Báo cáo khoa học: Characterization of the rice carotenoid cleavage dioxygenase 1 reveals a novel route for geranial biosynthesis ppt

12 497 0
Báo cáo khoa học: Characterization of the rice carotenoid cleavage dioxygenase 1 reveals a novel route for geranial biosynthesis ppt

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

Characterization of the rice carotenoid cleavage dioxygenase reveals a novel route for geranial biosynthesis Andrea Ilg, Peter Beyer and Salim Al-Babili Faculty of Biology, Institute of Biology II, Albert-Ludwigs University of Freiburg, Germany Keywords apocarotenoids; carotenoid cleavage; carotenoid dioxygenase; geranial; lycopene cleavage Correspondence S Al-Babili, Institute for Biology II, Cell Biology, Albert-Ludwigs University of Freiburg, Schaenzlestr 1, D-79104 Freiburg, Germany Fax: +49 761 203 2675 Tel: +49 761 203 8454 E-mail: salim.albabili@biologie.uni-freiburg.de (Received 19 September 2008, revised 25 November 2008, accepted 26 November 2008) doi:10.1111/j.1742-4658.2008.06820.x Carotenoid cleavage products – apocarotenoids – include biologically active compounds, such as hormones, pigments and volatiles Their biosynthesis is initiated by the oxidative cleavage of C–C double bonds in carotenoid backbones, leading to aldehydes and ⁄ or ketones This step is catalyzed by carotenoid oxygenases, which constitute an ubiquitous enzyme family, including the group of plant carotenoid cleavage dioxygenases (CCD1s), which mediates the formation of volatile C13 ketones, such as b-ionone, by cleaving the C9–C10 and C9¢–C10¢ double bonds of cyclic and acyclic carotenoids Recently, it was reported that plant CCD1s also act on the C5–C6 ⁄ C5¢–C6¢ double bonds of acyclic carotenes, leading to the volatile C8 ketone 6-methyl-5-hepten-2-one Using in vitro and in vivo assays, we show here that rice CCD1 converts lycopene into the three different volatiles, pseudoionone, 6-methyl-5-hepten-2-one, and geranial (C10), suggesting that the C7–C8 ⁄ C7¢–C8¢ double bonds of acyclic carotenoid ends constitute a novel cleavage site for the CCD1 plant subfamily The results were confirmed by HPLC, LC-MS and GC-MS analyses, and further substantiated by in vitro incubations with the monocyclic carotenoid 3-OH-c-carotene and with linear synthetic substrates Bicyclic carotenoids were cleaved, as reported for other plant CCD1s, at the C9–C10 and C9¢–C10¢ double bonds Our study reveals a novel source for the widely occurring plant volatile geranial, which is the cleavage of noncyclic ends of carotenoids Carotenoids are isoprenoid pigments synthesized by all photosynthetic organisms and some nonphotosynthetic bacteria and fungi In plants, carotenoids are essential in protecting the photosynthetic apparatus from photo-oxidation, and represent essential constituents of the light-harvesting and of the reaction center complexes [1–4] Carotenoids are also the source of apocarotenoids [5–7], which are physiologically active compounds, including the ubiquitous chromophore retinal, the chordate morphogen retinoic acid and the phytohormone abscisic acid as the best-known examples Further carotenoid-derived signaling molecules are represented by strigolactones, a group of C15 apoc- arotenoids attracting both symbiotic arbuscular mycorrhizal fungi and parasitic plants [8,9] and, as recently shown, exerting functions as novel plant hormones regulating shoot branching [10,11] In addition, the development of arbuscular mycorrhiza is also accompanied by accumulation of cyclohexenone (C13) and mycorradicin (C14) derivatives [12], all of which are apocarotenoids conferring a yellow pigmentation to infected roots [13] Apocarotenoids, such as bixin in Bixa orellana [14] and saffron in Crocus sativus [15], are plant pigments of economic value The synthesis of apocarotenoids is initiated by the oxidative cleavage of double bonds in carotenoid Abbreviations CCD, carotenoid cleavage dioxygenase; GST, glutathione S-transferase; NIST, National Institute of Standards and Technology; OsCCD1, Oryza sativa carotenoid cleavage dioxygenase 1; SPME, solid-phase microextraction 736 FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works A Ilg et al backbones, generally catalyzed by carotenoid oxygenases, nonheme iron enzymes that are common in all taxa [5–7,16] VP14 (viviparous14) from maize, which catalyzes the formation of the abscisic acid precursor xanthoxin by cleaving 9-cis-epoxy carotenoids [17], is the first identified member of this enzyme class On the basis of their substrate specificity, VP14 and its orthologs have been termed 9-cis-epoxy-carotenoid dioxygenases Plants possess a second group of carotenoid oxygenases, carotenoid cleavage dioxygenases (CCDs), which act on different carotenoid substrates The CCDs of higher plants contribute to diverse physiological processes, including the regulation of the outgrowth of lateral shoot buds [18–20] and plastid development [21] Plants release volatile apocarotenoids, including C13 ketones such as b-ionone and damascone [22], which constitute an essential aroma note in tea, grapes, roses, tobacco and wine [23] Such compounds may arise by unspecific oxidative degradation or lipid co-oxidation processes, involving lipoxygenases [24] Alternatively, they are produced by double bond-specific cleavage reactions mediated by peroxidases [25] or by CCDs such as members of the plant CCD1 subfamily Plant CCD1s cleave numerous cyclic and linear all-trans-carotenoids at the C9–C10 and C9¢–C10¢ double bonds into C14 dialdehydes, which are common to all carotenoid substrates, and two variable end-group-derived C13 ketones [6,7,16] The wide substrate specificity of plant CCD1s allows the production of divergent volatile C13 compounds, including b-ionophores, a-ionones, pseudoionone and geranylacetone The first member of the CCD1 subfamily was identified from Arabidopsis thaliana [26], and was later shown to act as a dioxygenase [27] Sequence homology then allowed the identification and charcterization of orthologs from several plant species, such as crocus, tomato, grape, melon, petunia and maize [15,28–32] The biological function of CCD1s was confirmed by loss-of-function experiments in tomato fruits and petunia flowers, leading to decreased emission of b-ionone [27,31] Moreover, recent studies on the CCD1 from maize indicated its involvement in the formation of cyclohexenone and mycorradicin in mycorrhizal roots [33] Underscoring a role of CCD1 in carotenoid catabolism, seeds of Arabidopsis ccd1 mutants contained elevated carotenoid levels [34] This suggested that the modification of CCD1 expression is instrumental for altering volatile production contributing to taste, or in increasing the carotenoid content in crop plant tissues where elevated provitamin A carotenoid levels are aimed for, such as high-b-carotene tomato [35,36], canola [37], golden rice [38] or golden potato A novel route for geranial formation [39] Hence, the identification of substrates and cleavage sites of CCD1s from crop plants is considered to be a worthwhile approach It has recently been discovered that plant CCD1s exert additional activity at the C5–C6 and ⁄ or the C5¢– C6¢ double bonds of acyclic carotenoids, leading to the formation of the C8 ketone 6-methyl-5-hepten-2-one [32] In this study, we investigated the enzymatic activities of the sole CCD1 [Oryza sativa CCD1 (OsCCD1)] occurring in rice Our study revealed the C7¢–C8¢ double bond of linear and monocyclic carotenoids to be an additional novel cleavage site of OsCCD1, leading to geranial and indicating a novel plant route for the formation of this widespread volatile compound Results Purified OsCCD1 cleaves acyclic apolycopenals at the C7–C8 double bond To investigate the activity of OsCCD1, the corresponding cDNA was cloned and expressed as a glutathione S-transferase (GST)-fusion protein in Escherichia coli cells However, the insolubility of the fusion protein, which could not be improved by modulating the expression conditions, hampered its purification Therefore, the GST–OsCCD1 fusion was expressed in BL21(DE3) E coli cells harboring the vector pGro7, which encodes the chaperones groES–groEL, enhancing correct folding This resulted in a striking improvement of the GST–OsCCD1 solubility, allowing protein purification (Fig S1) It has been shown that CCD1s from Arabidopsis and maize maintain their regional specificity in cleaving the C9–C10 double bond with the synthetic apocarotenoid b-apo-8¢-carotenal (C30), forming b-ionone (C13) and the C17 dialdehyde apo-8,10¢-carotendial [27,32] To test the cleavage activity of OsCCD1, in vitro assays were performed with this substrate, using purified enzyme Subsequent HPLC analyses (data not shown) revealed a cleavage pattern identical to that of the plant CCD1s mentioned above To determine the impact of the chain length and of the ionone ring on the cleavage pattern, purified OsCCD1 was incubated with b-apo-10¢-carotenal (C27), which is shorter than b-apo-8¢-carotenal (C30), and the acyclic substrates apo-10¢-lycopenal (C27) and apo-8¢-lycopenal (C30) HPLC analyses of the incubation with the cyclic b-apo-10¢-carotenal (Fig 1; substrate I) revealed an almost complete conversion of this substrate (Table S1) and the formation of the C14 dialdehyde apo-10,10¢-carotendial (rosafluene dialdehyde; product 1) and b-ionone (C13; product 2), as confirmed by FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works 737 A novel route for geranial formation A B C 738 A Ilg et al Fig (A) HPLC analyses of OsCCD1 in vitro assays with synthetic apocarotenoids The cyclic b-apo-10¢-carotenal (C27, I) was cleaved into apo-10,10¢-carotendial (C14, 1) and b-ionone (C13, 2); the lower amount of the former is probably a result of its instability The acyclic substrates apo-10¢-lycopenal (C27, II) and apo-8¢-lycopenal (C30, III) were converted into pseudoionone (3) and three dialdehydes of different chain lengths (1, and from II; 4, and from III), as indicated by their UV ⁄ visible-spectra (shown in the insets) and retention times (B) Structures of the synthetic substrates showing the cleavage I, b-apo-10¢-carotenal; II, apo-10¢-lycopenal; III, apo-8¢lycopenal The substrates were cleaved at three different double bonds, including the novel site (b, shaded) Cleavage of the C9–C10 double bond (a) results in products and from I, products and from II, and products and from III The cleavage of (b) leads to product from II, and product from III The cleavage of the C5–C6 double bond (c) leads to product from II, and product from III (C) Structures of the cleavage products detected: 1, apo-10,10¢-carotendial (C14 dialdehyde); 2, b-ionone (C13); 3, pseudoionone (C13); 4, apo-8,10¢-carotendial (C17 dialdehyde); 5, apo-6,10¢-carotendial (C19 dialdehyde); 6, apo-8,8¢-carotendial (C20 dialdehyde); 7, apo-6,8¢-carotendial (C22 dialdehyde) LC-MS and GC-MS analyses, respectively (data not shown) The relatively low amount of the C14 dialdehyde is probably a result of instability The formation of b-ionone from b-apo-8¢-carotenal (C30) and b-apo10¢-carotenal (C27) suggested that the cleavage of the C9–C10 double bond occurs independently of the chain length of monocyclic apocarotenals, pointing to the ionone ring as a determinant governing the reaction site The two acyclic substrates were cleaved almost completely within 30 (Table S1), proving them to be as suitable as the monocyclic apocarotenals However, as shown in the HPLC analyses (Fig 1), the cleavage of apo-10¢-lycopenal (C27; substrate II) led to a more complex mixture of products, including three compounds (products 1, and 5) identified as dialdehydes on the basis of the fine structure of the corresponding UV–visible spectra The three dialdehydes differed in their chain lengths, as indicated by their retention times and absorption maxima Products and were supposed to represent a C14 and a C19 dialdehyde, respectively These are expected to arise upon cleavage at the known plant CCD1 sites (C9–C10 and C5–C6) The retention time of product indicated a chain length between C14 and C19 This pointed to the recognition of a novel cleavage site, at the C7–C8 double bond, between the above mentioned C9–C10 and C5–C6 positions, yielding a C17 dialdehyde To confirm their nature, the dialdehyde products, 1, and 5, were purified and analyzed by LC-MS In order to stabilize the C14 dialdehyde (product 1), it was derivatized with O-methyl-hydroxylamine-hydrochloride prior to LC-MS analyses As shown in Fig 2, derivatized FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works A Ilg et al A novel route for geranial formation HPLC analyses of the incubation with apo-8¢-lycopenal (C30; substrate III in Fig 1) confirmed novel cleavage of the C7–C8 double bond As shown in Fig 1, the reaction led to an equivalent series of C17, C20 and C22 dialdehydes (products 4, and 7), corresponding to the cleavage of the C9–C10, C7–C8 and C5–C6 double bonds, respectively The nature of these dialdehyde products was confirmed by LC-MS analyses (data not shown) Cleavage of the three double bonds described above must also lead to the three different mono-oxygenated products pseudoionone (C13), geranial (C10) and 6-methyl-5-hepten-2-one (C8) (for structures, see Fig 4) Pseudoionone was found by HPLC analysis (Fig 1; product 3) and its presence was further demonstrated by GC-MS analyses, which also showed the formation of geranial and and 6-methyl-5-hepten-2-one (data not shown) OsCCD1 mediates double cleavage of different site combinations in 3-OH-c-carotene and lycopene Fig LC-MS identification of the dialdehydes produced in vitro The three dialdehydes formed from apo-10¢-lycopenal in vitro (Fig 1; products 1, and 5) represent a C14, a C17 and a C19 dialdehyde, respectively, as suggested by their molecular ions of mass 275.17 (I, [M + (NCH3)2 + H]+; product 1), 257.13 (II, [M + H]+; product 4) and 283.14 (III, [M + H]+; product 5) product exhibited an [M + (NCH3)2 + H]+ molecular ion of mass 275.17 (substrate I), consistent with the C14 dialdehyde structure, and the molecular ions for products and (Fig 2; substrates II and III) proved their identities as C17 and C19 dialdehydes, respectively To determine the cleavage sites in natural substrates, purified OsCCD1 was incubated with the bicyclic zeaxanthin, the monocyclic 3-OH-c-carotene and the acyclic lycopene As shown in Fig 3, OsCCD1 converted zeaxanthin (substrate I) into the two products 3-OH-b-ionone (C13; product 1) and rosafluene-dialdehyde (C14; product 2), as confirmed by LC-MS and GC-MS analyses, respectively (data not shown) This suggested that OsCCD1 cleaves the C9–C10 and C9¢–C10¢ double bonds of bicyclic carotenoids, like other plant orthologs Although it occurred at lower conversion rates than with the synthetic substrates (Table S1), we clearly observed the accumulation of the C17 and C19 dialdehydes (Fig 3; products and 5, respectively) from 3-OH-c-carotene (Fig 3; substrate II) Owing to its instability, the third dialdehyde (C14), expected from the cleavage of the C9–C10 and C9¢–C10¢ double bonds, occurred at low levels only (Fig 3; product 2) The formation of the C14, C17 and C19 dialdehydes (for structures, see Fig 3C) suggested a single cut at the C9–C10 double bond of the ring-bearing side of 3-OH-c-carotene in combination with several cleavage options in the linear half of the molecule, namely at the C9¢–C10¢, C7¢–C8¢ and C5¢–C6¢ double bond The occurrence of the C17 dialdehyde confirmed the novel site at the C7¢–C8¢ double bond observed with apolycopenals and implied the formation of geranial Accordingly, the GC-MS analyses of the in vitro assays pointed to the conversion of 3-OH-c-carotene into FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works 739 A novel route for geranial formation A Ilg et al Fig (A) HPLC analyses of OsCCD1 in vitro incubations with three natural carotenoids UV–visible spectra of the products are shown in the insets The bicyclic zeaxanthin (I) was converted into 3-OH-b-ionone (1) and apo-10,10¢-carotendial (C14, 2) The products 3-OH-b-ionone (C13, 1), apo-10,10¢-carotendial (C14 dialdehyde, 2), pseudoionone (C13, 3), apo-8,10¢-carotendial (C17 dialdehyde, 4) and apo-6,10¢-carotendial (C19 dialdehyde, 5) were obtained from 3-OHc-carotene (II) The cleavage of lycopene (III) led to pseudoionone (C13, 3), apo-8,10¢-carotendial (C17 dialdehyde, 4) and apo-6,10¢-carotendial (C19 dialdehyde, 5) Longer incubations with lycopene also resulted in the accumulation of the C14 dialdehyde apo-10,10¢-carotendial corresponding to compound (not shown) (B) Structures of the substrates showing the cleavage sites, including the novel C7–C8 ⁄ C7¢–C8¢ double bonds (b, b¢, shaded) I, zeaxanthin; II, 3-OH-c-carotene; III, lycopene OsCCD1 cleaves zeaxanthin at the C9–C10 ⁄ C9¢–C10¢ double bonds (a ⁄ a¢) 3-OH-c-Carotene (II) is cleaved at the C9–C10 double bond (a) in combination with the C9¢–C10¢ double bond (a¢), the C7¢–C8¢ double bond or the C5¢–C6¢ (c¢) double bond Lycopene (III) is cleaved either at the C9–C10 double bond (a) in combination with one of the three double bonds C9¢–C10¢ (a¢), C7¢–C8¢ (b¢) or C5¢–C6¢ (c¢), or at the C9¢–C10¢ double bond (a¢) in combination with one of the a, b or c sites (C) Structures of the cleavage products detected A substrate IV), which was correctly recognized by the National Institute of Standards and Technology (NIST) library (mass spectral search program Version 2.0) In addition, these GC-MS analyses (data not shown) revealed the formation of the known plant CCD1 mono-oxo products 6-methyl-5-hepten-2-one, 3-OH-b-ionone and pseudoionone, the latter two of which had already been detected in the HPLC analyses (Fig 3; products and 3) The high lipophilicity of lycopene did not allow the use of octyl-b-glucoside as detergent in the corresponding in vitro assays Therefore, lycopene micelles were produced with a Triton X mixture The products formed from this substrate (Fig 3; substrate III, products 3, and 5) suggested the cleavage of the double bond combinations C9–C10 ⁄ C7¢–C8¢, C9–C10 ⁄ C5¢–C6¢ and their symmetrical counterparts The C14 dialdehyde formed by cleavage of the C9–C10 ⁄ C9¢–C10¢ double bond combination was only detectable in longer incubations (data not shown) GC-MS analyses of the lycopene incubations demonstrated the formation of pseudoionone and 6-methyl-5-hepten-2-one However, geranial could not be detected, although the formation of the C17 dialdehyde confirmed the cleavage of lycopene at the novel C7–C8 or C7¢–C8¢ site B C geranial (Fig 4; substrate IV), as indicated by the detection of the expected [M]+ molecular ion of mass 152.3 and the fragmentation pattern (Fig 4; 740 OsCCD1 catalyzes the formation of three different volatiles from lycopene in vivo To confirm cleavage at the C7–C8 or C7¢–C8¢ double bond in vivo, OsCCD1 was expressed in lycopene- FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works A Ilg et al A novel route for geranial formation Fig GC-MS analyses of volatile OsCCD1 products Volatile compounds produced in lycopene-accumulating and OsCCD1-expressing cells were collected using SPME and subjected to GC-MS (I–III) As suggested by the [M]+ molecular ions (bold) and comparison of mass spectra with the NIST library, the in vivo activity of OsCCD1 led to the volatiles 6-methyl-5-hepten-2-one (C8, I), geraniol (C10, II) and pseudoionone (C13, III) IV represents the detection of geranial (C10) produced from 3-OH-c-carotene in vitro A B Fig Determination of the relative amounts of dialdehyde products (A) Incubations with apo-8¢-lycopenal (C30) The peak areas of the three dialdehydes (C17, C20 and C22) formed from apo-8¢-lycopenal were calculated at a max plot of 350–550 nm The values represent the proportion of each dialdehyde in the sum of the three peak areas (B) Incubations with apo-10¢-lycopenal (C27), 3-OH-c-carotene and lycopene The values represent ratios of the C17 and C19 dialdehydes in the sum of their peak areas calculated by integrating each peak at its individual kmax Data represent the average of six independent incubations accumulating E coli cells, and volatile compounds were collected from the medium and analyzed by GCMS As shown in Fig 4, the activity of the enzyme resulted in the accumulation of 6-methyl-5-hepten-2one (I), the reduced form of geranial, geraniol (II), and pseudoionone (III) The three compounds, undetectable in the controls, were identified by their correct [M]+ molecular ions and by comparing the mass specFEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works 741 A novel route for geranial formation A Ilg et al tra with the NIST library HPLC analyses of the corresponding cell pellets revealed the accumulation of a complex mixture of compounds, tentatively identified as dialcohols corresponding to three dialdehydes described above (data not shown) OsCCD1 exhibits different preferences for the three cleavage sites To gain insights into the preference of OsCCD1 for the three cleavage sites, we determined the relative amounts of the C17, C20 and C22 dialdehyde products formed upon incubation with apo-8¢-lycopenal (C30), corresponding to C9–C10, C7–C8 and C5–C6 double bond recognition, respectively (Fig 1B; substrate III) The enzyme exhibited by far the highest preference for the C9–C10 double bond in vitro, as suggested by the predominance of the C17 dialdehyde, which accounted for about 90% of the total dialdehyde products (Fig 5A) The relative amount (about 7%) of the C20 dialdehyde was much higher than of the C22 dialdehyde (about 2%), indicating a higher affinity for the novel C7–C8 double bond than for the C5–C6 double bond The instability of the C14 dialdehyde arising from the double cleavage at the C9–C10 ⁄ C9¢–C10¢ double bonds in lycopene and 3-OH-c-carotene hampered determination of the preference for these sites, and allowed only a comparison of the C9–C10 ⁄ C7¢–C8¢ and C9–C10 ⁄ C5¢–C6¢ cleavage products corresponding to the C17 and C19 dialdehydes, respectively The higher relative amount of the C17 dialdehyde indicated a higher preference for the C7–C8 than for the C5–C6 double bond in 3-OH-c-carotene, whereas the opposite tendency was observed with lycopene and apo-10¢-lycopenal (Fig 5B) Discussion Plant CCD1s are known to catalyze the cleavage of the C9–C10 and the C9¢–C10¢ double bonds of several carotenoids Recently, it was shown that these enzymes can also cleave at the C5–C6 and ⁄ or the C5¢–C6¢ double bonds in lycopene [32] This was deduced from GC-MS analyses of lycopene-accumulating E coli cells expressing different plant CCD1s and from in vitro incubations with a CCD1 from maize (ZmCCD1), showing in both cases the formation of 6-methyl-5hepten-2-one (C8), besides pseudoionone Geranial (C10) was not detected, and cleavage of the C7– C8 ⁄ C7¢–C8¢ double bonds was therefore excluded [32] On the basis of our recent work on a Nostoc carotenoid oxygenase producing geranial and derivatives thereof from monocyclic carotenoids [40], we assumed 742 that plant CCD1s may also be able to cleave the C7–C8 ⁄ C7¢–C8¢ double bonds Here, we demonstrate that the rice enzyme OsCCD1 cleaves linear ends of carotenoids at three different double bond positions, including the C7–C8 ⁄ C7¢–C8¢ double bonds, leading to geranial To avoid further metabolization of products that can occur in vivo, we relied first on in vitro incubations using purified enzyme, which allowed clear identification of the products formed In a first approach, we checked the site specificities using synthetic apocarotenals packed in octyl-b-glucoside micelles This enabled us to observe the cleavage of the C7–C8 double bond However, the confirmation of this novel activity with the highly lipophilic lycopene and c-carotene required the use of different detergents The best activities were obtained with micelles produced with a Triton X mixture, following the protocol of Prado-Cabrero et al [41] The accumulation of the C17 dialdehyde in the lycopene assays confirmed the cleavage at the C9–C10 ⁄ C7¢–C8¢ double bonds However, the activities determined were still low in comparison to the incubations with zeaxanthin, and did not allow clear identification of geranial Furthermore, we did not detect significant conversion of lycopene in the 30 incubations used to determine the substrate preferences of the enzyme (Table S1) This weak activity is probably due to the use of the Triton X mixture, which was necessary to soulibilize lycopene, but led to an overall reduction of enzyme activity (Table S1) Therefore, we used the more polar 3-OH-c-carotene, which could be incorporated into octyl-b-glucoside micelles and which was readily cleaved at the double bond combinations C9–C10 ⁄ C9¢–C10¢, C9–C10 ⁄ C7¢–C8¢ and C9–C10 ⁄ C5¢–C6¢ The formation of multiple dialdehyde products allows some conclusions to be drawn on the site preferences of OsCCD1 For instance, the C14, C17 and C19 dialdehydes formed from lycopene and 3-OH-ccarotene arise from cleavage of the C9–C10 double bond, which is combined with the C9¢–C10¢, C7¢–C8¢ or C5¢–C6¢ double bonds The C9–C10 ⁄ C9¢–C10¢ double bonds constitute the main site, as suggested by the determination of the relative amounts of the dialdehydes produced from apo-8¢-lycopenal; the C9–C10 ⁄ C9¢– C10¢ double bonds also constitute the sole cleavage site in ring-bearing moieties of substrates This preference may explain the absence of the C20, C24 and C22 dialdehydes in the lycopene incubation, which would be expected if the cleavage reactions occurred only at the C7–C8 ⁄ C7¢–C8¢ and ⁄ or C5 ⁄ C6 ⁄ C5¢–C6¢ double bonds A further conclusion is that the first cleavage site plays a role in the determination of the second one in acyclic FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works A Ilg et al substrates This is shown by the different preferences for the C7–C8 and C5–C6 double bonds (Fig 5) in apo-10¢-lycopenal (C27) and apo-8¢-lycopenal (C30), which mimic a lycopene molecule cleaved at the C9¢– C10¢ and C7¢–C8¢ double bonds, respectively Moreover, comparison of the relative amounts of the C19 and C17 dialdehydes accumulated in the incubations with the natural substrates lycopene and 3-OH-c-carotene indicates that the preference of OsCCD1 for the C5¢–C6¢ and C7¢–C8¢ double bonds depends on the nature of the substrate The cleavage of a sole double bond in the cyclic moiety of 3-OH-c-carotene and of monocyclic b-apocarotenals with different chain lengths indicates that the b-ionone end-group may determine the reaction site in the polyene chain This may provide an explanation for the ‘wobbling’ of the enzyme on linear substrate moieties, where three different double bonds are being recognized Similar results were obtained with the cyanobacterial enzyme SynACO, representing up to now the only carotenoid oxygenase with a known crystal structure [42] SynACO cleaved b-apocarotenoids with different chain lengths at a sole site, the C15–C15¢ double bond, leading to retinal and derivatives thereof [43] In contrast, apolycopenals were cleaved at multiple positions, including the C15–C15¢ double bond, and indicating ‘wobbling’ of the enzyme (S Ruch, S Al-Babili and P Beyer; unpublished data) Owing to the high sequence similarity of plant CCD1s, it appears likely that the cleavage of the C7–C8 ⁄ C7¢–C8¢ double bonds of linear substrates is not unique to OsCCD1 Apart from the formation of geranial, the OsCCD1 cleavage reactions were identical to those of other plant CCD1s, as supported by the formation of pseudoionone, 6-methyl-5-hepten-2-one and b-ionones Geranial is biologically active and known to exert antifungal and antimicrobial activities [44,45]; it represents a major volatile of tomato fruits [46] and roses [47] Moreover, citral, the mixture of geranial and its cis-isomer neral, is a major component of the aroma of lemon grass and other lemon-scented plants Geranial is synthesized from geranyl diphosphate by the enzymes geraniol synthase [48] and geraniol dehydrogenase [49] The enzymatic cleavage of monocyclic and acyclic carotenoids into geranial represents a novel biosynthetic route, and may provide an explanation for the impact of lycopene accumulation on the emission of geranial, as observed in the fruits of several tomato and watermelon varieties [46], as well as in transgenic tomato fruits, where elevated carotenoid levels were accompanied by an increased emission of citral [50] The possible synthesis of geranial by tomato CCD1s is now under investigation A novel route for geranial formation Experimental procedures Cloning procedures Five micrograms of total RNA, isolated from 14-day-old seedlings (O sativa var japonica cv TP309), was used for cDNA synthesis using SuperScript III RnaseH) (Invitrogen, Paisley, UK), according to the instructions of the manufacturer Two microliters of cDNA was then applied for the amplification of OsCCD1 (accession no AK066766, encoded by Os12g0640600), using the primers CCD-1 (5¢ATGGGAGGCGGCGATGGCGATGAG-3¢) and CCD-2 (5¢-TCACGCTGATTGTTTTGCCAGTTG-3¢) The PCR reaction was performed with 100 ng of each primer, 200 lm dNTPs and lL of Advantage cDNA Polymerase Mix (BD Biosciences, San Jose, CA, USA) in the buffer provided, as follows: of initial denaturation at 94 °C, followed by 32 cycles of 30 s at 94 °C, 30 s at 58 °C, and at 68 °C, and 10 of final polymerization at 68 °C The obtained PCR product was purified using GFX PCR DNA and a Gel Band Purification Kit (Amersham Biosciences, Piscataway, NJ, USA), and cloned into the pCR2.1–TOPO vector and pBAD ⁄ TOPO (Invitrogen) to yield pCR–OsCCD1 and pBAD–OsCCD1, respectively The nature of the cDNA was verified by sequencing To express OsCCD1 as a GST-fusion protein, the corresponding cDNA was excised as an EcoRI fragment from pCR–OsCCD1, and then ligated into accordingly digested and alkaline phosphatase-treated pGEX–5X-1 (Amersham Biosciences) to yield pGEX–OsCCD1 Protein expression and purification pGEX–OsCCD1 was transformed into BL21(DE3) E coli cells harboring the plasmid pGro7 (Takara Bio Inc.; Mobitec, Gottingen, Germany), which encodes the groESgroEL ă chaperone system under the control of an arabinose-inducible promoter Overnight cultures (2.5 mL) were inoculated into 50 mL of · YT medium containing 0.2% (w ⁄ v) arabinose, grown at 28 °C to a D600 nm of 0.7, and induced with 0.2 mm isopropyl thio-b-d-galactoside for h Cells were harvested by centrifugation (10 min, 6000 g), resuspended in mL of NaCl ⁄ Pi (pH 7.3), and lysed using a French press Six milliliters of NaCl ⁄ Pi (pH 7.3) containing 1% Triton X-100 was then added, and the suspension was incubated at room temperature for 30 After centrifugation for 10 at 12 000 g, the fusion protein was purified using glutathione– Sepharose 4B (Amersham Biosciences), according to the manufacturer’s instructions OsCCD1 was then released by overnight treatment with the protease factor Xa in NaCl ⁄ Pi (pH 7.3) at room temperature, according to the manufacturer’s instructions (Amersham Biosciences) The protein eluate contained approximately 50% purified OsCCD1 Purification steps and protein expression were monitored by SDS ⁄ PAGE The control strain expressed only GST FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works 743 A novel route for geranial formation A Ilg et al to the peak area of the internal standard, which was quantified at its absorption maximum of 285 nm Enzyme assays Synthetic substrates were kindly provided by BASF (Ludwigshafen, Germany) Lycopene was obtained from Roth (Karlsruhe, Germany) Zeaxanthin and 3-OH-c-carotene were isolated from E coli cells transformed with carotenoid biosynthetic genes (unpublished data) The substrates were purified using TLC, and quantified spectrophotometrically at their individual kmax values, using extinction coefficients calculated from E1% [51] Protein concentrations were determined using the BioRad protein assay kit (BioRad, Hercules, CA, USA) In vitro assays contained 40 lg of purified enzyme eluate at substrate concentrations of 80 lm (lycopene and 3-OHc-carotene) or 40 lm (zeaxanthin and synthetic substrates) For the production of lycopene micelles, dried substrate was resuspended in 200 lL of benzene and mixed with 150 lL of an ethanolic detergent mixture consisting of 0.7% (v ⁄ v) Triton X-100 and 1.6% (v ⁄ v) Triton X-405 The mixture was then dried using a vacuum centrifuge to produce a carotenoid-containing gel The gel was resuspended in 110 lL of 2· incubation buffer containing mm tris(2-carboxyethyl)phosphine, 0.6 mm FeSO4 and mgỈmL)1 catalase (Sigma, Deisenhofen, Germany) in 200 mm Hepes ⁄ NaOH (pH 7.8) One hundred microliters of this lycopene suspension was then used in the in vitro assay, which was started by adding water and purified OsCCD1 to obtain a final volume of 200 lL 3-OH-c-Carotene, zeaxanthin and apocarotenoids were solubilized using octyl-b-glucoside at a final concentration of 1% (v ⁄ v) For this purpose, substrates were mixed with 50 lL of a 4% octyl-b-glucoside ethanolic solution, dried using a vacuum centrifuge, and resuspended in 100 lL of the 2· incubation buffer mentioned above Water and purified OsCCD1 were then added to obtain the final volume of 200 lL Depending on the substrates, the incubations were performed at 28 °C for h (lycopene and 3-OH-c-carotene), h (zeaxanthin) or 30 (synthetic substrates) Reactions were stopped by adding two volumes of acetone Lipophilic compounds were partitioned against petroleum ether ⁄ diethyl ether : (v ⁄ v), vacuum-dried, and dissolved in 40 lL of chloroform HPLC analyses were then performed using 20 lL of the extracts For GC-MS analyses, volatile compounds were collected with solid-phase microextraction (SPME) fibers (100 lm polydimethylsiloxane; Sigma-Aldrich) for 30 Conversion rates were determined in 30 incubation assays using 30 lg of purified enzyme eluate For quantification, 200 lL of an acetonic solution of a-tocopherole acetate (1 mgỈmL)1) was added as internal standard to each assay prior to extraction The conversion rates were determined by calculating the decrease of substrate peak areas measured at their individual kmax values using the max plot function of the software empower pro (Waters, Eschborn, Germany) Peak areas were normalized relative 744 In vivo test using lycopene-accumulating E coli cells Lycopene-accumulating XL1-Blue E coli cells (unpublished data), harboring the corresponding biosynthetic genes from Erwinia herbicola, were transformed with pBAD–OsCCD1 or with pBAD–TOPO as a negative control Overnight cultures were used to inoculate 50 mL of LB medium Bacteria were grown at 28 °C to a D600 nm of 0.5, and induced with 0.08% (w ⁄ v) arabinose Cells were harvested after h, and volatile compounds were collected by introducing the SPME fiber into the cell-free medium for 30 For HPLC analyses, cells were harvested after h, and carotenoids were extracted and processed as described above Analytical methods For HPLC analyses, a Waters system equipped with a photodiode array detector (model 996) was used A C30-reversed phase column (YMC Europe, Schermbeck, Germany) was developed with solvent system B [MeOH ⁄ t-butylmethyl ether ⁄ water (60 : : 20, v ⁄ v ⁄ v)] and solvent system A [MeOH ⁄ t-butylmethyl ether (1 : 1, v ⁄ v)] at a flow rate of mLỈmin)1, using a linear gradient from 100% solvent B to 43% solvent B within 45 min, and then to 0% solvent B within The final conditions were maintained for 26 at a flow rate of mLỈmin)1, and this was followed by re-equilibration LC-MS analyses of compounds collected from HPLC were performed using a Thermo Finnigan LTQ mass spectrometer coupled to a Surveyor HPLC system consisting of a Surveyor Pump Plus, Surveyor PDA Plus and Surveyor Autosampler Plus (Thermo Electron, Waltham, MA, USA) Separations were carried out using a YMC-Pack C30reversed phase column (150 · mm internal diameter, lm) Separation and identification of the C17 and C19 dialdehydes was performed as described in [40] The oxime of the C14 dialdehyde was produced by adding 50 lL of O-methyl-hydroxylamine-hydrochloride (15 mgỈmL)1) to an MeOH solution of the HPLC-purified compound, and then incubating for 20 at 50 °C The product was then partitioned against petroleum ether ⁄ diethyl ether : (v ⁄ v) The identification of the oxime was carried out according to [40] GC-MS analyses were carried out with a Finnigan Trace DSQ mass spectrometer coupled to a Trace GC gas chromatograph equipped with a 30 m Zebron ZB column (5% phenylpolysilanoxane ⁄ 95% dimethylpolysilanoxane, 0.25 mm internal diameter, and 0.25 lm film thickness; Phenomenex, Aschaffenburg, Germany) The temperature program used was as follows: 50 °C held isocratically for min, followed by a ramp of 25 °CỈmin)1 to a final FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works A Ilg et al temperature of 340 °C, which was maintained for an additional The He carrier gas flow was maintained at mLỈmin)1 using a split flow of : 20 The splitless time was min, and the injector oven temperature was set at 220 °C Standard electrospray ionization was used at an ion source potential of 70 eV and with an ion source temperature of 200 °C Identification of compounds was done by comparing the mass spectra with the NIST database A novel route for geranial formation 12 13 Acknowledgements This work was supported by the HarvestPlus programme (http://www.harvestplus.org) and by the Deutsche Forschungsgemeinschaft (DFG), Grant 892 ⁄ 1-3 We are indebted to J Mayer for valuable discussions We thank H Ernst for providing the synthetic substrates and E Scheffer for skilful technical assistance 14 References 16 Cunningham FX & Gantt E (1998) Genes and enzymes of carotenoid biosynthesis in plants Annu Rev Plant Physiol Plant Mol Biol 49, 557–583 Hirschberg J (2001) Carotenoid biosynthesis in flowering plants Curr Opin Plant Biol 4, 210–218 Fraser PD & BramLey PM (2004) The biosynthesis and nutritional uses of carotenoids Prog Lipid Res 43, 228– 265 DellaPenna D & Pogson BJ (2006) Vitamin synthesis in plants: tocopherols and carotenoids Annu Rev Plant Biol 57, 711–738 Moise AR, von Lintig J & Palczewski K (2005) Related enzymes solve evolutionarily recurrent problems in the metabolism of carotenoids Trends Plant Sci 10, 178–186 Bouvier F, Isner JC, Dogbo O & Camara B (2005) Oxidative tailoring of carotenoids: a prospect towards novel functions in plants Trends Plant Sci 10, 187–194 Auldridge ME, McCarty DR & Klee HJ (2006) Plant carotenoid cleavage oxygenases and their apocarotenoid products Curr Opin Plant Biol 9, 315–321 Akiyama K (2007) Chemical identification and functional analysis of apocarotenoids involved in the development of arbuscular mycorrhizal symbiosis Biosci Biotechnol Biochem 71, 1405–1414 ´ Bouwmeester HJ, Roux C, Lopez-Raez JA & Becard G (2007) Rhizosphere communication of plants, parasitic plants and AM fungi Trends Plant Sci 12, 224–230 ` 10 Gomez-Roldan V, Fermas S, Brewer PB, Puech-Pages V, Dun EA, Pillot JP, Letisse F, Matusova R, Danoun S, Portais JC et al (2008) Strigolactone inhibition of shoot branching Nature 455, 189–194 11 Umehara M, Hanada A, Yoshida S, Akiyama K, Arite T, Takeda-Kamiya N, Magome H, Kamiya Y, Shirasu 15 17 18 19 20 21 22 23 24 K, Yoneyama K et al (2008) Inhibition of shoot branching by new terpenoid plant hormones Nature 455, 195–200 Schliemann W, Ammer C & Strack D (2008) Metabolite profiling of mycorrhizal roots of Medicago truncatula Phytochemistry 69, 112–146 Walter MH, Fester T & Strack D (2000) Arbuscular mycorrhizal fungi induce the non-mevalonate methylerythritol phosphate pathway of isoprenoid biosynthesis correlated with accumulation of the ‘yellow pigment’ and other apocarotenoids Plant J 21, 571–578 Bouvier F, Dogbo O & Camara B (2003) Biosynthesis of the food and cosmetic plant pigment bixin (annatto) Science 300, 2089–2091 Bouvier F, Suire C, Mutterer J & Camara B (2003) Oxidative remolding of chromoplast carotenoids: identification of the carotenoid dioxygenase CsCCD and CsCZD genes involved in Crocus secondary metabolite biogenesis Plant Cell 15, 47–62 Kloer DP & Schulz GE (2006) Structural and biological aspects of carotenoid cleavage Cell Mol Life Sci 3, 2291–2303 Schwartz SH, Tan BC, Gage DA, Zeevart JAD & McCarty DR (1997) Specific oxidative cleavage of carotenoids by VP14 of maize Science 276, 1872–1874 Schwartz SH, Qin X & Loewen MC (2004) The biochemical characterization of two carotenoid cleavage enzymes from Arabidopsis indicates that a carotenoidderived compound inhibits lateral branching J Biol Chem 279, 46940–46945 Booker J, Auldridge M, Wills S, McCarty D, Klee H & Leyser O (2004) MAX3 ⁄ CCD7 is a carotenoid cleavage dioxygenase required for the synthesis of a novel plant signaling molecule Curr Biol 14, 1232–1238 Alder A, Holdermann I, Beyer P & Al-Babili S (2008) Carotenoid oxygenases involved in plant branching catalyse a highly specific conserved apocarotenoid cleavage reaction Biochem J 416, 289–296 Naested H, Holm A, Jenkins T, Nielsen HB, Harris CA, Beale MH, Andersen M, Mant A, Scheller H, Camara B et al (2004) Arabidopsis VARIEGATED encodes a chloroplast-targeted, zinc-finger protein required for chloroplast and palisade cell development J Cell Sci 117, 4807–4818 Goff SA & Klee HJ (2006) Plant volatile compounds: sensory cues for health and nutritional value? Science 311, 815–819 ´ Rodrı´ guez-Bustamante E & Sanchez S (2007) Microbial production of C13-norisoprenoids and other aroma compounds via carotenoid cleavage Crit Rev Microbiol 33, 211–230 Wu Z & Robinson S (1999) Co-oxidation of b-carotene catalyzed by soybean and recombinant pea lipoxygenases J Agric Food Chem 47, 4899–4906 FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works 745 A novel route for geranial formation A Ilg et al 25 Zorn H, Langhoff S, Scheibner M, Nimtz M & Berger RG (2003) A peroxidase from Lepista irina cleaves beta,beta-carotene to flavor compounds Biol Chem 384, 1049–1056 26 Schwartz SH, Xiaoqiong Q & Zeevart JAD (2001) Characterization of a novel carotenoid cleavage dioxygenase from plants J Biol Chem 276, 25208–25211 27 Schmidt H, Kurtzer R, Eisenreich W & Schwab W (2006) The carotenase AtCCD1 from Arabidopsis thaliana is a dioxygenase J Biol Chem 281, 9845–9851 28 Simkin AJ, Schwartz SH, Auldridge M, Taylor MG & Klee HJ (2004) The tomato carotenoid cleavage dioxygenase genes contribute to the formation of the flavor volatiles b-ionone, pseudoionone, and geranylacetone Plant J 40, 882–892 29 Mathieu S, Terrier N, Procureur J, Bigey F & Gunata ă Z (2005) A carotenoid cleavage dioxygenase from Vitis vinifera L.: functional characterization and expression during grape berry development in relation to C13-norisoprenoid accumulation J Exp Bot 56, 2721–2731 30 Ibdah M, Azulay Y, Portnoy V, Wasserman B, Bar E, Meir A, Burger Y, Hirschberg J, Schaffer AA, Katzir N et al (2006) Functional characterization of CmCCD1, a carotenoid cleavage dioxygenase from melon Phytochemistry 67, 1579–1589 31 Simkin AJ, Underwood BA, Auldridge M, Loucas HM, Shibuya K, Schmelz E, Clark DG & Klee HJ (2004) Circadian regulation of the PhCCD1 carotenoid cleavage dioxygenase controls emission of beta-ionone, a fragrance volatile of petunia flowers Plant Physiol 136, 3504–3514 32 Vogel JT, Tan BC, McCarty DR & Klee HJ (2008) The carotenoid cleavage dioxygenase enzyme has broad substrate specificity, cleaving multiple carotenoids at two different bond positions J Biol Chem 283, 11364–11373 33 Sun Z, Hans J, Walter MH, Matusova R, Beekwilder J, Verstappen FW, Ming Z, van Echtelt E, Strack D, Bisseling T et al (2008) Cloning and characterisation of a maize carotenoid cleavage dioxygenase (ZmCCD1) and its involvement in the biosynthesis of apocarotenoids with various roles in mutualistic and parasitic interactions Planta 228, 789–801 34 Auldridge ME, Block A, Vogel JT, Dabney-Smith C, Mila I, Bouzayen M, Magallanes-Lundback M, DellaPenna D, McCarty DR & Klee HJ (2006) Characterization of three members of the Arabidopsis carotenoid cleavage dioxygenase family demonstrates the divergent roles of this multifunctional enzyme family Plant J 45, 982–993 35 Romer S, Fraser PD, Kiano JW, Shipton CA, Misawa N, Schuch W & Bramley PM (2000) Elevation of the provitamin A content of transgenic tomato plants Nat Biotechnol 18, 666–669 36 Rosati C, Aquilani R, Dharmapuri S, Pallara P, Marusic C, Tavazza R, Bouvier F, Camara B & 746 37 38 39 40 41 42 43 44 45 46 47 48 Giuliano G (2000) Metabolic engineering of betacarotene and lycopene content in tomato fruit Plant J 24, 413–419 Ravanello MP, Ke D, Alvarez J, Huang B & Shewmaker CK (2003) Coordinate expression of multiple bacterial carotenoid genes in canola leading to altered carotenoid production Metab Eng 5, 255–263 Ye X, Al-Babili S, Kloti A, Zhang J, Lucca P, Beyer P ă & Potrykus I (2000) Engineering the provitamin A (beta-carotene) biosynthetic pathway into (carotenoidfree) rice endosperm Science 287, 303–305 Diretto G, Al-Babili S, Tavazza R, Papacchioli V, Beyer P & Giuliano G (2007) Metabolic engineering of potato carotenoid content through tuber-specific overexpression of a bacterial mini-pathway PLoS ONE 2, e350, doi: 10.1371/journal.pone.0000350 Scherzinger D & Al-Babili S (2008) In vitro characterization of a carotenoid cleavage dioxygenase from Nostoc sp PCC 7120 reveals a novel cleavage pattern, cytosolic localization and induction by highlight Mol Microbiol 69, 231–244 Prado-Cabrero A, Estrada AF, Al-Babili S & Avalos J (2007) Identification and biochemical characterization of a novel carotenoid oxygenase: elucidation of the cleavage step in the Fusarium carotenoid pathway Mol Microbiol 64, 448–460 Kloer DP, Ruch S, Al-Babili S, Beyer P & Schulz GE (2005) The structure of a retinal-forming carotenoid oxygenase Science 308, 267–269 Ruch S, Beyer P, Ernst H & Al-Babili S (2005) Retinal biosynthesis in Eubacteria: in vitro characterization of a novel carotenoid oxygenase from Synechocystis sp PCC 6803 Mol Microbiol 55, 1015–1024 Park MJ, Gwak KS, Yang I, Choi WS, Jo HJ, Chang JW, Jeung EB & Choi IG (2007) Antifungal activities of the essential oils in Syzygium aromaticum (L.) Merr Et Perry and Leptospermum petersonii Bailey and their constituents against various dermatophytes J Microbiol 45, 460–465 Lee SB, Cha KH, Kim SN, Altantsetseg S, Shatar S, Sarangerel O & Nho CW (2007) The antimicrobial activity of essential oil from Dracocephalum foetidum against pathogenic microorganisms J Microbiol 45, 53–57 Lewinsohn E, Sitrit Y, Bar E, Azulay Y, Meir A, Zamir D & Tadmor Y (2005) Carotenoid pigmentation affects the volatile composition of tomato and watermelon fruits, as revealed by comparative genetic analyses J Agric Food Chem 20, 3142–3148 Gang DR (2005) Evolution of flavors and scents Annu Rev Plant Biol 56, 301–325 Iijima Y, Gang D, Fridman E, Lewinsohn E & Pichersky E (2004) Characterization of geraniol synthase from the peltate glands of sweet basil Plant Physiol 134, 370–379 FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works A Ilg et al 49 Croteau R, Kutchan TM & Lewis NG (2000) Natural products (secondary metabolites) In Biochemistry and Molecular Biology of Plants (Buchanan B, Gruissem W & Jones R, eds), pp 1250–1318 American Society of Plant Physiologists, Rockville, MD ´ 50 Simkin AJ, Gaffe J, Alcaraz JP, Carde JP, Bramley PM, Fraser PD & Kuntz M (2007) Fibrillin influence on plastid ultrastructure and pigment content in tomato fruit Phytochemistry 68, 1545–1556 51 Barua AB & Olson JA (2000) b-Carotene is converted primarily to retinoids in rats in vivo J Nutr 130, 1996– 2001 A novel route for geranial formation Fig S1 Coomassie-stained SDS ⁄ PAGE gel of OsCCD1 purification fractions Table S1 Conversion rates of the substrates This supplementary material can be found in the online version of this article Please note: Wiley-Blackwell is not responsible for the content or functionality of any supplementary materials supplied by the authors Any queries (other than missing material) should be directed to the corresponding author for the article Supporting information The following supplementary material is available: FEBS Journal 276 (2009) 736–747 Journal compilation ª 2008 FEBS No claim to original German government works 747 ... F, Matusova R, Danoun S, Portais JC et al (2008) Strigolactone inhibition of shoot branching Nature 455, 18 9? ?19 4 11 Umehara M, Hanada A, Yoshida S, Akiyama K, Arite T, Takeda-Kamiya N, Magome... Discussion Plant CCD1s are known to catalyze the cleavage of the C9–C10 and the C9¢–C10¢ double bonds of several carotenoids Recently, it was shown that these enzymes can also cleave at the C5–C6 and... linear substrates is not unique to OsCCD1 Apart from the formation of geranial, the OsCCD1 cleavage reactions were identical to those of other plant CCD1s, as supported by the formation of pseudoionone,

Ngày đăng: 07/03/2014, 03:20

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan