Skip to content
Publicly Available Published by De Gruyter April 26, 2017

New bioactive compounds from the marine-derived actinomycete Nocardiopsis lucentensis sp. ASMR2

  • Essam M. Eliwa , Ahmed S. Abdel-Razek , Marcel Frese , Daniel Wibberg , Ahmed H. Halawa , Ahmed M. El-Agrody , Ahmed H. Bedair , Jörn Kalinowski , Norbert Sewald and Mohamed Shaaban EMAIL logo

Abstract

In the search for new bioactive compounds from extremophilic actinomycetes, a new marine actinomycete strain, Nocardiopsis lucentensis sp. ASMR2 has been isolated and taxonomically identified from marine plants collected in the Red Sea at Hurghada coasts. A large-scale fermentation of the strain on modified rice solid medium was performed, followed by work-up and purification of the obtained extract using a series of chromatographic purifications, delivering the novel butenolide system 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl)propionamide (1a) along with the naturally new 4-methoxy-2H-isoquinolin-1-one (2). Furthermore, eight known bioactive compounds are also reported, namely, indole-3-carboxylic acid, indole-3-acetic acid, indole-3-acetic acid methyl ester, furan-2,5-dimethanol, tyrosol, glycerol linoleate, cyclo-(Tyr, Pro), and adenosine. The chemical structures of the new compounds (1a, 2) were confirmed by extensive one- and two-dimensional (1D and 2D) nuclear magnetic resonance (NMR) spectroscopy, electron ionization high resolution (EI-HR) mass spectrometry, and by comparison with literature data. The antimicrobial activity of the strain extract, as well as of compounds 1a and 2, were studied using a panel of pathogenic microorganisms. The in vitro cytotoxicity of the bacterial extract and compounds 1a and 2 were studied against the human cervix carcinoma cell line (KB-3-1) and its multidrug-resistant subclone (KB-V1).

1 Introduction

Microbial diversity constitutes an infinite pool of novel chemistry, making up a valuable source for innovative biotechnology [1]. The ocean floor has been recently demonstrated as an ecosystem with many unique forms of actinomycetes [2]. Actinomycetes are well-known producers of secondary metabolites [3]. They are widely distributed throughout the ocean and found in intertidal zones [4], seawater [5], animals [5], plants [6], sponges [7], and in ocean sediments [8]. This kind of unique adaptation characteristic of actinomycetes in the marine environment is a rich source for new species and a promising origin for pharmaceutically important compounds [1], [5], [9], [10], [11]. Many of these metabolites possess novel biological activities and have the potential to be developed as therapeutic agents [9]. Sequencing of marine actinomycete genomes may provide useful insights in the discovery of novel agents as well [12].

2 Results and discussion

In the course of our ongoing search for new bioactive compounds from microorganisms, a new marine actinomycete strain was isolated and classified as Nocardiopsis lucentensis sp. ASMR2, based on its 16S rDNA gene sequence, cultivation behavior, and morphological and physiological characteristics. The strain ASMR2 was isolated from a marine plant collected in the Red Sea at the coast of Hurghada, Egypt. Cultivation of the strain on solid rice medium was performed. Chemical studies on the extract obtained from the culture broth of this strain using different chromatographic techniques afforded the new butenolide derivative 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl) propanamide (1a), and the naturally new 4-methoxy-2H-isoquinolin-1-one (2), in addition to eight known metabolites: indole-3-carboxylic acid, indole-3-acetic acid, indole-3-acetic acid methyl ester, furan-2,5-dimethanol, tyrosol, glycerol linoleate, cyclo-(Tyr, Pro), and adenosine. The chemical structures of the new metabolites 1a and 2 were assigned on the basis of 1D and 2D nuclear magnetic resonance (NMR) spectroscopy and EI-HR mass spectrometry.

2.1 Morphological, cultural, and phenotypic characteristics

Cultures of strain ASMR2 grew well on certain International Streptomyces Project (ISP) media used, but light growth was observed on ISP4. The color of the aerial mycelium is mostly pale yellow to pale brown; white on ISP3 and Czapeck’s Dox agar, whereas the reverse sides of cultures were yellow on ISP3 and ISP4 and brown on ISP5. Yellow pigments have been observed on starch-nitrate agar and Czapeck’s Dox agar (Table 1). Based on the microscopic examinations (Fig. 1), spores of the strain ASMR2 are elongated and smooth. Physiologically, the strain ASMR2 is a weak H2S producer and utilizes d-glucose, d-xylose, d-mannose, l-arabinose, and sucrose for growth (Table 2).

Table 1:

Cultural characteristics of N. lucentensis sp. ASMR2.

MediumGrowthAerial myceliumReverse sidePigment
Yeast extract malt extract (ISP2)HeavyPale brownYellowish brown
Oat meal agar (ISP3)GoodWhiteYellow
Inorganic salt starch agar (ISP4)LightPale yellowPale yellow
Glycerol asparagine agar (ISP5)HeavyPale yellowBrown
Starch nitrate agarMediumPale brownReddish brownYellow
Czapeck’s dox agarHeavyWhitePale brownPale yellow

(–): no pigment produced.

Fig. 1: Spore chain structure of strain ASMR2 grown on ISP4 medium (50% seawater, 10 days, 37°C). (a) Light microscopy of sporulating mycelium (1200×) showing Rectiflexibilis; (b) transmission electron micrography of strain showing spore chain with smooth surface (bar=5 μm).
Fig. 1:

Spore chain structure of strain ASMR2 grown on ISP4 medium (50% seawater, 10 days, 37°C). (a) Light microscopy of sporulating mycelium (1200×) showing Rectiflexibilis; (b) transmission electron micrography of strain showing spore chain with smooth surface (bar=5 μm).

Table 2:

Morphological, physiological, and biochemical characteristics of N. lucentensis sp. ASMR2.

CharacteristicResults
Spore massWhite, yellow, or brown
Spore surfaceElongated smooth
Spore chain morphologyRf to zig-zag
Sugar patternNot detected
H2S production+
7% NaCl tolerance+++
Cellulose decomposition
Melanoid pigment production on
 Tryptone-yeast extract agar (ISP1)
 Peptone yeast extract medium (ISP6)
 Tyrosine agar medium (ISP7)
Utilization of sugar
d-Glucose+++
d-Xylose++
d-Mannose+
 Cellubiose++
 Starch++
 Sucrose++

(+) Weak; (++) good; (+++) very good; (–) negative; (Rf) Rectiflexibilis.

For taxonomic classification of strain ASMR2, its internal transcribed spacer (ITS) sequence was sequenced was amplified by applying polymerase chain reaction (PCR), and then sequenced and analyzed in a BLAST-based approach. Bioinformatic analysis of the 895-bp-long 16S rDNA sequence, amplified from a single colony of strain ASMR2, confirmed the highly closed relation of the isolate ASMR2 to N. lucentensis (99%). The results were supported by a neighbor-joining-based phylogenetic tree (Fig. 2). However, by comparing the characteristics of strain ASMR2 with those found in the literature, and the description manuals of actinomycetes [13], [14], [15], [16], [17], [18], [19], it has been confirmed that strain ASMR2 differed from the strain types of both phylogenetic neighbors and other closely similar Nocardiopsis species. The strain ASMR2 has been recorded in GenBank database with accession no. KX581217.

Fig. 2: Phylogenetic tree of strain ASMR2 based on 16S rDNA gene sequences, showing its close relationship to Nocardiopsis species. The tree was constructed using the neighbor-joining method.
Fig. 2:

Phylogenetic tree of strain ASMR2 based on 16S rDNA gene sequences, showing its close relationship to Nocardiopsis species. The tree was constructed using the neighbor-joining method.

2.2 Fermentation and structure elucidation

The N. lucentensis sp. ASMR2 was cultivated on modified solid rice medium containing malt extract (1%) and yeast extract (0.4%). The methanol extract of the strain exhibited strong activity against the yeast Candida albicans (14 mm), and low to moderate activity (9–11 mm) against the yeast Saccharomyces cerevisiae, Gram-positive Bacillus subtilis, and Staphylococcus aureus (Table 6). In the chemical screening monitored by thin layer chromatography (TLC), the bacterial extract exhibited numerous bands in a wide polarity range: some of them showed a group of ultraviolet (UV)-absorbing compounds that were detected as brown, orange, pink, violet, and blue bands by spraying with anisaldehyde/sulfuric acid; others were UV-nonabsorbing, showing violet-blue staining with anisaldehyde/sulfuric acid.

Separation of metabolites produced by the strain using a series of chromatographic techniques afforded the novel butenolide derivative 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl)propanamide (1a), and the naturally new 4-methoxy-2H-isoquinolin-1-one (2). Furthermore, the bacterial extract afforded additional eight known compounds: indole-3-carboxylic acid [20], indole-3-acetic acid [20], indole-3-acetic acid methyl ester [20], furan-2,5-dimethanol [20], tyrosol [20], glycerol linoleate [21], cyclo-(Tyr, Pro) [20], and adenosine [20]. Their structures were confirmed by comparison of the spectroscopic data and chromatographic properties with literature. The physico-chemical properties of 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl) propanamide (1a) and 4-methoxy-2H-isoquinolin-1-one (2) are listed in Table 3.

Table 3:

Physicochemical properties of the new compounds 1a and 2.

1a2
AppearanceColorless semisolidYellow solid
Rfa0.42b0.54c
Staining with anisaldehyde/sulfuric acidOrange
Molecular formulaC7H9NO4 (171)C10H9NO2(175)
(+)-ESI-MS: m/z (%)194 ([M+Na]+, 32), 365 ([2M+Na]+, 100)198 ([M+Na]+)
()-ESI-MS: m/z (%)170 ([MH] )327 ([2MNa] )
EI-MS: m/z (%)171.0 ([M]+, 100), 167.0 ([MCH2]+, 13), 153.0 ([MCO]+, 54), 149 (32), 143 (92), 141 (68), 125.0 (75), 123 (80), 117.0 (20), 113.0 (23), 112.0 (33)175 ([M]+, 24) 174 ([MH]+, 73), 159 (100), 149 (10), 131 (38), 115 (16), 105 (21), 103 (31)
EI-HRMS: m/z171.05241 (calc. 171.05261 for C7H9NO4)
UV/Vis: λmax (log ε), nm (MeOH)252 (4.16)218 (4.32), 260 (4.25)

aSilica gel G/UV254; b(CH2Cl2-10% MeOH); c(CH2Cl2-15% MeOH).

2.3 3′-Hydroxy-N-(2-oxo-2,5-dihydro-furan-4-yl)propionamide

Compound 1a was obtained as a middle-polar colorless semisolid showing UV absorbance on TLC and orange staining on spraying with anisaldehyde/sulfuric acid. The compound exhibited a UV absorbing peak at λmax=252 nm, referring to its aromatic/conjugated olefinic nature. Two quasimolecular ion peaks were revealed in ESI-MS-positive mode, confirming the molecular weight of 1a as 171 Da. EI-HRMS of 1a delivered the molecular formula C7H9NO4, bearing four double bond equivalents (DBE) (Table 3).

The NMR spectra (Table 4) of 1a exhibited two spin systems: one triplet 1H signal (J~1.4 Hz) in the olefinic region at δ=5.70 ppm (δC=94.6 ppm), which exhibited a weak H,H-COSY correlation (Fig. 2) with a 2H triplet signal (J~1.4 Hz) originating from an oxygenated methylene group visible at δ=5.15 ppm (δC=69.8 ppm). The second spin system consisted of two vicinal triplet (J~6.0 Hz) methylene signals at δ=3.88 ppm (δC=57.1 ppm) and δ=2.61 ppm (δC=39.0 ppm) as deduced by H,H-COSY connectivity, which are corresponding to an ethanediyl group flanked by oxygen and sp2 carbon systems, respectively. Based on the 13C NMR/HMQC spectra, compound 1a displayed seven carbon signals, as matched with the molecular formula, which have been classified into two quaternary carbons of ester/amide carbonyls (δ=175.8, 171.4 ppm), an oxygenated (or amide) quaternary sp2 carbon (δ=161.3 ppm), an olefinic methine carbon (δ=94.6 ppm, δH=5.70 ppm), and three methylene sp3 carbons (δ=69.8, 57.1, 39.0 ppm). Accordingly, the structure of compound 1a contains at least one ring system. A search in different databases [AntiBase [3], Dictionary of Natural Products [22], and Scifinder (https://scifinder.cas.org/scifinder)] considering the NMR results and the molecular formula of compound 1a confirmed its structural novelty.

Table 4:

13C (125 MHz) and 1H NMR data (500 MHz, CDCl3) of 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl) propanamide (1a).

No.δC (ppm)δH (ppm)
2175.8
394.66.70 (t, 1.4)
4161.3
569.85.15 (d, 1.4)
1′171.4
2′39.02.61 (t, 6.0)
3′57.13.88 (t, 6.0)

Consequently, an intensive study of the NMR data based on HMBC and H,H-COSY correlations was carried out (Fig. 3): based on the HMBC connectivities, the olefinic proton (H-3) at δ=5.70 ppm exhibited three HMBC correlations at the ester/amide-carbonyl (C-2, δ=175.8 ppm, 2J), oxygenated/amide-bounded carbon at δ=161.3 ppm (C-4, 2J), and the oxygenated methylene carbon at δ=69.8 ppm (C-5, 3J). Proton signals of the last methylene group (δ=5.15 ppm) displayed three alternative correlations at C-2 (δ=175.8 ppm, 3J), C-3 (δ=94.6 ppm, 3J), and C-4 (δ=161.3 ppm, 2J), suggesting one of two five-membered ring systems: (a) 3-amino-butyro-lactone or (b) 3-hydroxy-butyrolactame. The proton signals of the ethanediyl group (CH2-2′, and CH2-3′) system showed 2J and 3J correlations, respectively, at the respite ester/amide carbonyl group (C-1′, δ=171.4 ppm), constructing 3-hydroxy-propionic acid ester or 3-hydroxy-propanamide. Based on previous studies of HMBC, H,H-COSY correlations (Fig. 3), and the molecular formula of 1, two alternative structures have been considered: 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl)propanamide (1a) and 2-oxo-2,5-dihydro-1H-pyrrol-4-yl-3′-hydroxypropionate (1b). A search in different databases confirmed the novelty of both structure formulas. On the basis of the NMR chemical shifts of the methylene group CH2-5 (δH=5.15 ppm, δC=69.8 ppm), it must be included in a lactone five-membered ring 1a but not in a lactam moiety 1b. In the last structure 1b, the methylene group (CH2-5) is shifting mostly at δH=3.63 ppm (δC=45.2 ppm), which differs extremely from our NMR results, excluding such structural probability. Accordingly, 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl) propanamide (1a) is the sole reasonable structure in agreement with the chromatographic property and the NMR data discussed above.

Fig. 3: H,H-COSY (↔, ) and HMBC (→) correlations of 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl) propanamide (1a).
Fig. 3:

H,H-COSY (↔, ) and HMBC (→) correlations of 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl) propanamide (1a).

Butenolide derivatives are a group of important compounds containing a unique carbon skeleton of 2(5H) and 2(3H) furanones, consisting of unsaturated γ-lactone, which are also known as butenolactones/crotonolactones. They are widely present in many natural products [23], [24], for example, goniobutenolides A and B [23], [25], [26], digitoxigenin, lignans (e.g. podophyllotoxin), ascorbic acid, and antibiotics like patulin [27] and fissohamione [28]. Butenolides exhibit a variety of biological properties and have been considered as potential antitumor agents, fungicides, bactericides, insecticides, antibiotics, and anti-inflammatory and allergy inhibitors [23]. γ-Butyrolactones (GBL) are known to be neuropharmacologically active and exhibit anticonvulsant activity as well [23], [29].

2.4 4-Methoxy-2H-isoquinolin-1-one

As a middle-polar yellow solid, compound 2 was obtained displaying an UV absorbance (λ=254 nm) and blue fluorescence (λ=365 nm) during TLC. Nevertheless, it displayed no color staining with anisaldehyde/sulfuric acid. The UV spectrum of 2 displayed a strong absorbing peak at λmax=260 nm, referring to its aromatic nature. The molecular weight of 2 was proved as 175 Da according to electrospray ionization mass spectrometry, with a corresponding molecular formula of C10H9NO2 based on HR-EIMS, containing seven double bond equivalents.

The 1H NMR spectrum (Table 5) of 2 displayed five aromatic signals with each 1H, four among them are attributed to 1,2-disubstituted aromatic residue (J~8.3 Hz), which appeared at δ=8.30 (dd), 8.16 (dd), 7.60 (td), and 7.54 (td) ppm. This conclusion was further confirmed by H,H COSY (Fig. 4). The remaining 1H aromatic signal was visible as a singlet (δ=7.19 ppm). In the aliphatic region, a 3H singlet of an aromatic-bounded methyl ether signal was displayed at δ=3.97 ppm. According to the 13C NMR/HMQC spectra (Table 5), compound 2 displayed 10 carbon signals, which are identical with the revealed molecular formula. They were classified into four sp2 quaternary carbons, two among them are oxygenated (δ=154.9, 147.6 ppm), five are aromatic methines (δ=127.9–101.5 ppm), and one is an aromatic-attached methyl ether (δ=54.6 ppm). Based on its chromatographic properties, the number of unsaturation degrees (7DBE), as well as on the downfield 1HNMR shifting of the 1,2-disubstituted aromatic residue spectroscopic data, an indole structural nature of 2 must be excluded.

Table 5:

13C (125 MHz) and 1H NMR data (500 MHz, CD3OD) of 4-methoxy-2H-isoquinolin-1-one (2).

No.δC (ppm)δH (ppm)
1154.9
3101.57.19 (s)
4147.6
4a129.3
5121.48.16 (dd, 8.5, 1.3)
6127.97.60 (td, 8.3, 1.3)
7125.67.54 (td, 8.1, 1.3)
8123.08.30 (dd, 8.3, 1.4)
8a125.4
4-OCH354.63.97 (s)
Fig. 4: H,H-COSY (↔) and HMBC (→) correlations of 4-methoxy-2H-isoquinolin-1-one (2).
Fig. 4:

H,H-COSY (↔) and HMBC (→) correlations of 4-methoxy-2H-isoquinolin-1-one (2).

A consequent intensive study of compound 2 on the bases of HMBC and H,H-COSY correlations was done (Fig. 4). Based on the HMBC connectivities, the dd proton signal at δ=8.30 ppm (H-8) exhibited five correlations, among them a vital correlation at δ=154.9 ppm (C-1), confirming their location in preposition to each other. The last carbon (C-1, δ=154.9 ppm) is typically for an amide carbonyl present in the aromatized ring system. With respect to the H-8, the para-positioned (H-5) proton signal (δ=8.16 ppm) displayed five HMBC correlations including one 3J coupling with the second oxygenated carbon C-4 (δ=147.6 ppm), confirming their existence in preposition to each other as well. The last carbon (δ=147.6 ppm, C-4) displayed a 3J HMBC cross-section with the methyl ether signal (δ=3.97 ppm) proving their direct attachment. The methine carbon signal at δ=101.5 ppm (C-3, δH=7.19 ppm) is flanked by two resonance electron-donating hetero atoms (i.e. O and N). The corresponding singlet proton signal (δH=7.19 ppm) of C-3 showed 2J and 3J correlations versus C-4 and C-1, respectively (the remaining HMBC correlation of compound 2 is shown in Fig. 4), confirming our suggestion mentioned previously. In agreement with the chromatographic property and the NMR data discussed previously, structure 2 is deduced as 4-methoxy-2H-isoquinolin-1-one. Synthetically, compound 2 is known [30]; however, we have reported it herein for the first time as a new natural product with full assignment as well.

Isoquinolinone and isoquinolinequinone derivatives are commonly known as anticancer agents [31], [32], [33]. Particularly, isoquinolinone derivatives are anti-breast cancer agents targeting estrogen receptor alpha (ERα) and VEGF receptor 2 (VEGFR-2) through inhibition of ERα and VEGFR-2 simultaneously [34]. Further investigation also showed that they possessed good antiproliferation effects against MCF-7 breast cancer cells and potential antiangiogenesis effects in vivo. These kinds of compounds may provide a new and potential route to develop effective drugs for breast cancer [34], [35], [36].

2.5 Biological activities

Antimicrobial activity testing for the crude extract of the marine actinomycetes N. lucentensis sp. ASMR2 was carried out together with the two new compounds 1a and 2 against a set of microorganisms using the agar diffusion technique. Filtrate and cell extracts of strain ASMR2 showed similarity in their low to moderate activity against Gram-positive bacteria (8–12 mm). Remarkably, the microbial extract from bacteria cultivated on rice medium showed pronounced activity against the yeast C. albicans (14 mm), and low to moderate activity (9–11 mm) against the yeast S. cerevisiae, Gram-positive B. subtilis, and S. aureus. However, the compounds 1a and 2 exhibited no activity (Table 6). An in vitro cytotoxicity assay for the strain extracts and the produced compounds (1a, 2) was studied against the human cervix carcinoma cell line (KB-3-1) and its multidrug-resistant subclone (KB-V1), compared with cryptophycin-52 as reference sample, showing no cytotoxicity as well.

Table 6:

Antimicrobial activities of ASMR2 extract and the new compounds 1a, 2 in agar diffusion assays (in mm diameter).

PAaBSbSAcCAdSCeANf
Extract (filtrate)89
Extract (cells)129
Extract (rice medium)1011149
Compound 1a
Compound 2

aP. aeruginosa;bB. subtilis; cS. aureus; dC. albicans; eS. cerevisiae; fA. niger; (–)=no activity.

3 Experimental section

3.1 General experimental procedure

NMR spectra (1H NMR, 13C NMR, DEPT, COSY, HMQC, and HMBC) were measured on Bruker Avance DRX 500 and DRX 600 MHz (Bruker BioSpin, Billerica, MA, USA) spectrometers using standard pulse sequences and referenced to residual solvent signals. EI-HRMS was determined using GCT Premier Spectrometer (Waters, Milford, MA, USA). The UV and visible (UV/Vis) spectra were measured on Spectro UV/Vis Double Beam PC8 scanning auto Cell UVD-3200 (Labomed, Los Angeles, CA, USA), Labomed, Inc. Column chromatography was carried out on silica gel 60 (0.040–0.063 mm, Merck, Darmstadt, Germany) and Sephadex LH-20 (GE Healthcare, Little Chalfont, UK) as the stationary phases. Preparative TLC (0.5 mm thick) and analytical TLC were performed with precoated Merck silica gel 60 PF254+366. Rf values and visualization of chromatograms was carried out under UV light (λ=254 and 366 nm) and further by spraying with anisaldehyde/sulfuric acid followed by heating.

3.1.1 Isolation of the producing strain

The strain ASMR2 was obtained from a marine plant collected in the Red Sea, Hurghada, Egypt, at a depth of 30 m. It was cultivated in starch nitrate medium (g L−1) (starch, 20.0 g; KNO3, 2.0 g; K2HPO4, 1.0 g; MgSO4·7H2O, 0.5 g; NaCl, 0.5 g; FeSO4·7H2O, 0.01 g; CaCO3, 3.0 g; agar, 20.0 g; and 1000 mL of 50% seawater). The pH value was adjusted to 7.2 using 2N NaOH. For the isolation, the plant was washed three times with sterilized water to remove loosely attached salts. The outer surface was sterilized with 70% ethanol and sterilized water. Thereafter, the plant was cut with a sterile scalpel to reach the inner tissue surface. 5 mL of sterilized sea water were added to each sample and incubated for 30 min in a reciprocal water bath at 30°C. A series of 10-fold dilutions were made with sterile seawater and plated (100 μL) on prepared solid media. The plates were then incubated at 28°C for 6–8 weeks. The colonies with distinct morphological characteristics were selected and transferred onto freshly prepared solid media and stored in a refrigerator at 4°C until use. The strain was deposited at the Microbial Chemistry Department, National Research Centre, Egypt.

3.1.2 DNA isolation and 16S rDNA sequencing

The strain was inoculated in 100 mL Erlenmeyer flasks each containing 50 mL of ISP2 medium containing 10 gl−1 malt extract, 4 gl−1 yeast extract and 4 gl−1 glucose, pH 7.2 adjusted with 2N NaOH. The culture was incubated at 28°C for 3 days. 3 mL of culture were centrifuged at 5000×g for 10 min. The resulting cell pellet was used for genomic DNA isolation using a bacterial genome DNA isolation kit (Qiagen DNeasy Blood & Tissue kit, Cat. No: 69504, Hilden, Germany) following the manufacturer’s manual. DNA amplification was carried out with primer set 9F (5′-GAGTTTGATCCTGGCTCAG3′) and 1541R (5′-AGGAGGTGATCCAACC3′) (9F/1541R). The following amplification profile was used: an initial denaturation step at 94°C for 2 min was followed for 30 amplification cycles of 94°C for 1 min, 55°C for 1 min, and 72°C for 2 min and a final extension step of 72°C for 2 min. The PCR product was separated by agarose gel electrophoresis and visualized by UV fluorescence after ethidium bromide staining [37]. PCR cleanup was performed according to the description manual of the Qiagen kit: 28104. DNA sequences were determined by the CeBiTec DNA sequencing core facility on an Applied Biosystems AbiPrism 3730 sequencer using BigDye-terminator chemistry. Premixed reagents from Applied Biosystems were used. The results of 16S rDNA sequence were compared to the available database at GenBank by using BLAST software (blastn) from the National Centre Biotechnology Information (http://www.ncbi.nlm.nih.gov). The phylogenetic tree was constructed using the neighbor-joining tree method.

3.1.3 Morphological, cultural, and physiological characterization of strain ASMR2

The cultivation characteristics of the strain ASMR2 were studied according to the guidelines established in the International Streptomyces Project [38]. The aerial mycelium, pigmentation, and morphological features of the strain ASMR2 were examined after its growth on different ISP media, starch nitrate medium, and Czapeck’s Dox agar medium for 2, 3, and 4 weeks and the observations were recorded weekly. The micromorphology was studied on a culture grown at 37°C for 10 days on ISP4 medium [17]. The cells were examined for spore chain morphology under light microscope (Olympus CH-2) at a magnification of 1200×. Electron micrographs were made with transmission electron microscope at the National Research Centre, Egypt. Physiological and biochemical characteristics were studied by hydrogen sulfide production [39], cellulose decomposition [40], NaCl tolerance [41], and different carbon sources utilization [16].

3.2 Fermentation A (culture broth medium), work up and isolation

1 mL of prepared seed culture of strain ASMR2 was used to inoculate 18 1-L Erlenmeyer flasks containing 300 mL of ISP2 medium (50% seawater). The flasks were incubated in a shaking incubator (100 rpm) at 30°C for 7 days. The culture was harvested and centrifuged (4500 rpm at 5°C) to separate cells. The medium was extracted by ethyl acetate and the cells were extracted with acetone. Acetone and water were evaporated in vacuo. Finally, the residue was re-extracted with ethyl acetate. Chromatographic purification of the afforded bacterial extract (0.9 g) was carried out using silica gel column (100×2 cm), and eluted with petroleum ether-dichloromethane-methanol gradient (0.5 L petroleum ether, 0.5 L petroleum ether-CH2Cl2 [1:1], 0.5 L CH2Cl2, 0.5 L CH2Cl2-MeOH [97:2], 1 L CH2Cl2-MeOH [95:5], 0.5 L CH2Cl2-MeOH [93:7], 1 L CH2Cl2-MeOH [90:10], 0.5 L CH2Cl2-MeOH [80:20], 0.5 L CH2Cl2-MeOH [50:50]; 0.5 L MeOH). According to TLC monitoring, three fractions were obtained: FI (0.32 g), FII (0.28 g), and FIII (0.3 g). Further column chromatography of fraction III by Sephadex LH-20 (CH2Cl2/40% MeOH) and silica gel afforded furan-2,5-dimethanol (14 mg).

3.3 Fermentation B (rice medium), work up and isolation

100 mL of ISP2 medium was inoculated with a spore suspension of strain ASMR2 and cultivated at 28°C for 3 days as seed culture. A total of 5 mL of this seed culture was used to inoculate 1-L Erlenmeyer flasks (12 flasks) containing modified rice medium, consisting of 100 g of commercial rice; 150 mL of distilled water/seawater (1:1) supplemented with 0.4% yeast extract, and 1% malt extract. The flasks were incubated for 14 days at 37°C. After harvesting, 0.5 L methanol was added to each flask, and then applied to vigorous shaking for 5 min. The obtained aqueous methanol extract was then separated from rice by filtration under vacuum. After filtration, the water/methanol fraction was evaporated to remove methanol using a rotary evaporator (Heidolph). After complete evaporation of methanol, the water phase was re-extracted by ethyl acetate. The obtained ethyl acetate extracts were finally in vacuo concentrated to dryness and then applied to further work up.

The crude extract (6.0 g) was separated by column chromatography on silica gel (100×10 cm) with a cyclohexane-CH2Cl2-MeOH gradient (0.5 L cyclohexane, 0.5 L cyclohexane-CH2Cl2 [1:1], 0.5 L CH2Cl2, 0.5 L CH2Cl2-MeOH [97:2], 1 L CH2Cl2-MeOH [95:5], 0.5 L CH2Cl2-MeOH [93:7], 1 L CH2Cl2-MeOH [90:10], 0.5 L CH2Cl2-MeOH [80:20], 0.5 L CH2Cl2-MeOH [50:50]; 0.5 L MeOH). According to TLC monitoring, six fractions were obtained: FI (3 g), FII (0.54 g), FIII (0.1 g), FIV (0.47 g), FV (0.32 g), and FVI (1.57 g). Further column chromatography of fraction III on Sephadex LH-20 (CH2Cl2-40% MeOH) afforded indole-3 acetic acid methyl ester as a colorless solid (1 mg). An application of fraction IV to silica gel (column 100×2 cm) and elution with a cyclohexane-CH2Cl2-MeOH gradient resulted in five subfractions: FIVa (0.1 g), FIVb (0.07 g), FIVc (0.03 g), FIVd (0.19 g), and FIVe (0.08 g). Purification of subfraction FIVa on silica gel column and elution with DCM-MeOH gradient delivered indole-3 acetic acid as a colorless solid (10 mg). In a similar way, subfraction FIVb afforded indole-3-carboxylic acid (7 mg) as a colorless solid. Subfraction FIVc yielded, in the same way, tyrosol (5 mg) as a colorless solid. Purification of subfraction FIVd using Sephadex LH-20 (MeOH) resulted in the isolation of glyceryl monolinoleate as a colorless oil (54 mg). Fraction V was subjected to Sephadex LH-20 (MeOH) giving 4-methoxy-2H-isoquinolin-1-one (2, 12 mg) as a faint yellow solid, in addition to cyclo-(Tyro, Pro) (3 mg) and 3′-hydroxy-N-(2-oxo-2,5-dihydrofuran-4-yl)propanamide (1a, 5 mg) as colorless solids. Finally, the polar fraction FVI (1.57 g) was purified using silica gel column eluted with DCM-MeOH gradient followed by Sephadex LH-20 (MeOH) to afford adenosine (10 mg) as a colorless solid.

3.4 Antimicrobial assay using agar diffusion test

Antimicrobial activity testing of the crude extract of the marine isolate ASMR2 was carried out together with the two new compounds (1a, 2) against a set of microorganisms using the agar diffusion technique. Paper-disk diffusion assay [42], with some modifications, was followed to measure the antimicrobial activity. A total of 20 mL of medium seeded with test organism were poured into 9 cm sterile Petri dishes. After solidification, the paper disks (6 mm diameter) containing the extract or the purified compounds with a concentration of 10 μL of 25 mg mL−1 of stock solution (i.e. 250 μg per disk) were placed on inoculated agar plates and the loaded substances were allowed to diffuse into the bacterial lawn at 4°C for 2 h. Afterward, the plates were incubated for 24 h at 30°C. Both bacteria and yeasts were grown on nutrient agar medium containing 3 g L−1 beef extract, 10 g L−1 peptone, and 20 g L−1 agar. The pH value was adjusted to 7.2 using NaOH. Fungal strains were grown on potato dextrose agar medium containing 4 g L−1 potato extract, 20 g L−1 dextrose, 15 g L−1 agar no. 1 (pH 6). After incubation, the diameters of the inhibition zones were measured against a wide panel of test microorganisms comprising Gram-positive bacteria (B. subtilis, S. aureus), Gram negative bacteria (Pseudomonas aeruginosa ATCC 27853), yeasts (C. albicans ATCC 10231 and S. cerevisiae ATCC 9080), and the fungus Aspergillus niger.

3.5 Cytotoxicity assays

The KB-3-1 and KB-VI cells were cultivated as a monolayer in Dulbecco’s modified eagle medium with glucose (4.5 g L−1), l-glutamine, sodium pyruvate, and phenol red supplemented with 10% (KB-3-1) fetal bovine serum. The cells were maintained at 37°C and 5.3% CO2-humidified air. On the day before the test, the cells (70% confluence) were detached with trypsin-ethylenediamine tetra-acetic acid solution (0.05% and 0.02%, respectively, in Dulbecco’s phosphate-buffered saline) and placed in sterile 96-well plates at a density of 10,000 cells in 100 μL medium per well. The dilution series of the compounds were prepared from stock solutions in DMSO of concentrations of 100 mM, 50 mM, or 25 mM. The stock solutions were diluted with culture medium (10% fetal bovine serum) down to the picomolar range. The dilution prepared from the stock solution was added to the wells. Each concentration was tested in six replicates. Dilution series were prepared by pipetting liquid from well to well. The control contained the same concentration of DMSO as the first dilution. After incubation for 72 h at 37°C and 5.3% CO2-humidified air, 30 μL of an aqueous resazurin solution (175 μM) was added to each well. The cells were incubated under the same conditions for 5 h. Subsequently, the fluorescence was measured. The excitation was affected at a wavelength of 530 nm, whereas the emission was recorded at a wavelength of 588 nm. The IC50 values were calculated as a sigmoidal dose-response curve using GraphPad Prism 4.03 (GraphPad Software, San Diego, USA). The IC50 values equal the drug concentrations, in which vitality is 50% [43], [44].

4 Supplementary information

NMR spectra and other supplementary data associated with this article are given as Supporting Information available online (DOI: 10.1515/znb-2016-0250).

Acknowledgements

The authors are thankful to the NMR and MS Departments in Bielefeld University for the spectral measurements. We thank Carmela Michalek for biological activity testing and Marco Wißbrock for technical assistance. DNA sequences were determined by the CeBiTec DNA sequencing core facility. The bioinformatics support of the BMBF-funded project “Bielefeld-Gießen Resource Center for Microbial Bioinformatics – BiGi (Grant number 031A533)” within the German Network for Bioinformatics Infrastructure (de.NBI) is gratefully acknowledged. This research work has been financed by the German Academic Exchange Service (DAAD) with funds from the German Federal Foreign Office in the frame of the Research Training Network “Novel Cytotoxic Drugs from Extremophilic Actinomycetes” (Project ID57166072).

References

[1] W. Fenical, P. R. Jensen, Nat. Chem. Biol.2006, 2, 666.10.1038/nchembio841Search in Google Scholar PubMed

[2] P. R. Jensen, E. Gontang, C. Mafnas, T. J. Mincer, W. Fenical, Environ. Microbiol.2005, 7, 1039.10.1111/j.1462-2920.2005.00785.xSearch in Google Scholar PubMed

[3] H. Laatsch, AntiBase, Wiley-VCH, Weinheim Germany 2014. A Data Base for Rapid Structural Determination of Microbial Natural Products, and annual updates; see http://www.user.gwdg.de/~ucoc/laatsch/AntiBase.htm.Search in Google Scholar

[4] R. Subramani, W. Aalbersberg, Microbiol. Res.2012, 167, 571.10.1016/j.micres.2012.06.005Search in Google Scholar PubMed

[5] S. Ramesh, N. Mathivanan, World J. Microbiol. Biotechnol.2009, 25, 2103.10.1007/s11274-009-0113-4Search in Google Scholar

[6] N. M. Zin, N. I. M. Sarmin, N. Ghadin, D. F. Basri, N. M. Sidik, W. M. Hess, G. A. Strobel. FEMS Microbiol. Lett. 2007, 274, 83.10.1111/j.1574-6968.2007.00819.xSearch in Google Scholar PubMed

[7] W. Sun, S. Dai, S. Jiang, G. Wang, G. Liu, H. Wu, X. Li, Antonie Van Leeuwenhoek2010, 98, 65.10.1007/s10482-010-9430-8Search in Google Scholar PubMed

[8] J. Xiao, Y. Luo, S. Xie, J. Xu, Int. J. Syst. Evol. Microbiol.2011, 61, 16.10.1099/ijs.0.019976-0Search in Google Scholar PubMed

[9] L. A. Maldonado, W. Fenical, P. R. Jensen, C. A. Kauffman, T. J. Mincer, A. C. Ward, A. T. Bull, M. Goodfellow, Int. J. Syst. Evol. Microbiol.2005, 55, 1759.10.1099/ijs.0.63625-0Search in Google Scholar PubMed

[10] A. Kijjoa, P. Sawangwong, Mar. Drugs2004, 2, 73.10.3390/md202073Search in Google Scholar

[11] K. S. Lam, Curr. Opin. Microbiol.2006, 9, 245.10.1016/j.mib.2006.03.004Search in Google Scholar PubMed

[12] C. M. Rath, B. Janto, J. Earl, A. Ahmed, F. Z. Hu, L. Hiller, M. Dahlgren, R. Kreft, F. Yu, J. J. Wolff, H. K. Kweon, M. A. Christiansen, K. Håkansson, R. M. Williams, G. D. Ehrlich, D. H. Sherman. ACS Chem. Biol.2011, 6, 1244.10.1021/cb200244tSearch in Google Scholar PubMed PubMed Central

[13] E. B. Shirling, D. Gottlieb, Int. J. Syst. Bacteriol. 1968, 18, 69.10.1099/00207713-18-2-69Search in Google Scholar

[14] E. B. Shirling, D. Gottlieb, Int. J. Syst. Bacteriol.1968, 18, 279.10.1099/00207713-18-4-279Search in Google Scholar

[15] E. B. Shirling, D. Gottlieb, Int. J. Syst. Bacteriol.1972, 22, 265.10.1099/00207713-22-4-265Search in Google Scholar

[16] J. Meyer, Int. J. Syst. Bacteriol.1976, 26, 487.10.1099/00207713-26-4-487Search in Google Scholar

[17] A. F. Yassin, E. A. Galinsk, A. Wohlfarth, K. D. Jahnke, K. P. Schaal, H. G. Truper, Int. J. Syst. Bacteriol.1993, 43, 266.10.1099/00207713-43-2-266Search in Google Scholar

[18] S. A. Sabry, N. B. Ghanem, G. A. Abu-Ella, P. Schumann, E. Stackebrandt, R. M. Kroppenstedt. Int. J. Syst. Evol. Microbiol. 2004, 54, 453.10.1099/ijs.0.02814-0Search in Google Scholar PubMed

[19] W. Whitman, M. Goodfellow, P. Kampfer, H-J. Busse, M. Trujillo, W. Ludwig, K-I. Suzuki, A. Parte, Bergey’s Manual of Systematic Bacteriology, The Actinobacteria, Vol. 5, 2nd ed. New York, Springer, 2012.10.1007/978-0-387-68233-4Search in Google Scholar

[20] M. Shaaban, Dissertation, Georg-August University, Göttingen (Germany) 2004.Search in Google Scholar

[21] M. Shaaban, H. Nasr, A. Z. Hassan, M. S. Asker, Rev. Latinoam. Quím.2013, 41, 50.Search in Google Scholar

[22] Chapman & Hall Chemical Database, Dictionary of Natural Products on CD-ROM, 2016.Search in Google Scholar

[23] S. Kumar, R. Garg, A. Kabra, World J. Pharm. Res. Technol.2013, 1, 83.Search in Google Scholar

[24] N. Ito, Pure Appl. Chem.1991, 63, 13.10.1351/pac199163010013Search in Google Scholar

[25] T. K. M. Shing, H. C. Tsui, Z. H. Zhou, Org. Chem. 1995, 60, 3121.10.1021/jo00115a030Search in Google Scholar

[26] J. P. Smivet, J. M. Vatele, Tetrahedron Lett. 1996, 37, 4373.10.1016/0040-4039(96)00820-9Search in Google Scholar

[27] L. J. Haynes, Quart. Rev.1948, 2, 46.10.1039/qr9480200046Search in Google Scholar

[28] C. Yi-Chen, C. Fang-Rong, W. Yang-Chang, Tetrahedron Lett. 1999, 40, 7513.10.1016/S0040-4039(99)01597-XSearch in Google Scholar

[29] J. D. Doherty, S. E. Hattox, O. C. Snead, R. H. Roth, J. Pharmacol. Exp. Ther.1978, 207, 130.Search in Google Scholar

[30] L. R. Caswell, D. Ch-W. Yung, H. G. Reid, C. J. Linn, C. E. Ryan, C. A. Davidson, J. Heterocycl. Chem.1970, 7, 1205.10.1002/jhet.5570070539Search in Google Scholar

[31] R. P. Maskey, M. Shaaban, I. Gruen-Wollny, H. Laatsch, J. Nat. Prod. 2004, 67, 1131.10.1021/np0305425Search in Google Scholar PubMed

[32] U. W. Hawas, M. Shaaban, K. A. Shaaban, M. Speitling, A. Maier, G. Kelter, H. H. Fiebig, M. Meiners, E. Helmke, H. Laatsch, J. Antibiot. 2009, 72, 2120.Search in Google Scholar

[33] A. B. Abdelwahab, M. Shaaban, M. A. H. Ismail, K. A. M. Abouzid, A. G. Hanna, Indian J. Chem., Sect. B2014, 53B, 1098.Search in Google Scholar

[34] Z. Tang, S. Niu, F. Liu, K. Lao, J. Miao, J. Ji, X. Wang, M. Yan, L. Zhang, Q. You, H. Xiao, H. Xiang, Bioorg. Med. Chem. Lett.2014, 24, 2129.10.1016/j.bmcl.2014.03.042Search in Google Scholar PubMed

[35] L. Wang, Y. Zeng, T. Wang, H. Liu, H. Xiao, H. Xiang, PLoS One2014, 9, e86509. doi:10.1371/journal.pone.0086509.10.1371/journal.pone.0086509Search in Google Scholar PubMed PubMed Central

[36] X. Yang, S. Yang, H. Chai, Z. Yang, R. J. Lee, W. Liao, L. Teng, PLoS One2015, 10, e0136649; doi:10.1371/journal. pone.0136649.10.1371/journal. pone.0136649Search in Google Scholar

[37] V. Kumar, A. Bharti, O. Gusain, G. S. Bisht, J. Sci. Eng. Tech. Mgt.2010, 2, 10.Search in Google Scholar

[38] E. B. Shirling, D. Gottlieb, Int. J. Syst. Bacteriol.1966, 16, 313.Search in Google Scholar

[39] E. Kuster, S. T. Williams, Appl. Microbiol.1964, 12, 46.10.1128/am.12.1.46-52.1964Search in Google Scholar

[40] D. L. Crawford, E. McCoy, Appl. Microbiol. 1972, 24, 150.10.1128/am.24.1.150-152.1972Search in Google Scholar PubMed PubMed Central

[41] H. D. Tresner, J. A. Hayes, E. J. Backus, Appl. Microbiol.1968, 16, 1134.10.1128/am.16.8.1134-1136.1968Search in Google Scholar PubMed PubMed Central

[42] A. W. Baur, W. M. Kirby, J. C. Sherris, M. Truck, Am. J. Clin. Pathol.1966, 45, 493.10.1093/ajcp/45.4_ts.493Search in Google Scholar

[43] A. F. Awantu, B. N. Lenta, T. Bogner, Y. F. Fongang, S. Ngouela, J. D. Wansi, E. Tsamo, N. Sewald, Z. Naturforsch.2011, 66b, 624.10.1515/znb-2011-0610Search in Google Scholar

[44] B. Sammet, T. Bogner, M. Nahrwold, C. Weiss, N. Sewald, J. Org. Chem.2010, 75, 6953.10.1021/jo101563sSearch in Google Scholar PubMed


Supplemental Material:

The online version of this article (DOI: 10.1515/znb-2016-0250) offers supplementary material, available to authorized users.


Received: 2016-11-25
Accepted: 2017-3-19
Published Online: 2017-4-26
Published in Print: 2017-5-1

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 19.4.2024 from https://www.degruyter.com/document/doi/10.1515/znb-2016-0250/html
Scroll to top button