2013 Volume 61 Issue 5 Pages 511-515
Three new 3,6-disubstituted α-pyrones, nocapyrones H–J (1–3), were isolated from the marine actinomycete Nocardiopsis sp. KMF-001. Their structures were assigned to be 3-alkylated 6-(1-methyl-1-propenyl)-2H-pyran-2-ones on the basis of UV, MS, NMR, and high resolution (HR)-FAB-MS analyses. Nocapyrone H (1) reduced the pro-inflammatory factor such as nitric oxide (NO), prostaglandin E2 (PGE2) and interleukin-1β (IL-1β). Moreover, nocapyrone H showed 5.82% stronger inhibitory effect on NO production than chrysin at a concentration of 10 µm in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells.
In recent decades, marine natural products have been used in the production of new pharmaceuticals due to their great chemical diversity and potent biological activity in a variety of indications.1,2) Marine microorganisms have been considered fascinating sources for the discovery of new pharmacophores.3,4) Especially, the largest number of new biologically active metabolites, especially in antibiotic and antitumor components, have originated from actinomycetes.5,6) The Nocardiopsis is an actinobacterial genus consisting of former members of the genus Actinomadura that Meyer reclassified in 1976.7) Although Nocardiopsis does not represent a major group within actinomycetes, a ubiquitous distribution of Nocardiopsis strains has been reported in various environments, such as saline soil, marine sediment, salterns, honeybee guts, and marine sponges.8–13) Moreover, many secondary metabolites have been isolated from this genus, as for example, nocapyrones A–D,13) pendolmycin,14) lucentamycins A–D,15) 2-pyranone derivatives,16) nocardiopsins A and B,17) fijiolides A and B,18) nocazins A–C, and nocazoline A.19) In addition, Nocardiopsis has also been reported to possess various biologically activities, such as cytotoxicity against HeLa cells,16) tumor necrosis factor-α (TNF-α) inhibition,20) antimicrobial activity,20) protein kinase C inhibition,21) and staurosporine-like inhibition of cyclic AMP-dependent protein kinase.21)
We are screening the neuroprotective actinobacterial metabolites related to prevent of brain inflammation through inhibition of microglial activation. Although a low degree of brain inflammation can contribute to the recovery of damaged tissue from brain injury,22–24) excessive brain inflammation leads to neuronal cell death and is a cause of Parkinson’s disease, Alzheimer’s disease or cerebral ischemia.25–27) Inflammatory responses in the brain primarily result from the activation of astrocytes and microglia, which are the major immune effector cells in the central nervous system (CNS).28,29) Under normal conditions, microglia play a primary role in immune surveillance, and astrocytes serve to sustain the survival of neurons through the release of nerve growth factors and by buffering the activity of neurotransmitters.28,30) However, activated microglia release a variety of pro-inflammatory and cytotoxic factors, such as interleukin-1β (IL-1β), interleukin-6 (IL-6), TNF-α, nitric oxide (NO), reactive oxygen species (ROS) and arachidonic acid metabolites.31) The accumulation of pro-inflammatory and cytotoxic factors is injurious to neurons in vitro, and these factors are thought to actively contribute to the progression of neurodegenerative diseases in vivo.28,32–36)
In order to search for bioactive secondary metabolites from marine sources, we collected marine sediment in Ulleng Basin, the eastern sea of Korea. The actinomycete Nocardiopsis sp. KMF-001, was isolated from one sediment sample, and selected for further investigations due to anti-inflammatory activity of the organic extract. The liquid culture broth of bacterial strain KMF-001 was extracted with ethyl acetate. The ethyl acetate soluble fraction was subjected to vacuum column chromatography and repeated preparative reverse phase (RP)-HPLC to yield three new α-pyrone compounds. Their structures were established by extensive 2D-NMR experiments and high resolution (HR)-FAB-MS. These structures are characterized by 3-alkylated 6-(1-methyl-1-propenyl)-2H-pyran-2-ones, which were designated as nocapyrone H (1), I (2), and J (3). Compound 1 demonstrated neuroprotective effects in inflammation-related brain damage induced by microglial cell activation. Previously reported nocapyrones A–D are the first γ-pyrones reported from Nocardiopsis sp.,13) while nocapyrones E–G isolated from Nocardiopsis dassonvillei HR10-5 are composed of 3,6-disubstituted α-pyrones moiety.19) However, the structures of nocapyrones H (1), I (2), and J (3) are distinguished from the nocapyrones E–G by the functionality at C-6 position of the α-pyrones, which are 2-butenyl group in 1, 2, and 3, while 2-pentenyl group in the nocapyrones E–G.
Nocapyrone H (1) was isolated as a pale yellow oil and its molecular formula was assigned as C13H18O2 based on the HR-FAB-MS ([M+H]+ m/z 207.1389) and 13C-NMR data (Table 1). Its IR absorption band at 1713 cm−1 and UV absorption bands at λmax 234 and 332 nm are consistent with an extended α-pyrone chromophore.37) The 1H-NMR spectrum of 1 displayed signals from two ortho-coupled protons at δH 7.04 (1H, d br q, J=7.0, 1.0 Hz, H-4) and 6.05 (1H, d, J=7.0 Hz, H-5), one olefinic proton at δH 6.61 (1H, q br q, J=7.5, 1.0 Hz, H-8), one methylene at δH 2.29 (2H, d, J=7.0 Hz, H2-11), one methine at δH 1.95 (1H, tq, J=7.0, 7.0 Hz, H-12), two magnetically equivalent methyl doublet groups at δH 0.90 (6H, d, J=7.0 Hz, H3-13 and H3-14), and two other methyl groups at δH 1.81 (3H, d br t, J=7.5, 1.0 Hz, H3-9) and 1.84 (3H, br q, J=1.0 Hz, H-10). The heteronuclear single quantum coherence (HSQC) spectrum showed correlation of all proton signals with the corresponding 13C-NMR signals. The 13C-NMR spectrum also displayed signals for one carbonyl carbon (δC 163.0, C-2) and two quaternary sp2 carbons (δC 159.9 for C-6 and δC 126.0 for C-3); these signals, together with two proton signals at δH 7.04 and 6.05 in the 1H-NMR spectrum, suggested the presence of a 3,6-disubstituted α-pyrone system. The structure of an isobutyl group was assigned by sequential 1H–1H correlation spectroscopy (COSY) correlation between two magnetically equivalent methyl (δH 0.90, H3-13 and 14) signals, a methine (δH 1.95, H-12) signal, and a methylene (δH 2.29, H2-11) signal. In addition, the isobutyl group was connected through the C-3 position of the pyrone, which was determined by the heteronuclear multiple bond connectivity (HMBC) correlations from H-11 to C-2 (δC 163.0), C-3 (δC 126.0), and C-4 (δC 140.3). The 1H–1H COSY spectrum also showed correlations between an olefinic proton (δH 6.61, H-8) and two methyl signals (δH 1.81, H3-9 and δH 1.84, H3-10), which indicated the presence of a 2-butenyl side chain. This 2-butenyl functional group was connected through position 6 of the α-pyrone system based on the HMBC correlation from H-8 and H3-10 to C-6. The 2 dimensional nuclear Overhauser effect spectroscopy (2D NOESY) spectroscopic data could not be clear to assign the geometric configuration of the 2-butenyl chain. However, previously reported NMR data of gibepyrone A and fusalanipyrone proposed differences of 13C chemical shifts between E- and Z-butenyl group (E-form: δC-9 14.10/δC-10 12.05, Z-form δC-9 16.50/δC-10 15.70).38) Consequently, the geometric configuration of the 2-butenyl group in nocapyrone H (1) was consistent with the E-configuration. Thus, the structure of 1 was determined to be (E)-6-(but-2-en-2-yl)-3-isobutyl-2H-pyran-2-one.
Position | 1 | 2 | 3 | ||||
---|---|---|---|---|---|---|---|
δH, mult (J in Hz)a) | δCb) | δH, mult (J in Hz)a) | δCb) | δH, mult (J in Hz)a) | δCb) | δCc) | |
2 | 163.0, C | 163.0, C | 163.0, C | 165.1, C | |||
3 | 126.0, C | 126.8, C | 126.8, C | 128.5, C | |||
4 | 7.04, d br q (7.0, 1.0) | 140.3, CH | 7.08, d br q (7.0, 1.0) | 139.3, CH | 7.08, d br q (7.0, 1.0) | 139.1, CH | 142.1, CH |
5 | 6.05, d (7.0) | 100.4, CH | 6.06, d (7.0) | 100.5, CH | 6.06, d (7.0) | 100.5, CH | 102.5, CH |
6 | 159.9, C | 159.7, C | 159.7, C | 161.0, C | |||
7 | 126.8, C | 127.0, C | 127.2, C | 128.0, C | |||
8 | 6.61, q br q (7.5, 1.0) | 128.2, CH | 6.63, q br q (7.0, 1.0) | 128.2, CH | 6.62, q br q (7.0, 1.0) | 128.2, CH | 129.0, CH |
9 | 1.81, d br t (7.5, 1.0) | 14.1, CH3 | 1.83, d br t (7.0, 1.0) | 14.1, CH3 | 1.83, d br t (7.0, 1.0) | 14.1, CH3 | 14.2, CH3 |
10 | 1.84, br qd) (1.0) | 12.0, CH3 | 1.85, br qd) (1.0) | 12.0, CH3 | 1.86, br qd) (1.0) | 12.0, CH3 | 12.1, CH3 |
11 | 2.29, d (7.0) | 39.8, CH2 | 2.42, t (7.0) | 32.4, CH2 | 2.44, t (7.0) | 30.1, CH2 | 30.8, CH2 |
12 | 1.95, tq (7.0, 7.0) | 27.0, CH | 1.60, tq (7.5, 7.5) | 21.2, CH2 | 1.56, me) | 30.1, CH2 | 31.4, CH2 |
13 | 0.90, d (7.0) | 22.3, CH3 | 0.95, t (7.0) | 13.7, CH3 | 1.37, tq (7.5, 7.5) | 22.4, CH2 | 23.4, CH2 |
14 | 0.90, d (7.0) | 22.3, CH3 | 0.92, t (7.0) | 13.9, CH3 | 14.2, CH3 |
a) 500 MHz, CDCl3. b) 125 MHz, CDCl3. c) 125 MHz, CD3OD. d) Quintet. e) Overlapping signals with the H2O peak. Chemical shifts (δ) are given in ppm.
Nocapyrone I (2) was obtained as a pale yellow oil, and the molecular formula assigned was C12H16O2, which was based on the HR-FAB-MS ([M+H]+ m/z 193.1223) and 13C-NMR data (Table 1). The 1D and 2D NMR spectroscopic data for 2 were similar to those of 1. However, the NMR data of 2 displayed signals attributable to the linear propane chain connected to the α-pyrone moiety. The 1H-NMR spectrum showed two methylene signals at δH 2.42 (t, J=7.0 Hz, H-11)/δH 1.60 (tq, J=7.5, 7.5 Hz, H-12) and one methyl signal at δH 0.95 (t, J=7.0 Hz, H3-13). The 1H–1H COSY correlations between these signals also supported the presence of a propyl group in the structure of 2. The HMBC correlations indicated that the propyl unit was connected through the C-3 position of the α-pyrone moiety. The structure of 2 was accordingly assigned as (E)-6-(but-2-en-2-yl)-3-propyl-2H-pyran-2-one.
Finally, nocapyrone J (3) was isolated as a pale yellow oil, and the molecular formula assigned as C13H18O2, by interpretation of the HR-FAB-MS ([M+H]+ m/z 207.1383) and 13C-NMR data (Table 1). Overall, the 1H- and 13C-NMR spectroscopic of 3 were very similar to those of nocapyrone I (2) except for an additional methylene signal at δH 1.37 (2H, dq, J=7.5, 7.5 Hz, H-13), which indicated the presence of a butyl functionality in the structure of 3. The structure and position of the butyl group was assigned on the basis of a 1H–1H COSY correlation between the methylene signal (H2-13) and a methyl signal at δH 0.92, and an HMBC correlation from the H2-12 signal to C-3 signal. Although the HSQC spectrum showed a clear correlation between H2-13 and the carbon signal at δC 22.4, the other two methylene carbon signals (C-11 and C-12) could not be clearly identified. These C-11 and C-12 methylene signals observed at the same chemical shift (δC 30.1) in the 13C-NMR spectrum of 3 using CDCl3. However, these carbon signals were clearly distinguishable in the 13C-NMR spectrum using CD3OD. H2-11 and H2-12 signals were observed at δH 2.46 and 1.58 in the 1H-NMR spectrum using CD3OD, while the chemical shifts of the C-11 and C-12 signals were δC 30.8 and 31.4, respectively, in the 13C-NMR spectrum using CD3OD (Table 1). Therefore, the structure of compound 3 was determined to be (E)-6-(but-2-en-2-yl)-3-butyl-2H-pyran-2-one.
We investigated the effects of nocapyrones H (1), I (2), and J (3) on the inhibition of NO production in BV-2 microglial cells. Among these compounds, 1 showed significant inhibition on NO production in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells, while 2 and 3 possessed week effect on the NO production (Fig. 2A). We also compared the effect of 1, 2, and 3 with that of chrysin, which is known to reduce NO production.39) As the results, 1 exhibited 5.82% stronger inhibitory effect on NO production than chrysin at a concentration of 10 µm in LPS-stimulated BV-2 microglial cells (Fig. 2A). In addition, 1 increased cell viability in a dose-dependent manner (Fig. 2B). Treatment of BV-2 cells with 1 led to a modest decrease in iNOS protein levels at 10 µm (Fig. S25). The ability of 1 to decrease the production of prostaglandin E2 (PGE2) was investigated using BV-2 microglial cells treated with LPS (100 ng/mL). Pretreatment of primary microglial cells with 1 significantly decreased LPS-induced PGE2 production in a dose-dependent manner (Fig. 3A). We also measured the effects of 1 on cytokines, such as TNF-α and IL-1β, which are related to the LPS-induced inflammatory response in BV-2 microglial cells. 1 suppressed IL-1β secretion in BV-2 cells (Fig. 3B), while 1 increased TNF-α secretion in a dose-dependent manner (Fig. 3C). 1 also showed neuro-protective effects on LPS-induced, microglia-mediated neurotoxicity in N2a cells (Fig. S26-A). Caspase-3 activation was blocked by 1 in a dose-dependent manner, while treatment of 1 did not change expression of the anti- and pro-apoptotic proteins Bcl-2 and Bax, respectively, induced by activated microglia in N2a cells (Fig. S26-B). Even though 1, 2, and 3 have nearly the same structures except alkyl functionalities attached at C-3 position of the α-pyrone, 1 possesses much stronger inhibitory activity on NO production in LPS-stimulated BV-2 cells than 2 and 3. Previously reported literatures demonstrated that the form of alkyl functionalities (e.g., chain length, branch, and attached position) conjugated with aromatic compounds could cause remarkable differences of biological activities.40–43) In this case, nocapyrone H, which has a branched alkyl chain, possesses significant neuro-protective effect.
BV-2 microglial cells were pretreated with 1, 5 and 10 µm nocapyrones 1–3 for 30 min and stimulated with 100 ng/mL of LPS for 24 h. Nocapyrone H (1) inhibited LPS-induced production of NO (A). Nocapyrone H (1) increased cell viability (B). All data are presented as the mean±S.E.M. of three independent experiments. * p<0.05 indicates a significant difference compared with treatment with LPS alone.
BV-2 cells were pretreated with nocapyrone H (1) for 30 min and then stimulated with LPS (100 ng/mL) for 24 h. All data are presented as the mean±S.E.M. of three independent experiments. * p<0.05 indicates a significant difference compared with treatment with LPS alone.
To summarize, we isolated of three new 3,6-disubstitute α-pyrone, nocapyrones H–J (1–3), from marine actinomycete Nocardiopsis sp. KMF-001. In addition, nocapyrone H (1) showed stronger inhibitory effect on NO production at a concentration of 10 µm in LPS-stimulated BV-2 microglial cells. Moreover, nocapyrone (1) reduced the pro-inflammatory factor such as PGE2 and IL-1β in LPS-stimulated BV-2 microglial cells. We suggest that nocapyrone H (1) has neuro-protective effects in the setting of inflammation-related brain damage induced by microglial cell activation.
UV spectra were obtained with a PerkinElmer Lambda 35 UV/Vis spectrophotometer. FT-IR spectra were recorded on a Thermo Scientific Nicolet iS10 spectrometer. 1H- and 13C-NMR data were obtained using a Varian 500 MHz spectrometer with a carbon-enhanced cold probe in CDCl3 and CD3OD. The chemical shift values are reported in ppm and the coupling constants in Hz. Low-resolution electrospray ionization (ESI)-MS was measured on an Agilent Technologies VS/Agilent 1100 system. HR-FAB-MS data were obtained with the aid of a JEOL/JMS-AX505WA instrument. Lichroprep RP-18 (Merck, 40–63 µm) was used for vacuum flash chromatography. A Gilson 321 HPLC system, Luna 10 µm C18 column (250×21.20 mm, 10 µm, Phenomenex®) and Luna 10 µm C18 column (250×10.00 mm, 10 µm, Phenomenex®) with a Gilson UV/Vis-151 detector were used for the HPLC separation of compounds. A Waters 1525 HPLC-PDA system and Luna 5 µm C18 column (150×4.6 mm, 5 µm, Phenomenex®) were used to perform the HPLC analysis.
Identification and Cultivation of Strain KMF-001Strain KMF-001 was isolated from marine sediment collected at a depth of 1198 m in the Ulleung Basin, the eastern sea of Korea. Nearly complete 16S ribosomal RNA (rRNA) gene sequencing of the KMF-001 strain revealed a 99.9% sequence identity with Nocardiopsis dassonvillei. The cultured Nocardiopsis strain KMF-001 was deposited with the Korean Culture Center of Microorganisms (KFCC11531P). The bacterial strain KMF-001 was cultured in 16 replicates at a volume of 500 mL using an A1 liquid culture medium (10 g of starch, 4 g of peptone, and 2 g of yeast extract in 1 L of sea water) for 15 d at 27°C while shaking at 200 rpm. After 15 d, the whole culture broth were extracted with ethyl acetate (16 L overall). The ethyl acetate extract was concentrated to yield 1.6 g of crude extract.
Isolation of Compounds 1–3The crude extract was fractionated by Lichroprep RP-18 (40–63 µm, Merck, NJ, U.S.A.) flash column chromatography using step gradient elution with H2O and CH3OH (20%, 40%, 60%, 80%, and 100%) to give five subfractions. The 100% methanol fraction containing nocapyrones H (1) and J (3) was dried under a vacuum and subjected to reversed phase Prep HPLC separation [Gilson 321; Phenomenex Luna C18 (2) 10 µm column (21.2×250 mm); 10 mL/min; UV detection at 330 nm] using gradient elution with 10% to 100% MeCN/H2O in 0.02% trifluoroacetic acid (TFA) for 120 min. Nocapyrone I (2) was isolated from the 80% methanol fraction by reversed phase Prep HPLC [Gilson 321; Phenomenex Luna C18 (2) 10 µm column (21.2×250 mm); 10 mL/min; UV detection at 330 nm] using gradient elution with 10% to 100% MeCN/H2O in 0.02% TFA for 120 min. 1 (40 mg, tR 98 min), 2 (1.4 mg, tR 90 min), and 3 (0.8 mg, tR 105 min) were purified by reversed phase HPLC [Gilson 321; Phenomenex Luna C18 (2) 10 µm column (10×250 mm), 4 mL/min; UV detection at 330 nm] with an MeCN/H2O (in 0.02% TFA) gradient elution system from 50% to 100% for 60 min.
Nocapyrone H (1)pale yellow oil; 1H-NMR (CDCl3, 500 MHz) and 13C-NMR (CDCl3, 125 MHz) data: see Table 1; IR (film) νmax 2955, 2926, 2866, 1713, 1644, 1558, 1463, 1382, 1087, 805 cm−1; UV (MeCN) λmax (log ε) 203 (3.91), 234 (3.99), 332 (4.00) nm; HR-FAB-MS [M+H]+ m/z 207.1389 (calcd for C13H19O2, 207.1385)
Nocapyrone I (2)pale yellow oil; 1H-NMR (CDCl3, 500 MHz) and 13C-NMR (CDCl3, 125 MHz) data: see Table 1; IR (film) νmax 2951, 2926, 2872, 1713, 1641, 1555, 1454, 1378, 1128, 1112, 811 cm−1; UV (MeCN) λmax (log ε) 200 (3.17), 234 (4.03), 328 (3.96) nm; HR-FAB-MS [M+H]+ m/z 193.1223 (calcd for C12H17O2, 193.1229)
Nocapyrone J (3)pale yellow oil; 1H-NMR (CDCl3, 500 MHz) and 13C-NMR (CDCl3 or CD3OD, 125 MHz) data: see Table 1; IR (film) νmax 2954, 2929, 2870, 1713, 1641, 1562, 1376, 1257, 1130, 1082, 1022, 800 cm−1; UV (MeCN) λmax (log ε) 201 (3.94), 234 (3.97), 327 (3.95) nm; HR-FAB-MS [M+H]+ m/z 207.1383 (calcd for C13H19O2, 207.1385)
Cell Culture39)The BV-2 cell line, derived from murine microglia, was originally developed by Dr. V. Bocchini at the University of Perugia (Perugia, Italy). BV-2 cells have both the phenotypic and functional characteristics of reactive microglia cells. BV-2 cells were grown in Dulbecco’s modified Eagle’s medium (DMEM), supplemented with 5% fetal bovine serum (FBS) and 1% penicillin–streptomycin (PS). N2a cells were maintained in DMEM supplemented with 10% FBS and 1% PS. The cells were cultured in a humidified atmosphere of 5% CO2 at 37°C.
Measurement of NO Production39)To measure NO production, BV-2 cells were plated onto 96-well plates (3×104 cells/well) and treated with 100 ng/mL of LPS in the presence or absence of compound 1 (1, 5, and 10 µm) for 24 h. The release of nitrite, a soluble oxidation product of NO, into the culture medium was determined using the Griess reaction. The supernatant (50 µL) was harvested and mixed with an equal volume of the Griess reagent (1% sulfanilamide and 0.1% N-1-napthylethylenediamine dihydrochloride in 5% phosphoric acid). After 10 min, the absorbance at 570 nm was measured using a microplate reader. Sodium nitrite was used as a standard to calculate the NO2 concentration. Cell viability was measured using a 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT) assay.
Determining Neuronal Cell ViabilityTo determine neuronal cell viability, LPS-challenged BV-2 microglial cells were treated with compound 1, 2, or 3. After 24 h, conditioned medium was collected and used to treat N2a cells. Cell viability was measured using a MTT assay
Measurement of IL-1β, TNF-α, and PGE244–46)To measure IL-1β, PGE2, and TNF-α, cells were plated in a six-well plate at a density of 15×105 cells/well in DMEM and incubated for 24 h before sample treatment. Medium was collected and centrifuged 24 h after treatment with LPS (100 ng/mL) in the presence or absence of compound 1. PGE2, IL-1β, and TNF-α were measured by a competitive enzyme immunoassay kit (specific enzyme-linked immunosorbent assay (ELISA) kit for PGE2, IL-1β, and TNF-α, R&D Systems, Minneapolis, MN, U.S.A.) according to the manufacturer’s protocol.
Western Blot Analysis39,47,48)For the Western blot analysis, cell lysates were separated by 8–12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to a nitrocellulose membrane. After transfer, the membranes were blocked with 5% skim milk and incubated for 12 h with primary antibodies against iNOS, cyclooxygenase-2, Bax, Bcl-2, or cleaved caspase-3 at 4°C, followed by incubation for 1 h with horseradish peroxidase (HRP)-conjugated secondary antibodies at room temperature. Blots were developed using ECL Western blot detection reagents (Amersham Pharmacia Biotech).
Statistical AnalysisAll data are expressed as the mean±S.E.M. Statistical comparisons between the different treatment groups were performed using a one-way ANOVA with Tukey’s multiple comparison post-test. *p-Values <0.05 were considered statistically significant.
Supplemental Information AvailableThe spectroscopic data of nocapyrones H, I and J, as well as the biological activity data of nocapyrone H, are available.
This study was supported by the Korea Institute of Science and Technology institutional program, Grant number 2Z03840, and the Global Leading Technology Program of the Office of Strategic R&D Planning (10039303) funded by the Ministry of Knowledge Economy, Republic of Korea.