10.1, 4.6)
18.9, CH21.50, m 1.42, m
10178.1, C 176.7, C 13.0, CH30.96, t (7.4)
1119.6, CH31.78, s20.8, CH31.80, s62.6, CH23.69, d (11.3)
3.51, d (11.3)
1221.5, CH31.37, s23.1, CH31.37, s
OH 3.65, brs
Table 3. Cytotoxic activities of Compounds 68 (half maximal inhibitory concentration, IC50 in μM).
Table 3. Cytotoxic activities of Compounds 68 (half maximal inhibitory concentration, IC50 in μM).
CompoundBGC-823H460PC-3SMMC-7721
66.823.628.17.9
748.087.855.140.2
8a/8b53.848.266.141.5
Adriamycin1.51.01.82.2
Table 4. Antimicrobial activities of Compounds 13, 68 (minimum inhibitory concentration, MIC μg/mL).
Table 4. Antimicrobial activities of Compounds 13, 68 (minimum inhibitory concentration, MIC μg/mL).
MIC123678a/8bControl
Bacillus subtilis5025100350500.25 a
Staphylococcus aureus2525100325500.13 a
Escherichia coli------0.13 a
Candida albicans100-----1.0 b
Candidad parapsilosis100-100---2.0 b
Cryptococcus neoformans100100100---2.0 b
a Ciprofloxacin; b Amphotericin B.

Share and Cite

MDPI and ACS Style

Lei, H.; Lin, X.; Han, L.; Ma, J.; Ma, Q.; Zhong, J.; Liu, Y.; Sun, T.; Wang, J.; Huang, X. New Metabolites and Bioactive Chlorinated Benzophenone Derivatives Produced by a Marine-Derived Fungus Pestalotiopsis heterocornis. Mar. Drugs 2017, 15, 69. https://doi.org/10.3390/md15030069

AMA Style

Lei H, Lin X, Han L, Ma J, Ma Q, Zhong J, Liu Y, Sun T, Wang J, Huang X. New Metabolites and Bioactive Chlorinated Benzophenone Derivatives Produced by a Marine-Derived Fungus Pestalotiopsis heterocornis. Marine Drugs. 2017; 15(3):69. https://doi.org/10.3390/md15030069

Chicago/Turabian Style

Lei, Hui, Xiuping Lin, Li Han, Jian Ma, Qingjuan Ma, Jialiang Zhong, Yonghong Liu, Tiemin Sun, Jinhui Wang, and Xueshi Huang. 2017. "New Metabolites and Bioactive Chlorinated Benzophenone Derivatives Produced by a Marine-Derived Fungus Pestalotiopsis heterocornis" Marine Drugs 15, no. 3: 69. https://doi.org/10.3390/md15030069

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop