BIOLOGICALLY ACTIVE NATURAL PRODUCTS: AGROCHEMICALS - CHAPTER 8 potx

13 240 0
BIOLOGICALLY ACTIVE NATURAL PRODUCTS: AGROCHEMICALS - CHAPTER 8 potx

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

8 Coronatine: Chemistry and Biological Activities Akitami Ichihara and Hiroaki Toshima CONTENTS 8.1 Introduction 8.2 Structures of Coronatine and Analogs 8.3 Biosynthesis of Coronatine 8.4 Syntheses of Coronatine and Biosynthetic Analogs of Jasmonic Acid 8.4.1 Asymmetric Total Synthesis of Coronatine 8.4.2Synthesis of Analogs of Jasmonic Acid and its Biosynthetic Intermediates 8.5 Biological Activities of Coronatine 8.5.1 Allocoronamic Acid as Inhibitors of Ethylene Biosynthesis 8.5.2 Similar Biological Activities of Coronatine to Those of Jasmonic Acid 8.6 Perspective References 8.1 Introduction The phytotoxin, coronatine (1), was isolated from the phytopathogenic bacterium Pseudomonas syringae pv. atropurpurea which causes chocolate spot disease on the leaves of Italian ryegrass. 1 After structural determination of 1, it was found that several pathovars of Pseudomonas syringae and Xanthomonas campestris pv. phormiicola produce 1 as a phytotoxic compound. Closely related analogues of 1 were isolated from Pseudomonas syringae pv. gly- cinea, pv. tomato, and Xanthomonas campestris pv. phormiicola. Recently, there has been much interest in 1, since various biological activities similar to those of jasmonic acid were shown in assays for tuber-inducing activity, etc. Furthermore, the potent activities of 1 are 100 to 10,000 times higher than those of jasmonic acid. Therefore, 1 is the most valuable probe in the area of plant physiological studies relating to jasmonic acid. In order to supply a suffi- cient amount of 1, an efficient synthetic method is urgently needed, since the productivity of 1 by P. syringae pathovars is rather fluctuated depending on the culture conditions and/or mutation. The total synthesis of optically active coronatine (1) was accomplished and made it possible to supply a practical amount of (+)-1. Analogues of 1 and jasmonic acid, and the compounds related to biosynthetic intermediates and amino acid conjugates of jasmonic acid were also synthesized and their biological activities were evaluated in some bioassay systems. In addition to the hormonal activities of coronatine, the similarity © 1999 by CRC Press LLC of the structure of coronamic acid with that of 1-amino-1-cyclopropanecarboxylic acid (ACC), the direct precursor of the plant hormone ethylene, made it possible to develop inhibitors of ethylene biosynthesis. 8.2 Structures of Coronatine and Analogs The structure and stereochemistry of coronatine (1) were determined by spectroscopic methods and x-ray analyses of coronafacic acid (2) and (–)-N-acetylcoronamic acid. 2-4 The structure of coronatine (1) is quite unique in that it consists of coronafacic acid (2) and coro- namic acid (3) (Figure 8.1). After the discovery of 1, it was found that several P. syringae pathovars (pv. maculicola, 5 pv. glycinea, 6 pv. morsprunorum 7 and pv. tomato 8 ) produce 1. Besides coronatine, Pseudomonas syringae pv. glycinea produces several coronatine ana- logues, norcoronatine (4), N-coronafacoyl-L-valine (5), N-coronafacoyl-L-isoleucine (6), and N-coronafacoyl-L-alloisoleucine (7) which are all biologically active. 9,10 The absolute configuration of norcoronatine (4) was confirmed by the comparison of physicochemical data with those of natural and synthetic specimens. 11 Furthermore, Pseudomonas syringae pv. tomato produces N-caronafacoyl-L-valine (5) 12 and Xanthomonas campestris pv. phormii- cola produces N-coronafacoyl-L-valine (5) and N-coronafacoyl-L-isoleucine produces (6). 13 It also was proved that Xanthomonas campestris pv. phormiicola produces coronatine (1) itself. 4 8.3 Biosynthesis of Coronatine The two components of coronatine (1) are biosynthesized from two different pathways respectively (Scheme 8.1). Coronafacic acid (2) was shown to be a novel polyketide derived FIGURE 8.1 Structures of (+)-coronatine and the related compounds. © 1999 by CRC Press LLC from three acetate units, one butyrate unit, and one unit of pyruvate. 15 Recently, it was found that the labeled pyruvate was carboxylated to oxaloacetate which was further con- verted into α-ketoglutarate through the TCA cycle. Further support of the intermediacy of α-ketoglutarate was provided by incorporation experiments of a labeled precursor, [3,4- 13 C 2 ]-DL-glutamate to give a labeled 2 at C-2 and C-3. Interestingly, Mitchell et al. 16 have described the isolation of the cyclopentenone derivative 8 from Pseudomonas syringae pv. tomato which is able to produce coronatine as a major component. Feeding experiments of the [3,4- 13 C 2 ]-DL-glutamate into this bacterium gave a labeled cyclopentenone 8 which exhibits 13 C- 13 C coupling between C-2 and C-3 with an enrichment at each carbon of ca. 0.89%. The coupling pattern is completely consistent with that observed in coronafacic acid (2). The results also are compatible with the previous biosynthetic studies of coronafacic acid, and the cyclopentenone 8 would be a plausible intermediate that would be transformed by Michael’s addition of malonyl CoA to an indanone derivative 8a and successive decarboxylation, reduction of the C-5 carbonyl, and dehydration to give coronafacic acid (2) (Scheme 8.2). The biosynthetic studies of coronamic acid (3) showed that both L-isoleucine and L-alloi- soleucine are specifically incorporated into the amino acid (3), the latter L-alloisoleucine is a much more efficient precursor. Since the absolute configuration of coronamic acid (3) is consistent with that of L-alloisoleucine, the incorporation efficiency is easily understand- able, though the cyclization mechanism has been completely elucidated. 17 Previously, coro- natine analogues, N-coronafacoyl-L-isoleucine (6) and N-coronafacoyl-L-alloisoleucine, were regarded as promising intermediates in the biosynthesis of coronatine (1). However, this is not the case. Since biosynthetic condensation of coronafacic acid (2) and coronamic acid (3) has been made by the incorporation of 14 C-coronamic acid (3) into coronatine (1) in Pseudomonas syringae pv. glycinea PG 4180. 18 Extensive genetic studies have been done with the same strain and, using genetic complementation, substrate feeding, and co-culturing experiments, regions involved in the biosynthesis of major intermediates, coronafacic acid (2) and coronamic acid (3) and the ligase of these components were identified in the coro- natine production gene cluster. 19 SCHEME 8.1 Biosynthesis of (+)-coronatine. SCHEME 8.2 Plausible biosynthesis of (+)-coronafacic acid. © 1999 by CRC Press LLC 8.4 Syntheses of Coronatine and Biosynthetic Analogs of Jasmonic Acid 8.4.1 Asymmetric Total Synthesis of Coronatine We have reported the partial synthesis of (+)-coronatine (1) by coupling between the acid chlo- ride (2c) derived form natural (+)-coronafacic acid (2) and synthetic (+)-coronamic acid (3) as a free amino acid. 3 This route is not efficient because the optical resolution of racemic 3 is required and the coupling yield is low (see Scheme 8.5). In order to supply a sufficient amount of (+)-1, we have developed a new route via the asymmetric syntheses of (+)-2 and (+)-3. We chose a most straightforward route for (+)-3 involving tandem alkylation of the mal- onate anion with a dielectrophile derived from (R)-1, 2-butanediol (9) (Scheme 8.3). 20 (R)-Malic acid was used as a chiral source and converted to 9 in 52% of overall yield in five steps. By treating 9 with thionyl chloride and then ruthenium tetraoxide, cyclic sulfate 10 was quantitatively obtained. Dibenzyl malonate anion, generated with 2 eq. of sodium hydride, was alkylated with 10 to give the cyclopropane derivative 11 in 93% yield. The first intermolecular substitution occurred at a primary site to give a b-sulfate which was smoothly cyclized to 11 by intramolecular substitution with inversion of the stereogenic center. Alkaline hydrolysis of 11 gave the half-acid 12 in 91% yield and 96% d.e., because the sterically less-hindered ester was selectively hydrolyzed. Treatment of 12 with diphe- nylphosphoryl azide (DPPA) by refluxing in tert-butanol gave the protected amino acid derivative 13 via Curtius rearrangement in 76% yield. Recrystallization of 13 from ethyl acetate/hexane increased the purity. One pot (alkaline and subsequent acidic) hydrolysis of 13, ion exchange, and crystallization from acetone/H 2 O gave (+)-3 (89% yield, >99.8% e.e., 96.6% d.e.); mp > 190°C (sublimation); [α] D 23 + 15.8° (c 1.10, H 2 O) [lit. [α] D 20 + 14.7° (c 1.67, H 2 O)]. Of course, 13 can be used as an amine component for amide formation after SCHEME 8.3 Syntheses of all four stereoisomers of coronamic acid. © 1999 by CRC Press LLC removal of the Boc group under usual acidic conditions. Furthermore, esterification of 12 with diazomethane and subsequent hydrogenolysis gave another half-acid 14 which could be converted to (+)-allocoronamic acid [(+)-16]. Antipodes, (–)-coronamic acid [(–)-3] and (–)-allocoronamic acid [(–)-16], were synthesized starting from (S)-malic acid. In this way, we could develop a practical route for all four stereoisomers of coronamic acid in high chemical yields and high optical purities without the optical resolution step. Several grams of the four isomers were synthesized by our new route. We have already developed a new approach using intramolecular 1,6-conjugate addition for construction of a 1-hydrindane framework such as 2 on a racemic substrate (Scheme 8.4). 21 By this methodology, the C 6 -non, C 6 -methyl, and C 6 -butyl substituted ana- logs [(±)-19b, -19c, -19d, -20a, -20b, -20c, and -20d] of 2, could be synthesized from (±)-18a-d (Scheme 8.4). 22 In principle, this method would be applied to synthesize the optically active form. 23,24 For our synthesis of (+)-2, the efficient preparation of the optically active ester [(+)-17] is required. This problem was solved using the catalytic asymmetric Michael reac- tion promoted by the heterobimetallic BINOL complex developed by Shibasaki and co- workers. 25 Michael reaction between 2-cyclopenten-1-one and diethyl malonate was car- ried out in the presence of Ga-Na-(S)-BINOL complex (10 mol%) and sodium tert-butoxide (7 mol%) to give a keto-diester in 95% yield. Acetalization and subsequent decarboxylation gave desired the ester (+)-17 in 74% yield. The optical purity of (+)-17 was determined as 95% e.e. by an HPLC analysis using a chiral column which was sufficiently pure for subse- quent reactions. The total synthesis of (+)-2 was carried out according to the synthetic route of racemic 2. Aldol condensation between the lithium enolate of (+)-17 and 2-ethylacrolein proceeded stereospecifically in the newly formed stereogenic centers to give a ca. 1:1 mix- ture of 21a and 21b which possesses a syn-relationship between the ethoxycarbonyl and hydroxyl groups. Mesylation of the mixture of 21a and 21b and subsequent β-elimination with DBU gave α, β, δ, γ-unsaturated ester possessing (E)-geometry as the sole product. Further deacetalization with p-TsOH in acetone gave (–)-18a, the precursor of intramolec- ular 1,6-conjugate addition in 78% yield [four steps from (+)-17]. Treatment of (–)-18a with NaOEt (3 eq.) in EtOH gave 22a corresponding to the ethyl ester of coronafacic acid and its C 6 -epimer 22b in 71% yield (diastereoselectivity; ca. 3:1). Isomerization of 22b with DBA also was possible to give a 1:1 mixture of 22a and 22b. The optical purity of 22a was deter- mined as 98% e.e. by an HPLC analysis using a chiral column. This result demonstrates that the optical purity of (+)-17 was completely retained in 22a without any loss through the sequence of reactions including intramolecular 1,6-conjugate addition. The effectiveness of our new approach, on both racemic and optically active forms, was proven. Acidic hydrol- ysis of 22a gave (+)-2 in 70% yield after recrystallization; mp 142-143°C; [α] D 23 +122° (c 1.00, MeOH) [lit. mp 141-142°C; [α] D 20 +119° (c 3.30, MeOH)], whose spectral data were identical with those of natural 2 in all respects. The overall yield via our new route was 24% in nine steps from 2-cyclopenten-1-one. The optical purity of (+)-2 is estimated to be at least >98% e.e. based on 98% e.e. of 22a. In practice, judging from the specific rotation, (+)-2 can be regarded as optically pure. After deprotection of the Boc group of 13 with TFA, the resulting amine TFA salt 13a (evaporated to dryness and used without purification) was coupled with (+)-2 in the pres- ence of a water-soluble carbodiimide to give coronatine benzyl ester (1a) in 89% yield (Scheme 8.5). In a preliminary experiment, coupling between racemic 2 and optically active 13a gave a mixture of 1a and its diastereomer (inseparable on TLC), whose 1 H-NMR spec- trum gave partially separated signals based on the respective diastereomers (ratio ca. 1:1). However, in the case of coupling (+)-2, the 1 H-NMR spectrum of 1a was observed as the sin- gle diastereomer. This result means that the synthetic (+)-2 has practical enantiomeric purity and 1a can be regarded as optically pure. Deprotection of 1a by hydrogenolysis in the pres- ence of 10% Pd-C in ethyl acetate provided (+)-1 in 80% yield; mp 162-164°C [α] D 22 +76.6° © 1999 by CRC Press LLC SCHEME 8.4 Syntheses of coronafacic acid and its analogs. © 1999 by CRC Press LLC (c 2.20, MeOH) [lit. mp 161-163°C; [α] D 20 +68.4° (c 2.20, MeOH)]. The spectral data of syn- thetic (+)-1 were identical with those of natural 1 in all respects including the specific rota- tion. In this way, the first asymmetric total synthesis of (+)-1 has been accomplished. The yield was remarkably improved in contrast to the previous synthesis of (+)-1 from natural 2. 8.4.2 Syntheses of Analogs of Jasmonic Acid and its Biosynthetic Intermediates Even-numbered OPC homologs (OPC-8:0, -6:0, and -4:0) as biosynthetic precursors of jas- monic acid (JA) (Scheme 8.6) and odd-numbered OPC homologues (OPC-7:0, -5:0, -3:0, and SCHEME 8.5 Total synthesis of coronatine. SCHEME 8.6 Biosynthesis of jasmonic acid and structural similarity to coronatine. © 1999 by CRC Press LLC -1:0) were synthesized from 2-[(Z)-2-pentenyl]cyclopenten-1-one (23) as the common starting material in short steps and with high yields via conjugate addition (Scheme 8.7). 26 Only in the case of synthesizing OPC-1:0, tris(phenylthio)methyl lithium as a C 1 -synthon was used for the conjugate addition to give the orthothioester which was converted to OPC-1:0 (24a) via ethanolysis and subsequent hydrolysis. In the other cases, zinc–copper reagents which were prepared from iode-esters possessing the requisite carbon chain, were applied for the conjugate addition. The resulting esters were hydrolyzed to OPC homologues [OPC-3:0 (24b), -4:0 (24c); -5:0 (24d), -6:0 (24e), -7:0 (24f), and -8:0 (24g)]. Each acid was observed as a trans-rich mixture in the 13 C-NMR spectra. Esters of odd-numbered OPC homologues (25a, 25b, 25d, and 25f) are convenient sub- strates for synthesizing analogs which are not structurally subject to β-oxidation. 27 Protec- tion of the carbonyl group and subsequent LAH reduction gave an alcohol which was treated with tert-butyl bromoacatate to give a 3-oxa-OPC ester. Acidic hydrolysis gave 3-Oxa-OPC-8:0 (26d), -6:0 (26b), and -4:0 (26a). Instead of LAH reduction, DIBAL reduction gave an aldehyde which was treated with trichloroacetic acid/sodium trichloroacetate to give a trichloromethylcarbinol. Fluorination and hydrolysis gave 2-fluoro-OPC-8:0 (27f), -6:0 (27d), and -4:0 (27b). Each acid also was observed as trans-rich mixture in the 13 C-NMR spectra. N-(Jasmonyl)amino acid conjugates (JA-AAs) were also synthesized from JA (racemic and trans-rich) and the appropriate amino acids. 28 In addition to naturally occurring JA- Leu, JA-Ile, JA-Val (derived from aliphatic amino acids), JA-Phe, JA-Tyr, and JA-Trp (derived from aromatic amino acids), not naturally occurring JA-Gly, JA-β-Ala, JA-ACC, JA-(+)-coronamic acid were also synthesized. Among them, JA-Leu, JA-Ile, JA-Val, JA-ACC, and JA-(+)-coronamic acid are structurally similar to coronatine. SCHEME 8.7 Syntheses of OPC-homologous series, 3-oxa-OPC- and 2-fluoro-OPC-analogs. © 1999 by CRC Press LLC 8.5 Biological Activities of Coronatine 8.5.1 Allocoronamic Acid as Inhibitors of Ethylene Biosynthesis Although naturally occurring coronamic acid [(+)-3] has the (1S, 2S)-configuration, three other possible stereoisomers [(–)-3, (+)-16, and (–)-16, see Scheme 8.3] are considered and have been prepared via optical resolution or asymmetric synthesis as described above. 3,20,29 Coronamic acids are regarded as analogs of ACC and correspond to substituted ACC with an ethyl group. ACC is well known as the direct precursor of a plant hormone, ethylene. Therefore, coronamic acids were applied to elucidating stereospecific recognition of ACC by the ethylene-forming enzyme (EFE). 30 One stereoisomer, (1R, 2S)-allocoronamic acid [(+)-16] has been found to be converted to 1-butene at a 50 to 250 times higher rate than any of the other stereoisomers (Scheme 8.8). Since 1-butene does not exhibit ethylene-like actions, (+)-16 would be used as a competitive inhibitor of ethylene biosynthesis. There- fore, we examined the inhibitory effect of coronomic acids on senescence in cut carnation flowers (Dianthus caryophyllus L. cv. Lightpink Barbara) (Table 8.1). 31 Although both race- mic and optically active allocoronamic acids exhibited antisenescent activity, (+)-16 exhib- ited the strongest activity at 10 mM (longevity, 12 to 13 days). The longevity was slightly shorter than that of silver thiosulfate complex (STS) used as a positive control. In the case of using (–)-16, the stems changed to yellow and broke in extreme cases. In addition, we measured the relative EFE activity using a tomato suspension-cell culture. The relative EFE activity was decreased by both (+)-16 (78.2%) and (–)-16 (85.8%). α-aminoisobutyric acid (AIB) which is a known competitive inhibitor of EFE, also decreased the relative EFE activ- ity with similar intensity (84.7%) to those of allocoronamic acids. Thus, it was proven that allocoronamic acids acted as inhibitors of ethylene biosynthesis. SCHEME 8.8 Biosynthesis of ethylene and stereospecific recognition of ACC/(+)-allocoronamic acid by EFE. © 1999 by CRC Press LLC 8.5.2 Similar Biological Activities of Coronatine to Those of Jasmonic Acid A chlorosis-inducing phytotoxin, 1, was isolated with the guidance of the potato cell expansion-inducing activity, and the hormone-like activities of 1 already have been stud- ied. Recently, there has been strong interest in 1 which has been shown to exhibit various biological activities similar to those of JA, known as an endogenous plant-growth regulator and signal transmitter. 32 JA and JA-Me (or as jasmonoid) are recognized as a kind of phy- tohormone and known to exhibit various biological activities in higher plants: senescence- promoting activity, growth-inhibitory activity, potato tuber-inducing activity, potato cell expansion-inducing activity, etc. These biological activities also are induced by 1 and the latter two activities were the first discovery in the similarity of the biological activities of coronatine to those of JA. Furthermore, the potent activities of 1 are 100 to 10,000 times higher than those of JA. 33-35 There are other examples that revealed that 1 and JA (or JA-Me) exhibited similar biological activities. Both 1 and JA-Me caused similar growth-inhibitory effects on Arabidopsis (seedling and root), stimulated anthocyanin accumulation, and increased the level of two proteins (31 and 29 kD). Arabidopsis mutants selected for resis- tance to 1 are male sterile are also insensitive to JA-Me and resistant to bacterial patho- gens. 36 It was proven with a tomato suspension culture that both 1 and JA played a central role in regulating EFE activity in ethylene biosynthesis. 33,34 Both 1 and JA stimulated the biosynthesis and emission of volatiles (terpenes, acetogenins, and aromatics) in many plant species. 37 Antitumor taxane-type diterpenes were increased ca. 10 times by treating Taxus baccta with 1 and JA-Me. 38 In the tendril coiling assay of Bryonia dioica, 1, the 12-oxo-PDA methyl ester, the OPC-8:0 methyl ester, and JA-Me exhibited activity that decreased in that order. 39 This result suggested the importance of not only JA, but octadecanoid precursors as signaling molecules of higher plants. In our recent study (Figures 8.2 and 8.3), the following results were obtained: (1) JA-amino acid conjugate (JA-Leu, JA-Ile, JA-Val) exhibited weaker TABLE 8.1 Inhibitory Effect of Coronamic Acids on Senescence in Cut Carnation Flowers (Dianthus caryophyllus L. cv. Lightpink Barbara) Concentration Longevity Compound a (mM) (Days) Control (H 2 O) – 6 STS (Koto fresh K-20) 1/1000 c 15 (±)-CA 10 6 (+)-(1S,2S)-CA 5 6–7 (–)-(1R,2R)-CA 5 6 (±)-AlloCA 10 8–10 (±)-AlloCA b 10 10–11 (+)-(1R,2S)-AlloCA 10 12–13 5 10–11 1.67 9 (–)-(1S,2R)-AlloCA 10 8–9 5 9–10 1.67 10–11 a Three flowers were used for each compound tested. b After treating the flowers in the reagent for 24 h, the solution was changed to distilled water. c The original solution of a commercially available STS reagent was used after diluting 1000 times. © 1999 by CRC Press LLC [...]... OPC-7:0 and OPC-5:0 also exhibited tuber-inducing activity; (4) the C6-alkyl-substituted analogs of 2 exhibited weak tuber-inducing activity;22 (5) a unique activity, decreasing the weight of potato tissue by 2- uoro-OPC -8 : 0 in the cell expansion-inducing assay was first detected; and (6) 3-oxa-OPC -8 : 0, -6 :0, and -4 :0 also exhibited weak tuber-inducing activity The last result suggests that β-oxidation...FIGURE 8. 2 Structure–activity relationship in potato cell expansion-inducing assay FIGURE 8. 3 Structure–activity relationship in potato tuber-inducing assay © 1999 by CRC Press LLC potato cell expansion-inducing activity than that of JA; (2) OPC -8 : 0, -6 :0, and -4 :0 exhibited both potato cell expansion-inducing and tuber-inducing activity at a similar concentration to JA; (3) not naturally occurring OPC-7:0... of JA, OPC -8 : 0, -6 :0, and -4 :0 in themselves exhibit tuber-inducing activity In all the examined results of comparative bioassays of 1, JA, biosynthetic precursors and analogs, 1 was always the most active compound Since in vivo coronatine has not been isolated as a secondary product, 12-oxo-PDA and/or OPC-homologs would play an important role as signaling compounds in higher plants.4 0-4 4 8. 6 Perspective... 19 78 6 Mitchell, R E and Young, H., Phytochemistry, 17, 20 28, 19 78 7 Mitchell, R E., Physiol Plant Pathol., 20, 83 , 1 982 8 Mitchell, R E., Hale, C N., and Shanks, J C., Physiol Plant Pathol., 23, 315, 1 983 9 Mitchell, R E., Phytochemistry, 24, 1 485 , 1 985 10 Mitchell, R E and Young, H., Phytochemistry, 24, 2716, 1 985 11 Mitchell, R E., Pirrung, M C., and McGeehan, M., Phytochemistry, 26, 2695, 1 987 ... Engl., 34, 1600, 1995 38 Hara, Y and Yukimune, T., JP 081 988 63A2 39 Weiler, E W., Kutchan, T M., Gorba, T., Brodschelm, W., Niesel, U., and Bublitz, F., FEBS Lett., 345, 9, 1994 40 Schierle, K., Hopke, J., Niedt, M.-L., Boland, W., and Stechan, E., Tetrahedron Lett., 37, 87 15, 1996 41 Blechert, S., Brodshelm, W., Holder, S., Kammerer, L., Kutchan, T M., Mueller, M J., Xia, Z.-Q., and Zenk, M H., Proc... the JA-(jasmonoid- and octadecanoid-) receptor family is the remaining biggest problem, and coronatine would play an important role in solving the problem Our asymmetric synthesis is able to provide a certain amount of coronatine; however, further synthetic studies on more structurally simple and biologically active analogs of coronatine should continue, and might result in developing a new plant-regulatory... Shibasaki, M., Chem Eur J., 2, 1 386 , 1996 26 Toshima, H., Nara, S., Aramaki, H., Ichihara, A., Koda, Y., and Kikuta, Y., Biosci Biotech Biochem., 61, 1724, 1997 27 Toshima, H., Fujino, Y., Ichihara, A., Koda, Y., and Kikuta, Y., Biosci Biotech Biochem (in press) 28 Kramell, R., Schmidt, J., Schneider, G., Sembdner, G., and Schreiber, K., Tetrahedron Lett., 44, 5791, 1 988 29 Shiraishi, K., Ichihara, A.,... C L., Malvick, D K., and Mitchell, R E., J Bact., 171, 80 7, 1 989 © 1999 by CRC Press LLC 13 Mitchell, R E., Phytochemistry, 30, 3917, 1991 14 Tamura, K., Takikawa, Y., Tsuyumu, S., Goto, M., and Watanabe, M., Ann Phytopathol Soc Jpn., 58, 276, 1992 15 Parry, R J., Jiralerspong, S., Mhaskar, S., Alemany, L., and Willcott, R., J Am Chem Soc., 1 18, 703, 1996 16 Mitchell, R E., Young, H., and Liddell,... and Sakamura, S., Plant Physiol., 70, 195, 1 982 31 Toshima, H., Niwayama, Y., Nagata, H., Greulich, F., and Ichihara, A., Biosci Biotech Biochem., 57, 1394, 1993 32 Sembdner, G and Parthier, B., Annu Rev Plant Physiol Plant Mol Biol., 44, 596, 1993 33 Greulich, F., Yoshihara, T., Toshima, H., and Ichihara, A., XV Int Bot Congress, Yokohama Abstr., 4154, 388 , 1993 34 Greulich, F., Yoshihara, T., and... Alemany, L., and Willcott, R., J Am Chem Soc., 1 18, 703, 1996 16 Mitchell, R E., Young, H., and Liddell, M J., Tetrahedron Lett., 36, 3237, 1995 17 Parry, R J., Lin, M.-T., Walker, A E., and Mhaskar, S., J Am Chem Soc., 113, 184 9, 1991 18 Mitchell, R E., Young, S A., and Bender, C L., Phytochemistry, 35, 343, 1994 19 Lydon, J., PGRSA Quarterly, 24, 111, 1996 20 Toshima, H and Ichihara, A., Biosci Biotech . (Koto fresh K-20) 1/1000 c 15 (±)-CA 10 6 (+ )-( 1S,2S)-CA 5 6–7 (– )-( 1R,2R)-CA 5 6 (±)-AlloCA 10 8 10 (±)-AlloCA b 10 10–11 (+ )-( 1R,2S)-AlloCA 10 12–13 5 10–11 1.67 9 (– )-( 1S,2R)-AlloCA 10 8 9 5 9–10 1.67. aromatic amino acids), not naturally occurring JA-Gly, JA-β-Ala, JA-ACC, JA-(+)-coronamic acid were also synthesized. Among them, JA-Leu, JA-Ile, JA-Val, JA-ACC, and JA-(+)-coronamic acid are structurally. ana- logs [(± )-1 9b, -1 9c, -1 9d, -2 0a, -2 0b, -2 0c, and -2 0d] of 2, could be synthesized from (± )-1 8a-d (Scheme 8. 4). 22 In principle, this method would be applied to synthesize the optically active form. 23,24

Ngày đăng: 11/08/2014, 12:21

Từ khóa liên quan

Mục lục

  • BIOLOGICALLY ACTIVE NATURAL PRODUCTS: Agrochemicals

    • Table of Contents

    • Chapter 8: Coronatine: Chemistry and Biological Activities

      • 8.1 Introduction

      • 8.2 Structures of Coronatine and Analogs

      • 8.3 Biosynthesis of Coronatine

      • 8.4 Syntheses of Coronatine and Biosynthetic Analogs of Jasmonic Acid of Jasmonic Acid

        • 8.4.1 Asymmetric Total Synthesis of Coronatine

        • 8.4.2 Syntheses of Analogs of Jasmonic Acid and its Biosynthetic Intermediates

        • 8.5 Biological Activities of Coronatine

          • 8.5.1 Allocoronamic Acid as Inhibitors of Ethylene Biosynthesis

          • 8.5.2 Similar Biological Activities of Coronatine to Those of Jasmonic Acid

          • 8.6 Perspective

          • References

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan