Academia.eduAcademia.edu
Volume 5. Issue 6. Pages 841-992. 2010 ISSN 1934-578X (printed); ISSN 1555-9475 (online) www.naturalproduct.us NPC Natural Product Communications EDITOR-IN-CHIEF DR. PAWAN K AGRAWAL Natural Product Inc. 7963, Anderson Park Lane, Westerville, Ohio 43081, USA agrawal@naturalproduct.us HONORARY EDITOR PROFESSOR GERALD BLUNDEN The School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT U.K. axuf64@dsl.pipex.com EDITORS PROFESSOR ALESSANDRA BRACA Dipartimento di Chimica Bioorganicae Biofarmacia, Universita di Pisa, via Bonanno 33, 56126 Pisa, Italy braca@farm.unipi.it PROFESSOR DEAN GUO State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100083, China gda5958@163.com PROFESSOR J. ALBERTO MARCO Departamento de Quimica Organica, Universidade de Valencia, E-46100 Burjassot, Valencia, Spain alberto.marco@uv.es PROFESSOR YOSHIHIRO MIMAKI School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Horinouchi 1432-1, Hachioji, Tokyo 192-0392, Japan mimakiy@ps.toyaku.ac.jp PROFESSOR STEPHEN G. PYNE Department of Chemistry University of Wollongong Wollongong, New South Wales, 2522, Australia spyne@uow.edu.au PROFESSOR MANFRED G. REINECKE Department of Chemistry, Texas Christian University, Forts Worth, TX 76129, USA m.reinecke@tcu.edu PROFESSOR WILLIAM N. SETZER Department of Chemistry The University of Alabama in Huntsville Huntsville, AL 35809, USA wsetzer@chemistry.uah.edu PROFESSOR YASUHIRO TEZUKA Institute of Natural Medicine Institute of Natural Medicine, University of Toyama, 2630-Sugitani, Toyama 930-0194, Japan tezuka@inm.u-toyama.ac.jp PROFESSOR DAVID E. THURSTON Department of Pharmaceutical and Biological Chemistry, The School of Pharmacy, University of London, 29-39 Brunswick Square, London WC1N 1AX, UK david.thurston@pharmacy.ac.uk ADVISORY BOARD Prof. Berhanu M. Abegaz Gaborone, Botswana Prof. Viqar Uddin Ahmad Karachi, Pakistan Prof. Øyvind M. Andersen Bergen, Norway Prof. Giovanni Appendino Novara, Italy Prof. Yoshinori Asakawa Tokushima, Japan Prof. Lee Banting Portsmouth, U.K. Prof. Julie Banerji Kolkata, India Prof. Anna R. Bilia Florence, Italy Prof. Maurizio Bruno Palermo, Italy Prof. Josep Coll Barcelona, Spain Prof. Geoffrey Cordell Chicago, IL, USA Prof. Cristina Gracia-Viguera Murcia, Spain Prof. Duvvuru Gunasekar Tirupati, India Prof. A.A. Leslie Gunatilaka Tucson, AZ, USA Prof. Kurt Hostettmann Lausanne, Switzerland Prof. Martin A. Iglesias Arteaga Mexico, D. F, Mexico Prof. Jerzy Jaroszewski Copenhagen, Denmark Prof. Leopold Jirovetz Vienna, Austria Prof. Teodoro Kaufman Rosario, Argentina Prof. Norbert De Kimpe Gent, Belgium Prof. Karsten Krohn Paderborn, Germany Prof. Hartmut Laatsch Gottingen, Germany Prof. Marie Lacaille-Dubois Dijon, France Prof. Shoei-Sheng Lee Taipei, Taiwan Prof. Francisco Macias Cadiz, Spain Prof. Imre Mathe Szeged, Hungary Prof. Joseph Michael Johannesburg, South Africa Prof. Ermino Murano Trieste, Italy Prof. M. Soledade C. Pedras Saskatoon, Cnada Prof. Luc Pieters Antwerp, Belgium Prof. Om Prakash Manhattan, KS, USA Prof. Peter Proksch Düsseldorf, Germany Prof. Phila Raharivelomanana Tahiti, French Plynesia Prof. Satyajit Sarker Wolverhampton, UK Prof. Monique Simmonds Richmond, UK Prof. Valentin Stonik Vladivostok, Russia Prof. Winston F. Tinto Barbados, West Indies Prof. Karen Valant-Vetschera Vienna, Austria Prof. Peter G. Waterman Lismore, Australia INFORMATION FOR AUTHORS Full details of how to submit a manuscript for publication in Natural Product Communications are given in Information for Authors on our Web site http://www.naturalproduct.us. Authors may reproduce/republish portions of their published contribution without seeking permission from NPC, provided that any such republication is accompanied by an acknowledgment (original citation)-Reproduced by permission of Natural Product Communications. Any unauthorized reproduction, transmission or storage may result in either civil or criminal liability. The publication of each of the articles contained herein is protected by copyright. Except as allowed under national “fair use” laws, copying is not permitted by any means or for any purpose, such as for distribution to any third party (whether by sale, loan, gift, or otherwise); as agent (express or implied) of any third party; for purposes of advertising or promotion; or to create collective or derivative works. Such permission requests, or other inquiries, should be addressed to the Natural Product Inc. (NPI). A photocopy license is available from the NPI for institutional subscribers that need to make multiple copies of single articles for internal study or research purposes. To Subscribe: Natural Product Communications is a journal published monthly. 2010 subscription price: US$1,695 (Print, ISSN# 1934-578X); US$1,695 (Web edition, ISSN# 1555-9475); US$2,095 (Print + single site online); US$595 (Personal online). Orders should be addressed to Subscription Department, Natural Product Communications, Natural Product Inc., 7963 Anderson Park Lane, Westerville, Ohio 43081, USA. Subscriptions are renewed on an annual basis. Claims for nonreceipt of issues will be honored if made within three months of publication of the issue. All issues are dispatched by airmail throughout the world, excluding the USA and Canada. NPC 2010 Vol. 5 No. 6 853 - 858 Natural Product Communications Antimicrobial and Antiparasitic Abietane Diterpenoids from the Roots of Clerodendrum eriophyllum Francis Machumia, Volodymyr Samoylenkob, Abiy Yenesewa, Solomon Deresea, Jacob O. Midiwoa, Frank T. Wiggersb, Melissa R. Jacobb, Babu L. Tekwanib,c, Shabana I. Khanb, Larry A. Walkerb,c and Ilias Muhammadb,* a Department of Chemistry, University of Nairobi, P.O. Box 30197 (00100), Nairobi, Kenya National Center for Natural Products Research and cDepartment of Pharmacology, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, USA b milias@olemiss.edu Received: February 4th, 2010; Accepted: March 27th, 2010 Chromatographic separation of the roots of a Kenyan medicinal plant, Clerodendrum eriophyllum, led to the isolation of ten abietane diterpenoids (1-10), one of which (1) was isolated for the first time from a natural source. Using spectroscopic data, the structure of 1 was determined to be 12-hydroxy-8,12-abietadiene-3,11,14-trione. Circular dichroism (CD) spectra showed that the stereochemistry of compounds 1, 3, and 6-8 belongs to the normal series of abietane diterpenes, which confirmed the absolute stereochemistry of the isolated compounds. Compounds 1-10 were evaluated for their in vitro antiplasmodial, antileishmanial, antifungal and antibacterial activities. Compounds 3 and 7 exhibited potent antifungal activity (IC50/MIC 0.58/1.25 and 0.96/2.5 g/mL, respectively) against C. neoformans, whereas 3, 6 and 7 showed strong antibacterial activity against Staphylococcus aureus and methicillin-resistant S. aureus with IC50/MIC values between 1.33-1.75/2.5-5 and 0.961.56/2.5 g/mL, respectively. In addition, compounds 3 and 9 exhibited potent antileishmanial activity (IC50 0.08 and 0.20 g/mL, respectively) against L. donovani, while 3 and 7 displayed weak antimalarial activity against Plasmodium falciparum, but 9 was inactive. Keywords: Clerodendrum eriophyllum, Verbenaceae, abietane diterpenoids, antimicrobial, antileishmanial, antimalarial. Clerodendrum eriophyllum Gürke (Verbenaceae), a small tree 0.5 – 2 m high, is scattered in the dry bushlands of Eastern Kenya where the plant is used by local communities for the treatment of malaria [1]. The plant has no record of previous phytochemical analysis. However, the methanol extract of C. eriophyllum root bark previously showed weak in vitro activity against Plasmodium falciparum D6 and W2 clones (IC50 9.5110.56 g/mL); while its methanol and aqueous extracts exhibited significant in vivo chemosuppression (i. e., 90.1% and 61.5%, respectively) against P. berghei infected mice treated intraperitoneally at a dose of 100 mg/kg body weight [2]. The genus Clerodendrum is known to contain iridoids [3, 4], abietane diterpenoids [5-7] and steroids [8]. In the quest for antiplasmodial compounds from Kenyan plants, we have investigated the roots of C. eriophyllum collected from Eastern Kenya. In this paper we report the isolation and structure elucidation of a new abietane diterpenoid, 12hydroxy-8,12-abietadiene-3,11,14-trione (1), obtained alongside nine other known abietane diterpenoids (2-10). OH O 15 O 11 20 16 1 10 OH HO 8 O 5 R R 3 R=O 4 R = H2 1 R=O 2 R = H2 OH HO HO O O OH HO HO R O OH 7 R=H 8 R = OH 5 R=O 6 R = H2 O HO R H H H 18 OH O OH 9 H 10 Figure 1: Chemical structures of compounds 1-10 isolated from C. eriophyllum. We also report the antiplasmodial, antileishmanial, antibacterial, and antifungal activities of the isolated compounds. 854 Natural Product Communications Vol. 5 (6) 2010 The 1:1 MeOH/CH2Cl2 extract of roots of C. eriophyllum showed moderate antiplasmodial activity with IC50 values of 8.8 g/mL against chloroquinesensitive (D6) and -resistant (W2) strains of P. falciparum. Repeated chromatographic purification of this extract gave 12-hydroxy-8,12-abietadiene-3,11,14trione (1), as well as nine known abietane diterpenoids, namely royleanone (2) [9], taxodione (3) [10], 11hydroxy-7,9(11),13-abietatrien-12-one (4) [11], sugiol (5) [12], ferruginol (6) [13], 6-hydroxysalvinolone (7) [14], 6,11,12,16-tetrahydroxy-5,8,11,13-abietatetra-en7-one (8) [15], uncinatone (9) [16], and 11-hydroxy8,11,13-abietatriene-12-O- -xylopyranoside (10) [15]. The molecular formula of compound 1 was established as C20H26O4 (m/z 331.1910 [M+H]+; calculated for m/z 331.1909) by HRESIMS. The UV absorption maxima at λmax 273 and 378 nm closely matched those of the p-quinone chromophore of royleanone 2 [9]. The IR spectrum indicated the presence of hydroxyl group(s) (νmax 3080-3430 cm-1), unconjugated carbonyl (νmax 1704 cm-1), together with olefinic and conjugated carbonyl absorptions of the p-quinone moiety (νmax 1609, 1633 and 1650 cm-1). The 13C NMR spectrum showed 20 signals, with the sp2 region displaying four olefinic quaternary carbons (δC 124.3, 144.1, 145.8, 150.6) and two conjugated carbonyls (δC 183.2, 186.9) assignable to the p-quinone moiety. The 1H NMR spectrum did not show signals in either the olefinic or aromatic regions, but did show signals for three methyl singlets at δH 1.09, 1.13 and 1.24, assignable to C-18, C-19 and C-20, respectively, of an abietane skeleton, and an isopropyl group, with two methyl doublets at δH 1.19 and 1.20 (each 3H, J = 7.0 Hz) and a methine septet at δH-15 3.15. The position of the isopropyl group was deduced from HMBC correlations (Table 1) between δH-15 3.15 and δC-12 150.6, δC-13 124.3, and δC-14 186.9. The unconjugated carbonyl at δC 216.7 was established to be at C-3 from HMBC correlations between δH-18 1.13, δH-19 1.09 and the carbonyl carbon signal. Furthermore, a comparison of the 13C NMR spectral data of 1 with those of the known compound royleanone 2 showed close similarities of the carbon signals, except for the differences associated with C-2 – C-5 due to the presence of a carbonyl group at the C-3 position (δC 33.8, 216.7, 46.9 and 50.8 vs. 18.5, 41.2, 33.8 and 44.4 for 2, respectively). A complete set of 2D NMR experiments [1H-1H COSY, 1H-13C HMQC, 1H-13C HMBC (Table 1), 1H-1H NOESY] allowed the unambiguous establishment of the structure of 1 as 12-hydroxy-8,12-abietadiene-3,11,14- trione. Circular dichroism (CD) spectra showed that the stereochemistry of compounds 1, 3, and 6-8 belong to the normal series (A/B trans) of diterpenes. The Machumi et al. Table 1: 1H and 13C NMR spectroscopic data (J values in Hz, in parenthesis), and 1H-13C HMBC correlations of compound 1. H/C 1 H C 1.76, m; 2.81, m 34.5, t 2.59, ddd (15.6, 9.2, 5.4); 2.51, dt (15.6, 7.2) 1.76, m 33.8, t 6 1.46, ddd (21.8, 12.0, 4.5); 1.76,m 18.6, t 7 2.31, ddd (19.6, 12.0, 6.0); 2.84, br dd (19.6, 5.4) 26.0, t 8 9 10 11 12 13 14 15 3.15, sept (7.0) 145.8, s 144.1, s 37.3 ,s 183.2 ,s 150.6 ,s 124.3, s 186.9, s 24.1, d 16 17 18 1.20, d (7.0) 1.19, d (7.0) 1.13, s 19.9, q 19.8, q 27.7, q 2 3 4 5 19 1.09, s 20 1.24, s 12-OH 7.21, s 216.7, s 46.9, s 50.8, d 20.0, q 20.6, q - HMBC C-2, C-3, C-5, C-10, C-20 C-1, C-3, C-4, C-10 C-1, C-3, C-4, C-6, C-7, C-10, C-18, C-19, C-20 C-4, C-5, C-7, C-8, C-10 C-5, C-6, C-8, C-9. C-14 C-12, C-13, C-14, C-16, C-17 C-13, C-15, C-17 C-13, C-15, C-16 C-3, C-4, C-5, C-19 C-3, C-4, C-5, C-18 C-1, C-5, C-9, C-10 C-11, C-12, C-13 positive Cotton effect at 275 and 284 nm for compound 1 supports the β-orientation of the methyl group at C-10, i.e. (10S)-Me configuration, according to the rule for π-π* transition of an α,β-unsaturated ketone [17]. The CD spectra of compounds 3 and 6 are in agreement with those of the known taxodione analog [18] and ferruginol [19], respectively, confirming their absolute stereochemistry. CD spectra of 7 and 8, not previously reported, are similar to the related abietane diterpene cyrtophyllone A, whose absolute stereochemistry was determined by X-ray crystallography [17]. Only recently, the absolute configuration of 6hydroxysalvinolone (7) was determined as (10R)-Me by enhanced X-ray crystallography [20], thus, supporting its stereochemistry deduced from CD spectra. The antiplasmodial, antileishmanial, antifungal, antibacterial and cytotoxic activities are summarized in Tables 2-4. Compounds 3 and 9 demonstrated potent antileishmanial activities with IC50 values of 0.08 and 0.20 g/mL, respectively, against L. donovani, compared with those observed for the standard drug amphotericin B (IC50 0.13 g/mL). On the other hand, the antiplasmodial activities of compounds 3, 7 and 8 were found to be very weak, with IC50 values of 1.2 - 4.8 g/mL, when compared with the standard artemisinin (IC50 <0.026 g/mL). Strong antifungal activities were also displayed by 3 and 7, Diterpenoids from Clerodendrum eriophyllum Natural Product Communications Vol. 5 (6) 2010 855 Table 2: Antiplasmodial, antileishmanial and cytotoxic activity of compounds 2-10. Compound/extract C.eriophyllum extract 1 2 3 6 7 8 9 10 Chloroquine Artemisinin Pentamidine Amphotericin B P. falciparum D6a W2b IC50, μg/mL 8.8 8.8 1.2 1.8 3.0 <0.026 <0.026 NT NT 1.2 2.5 4.8 0.14 <0.026 NT NT VERO TC50 μg/mL NC NC NC NC NC 4.5 NC NC NC NC NC NT NT L. donovani IC50 IC90 μg/mL μg/mL NT NT 16 NT 0.08 4 3.2 12 0.2 NT NT NT 1.4 0.13 32 NT 0.21 13 6.5 22 0.9 NT NT NT 6 0.3 a Chloroquine-sensetive clone; bChloroquine-resistant clone ;- =Not Active; NT = Not Tested; NC = Not cytotoxic (up to the maximum dose tested; 4.76 μg/mL for pure compounds and 47.6 mg/ml for crude extracts). IC50 is the concentration that affords 50% inhibition of growth. Table 3: Antifungal activities of compounds 2-10. Compound 1 2 3 6 7 8 9 10 Amphotericin B C. glabrata -/-/5.2/10 -/-/14.9/20 -/-/0.31/0.65 IC50/MIC, g/mL C. krusei C. A. C. albicans neoformans fumigatus -/-/-/-/-/-/-/-/12.0/0.58/1.25 8.9/12.5/-/-/-/-/-/0.96/2.5 11.2/-/14.5/20 5.9/20 -/-/-/-/-/-/-/-/-/-/0.95/1.25 0.44/1.25 1.29/2.50 0.43/1.25 - =Not Active; NT = Not Tested; IC50 is the concentration that affords 50% inhibition of growth; MIC is the lowest test concentration that allows no detectable growth. Table 4: Antibacterial activities of compounds 2-10. IC50/MIC, g/mL Compound S. aureus 1 2 3 6 7 8 9 10 Ciprofloxacin -/-/1.35/5 1.33/2.5 1.75/5 6.8/20 -/-/0.1/0.25 MRS E. coli P. M. aureginosa intracellulare -/-/-/-/-/-/-/-/1.47/2.5 -/-/11.9/0.96/2.5 -/-/14.5/1.56/2.5 -/-/-/8.44/20 -/-/-/-/-/-/-/-/-/-/-/0.08/0.25 0.004/0.008 0.06/0.25 0.30/1.00 =Not Active; NT = Not Tested; IC50 is the concentration that affords 50% inhibition of growth; MIC is the lowest test concentration that allows no detectable growth. showing IC50 values of 0.58 and 0.96 g/mL, respectively, against C. neoformans, as compared with 0.44 g/mL of the standard amphotericin B. Compounds 3, 6 and 7 showed strong antibacterial activity against Staphylococcus aureus and methicillinresistant S. aureus with IC50/MIC values between 1.331.75/2.5-5 and 0.96-1.56/2.5 g/mL, respectively. With regard to cytotoxicity, only 6-hydroxysalvinolone (7) showed moderate cytotoxic activity with an IC50 value of 4.5 g/mL against monkey kidney fibroblasts (VERO). Finally, compound 1 showed weak in vitro antileishmanial activity against L. donovani (IC50 16 μg/mL), but found to be inactive in antimalarial and antimicrobial assays. Experimental General: Optical rotations were measured in CHCl3 or MeOH using an AUTOPOL IV® instrument at ambient temperature. Circular dichroism (CD) spectra were recorded in MeCN using an Olis DCM 20 CD spectrometer at ambient temperature. IR spectra were taken as films on a Bruker Tensor 27 FTIR instrument. UV spectra were obtained in MeCN using a HewlettPackard 8453 spectrophotometer; 1D and 2D NMR data were acquired on a Bruker BioSpin instrument at 600 MHz (1H), 150 (13C) in CDCl3 using the residual solvent as int. standard. HRMS were obtained by direct injection using a Bruker Bioapex-FTMS with electrospray ionization (ESI). For column chromatography (CC), Merck silica gel 60 (0.063-0.200 mm) and Fluka Sephadex LH-20 were used as stationary phases; For PTLC, Merck silica gel 60 PF254+366, coated on glass plates to make 1.0 mm layers was used; Analytical TLC was carried out using factory prepared aluminum plates (0.25 mm) coated with silica gel (60 F254, Merck); The isolated compounds were visualized by observing under UV light at 254 or 365 nm, followed by spraying with 1% vanillin-H2SO4 spray reagent. Plant material: The roots of Clerodendrum eriophyllum were collected from Machakos, Eastern Kenya in November 2007 and identified at the Department of Botany, University of Nairobi, Kenya, where a voucher specimen No. JMFM/2007/11 has been deposited. Extraction and isolation: The roots of C. eriophyllum were air dried and pulverized to give 1.8 kg of material. This was extracted by cold percolation at room temperature using 1:1 MeOH/ CH2Cl2 (3×4 L, 24 h each), followed by 100% methanol (1×4 L, 24 h) to give 65 g of brown gummy extract, of which 35 g was adsorbed onto 40 g of silica gel and subjected to CC on a silica gel column (300 g, 5×35 cm), eluted with nhexane/CH2Cl2 (95:5, 3.5 L; 9:1, 1.25 L; 3:1, 2 L; 1:1, 3 L; 1:3, 1 L; 100% CH2Cl2, 1.5 L) followed by CH2Cl2/MeOH (99:1, 1.25 L; 98:2, 1 L; 95:5, 1 L). Sixty-two fractions of eluents, collected in 250 mL aliquots, were concentrated using a rotary evaporator and similar fractions were combined on the basis of TLC analysis. Combination of fraction 5-9 crystallized in n-hexane/CH2Cl2 (95:5) gave 2 (260 mg). Fractions 11-19 (640 mg) were rechromatographed over silica gel (50 g, 2.0×30 cm) and eluted with n-hexane/CH2Cl2 (95:5) to give 4 (8.7 mg) after 0.6 L of elution, and 6 (65 mg) after 1.4 L of elution. The latter was further 856 Natural Product Communications Vol. 5 (6) 2010 purified by PTLC developed with n-hexane/CH2Cl2 (8:2). Fraction 22-28 (160 mg) was purified on a Sephadex LH 20 column (100 g, 2.5×30 cm), eluted with MeOH/ CH2Cl2 1:1 (0.3 L), followed by PTLC developed with n-hexane/CH2Cl2 7:3 to give 3 (28 mg). Fraction 34-45 (600 mg) was subjected to silica gel CC (50 g, 2.0×30 cm), eluted with CH2Cl2/ n-hexane (8:2) to give 1 (4.0 mg) after 0.28 L of elution, 9 (6.7 mg) after 0.35 L of elution, and 5 (10.3 g) after 0.8 L of elution. Fractions 52-57 (580 mg) were rechromatographed over silica gel (50 g, 2.0×30 cm) and eluted with CH2Cl2 to yield 7 (6.5 mg) and 8 (64.2 mg), both of which crystallized from CH2Cl2, after 0.52 L and 1.3 L of elution, respectively. Compound 10 (15 mg) was obtained after purifying fractions 58-60 on a Sephadex LH 20 column (100 g, 2.5×30 cm) eluted with MeOH/ CH2Cl2 1:1 (0.3 L). Royleanone (2) [9], taxodione (3) [10], 11-hydroxy7,9(11),13-abietatrien-12-one (4) [11], sugiol (5) [12], ferruginol (6) [13], 6-hydroxysalvinolone (7) [14], 6,11,12,16-tetrahydroxy-5,8,11,13-abietatetra-en-7-one (8) [15], uncinatone (9) [16] and 11-hydroxy-8,11,13abietatriene-12-O- -xylopyranoside (10) [15] were identified by comparison of their full physical (mp and optical rotation) and spectral data (UV, IR, 1H and 13C NMR, and MS) with those reported in the literatures. 12-Hydroxy-8,12-abietadiene-3,11,14-trione (1) Yellow solid Rf 0.5 (n-hexane/CH2Cl2/MeOH 60:39:1) [α]D25: +184 (c 0.16, CHCl3) IR (film) νmax, cm-1: 3430-3080 (OH), 2965 (C-H), 2934 (C-H), 2873 (C-H), 1704 (C=O), 1650 (C=O), 1633 (C=O), 1609 (C=C), 1462 (C-H), 1271 (C-O). UV (MeCN) λmax (log ε), nm: 201 (3.94), 205 (3.86), 273 (3.98), 378 (2.78) CD (MeCN) λmax ([θ], deg·cm2/dmol), nm: 208 (+6.4·103), 275 (+18.9·103), 284 (+20.2·103), 330 (-2.3·103). 1 H and 13C NMR: Table 1. HRESIMS: m/z 331.1910 [M+H]+ (calcd. for C20H27O4, 331.1909), 329.1758 [M-H]- (calcd. for C20H25O4, 329.1753). Taxodione (3) UV (MeCN) λmax (log ε), nm: 211 (3.81), 315 (4.13), 325 (4.13), 334 (4.12), 337 (4.11), 394 (3.38). CD (MeCN) λmax ([θ], deg·cm2/dmol), nm: 205 (20.2·103), 261 (-12.7·103), 321 (-18.7·103), 337 (19.2·103), 445 (+13.6·103). Ferruginol (6) UV (MeCN) λmax (log ε), nm: 211 (3.90), 219 (3.87), 281 (3.58). Machumi et al. CD (MeCN) λmax ([θ], deg·cm2/dmol), nm: 206 (-2.8·103), 211 (+12.9·103), 227 (+9.5·103), 265 (-0.9·103), 301 (-2.3·103). 6-Hydroxysalvinolone (7) UV (MeCN) λmax (log ε), nm: 219 (3.97), 250 (3.98), 284 (3.94), 335 (4.00). CD (MeCN) λmax ([θ], deg·cm2/dmol), nm: 213 (+36.3·103), 281 (+30.4·103), 338 (-19.7·103). 6,11,12,16-Tetrahydroxy-5,8,11,13-abietatetra-en-7one (8) UV (MeCN) λmax (log ε), nm: 218 (3.98), 251 (4.01), 285 (3.89), 339 (3.97), 407 (2.90). CD (MeCN) λmax ([θ], deg·cm2/dmol), nm: 212 (+48.9·103), 284 (+29.7·103), 339 (-19.7·103). Uncinatone (9) UV (MeCN) λmax (log ε), nm: 220 (4.06), 228 (4.08), 232 (4.09), 283 (4.17), 298 (4.17), 334 (4.01), 376 (3.95). CD (MeCN) λmax ([θ], deg·cm2/dmol), nm: 205 (-18.8·103), 213 (-18.9·103), 234 (+24.5·103), 241 (+23.9·103), 271 (sh) (-4.3·103), 297 (-17.1·103), 323 (+12.7·103), 350 (-2.7·103), 381 (-5.6·103). Antimicrobial assay: All organisms were obtained from the American Type Culture Collection (Manassas, VA) and included the fungi Candida albicans ATCC 90028, C. glabrata ATCC 90030, C. krusei ATCC 6258, Cryptococcus neoformans ATCC 90113 and Aspergillus fumigatus ATCC 90906; the bacteria Staphylococcus aureus ATCC 29213, methicillinresistant S. aureus ATCC 43300 (MRS), Escherichia coli ATCC 35218, Pseudomonas aeruginosa ATCC 27853 and Mycobacterium intracellulare ATCC 23068. Susceptibility testing was performed using a modified version of the CLSI methods [21,22], as described by Samoylenko et al [23]. Drug controls, ciprofloxacin (ICN Biomedicals, Ohio) for bacteria and amphotericin B (ICN Biomedicals, Ohio) for fungi, were included in each assay. Antimalarial/ parasite LDH assay: The in vitro antimalarial activity was measured by a colorimetric assay that determines the parasitic lactate dehydrogenase (pLDH) activity [23, 24]. The assay was performed in a 96-well microplate and included two P. falciparum strains [Sierra Leone D6 (chloroquinesensitive) and Indochina W2 (chloroquine-resistant)]. The IC50 values were computed from the dose response curves generated by plotting percent growth against test concentrations. DMSO, artemisinin and chloroquine were included in each assay as vehicle and drug controls, respectively. Diterpenoids from Clerodendrum eriophyllum Antileishmanial assay: Antileishmanial activity of the compounds was tested in vitro on a culture of Leishmania donovani promastigotes. In a 96 well microplate assay, compounds with appropriate dilution were added to the Leishmania promastigotes culture (2×106 cells/mL). The plates were incubated at 26°C for 72 h and growth of Leishmania promastigotes was determined by Alamar blue assay [25]. Pentamidine and amphotericin B were used as standard antileishmanial agents. IC50 values for each compound were computed from the growth inhibition curve. Cytotoxicity assay: The in vitro cytotoxic activity was determined against monkey kidney fibroblasts (VERO) Natural Product Communications Vol. 5 (6) 2010 857 following the method described by Samoylenko et al [23]. Doxorubicin was used as the positive and DMSO as the negative (vehicle) control. Acknowledgments - The authors sincerely thank Mr John P. Hester for database management and technical assistance, Mr John Trott and Ms Marsha Wright for assistance in biological work and Dr Bharathi Avula for recording mass spectra. One of our authors (FM) thanks DAAD-NAPRECA for a scholarship. This work was supported in part by the United States Department of Agriculture, Agricultural Research Service Specific Cooperative Agreement No. 58-6408-2-0009 and NIH, NIAID, Division of AIDS, Grant No. AI 27094. References [1] Beenje HK. (1994) Kenyan trees, shrubs and lianas. National Museums of Kenya. Nairobi, Kenya, 613. [2] Muthaura CN, Rukunga GM, Chhabra SC, Omar SA, Guantai AN, Gathirwa JW, Tolo FM, Mwitari PG, Keter LK, Kirira PG, Kimani CW, Mungai GM, Njagi ENM. (2007) Antimalarial activity of some plants traditionally used in Meru district of Kenya. Phytotherapy Research, 21, 260-267. [3] Tian J, Zhao QS, Zhang HJ, Lin ZW, Sun HD. (1997) New cleroindicins from Clerodendrum indicum. Journal of Natural Products, 60, 766-769. [4] Yang H, Hou AJ, Mei SX, Sun HD, Che CT. (2002) Constituents of Clerodendrum bungei. Journal of Asian Natural Products Research, 4, 165-169. [5] Liu S, Zhu H, Zhang S, Zhang X, Yu Q, Xuan L, (2008) Abietane diterpenoids from Clerodendrum bungei. Journal of Natural Products, 71, 755-759. [6] Fan TP, Min Z, Song G, Iinuma M, Tanaka T. (1999) Abietane diterpenoids from Clerodendrum mandarinorum. Phytochemistry, 51, 1005-1008. [7] Fan TP, Min Z, Iinuma M, Tanaka T. (2000) Rearranged abietane diterpenoids from Clerodendrum mandarinorum. Journal of Asian Natural Products Research, 2, 237-243. [8] Pandey R, Verma RK, Singh SC, Gupta MM. (2003) 4α-Methyl-24 -ethyl-5α-cholesta-14,25-dien-3 -ol and 24 -ethylcholesta5,9(11),22E-trien-3 -ol, sterols from Clerodendrum inerme. Phytochemistry, 63, 415-420. [9] Edwards OE, Feniak G, Los M. (1962) Diterpenoid quinones of Inula royleana D.C. Canadian Journal of Chemistry, 40, 15401546. [10] Kupchan SM, Karim A, Marcks C (1968) Taxodione and taxodone, two novel diterpenoid quinone methide tumor inhibitors from Taxodium distichum. Journal of the American Chemical Society, 90, 5923-5924. [11] Dellar JE, Core MD, Waterman PG. (1996) Antimicrobial abietane diterpenoids from Plectranthus elegans. Phytochemistry, 41, 735-738. [12] Ying BP, Kubo I. (1991) Complete proton and carbon-13 NMR assignments of totarol and its derivatives. Phytochemistry, 30, 1951-1955. [13] Samoylenko V, Dunbar DC, Gafur MA, Khan SI, Ross SA, Mossa JS, El-Ferali FS, Tekwani BL, Bosselaers J, Muhammad I. (2008) Antiparasitic, nematicidal and antifouling constituents from Juniperus berries. Phytotherapy Research, 22, 1570-1576. [14] Hueso-Rodriguez JA, Jimeno ML, Rodriguez B, Savona G, Bruno M. (1983) Abietane diterpenoids from the root of Salvia phlomoides. Phytochemistry, 22, 2005-2009. [15] Han L, Huang X, Dahse H, Moellmann U, Grabley S, Lin W, Satler I. (2008) New abietane diterpenoids from the mangrove Avicennia marina. Planta Medica, 74, 432-437. [16] Dorsaz A, Marston A, Stoeckli-Evans H, Msonthi JD, Hostettmann K. (1985) Uncinatone, a new antifungal hydroquinone diterpenoid from Clerodendrum uncinatum. Helvetica Chimica Acta, 68, 1605-1610. [17] Tian X, Min Z, Xie N, Lei Y, Tian Z, Zheng Q, Xu R, Tanaka T, Iinuma M, Mizuno M. (1993) Abietane diterpenes from Clerodendron cyrtophyllum. Chemical & Pharmaceutical Bulletin, 41, 1415-1417. [18] Katti SB, Ruedi P, Eugster CH. (1982) Diterpenoid quinomethans, vinylogous quinones and a phyllocladene derivative from Plectranthus purpuratus Harv. (Labiatae). Helvetica Chimica Acta, 65, 2189-2197. [19] Briggs LH, Cain BF, Davis BR, Wilmshurst JK. (1959) Absolute configuration of phyllocladene, mirene, rimuene, cupressene, and kaurene. Tetrahedron Letters, 8, 13-16. 858 Natural Product Communications Vol. 5 (6) 2010 Machumi et al. [20] Fun H, Quah CK, Chantrapromma S. (2010) Redetermination and absolute configuration of 6-hydroxysalvinolone. Acta Crystallographica, Section E, E66 (1), o146-o147. [21] NCCLS (1998) Reference method for broth dilution antifungal susceptibility testing of conidium-forming filamentous fungi; Proposed Standard, M38-P. National Committee on Clinical Laboratory Standards, 18 (13). [22] NCCLS (2000) Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically M7-A5. National Committee on Clinical Laboratory Standards, 20 (2). [23] Samoylenko V, Jacob MR, Khan SI, Zhao J, Tekwani BL, Midiwo JO, Walker LA, Muhammad I. (2009) Antimicrobial, antiparasitic and cytotoxic spermine alkaloids from Albizia schimperiana. Natural Product Communications, 4, 791-796. [24] Makler MT, Ries JM, Williams JA, Bancroft JE, Piper RC, Gibbins BL, Hinriches DJ. (1993) Parasite lactate dehydrogenase as an assay for Plasmodium falciparum drug sensitivity. American Journal of Tropical Medicine Hygiene, 48, 739-741. [25] Mikus J, Steverding D. (2000) A simple colorimetric method to screen drug cytotoxicity against Leishmania [by] using the dye Alamar Blue. Parasitology International, 48, 265-269. Natural Product Communications Vol. 5 (6) 2010 Published online (www.naturalproduct.us) RP-HPLC Analysis and Antidiabetic Activity of Swertia paniculata Jagmohan S. Negi, Pramod Singh, Geeta Joshi née Pant and Mohan S. M. Rawat 907 Antioxidants from the Leaves of Cinnamomum kotoense Kuo-Chen Cheng, Man-Chun Hsueh, Hou-Chien Chang, Alan Yueh-Luen Lee, Hui-Min Wang and Chung-Yi Chen 911 Chemical Constituents and Antimicrobial Activities of Canthium horridum Biao Yang, Guangying Chen, Xiaoping Song, Zhong Chen, Xinming Song and Jing Wang 913 Colon Targeted Curcumin Delivery Using Guar Gum Edwin J. Elias, Singhal Anil, Showkat Ahmad and Anwar Daud 915 Formadienoate-A and B: Two New Long Chained Feruloyl Esters from Clerodendrum formicarum (Lamiaceae) of Cameroon Muhammad Shaiq Ali, Zeeshan Ahmed, Muhammad Imran Ali and Joseph Ngoupayo 919 Free-radical Scavenging Activity of some European Polyporales Kateřina Macáková, Lubomír Opletal, Miroslav Polášek and Věra Samková 923 In vitro Plant Regeneration from Callus of Citrus x monstruosa (Pompia), an Endemic Citrus of Sardinia Daniele Fraternale, Laura Giamperi, Anahi Bucchini, Pierpaolo Cara and Donata Ricci 927 Purification and Biochemical Characterization of Alkaline Serine Protease from Caesalpinia bonducella Hidayatullah Khan, Irshad Ali, Arif-ullah Khan, Mushtaq Ahmed, Zamarud Shah, Ahmad Saeed, Rubina Naz, Mohamad Rais Mustafa and Atiya Abbasi 931 Composition of the Essential Oil of Argania spinosa (Sapotaceae) Fruit Pulp Hicham Harhar, Said Gharby, Mohamed Ghanmi, Hanae El Monfalouti, Dominique Guillaume and Zoubida Charrouf 935 The Volatile Constituents of Salvia leucantha Luis B. Rojas, Tomas Visbal, Marielba Morillo, Yndra Cordero de Rojas, Juan Carmona Arzola and Alfredo Usubillaga 937 Terpenoid Composition of the Essential Oils of Teucrium royleanum and T. quadrifarium Lalit Mohan, Charu C. Pant, Anand B. Melkani and Vasu Dev 939 Influence of Some Environmental Factors on the Essential Oil Variability of Thymus migricus Alireza Yavari, Vahideh Nazeri, Fatemeh Sefidkon and Mohammad Esmail Hassani 943 Chemical Investigations of Essential Oils from Endemic Cupressaceae Trees from New Caledonia Nicolas Lebouvier, Chantal Menut, Edouard Hnawia, Audrey Illinger, Pierre Cabalion and Mohammed Nour 949 Comparative Composition of Four Essential Oils of Oregano Used in Algerian and Jordanian Folk medicine Djemaa Berrehal, Tarek Boudiar, Lakhal Hichem, Assia Khalfallah, Ahmed Kabouche, Ahmad Al-Freihat, Alireza Ghannadi, Ebrahim Sajjadi, Mitra Mehrabani, Jawad Safaei-Ghomi and Zahia Kabouche 957 Chemical Composition and Biological Activities of Santiria trimera (Burseraceae) Essential Oils from Gabon Raphaël Bikanga, Thomas Makani, Huguette Agnaniet, Louis Clément Obame, Fatouma Mohamed Abdoul-Latif, Jacques Lebibi and Chantal Menut 961 Chemical Composition and Larvicidal Activity of Eugenia triquetra Essential Oil from Venezuelan Andes Flor D. Mora, Jorge L. Avila, Luis B. Rojas, Rosslyn Ramírez, Alfredo Usubillaga, Samuel Segnini, Juan Carmona and Bladimiro Silva 965 Evaluation of Bioactivity of Linalool-rich Essential Oils from Ocimum basilucum and Coriandrum sativum Varieties Ahmet D. Duman, Isa Telci, Kenan S. Dayisoylu, Metin Digrak, İbrahim Demirtas and Mehmet H. Alma 969 Constituents, Antileishmanial Activity and Toxicity Profile of Volatile Oil from Berries of Croton macrostachyus Yinebeb Tariku, Ariaya Hymete, Asrat Hailu and Jens Rohloff 975 Free Radical Scavenging and Antibacterial Activities, and GC/MS Analysis of Essential oils from Different Parts of Falcaria vulgaris from Two Regions Ali Shafaghat 981 The Effects of Maturity on Chilli Pepper Volatile Components Determined by SDE, GC-MS and HPLC Rong Liu, Ke Xiong, Xiongze Dai, Li Wang, Zhimin Liu and Wentong Xue 985 Natural Product Communications 2010 Volume 5, Number 6 Contents Original Paper Page α-Glucosidase Inhibitory Constituents of Linaria kurdica subsp. eriocalyx İrfan Aydoğdu, Figen Zihnioğlu, Tamer Karayildirim, Derya Gülcemal, Özgen Alankuş-Çalışkan and Erdal Bedir 841 Synthesis and Insecticidal Activities of New Ether-Derivatives of Celangulin-V Jiwen Zhang, Zhaonong Hu, Hua Yang and Wenjun Wu 845 New Sesquiterpene Lactone and Other Constituents from Centaurea sulphurea (Asteraceae) Hichem Lakhal, Tarek Boudiar, Ahmed Kabouche, Zahia Kabouche, Rachid Touzani and Christian Bruneau 849 Protective Effects of Isoatriplicolide Tiglate from Paulownia coreana against Glutamate-induced Neurotoxicity in Primary Cultured Rat Cortical Cells Ill-Min Chung, Eun-Hye Kim, Hyun-Seok Jeon and Hyung-In Moon 851 Antimicrobial and Antiparasitic Abietane Diterpenoids from the Roots of Clerodendrum eriophyllum Francis Machumi, Volodymyr Samoylenko, Abiy Yenesew, Solomon Derese, Jacob O. Midiwo, Frank T. Wiggers, Melissa R. Jacob, Babu L. Tekwani, Shabana I. Khan, Larry A. Walker and Ilias Muhammad 853 Tetranortriterpenoids from Spathelia sorbifolia (Rutaceae) Denise S. Simpson, Stewart McLean, William F. Reynolds and Helen Jacobs 859 A Validated Method for Standardization of the Bark of Clerodendron serratum Arunava Gantait, Payel Roy, Neelesh Kumar Nema, Pradip Kumar Dutta and Pulok Kumar Mukherjee 863 Activity of Extracts and Procesterol from Calotropis gigantea against Entamoeba histolytica Shailendra Singh, Neelam Bharti, Manoj Chugh, Fehmida Naqvi and Amir Azam 867 Ampullosine, a new Isoquinoline Alkaloid from Sepedonium ampullosporum (Ascomycetes) Dang Ngoc Quang, Jürgen Schmidt, Andrea Porzel, Ludger Wessjohann, Mark Haid and Norbert Arnold 869 HPLC - DAD Analysis of Lycorine in Amaryllidaceae Species Gulen Irem Kaya, Derya Cicek, Buket Sarıkaya, Mustafa Ali Onur and Nehir Unver Somer 873 Simultaneous HPLC Determination of Three Bioactive Alkaloids in the Asian Medicinal Plant Stephania rotunda Sothavireak Bory, Sok-Siya Bun, Béatrice Baghdikian, Fathi Mabrouki, Sun Kaing Cheng, Riad Elias, Hot Bun and Evelyne Ollivier 877 Impact of Cruciferous Phytoalexins on the Detoxification of Brassilexin by the Blackleg Fungus Pathogenic to Brown Mustard M. Soledade C. Pedras and Ryan B. Snitynsky 883 Flavonoids from Erythrina vogelii (Fabaceae) of Cameroon Muhammad Imran Ali, Zeeshan Ahmed, Alain Francois Kamdem Waffo and Muhammad Shaiq Ali 889 HPLC/DAD Comparison of Sixteen Bioactive Components Between Da-Cheng-Qi Decoction and its Parent Herbal Medicines Fengguo Xu, Ying Liu, Rui Song, Haijuan Dong and Zunjian Zhang 893 Secondary Metabolites of Hypericum confertum and their Possible Chemotaxonomic Significance Cüneyt Çırak, Jolita Radušienė, Valdimaras Janulis and Liudas Ivanauskas 897 Antioxidant Effects of Secondary Metabolites from Geranium psilostemon Didem Şöhretoğlu, Suna Atasayar Sabuncuoğlu, M. Koray Sakar, Hilal Özgüneş and Olov Sterner 899 Bioactive Isoflavones from Dalbergia vacciniifolia (Fabaceae) Ester Innocent, Joseph J. Magadula, Charles Kihampa and Matthias Heydenreich 903 Continued inside backcover