Next Article in Journal
New Hydrazinothiazole Derivatives of Usnic Acid as Potent Tdp1 Inhibitors
Next Article in Special Issue
New Morphiceptin Peptidomimetic Incorporating (1S,2R,3S,4S,5R)-2-Amino-3,4,5-trihydroxycyclopen-tane-1-carboxylic acid: Synthesis and Structural Study
Previous Article in Journal
Black Chokeberry Aronia Melanocarpa L.—A Qualitative Composition, Phenolic Profile and Antioxidant Potential
Previous Article in Special Issue
Synthesis of a Di-Mycoloyl Tri-Arabinofuranosyl Glycerol Fragment of the Mycobacterial Cell Wall, Based on Synthetic Mycolic Acids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Glycosidase Inhibition of Broussonetine M and Its Analogues

1
Beijing National Laboratory for Molecular Science (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
2
University of Chinese Academy of Sciences, Beijing 100049, China
3
Department of Hospital Pharmacy, University of Toyama, 2630 Sugitani, Toyama 930–0194, Japan
4
Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX13TA, UK
5
National Engineering Research Center for Carbohydrate Synthesis, Jiangxi Normal University, Nanchang 330022, China
*
Authors to whom correspondence should be addressed.
Molecules 2019, 24(20), 3712; https://doi.org/10.3390/molecules24203712
Submission received: 17 September 2019 / Revised: 10 October 2019 / Accepted: 14 October 2019 / Published: 15 October 2019
(This article belongs to the Special Issue Carbohydrates in Synthesis)

Abstract

:
Cross-metathesis (CM) and Keck asymmetric allylation, which allows access to defined stereochemistry of a remote side chain hydroxyl group, are the key steps in a versatile synthesis of broussonetine M (3) from the d-arabinose-derived cyclic nitrone 14. By a similar strategy, ent-broussonetine M (ent-3) and six other stereoisomers have been synthesized, respectively, starting from l-arabino-nitrone (ent-14), l-lyxo-nitrone (ent-3-epi-14), and l-xylo-nitrone (2-epi-14) in five steps, in 26%–31% overall yield. The natural product broussonetine M (3) and 10’-epi-3 were potent inhibitors of β-glucosidase (IC50 = 6.3 μM and 0.8 μM, respectively) and β-galactosidase (IC50 = 2.3 μM and 0.2 μM, respectively); while their enantiomers, ent-3 and ent-10’-epi-3, were selective and potent inhibitors of rice α-glucosidase (IC50 = 1.2 μM and 1.3 μM, respectively) and rat intestinal maltase (IC50 = 0.29 μM and 18 μM, respectively). Both the configuration of the polyhydroxylated pyrrolidine ring and C-10’ hydroxyl on the alkyl side chain affect the specificity and potency of glycosidase inhibition.

Graphical Abstract

1. Introduction

In the past several decades, numerous polyhydroxylated pyrrolidine iminosugars have been isolated and synthesized, which has enriched the extended family of iminosugars [1,2,3,4,5,6,7]; these natural products, together with their synthetic analogues, have exhibited a variety of important biological activities and shown great potential as chemotherapeutic agents for an ever widening number of diseases [6,8]. DAB (1,4-dideoxy-1,4-imino-d-arabinitol, 1) [9] and DMDP (2,5-dihydroxymethyl-3,4-dihydroxypyrrolidine, 2) [10] are among the most widespread and studied iminosugars (Figure 1). DAB (1), isolated in 1985, is a powerful α-glucosidase inhibitor [11,12,13,14,15], whereas DMDP (2) is a potent inhibitor of bovine liver β-glucosidase and β-galactosidase (IC50 = 9.7 μM and 3.3 μM, respectively) [16]. DAB (1) was also found to be a potent inhibitor of glycogen phosphorylase, which made it a potential therapeutic agent for the treatment of diabetes [17]. In contrast, the enantiomers LAB (1,4-dideoxy-1,4-imino-l-arabinitol) (ent-1) [15] and l-DMDP (ent-2) are both potent and specific α-glucosidase inhibitors [16,18].
Many efforts have been devoted to the synthesis and glycosidase inhibition studies of DAB (1) [19,20] and various analogues [13,21,22,23,24,25,26,27,28]. Among these studies, the biological evaluation results of α-C-1-alkyl-DABs and their l-enantiomers [29] attracted our attention. While α-C-1-alkyl-DABs shifted to be β-glucosidase inhibitors with improved potency as the length of the C-1 alkyl chain increased, α-C-1-alkyl-LABs upheld similar α-glucosidase inhibitory activities to the parent compound. The above trend in changes of glycosidase inhibition reminded us of a special class of polyhydroxylated pyrrolidine alkaloids, broussonetines, which are a subgroup of natural iminosugars with more than 30 members (Figure 2). Broussonetines were isolated from the branches of the deciduous tree Broussonetia Kazinoki SIEB, which is widely distributed in several Asian countries and has been used as a folk medicine [30,31,32,33,34,35,36,37]. Most broussonetines share a common (2R, 3R, 4R, 5R)-pyrrolidine moiety with a 13-carbon chain containing various functional groups on the C-5 position. Therefore, they can be structurally regarded as DAB-related iminosugars or α-1-C-alkylated-DABs. However, with respect to glycosidase inhibitory activities, broussonetines are better to be considered as analogues of DMDP (2), of which most were found to be good β-glucosidase and β-galactosidase inhibitors. For example, broussonetine E (4), F (5), and G (6) show potent β-glycosidase inhibition and have therapeutic potential as antitumor and anti-HIV agents [38]. Broussonetine M (3), isolated in 2000 by Kusano’s group, was a potent inhibitor of β-galactosidase from bovine liver (IC50 = 8.1 μM) [36].
According to our previous study on the synthesis and glycosidase inhibition of broussonetines [39,40], enantiomers of the above broussonetines would probably demonstrate a similar inhibitory profile by analogy with LAB (ent-1) and l-DMDP (ent-2), and as such [22,26,41,42], these compounds may have potential in the treatment of type II diabetes, cancers, and viral infections [43,44,45,46]. Hence, in this work, broussonetine M (3) was selected as the research objective; synthesis and glycosidase inhibition of the natural product and its analogues, including l-enantiomers and pyrrolidine core stereoisomers, were finished, aiming for a better understanding of structure–activity relationship (SAR) of this interesting class of pyrrolidine iminosugars.
Though the first synthesis of broussonetine M (3) was accomplished with d-serine as the starting material [47], we have shown that most broussonetines can be efficiently constructed via a general synthetic strategy employing sugar-derived cyclic nitrones [39,40]. The pyrrolidine core of this class of iminosugars can be derived from cyclic sugar nitrones [44,48,49,50] with the corresponding stereochemistry in the hydroxylated pyrrolidine ring, while the various side chains could be installed via cross-metathesis (CM) reactions [46,47,51,52]. This general strategy is capable of synthesizing a number of natural broussonetines, as well as a variety of broussonetine analogues for SAR study; it has been successful in the synthesis of broussonetine I (7), J2 (10), and W (11) [39,40]. Therefore, this strategy was applied in the construction of broussonetine M (3) and its analogues.

2. Results and Discussion

2.1. Synthesis of Broussonetine M

Our retrosynthesis for broussonetine M (3) is presented in Scheme 1. The precursor 12 of broussonetine M was obtained by the CM reaction between the pyrrolidine 13 and the alcohol 15. The pyrrolidine 13 was conveniently prepared from d-arabinose-derived cyclic nitrone 14 [53,54,55,56,57]. The alcohol 15, which contains one stereocenter, was synthesized through asymmetric Keck allylation of aldehyde 16. In this synthetic route, only the stereocenter in alcohol 15 is constructed by virtue of an asymmetric reaction; of the four stereocenters on the pyrrolidine ring, three were determined by sugar-derived cyclic starting nitrone 14 and the fourth was formed by the high diastereoselectivity of organometallic addition to the nitrone 14.
For the synthesis of broussonetine M (3), addition of Grignard reagent 18, prepared from 8-bromo-1-octene, to d-arabino-nitrone (14) afforded the hydroxylamine 19 in high yield and excellent diastereoselectivity; none of the other diastereomers were formed (Scheme 2) [54]. Due to its chemical instability, the hydroxylamine 19 was directly used in the next step of reaction without further purification. Successive zinc reduction and N-Cbz protection of the crude hydroxylamine 19 provided the pyrrolidine 13, as the required CM reaction precursor, in 64% overall yield in three steps. The configuration of the newly formed C-5 chiral center was determined as R by nuclear Overhauser effects experiment on 19, with the observation of the strong correlation between H-5 and H-6a, H-6b (Scheme 2).
The key step in the synthesis of the chiral alkyl alcohol 15 was the asymmetric Keck allylation (Scheme 3) [58]. Treatment of butyl glycol 20 with BnBr/NaH/TBAI in DMF-THF gave the mono-O-benzylated alcohol 21 in 91% yield. Swern oxidation of alcohol 21 afforded aldehyde 16 (89%), which on enantioselective Keck allylation using (S)-BINOL produced alcohol 15 (93% ee) [59]. Using (R)-BINOL as the ligand, alcohol ent-15 was also prepared by the same method (See the Supplementary Materials for HPLC analysis of compounds 15 and ent-15).
CM reaction between pyrrolidine 13 and alcohol 15 completed the synthesis of broussonetine M (3). In this case, the free hydroxyl group of the alcohol 15 was tolerated for the CM reaction with no need for O-protection. CM reaction of pyrrolidine 13 and alcohol 15 promoted by Grubbs II catalyst produced olefin 12 as an inseparable Z/E mixture in moderate yield (43%). Pd/C-catalyzed hydrogenation of compound 12 in acidic methanol afforded the target product broussonetine M (3) in quantitative yield (Scheme 4). Thus, broussonetine M (3) was synthesized in five linear steps starting from d-arabino-nitrone (14) in 28% total yield. The 1H- and 13C-NMR spectra and the specific rotation of the synthetic broussonetine M (3) were all consistent with those reported for the synthetic broussonetine M (3) [47], but had some differences with those of natural products [36] (See the Supplementary Materials for comparison of NMR data). Since the structure and configuration of product 3 were ensured by all the synthetic materials and procedures, as in the work of Alberto Marco et al. [47], the NMR spectra differences may be explained by minute pH variation or metal impurities [60,61].

2.2. Synthesis of Analogues of Broussonetine M

Glycosidase inhibition by pyrrolidine iminosugars are among those that are most difficult to predict [16]; minor modification of the iminosugar can lead to a distinct change of the inhibition profile. In order to explore the preliminary structure–activity relationship of this type of iminosugar, seven analogues of broussonetine M (3), including its l-enantiomer and other six stereoisomers, were then prepared. By the same strategy as that for pyrrolidine 13, synthesis of ent-13, ent-3-epi-13, and 2-epi-13 were accomplished from the corresponding sugar-derived cyclic nitrones ent-14, ent-3-epi-14, and 2-epi-14. The configurations of the newly constructed chiral centers in these compounds were all confirmed by NOE experiments. With these pyrrolidines in hand, CM reaction with alcohol 15, and subsequent hydrogenation, provided the target products, i.e., ent-10’-epi-3, ent-3,10’-di-epi-3, and 2-epi-3, of which, the C10’-hydroxyls retained S configuration as that of the natural product (Table 1).
In order to evaluate the influence of the C10’-hydroxyl on glycosidase inhibition, the C10’-epimers of broussonetine M (3), including 10’-epi-3, ent-3, ent-3-epi-3, and 2,10’-di-epi-3, were also synthesized from the above four pyrrolidines and alcohol ent-15 by the same strategy.

2.3. Glycosidase Inhibition

The synthetic broussonetine M (3) and its analogues were all assayed as potential glycosidase inhibitors of a range of enzymes, as shown in Table 2 and Table 3.
According to the results, all the compounds showed moderate to potent inhibition of β-galactosidase from bovine liver; 10’-epi-3 was the most potent inhibitor (IC50 = 0.2 μM). The natural product broussonetine M (3) and 10’-epi-3 showed potent inhibition toward β-glucosidase (IC50 = 6.3 μM and 0.8 μM, respectively) and β-galactosidase (IC50 = 2.3 μM and 0.2 μM, respectively) from bovine liver, which were consistent with DMDP (2) and other DAB or DMDP derivatives with a long alkyl side chain on C-1 position [29]. However, different from DMDP (2), compounds 3 and 10’-epi-3 lost their inhibitions against α-glucosidase, α-mannosidase, and trehalase, whereas they showed moderate β-glucuronidase inhibition. Furthermore, 10’-epi-3 with C-10’ having R configuration was about 4 times better as an inhibitor than broussonetine M (3) with C-10’ having S configuration.
Ent-broussonetine M (ent-3) and ent-10’-epi-3 exhibited potent and more selective inhibition of α-glucosidase from rice (IC50 = 1.2 μM and 1.3 μM) and rat intestinal maltase (IC50 = 0.29 μM and 18 μM), analogous to the inhibition by l-DMDP [16,18], ent-broussonetine I, J2 [39], and other LAB or l-DMDP derivatives with a long alkyl chain on the C1 position [29,62]. Ent-3-epi-3 and ent-3,10-di-epi-3 showed moderate and broad inhibition against α-glucosidase, β-glucosidase, β-galactosidase, and α-l-fucosidase, while l-altro-DMDP was a selective moderate α-glycosidase inhibitor. d-gluco-DMDP exhibited moderate inhibition against α-glucosidase. For d-gluco-DMDP-related broussonetine M analogs, compounds 2-epi-3 and 2,10-di-epi-3 both showed moderate inhibition against β-glucosidase, β-galactosidase, and β-glucuronidase; 2,10’-di-epi-3 was a potent inhibitor of α-glucosidase, while 2-epi-3 did not show such inhibition. The results indicate that the correct configuration at C-10’ is essential for α-glucosidase inhibition.
Therefore, configurations of both the pyrrolidine ring and C-10’ have significant influences on glycosidase inhibition of broussonetine M and its analogs. In detail, comparison of the inhibitory profiles of ent-broussonetine M (ent-3) and ent-10’-epi-3 with those of broussonetine M (3) and 10’-epi-3 showed high similarity to those of DAB and LAB derivatives (or DMDP and l-DMDP derivatives), of which we have previously reported opposite inhibitions toward α- and β-glycosidases for enantiomers [16,18,42]. In contrast, inhibitory activities of other broussonetine analogues with l-altro-DMDP and d-gluco-DMDP pyrrolidine cores are more difficult to predict, but the presence of the 13-carbon chains basically narrowed down the inhibitory profiles.
The structure–activity relationship uncovered in this work would be helpful in the research and development of new glycosidase inhibitors.

3. Materials and Methods

3.1. General Methods

NMR spectra was recorded at 300 MHz, 400 MHz, or 500 MHz (1H-NMR) and 75 MHz, 100 MHz, or 125 MHz (13C-NMR) in CDCl3 (with TMS as internal standard), C5D5N, or CD3OD (with solvent signal as internal standard). High-resolution mass spectra (HRMS) were performed on a LTQ/FT linear ion trap mass spectrometer. All reagents were used as received without any further purification or prepared, as described in the literature. CH2Cl2 was freshly distilled from CaH2. Tetrahydrofuran was distilled from sodium benzophenone. TLC plates were visualized by ultraviolet light or by treatment with a spray of Pancaldi reagent ((NH4)6MoO4, Ce(SO4)2, H2SO4, H2O) or a 0.5% solution of KMnO4 in acetone. Column chromatography was performed on a flash column chromatography with silica gel (200–300 mesh). Polarimetry was determined using an Optical Activity AA-10R polarimeter with concentrations (c) given in grams per 100 mL. IR data were measured as films on KBr plates and are given only when relevant functions are present. Chiral HPLC analyses were performed on an Agilen 1100 Series using a Daicel Chiralpak (OD-H) column with hexanes/i-PrOH as the eluent.

3.2. Material and Methods for the Enzyme Inhibition Assay

With rat intestinal maltase as an exception, other enzymes were purchased from Sigma-Aldrich Chemical Co. (St. Louis, Mo. USA). Brush border membranes prepared from rat small intestine according to the method of Kessler et al. [63] were assayed at pH 6.8 for rat intestinal maltase using maltose. The released d-glucose was determined colorimetrically using the Glucose CII-test Wako (Wako Pure Chemical Ind.; Osaka, Japan). Other glycosidase activities were determined using an appropriate p-nitrophenyl glycoside as substrate in a buffer solution at the optimal pH value of each enzyme. The reaction was stopped by adding 400 mM Na2CO3. The released p-nitrophenol was measured spectrometrically at 400 nm [16].

3.3. Chemistry

3.3.1. Synthesis of 4-(benzyloxy)butan-1-ol (21)

NaH (60%, 11.52 g, 0.48 mol) was added slowly to a solution of butane-1,4-diol 20 (36.05 g, 0.4 mol) in dry THF (300 mL) at 0 °C. After stirring at room temperature for 0.5 h, tetrabutylammonium iodide (0.74 g, 8.0 mmol) was added, followed by a dropwise solution of benzyl bromide (68.41 g, 0.4 mol) in THF while the temperature was kept between 45 °C and 50 °C. The reaction mixture was stirred for 2 h and then quenched by aq. NH4Cl solution (20 mL). Water (200 mL) was added and the organic layer separated; the aqueous layer was extracted with EtOAc (200 mL × 3). The combined organic phases were dried over MgSO4, filtered, and concentrated in vacuo. The crude product was purified by flash column chromatography (silica gel, petroleum ether/EtOAc = 3/1) to give 4-(benzyloxy)butan-1-ol 21 (light yellow oil, 69.9 g, 97% yield). 1H-NMR (300 MHz, CDCl3) δ 7.35–7.28 (m, 1H), 4.52 (s, 2H), 3.68–3.63 (d, J = 5.8 Hz, 2H), 3.54–3.50 (d, J = 5.9 Hz, 2H), 1.81–1.60 (m, 4H); 13C-NMR (75 MHz, CDCl3) δ 138.2, 128.4, 127.7, 127.7, 73.1, 70.4, 62.7, 30.1, 26.7.

3.3.2. Synthesis of 4-(benzyloxy)butanal (16)

A solution of DMSO (8.74 mL, 0.26 mol) in dry CH2Cl2 (20 mL) was added dropwise to a solution of (COCl)2 (24.57 mL, 0.29 mol) in dry CH2Cl2 (100 mL) at –78 °C. The mixture was stirred for 5 min. A solution of 4-(benzyloxy)butan-1-ol 21 (43.3 g, 0.24 mol) in dry CH2Cl2 (50 mL) was then added dropwise while the temperature was kept below −65 °C. After 15 min, NEt3 (166.94 mL, 1.2 mol) was added dropwise. After stirring for 10 min at −78 °C, the reaction mixture was allowed to warm to room temperature and diluted with CH2Cl2 (200 mL). The organic layer was washed with brine (2 × 100 mL). The combined organic extracts were dried over MgSO4, filtered, and concentrated under reduced pressure. Purification by flash chromatography on silica gel (petroleum ether/EtOAc = 10/1) afforded 4-(benzyloxy)butanal 16 (39.8 g, 93% yield) as a yellow oil. 1H-NMR (300 MHz, CDCl3) δ 9.77 (t, J = 1.6 Hz, 1H), 7.35–7.30 (m, 5H), 4.48 (s, 2H), 3.50 (t, J = 5.9 Hz, 2H), 2.54 (dt, J = 7.2, 1.6 Hz, 1H), 1.98–1.91 (m, 2H); 13C-NMR (75 MHz, CDCl3) δ 202.4, 138.3, 128.4, 127.7, 73.0, 69.1, 41.0, 22.6.

3.3.3. General Procedure for Synthesis of (R)-7-(benzyloxy)hept-1-en-4-ol (15) and (S)-7-(benzyloxy)hept-1-en-4-ol (ent-15), with Alcohol 15 as an Example

Under Ar atmosphere, (S)-BINOL (52 mg, 0.2 mmol) and Ti(OiPr)4 (45 mg, 0.2 mmol) were added to a solution of dried 4 Å molecular sieves (2.2 g) in CH2Cl2 (20 mL). The reaction mixture was heated at reflux for 1 h, and then allowed to cool to room temperature. A solution of aldehyde 16 (356 mg, 2 mmol) in CH2Cl2 (15 mL) was added to the reaction mixture. After stirring for 0.5 h at room temperature, the solution was cooled to −78 °C and allyltributyltin (993 mg, 3 mmol) was added dropwise. The reaction mixture was stirred for an additional 20 min at −78 °C, then kept at −20 °C. After 12 h, the reaction mixture was filtered through a pad of celite into a 200 mL flask that contained a stirred sat. aq. NaHCO3 solution (50 mL); the resulting reaction mixture was stirred for 1 h. Then, the layers were separated and the aqueous layer was extracted with CH2Cl2 (3 × 50 mL). The combined organic extracts were dried with MgSO4; subsequent removal of all volatiles under reduced pressure and column chromatography of the residue on silica gel (petroleum ether/EtOAc = 20/1) afforded homoallylic alcohol 15 (729.7 mg, 83% yield) as colorless oil.
Data for 15: [α]D 20 +4.14 (c 3.85 in CH2Cl2); HPLC analysis: 92.6% ee [Daicel CHIRALPAK OD-H column, 20 °C, 220 nm, hexane/i-PrOH = 95:5, 1 mL/min, 20.8 min (major), 23.9 min (minor)]; νmax/cm−1: 3401 (s), 3068 (w), 2924 (s), 1679 (w), 1453 (m), 1096 (s), 1021 (m), 697 (m); 1H-NMR (300 MHz, CDCl3) δ 7.39–7.23 (m, 5H), 5.93–5.75 (m, 1H), 5.15–5.10 (m, 1H), 5.08 (t, J = 1.2 Hz, 1H), 4.50 (s, 2H), 3.64 (tt, J = 8.1, 4.5 Hz, 1H), 3.50 (t, J = 6.0 Hz, 2H), 2.52 (br, 1H), 2.26–2.15 (m, 2H), 1.77–1.60 (m, 3H), 1.54–1.45 (m, 1H); 13C-NMR (75 MHz, CDCl3) δ 138.3, 135.1, 128.4, 127.7, 127.7, 117.7, 73.0, 70.6, 70.5, 42.0, 34.0, 26.2; HRMS(ESI) calcd for C14H21O2+ [M + H]+ 243.13555, found 243.13564.
Data for ent-15: colorless oil; yield: 86%; [α]D 20 +4.08 (c 4.45 in CH2Cl2); HPLC analysis: 92.8% ee [Daicel CHIRALPAK OD-H column, 20 °C, 220 nm, hexane/i-PrOH = 95:5, 1 mL/min, 21.3 min (minor), 23.1 min (major)]; νmax/cm−1: 3401 (s), 3068 (w), 2924 (s), 1679 (w), 1453 (m), 1096 (s), 1021 (m), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.38–7.31 (m, 4H), 7.31–7.26 (m, 1H), 5.90–5.76 (m, 1H), 5.15–5.12 (m, 1H), 5.10 (s, 1H), 4.50 (s, 2H), 3.7–3.61 (m, 1H), 3.51 (t, J = 6.0 Hz, 2H), 2.36 (d, J = 3.2 Hz, 1H), 2.32–2.24 (m, 1H), 2.23–2.14 (m, 1H), 1.82–1.70 (m, 2H), 1.70–1.60 (m, 1H), 1.50 (m, 1H); 13C-NMR (100 MHz, CDCl3) δ 138.3, 135.1, 128.4, 127.7, 127.7, 117.7, 73.0, 70.6, 70.5, 42.0, 34.0, 26.2; HRMS(ESI) calcd for C14H21O2Na+ [M + Na]+ 243.13555, found 243.13544.

3.3.4. General Procedure for Synthesis of Compounds 19, ent-19, ent-3-epi-19, and 2-epi-19, with 19 as an Example

Part of the solution of 8-bromo-1-octene (573.3 mg, 3.0 mmol) in THF (2 mL) was quickly added via syringe to a stirred solution of Mg (1.16 g, 5.0 mmol) and I2 (cat.) in THF (5 mL) under Ar atmosphere. The mixture was heated until the color disappeared; then, the remaining 8-bromo-1-octene was added dropwise. After the addition was completed, the resulting reaction mixture was heated to reflux for 1 h and then was allowed to cool to room temperature. The prepared Grignard reagent was added slowly to a solution of d-arabino-nitrone (14) (417.5 mg, 1.0 mmol) in THF (10 mL) via syringe at 0 °C under Ar atmosphere. The reaction mixture was stirred for 0.5 h; then sat. aq. NH4Cl was added to quench the reaction. The organic layer was separated and the aqueous layer was extracted with EtOAc (3 × 20 mL). The combined organic phases were dried over MgSO4 and filtered; the solvent was removed under reduced pressure to give the crude product hydroxylamine 19, which was used without further purification because of its instability. The sample for structure characterization was purified by flash column chromatography on silica gel (petroleum ether/EtOAc = 5/1) as a colorless syrup.
Data for (2R,3R,4R,5R)-3,4-bis(benzyloxy)-2- ((benzyloxy)methyl)-1-hydroxyl-5-(oct-7-en-1-yl)pyrrolidine (19): [α]D 20 -8.6 (c 1.2 in CH2Cl2); νmax/cm−1: 3030 (w), 2926 (s), 2855 (s), 1454 (m), 1362 (w), 1097 (s), 735 (m), 697 (s); 1H-NMR (400 MHz, CDCl3) δ 7.32–7.24 (m, 15H), 5.80 (ddt, J = 16.9, 10.2, 6.6 Hz, 1H), 5.01–4.91 (m, 2H), 4.56–4.42 (m, 6H), 3.95–3.92 (m, 1H), 3.80–3.76 (m, 2H), 3.58 (dd, J = 9.2, 6.9 Hz, 1H), 3.54–3.50 (m, 1H), 3.17 (dt, J = 7.5, 5.4 Hz, 1H), 2.04–1.99 (m, 2H), 1.88–1.83 (m, 1H), 1.50–1.43 (m, 1H), 1.42–1.28 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 139.3, 138.3, 138.2, 138.2, 128.5, 128.5, 128.1, 128.0, 127.8, 127.8, 127.7, 114.3, 86.8, 84.7, 73.5, 71.8, 71.8, 70.2, 70.1, 68.4, 33.9, 29.8, 29.2, 29.0, 26.7; HRMS(ESI) calcd for C34H44O4N+ [M + H]+ 530.32649, found 530.32565.
Data for (2S,3S,4S,5S)-3,4-bis(benzyloxy)-2-((benzyloxy)methyl)-1-hydroxyl-5-(oct-7-en-1-yl) pyrrolidine (ent-19): colorless syrup; [α]D 20 + 7.8 (c 2.8 in CH2Cl2); νmax/cm−1: 3030 (w), 2926 (s), 2856 (s), 1454 (m), 1362 (w), 1099 (s), 735 (m), 697 (s); 1H-NMR (400 MHz, CDCl3) δ 7.31–7.25 (m, 15H), 6.60 (br, 1H), 5.80 (ddt, J = 16.9, 10.2, 6.7 Hz, 1H), 5.00–4.91 (m, 2H), 4.56–4.42 (m, 6H), 3.94 (t, J = 3.2 Hz, 1H), 3.80–3.77 (m, 2H), 3.60–3,56 (dd, J = 9.2, 6.9 Hz, 1H), 3.54–3.50 (m, 1H), 3.16 (dt, J = 7.2, 5.4 Hz, 1H), 2.02 (q, J = 6.9 Hz, 2H), 1.89–1.84 (m, 1H), 1.59–1.43 (m, 1H), 1.37–1.22 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 139.3, 138.3, 138.2, 138.2, 128.5, 128.4, 128.4, 128.1, 128.0, 127.8, 127.8, 127.7, 114.3, 86.8, 84.7, 73.4, 71.8, 71.7, 70.2, 70.0, 68.4, 33.9, 29.8, 29.2, 29.0, 26.6; HRMS(ESI) calcd for C34H44O4N+ [M + H]+ 530.32649, found 530.32550.
Data for (2S,3R,4S,5S)-3,4-bis(benzyloxy)-2-((benzyloxy)methyl)-1-hydroxyl--5-(oct-7-en-1-yl) pyrrolidine (ent-3-epi-19): colorless syrup; [α]D 20 −24.5 (c 4.5 in CH2Cl2); νmax/cm−1: 3030 (w), 2926 (s), 2855 (s), 1454 (m), 1363 (w), 1098 (br), 734 (m), 697 (s); 1H-NMR (400 MHz, CDCl3) δ 7.30–7.24 (m, 15H), 6.64 (br, 1H), 5.84–5.74 (ddt, J = 16.9, 10.2, 6.7 Hz, 1H), 5.03–4.88 (m, 2H), 4.68–4.43 (m, 6H), 4.19 (t, J = 5.1 Hz, 1H), 3.86–3,76 (m, 2H), 3.68 (dd, J = 6.8, 5.3 Hz, 1H), 3.56–3.51 (m, 1H), 3.29–3.24 (m, 1H), 2.04–1.99 (m, 2H), 1.75–1.69 (m, 1H), 1.49–1.28 (m, 9H); 13C-NMR (100 MHz, CDCl3) δ 139.2, 138.5, 138.4, 138.2, 128.6, 128.4, 128.4, 128.2, 127.9, 127.9, 127.8, 127.8, 127.7, 127.7, 127.0, 114.3, 82.9, 76.8, 73.6, 73.5, 72.7, 70.2, 69.3, 67.4, 33.9, 30.1, 29.8, 29.2, 29.0, 26.9; HRMS(ESI) calcd for C34H44O4N+ [M + H]+ 530.32649, found 530.32573.
Data for (2S,3R,4R,5R)-3,4-bis(benzyloxy)-2-((benzyloxy)methyl)-1-hydroxyl-5-(oct-7-en-1-yl) pyrrolidine (2-epi-19): colorless syrup; [α]D 20 + 22.8 (c 2.5 in CH2Cl2); νmax/cm−1: 3290 (br), 3028 (w), 2925 (s), 2856 (s), 1453 (m), 1353 (w), 1112 (s), 729 (s), 693 (s); 1H-NMR (400 MHz, CDCl3) δ 7.36–7.23 (m, 15H), 5.85–5.75 (ddt, J = 16.9, 10.2, 6.7 Hz, 1H), 5.32 (s, 1H), 5.01–4.91 (m, 2H), 4.57–4.50 (m, 4H), 4.42–4.32 (m, 2H), 3.94–3.87 (m, 2H), 3,81–3.78 (m, 1H), 3,53–3.52 (m, 1H), 3.36–3.31 (dt, J = 8.0, 5.3 Hz, 1H), 2.85–2.80 (dt, J = 8.0, 5.3 Hz, 1H), 2.05–2.00 (dd, J = 7.1, 6.6 Hz, 1H), 1.81–1.75 (m, 1H), 1.57–1.50 (m, 1H), 1.42–1.28 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 139.3, 138.3, 138.2, 138.0, 128.5, 128.0, 127.9, 127.9, 127.8, 127.8, 114.3, 85.4, 79.8, 73.6, 73.1, 72.3, 71.6, 69.7, 68.0, 33.9, 32.8, 29.8, 29.2, 29.0, 26.2; HRMS(ESI) calcd for C34H44O4N+ [M + H]+ 530.32649, found 530.32577.

3.3.5. General Procedure for Synthesis of Compounds 13, ent-13, 3-epi-ent-13, and 2-epi-13, with 13 as an Example

Zinc powder (653.8 mg, 10 mmol) was added to a suspension of Cu(OAc)2 (18.2 mg, 0.1 mmol) in AcOH (10 mL) and the reaction mixture was stirred for 0.5 h. Then, a solution of the crude hydroxylamine 19 in AcOH (5 mL) was added and the reaction mixture was stirred for 10 h. The solid was removed by filtration and all volatiles were removed under reduced pressure. The residue was dissolved in EtOAc (20 mL) and sat. aq. NaHCO3 was added to neutralize the solution. The resulting precipitate was removed by filtration and the organic layers were collected; the aqueous layer was extracted with EtOAc (3 × 10 mL). The combined organic layer was dried over MgSO4, filtered, and concentrated under reduced pressure to give the crude amine, which was used in the next step without further purification. NaHCO3 (252.0 mg, 3.0 mmol) and CbzCl (255.9 mg, 1.5 mmol) were added slowly to a stirred solution of the crude amine in methanol (10 mL) and the reaction mixture was stirred at room temperature for 6 h. Then sat. aq. NaHCO3 (20 mL) was added to quench the reaction and EtOAc (20 mL) was added. The organic layer was separated and the aqueous layer was extracted by EtOAc (3 × 10 mL). The combined organic layers were dried over MgSO4, filtered, and the solvent was removed under reduced pressure. Purification by flash chromatography on silica gel (petroleum ether/EtOAc = 15/1) afforded the carbamate 13 as light yellow syrup (423 mg, yield: 64% for 3 steps).
Data for (2R, 3R, 4R, 5R)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-2-((benzyloxy)methyl)-5-(oct-7-en-1-yl)pyrrolidine (13): [α]D 20 −36.4 (c 2.25 in CH2Cl2); νmax/cm−1: 2926 (m), 2855 (w), 1700 (vs), 1407 (m), 1347 (w), 1093 (m), 696 (m); 1H-NMR (400 MHz, CDCl3) δ 7.38–7.23 (m, 17H), 7.23–7.15 (m, 3H), 5.86–5.72 (m, 1H), 5.22–5.15 (m, 1H), 5.05 (s, 0.5H), 5.02–5.01 (m, 1H), 4.96–4.92 (m, 1.5H), 4.65–4.57 (m, 1.5H), 4.47–4.32 (m, 4.5H), 4.27–4.23 (m, 0.5H),4.15–4.12 (m, 1.5H), 4.07–4.03 (m, 0.5H), 3.86–3.71 (m, 2.5H), 3.50–3.44 (m, 1H), 2.09–1.94 (m, 2.5H), 1.78– 1.48 (m, 1.5H), 1.29 (m, 4.5H), 1.16 (m, 3.5H); 13C-NMR (100 MHz, CDCl3) δ 153.6, 153.2, 138.1, 138.1, 137.5, 137.3, 136.9, 136.9, 136.7, 136.6, 135.6, 135.6, 127.5, 127.4, 127.3, 127.3, 127.1, 126.99, 126.95, 126.69, 126.65, 126.60, 126.57, 126.52, 126.46, 113.2, 113.1, 83.4, 82.3, 82.1, 81.0, 72.0, 71.9, 70.1, 70.0, 69.8, 67.7, 66.8, 65.8, 65.7, 64.0, 63.6, 61.8, 61.5, 32.7, 32.7, 30.4, 29.1, 28.2, 28.0, 27.8, 27.8, 27.7, 25.5, 25.5; HRMS(ESI) calcd for C42H49O5NNa+ [M + Na]+ 670.35029, found 670.34943.
Data for (2S,3S,4S,5S)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-2-((benzyloxy)methyl)-5-(oct-7-en- 1-yl)pyrrolidine (ent-13): light yellow syrup; yield for 3 steps: 65%; [α]D 20 + 35.0 (c 6.95 in CH2Cl2); νmax/cm−1: 3031 (w), 2926 (m), 1701 (vs), 1408 (m), 1095 (vs), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.37–7.14 (m, 20H), 5.80–5.73 (m, 1H), 5.22–5.14 (m, 1H), 5.02–4.92 (m, 3H), 4.62–4.52 (m, 1.5H), 4.44–4.29 (m, 5H), 4.18 (d, J = 8.4 Hz, 1H), 4.11–4.08 (m, 1H), 3.93–3.72 (m, 2.5H), 3.51–3.46 (m, 1H), 2.18–1.84 (m, 2.5H), 1.81–1.48 (m, 1.5H), 1.41–1.05 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 154.8, 154.4, 139.2, 139.2, 138.8, 138.5, 138.2, 138.1, 137.9, 137.9, 136.9, 136.8, 128.7, 128.62, 128.59, 128.54, 128.51, 128.33, 128.25, 128.2, 128.1, 127.94, 127.88, 127.82, 127.76, 127.7, 127.6, 114.54, 114.49, 84.6, 83.5, 83.3, 82.1, 73.2, 73.1, 71.3, 71.2, 71.0, 69.0, 68.0, 67.0, 66.9, 65.3, 64.9, 63.2, 62.8, 34.0, 33.9, 31.6, 30.3, 29.5, 29.2, 29.1, 28.99, 28.95, 26.69, 26.65; HRMS(ESI) calcd for C42H49O5NNa+ [M + Na]+ 670.35029, found 670.34998.
Data for (2S,3R,4S,5S)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-2-((benzyloxy)methyl)-5-(oct-7-en- 1-yl)pyrrolidine (ent-3-epi-13): light yellow syrup; yield for 3 steps: 64%; [α]D 20 + 5.1 (c 3.45 in CH2Cl2); νmax/cm−1: 3030 (w), 2926 (m), 1701 (vs), 1406 (m), 1094 (s), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.35–7.19 (m, 20H), 5.82–5.75 (m, 1H), 5.23–4.92 (m, 4H), 4.82–4.51 (m, 5H), 4.40–4.26 (m, 3H), 4.13 (br, 1H), 3.87–3.67 (m, 2.5H), 3.60 (m, 0.5H), 2.08–1.86 (m, 2.5H), 1.64 (s, 0.5H), 1.41–0.92 (m, 9H); 13C-NMR (100 MHz, CDCl3) δ 154.7, 154.6, 139.2, 138.8, 138.5, 138.4, 136.8, 128.6, 128.5, 128.3, 128.2, 128.1, 128.0, 127.82, 127.75, 127.6, 127.5, 127.4, 127.3, 114.5, 81.1, 79.9, 78.1, 77.7, 77.4, 77.1, 73.1, 73.00, 72.8, 72.3, 72.3, 72.0, 70.3, 69.2, 67.0, 66.9, 62.8, 62.40, 58.0, 57.9, 33.9, 33.3, 32.0, 29.4, 29.1, 29.0, 28.9, 26.7, 26.5; HRMS(ESI) calcd for C42H49O5NNa+ [M + Na]+ 670.35029, found 670.35004.
Data for (2S,3R,4R,5R)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-2-((benzyloxy)methyl)-5-(oct-7- en-1-yl)pyrrolidine (2-epi-13): light yellow syrup; yield for 3 steps: 71%; [α]D 20 −4.2 (c 1.95 in CH2Cl2); νmax/cm−1: 3031 (w), 2927 (m), 1702 (vs), 1406 (m), 1095 (s), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.35–7.17 (m, 20H), 5.83–5.73 (m, 1H), 5.10 (s, 2H), 5.00–4.91 (m, 2H), 4.61–4.59 (m, 2H), 4.62–4.33 (m, 5H), 4.15–4.12 (m, 1H), 3.92–3.90 (m, 1H), 3.80–3.65 (m, 3H), 2.00–1.98 (m, 2H), 1.78 (m, 1H), 1.62–1.54 (m, 1H), 1.31–1.105 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 155.8, 139.3, 138.6, 138.2, 138.1, 136.9, 128.6, 128.5, 128.4, 128.0, 127.9, 127.83, 127.79, 127.7, 127.6, 114.3, 84.9, 82.5, 73.4, 73.0, 71.7, 68.2, 67.0, 62.3, 59.1, 33.9, 33.5, 29.5, 29.1, 29.0, 26.0; HRMS(ESI) calcd for C42H49O5NNa+ [M + Na]+ 670.35029, found 670.34973.

3.3.6. General Procedure for Synthesis of Compounds 12, 10’-epi-12, ent-10’-epi-12, ent-12, ent-3,10’-di-epi-12, ent-3-epi-12, 2-epi-12, and 2,10’-di-epi-12, with 12 as an Example

Grubbs II catalyst (6.8 mg, 0.008 mmol) was added to a solution of 13 (50 mg, 0.077 mmol) and 15 (25.4 mg, 0.116 mmol) in dry CH2Cl2 and the resulting reaction mixture was heated to reflux for 4 h. Then the solvent was removed under reduced pressure and purification of the residue by flash chromatography on silica gel (petroleum ether/EtOAc = 5/1) afforded 12 (Z/E mixture) as a yellow syrup (28 mg, 43% yield).
Data for (2R,3R,4R,5R)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5- ((R)-13-(benzyloxy)-10-hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (12): [α]D 20 −20.0 (c 1.50 in CH2Cl2); νmax/cm−1: 3079 (w), 2925 (s), 2854 (m), 1699 (s), 1409 (m), 1094 (s), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.45–7.13 (m, 25H), 5.49–5.39 (m, 2H), 5.22–5.15 (dd, J = 14.8, 12.6 Hz, 1H), 5.05–5.02 (d, J = 12.3 Hz, 1H), 4.65–4.57 (m, 1.5H), 4.50–4.32 (m, 6.5H), 4.2–4.23 (dd, J = 10.5, 4.0 Hz, 0.5H), 4.18–4.12 (m, 1.5H), 4.07–4.03 (dd, J = 8.7, 4.1 Hz, 0.5H), 3.85–3.71 (m, 1.5H), 3.62–3.58 (d, J = 3.8 Hz, 1H), 3.52–3.44 (m, 3H), 2.26–1.87 (m, 5H), 1.79–1.40 (m, 5H), 1.38–1.14 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 153.6, 153.2, 137.5, 137.3, 136.93, 136.88, 136.63, 135.58, 133.32, 133.25, 131.9, 127.5, 127.4, 127.32, 127.28, 127.1, 126.98, 126.95, 126.64, 126.56, 126.5, 125.0, 83.4, 82.3, 82.1, 81.0, 76.3, 76.0, 75.7, 72.0, 71.9, 71.7, 70.1, 70.0, 69.8, 69.5, 69.4, 67.7, 66.8, 65.6, 64.0, 63.6, 61.9, 61.6, 39.8, 34.4, 32.8, 31.6, 31.1, 30.4, 29.1, 28.7, 28.4, 28.2, 28.1, 28.0, 26.4, 25.5, 25.2, 25.0; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46423.
Data for (2R,3R,4R,5R)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5-((S)-13-(benzyloxy)-10- hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (10’-epi-12): yellow syrup; yield: 43%; [α]D 20 −20.0 (c 1.50 in CH2Cl2); νmax/cm−1: 3080 (w), 2927 (s), 2855 (m), 1699 (s), 1409 (m), 1095 (s), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.33–7.18(m, 25H), 5.54–5.38 (m, 2H), 5.22–5.15(m, 1H), 5.05–5.02 (m, 1H), 4.65–4.57 (m, 1.5H), 4.51–4.32 (m, 6.5H), 4.26–4.22 (m, 0.5H), 4.16–4.11 (m, 1.5H), 4.07–4.03 (m, 0.5H), 3.86–3.71 (m, 2.5H), 3.62–3.58 (m, 1H), 3.53–3.44 (m, 3H), 2.24–1.86 (m, 5H), 1.74–1.61(m, 4H), 1.52–1.38 (m, 1H), 1.36–1.16 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 154.7, 154.3, 138.61, 138.36, 138.03, 137.98, 137.76, 136.67, 134.30, 134.23, 132.98, 130.02, 128.6, 128.49, 128.46, 128.4, 128.2, 128.10, 128.06, 127.8, 127.74, 127.69, 127.65, 127.6, 126.1, 84.5, 83.4, 83.2, 82.0, 73.1, 73.0, 72.7, 71.2, 71.1, 70.9, 70.6, 70.5, 68.8, 67.9, 66.91, 66.87, 65.1, 64.8, 63.0, 62.7, 40.9, 33.9, 32.7, 32.6, 31.5, 30.2, 29.5, 29.2, 29.1, 27.5, 26.6, 26.3; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46411.
Data for (2S,3S,4S,5S)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5-((R)-13-(benzyloxy)-10- hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (ent-10’-epi-12): yellow syrup; yield: 41%; [α]D 20 + 28.6 (c 1.85 in CH2Cl2); νmax/cm−1: 3470 (m), 3031 (w), 2925 (s), 1699 (vs), 1409 (m), 1096 (vs), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.35–7.17 (m, 25H), 5.54–5.34 (m, 2H), 5.22–5.15 (m, 1H),5.05–5.02 (m, 1H), 4.65–4.56 (m, 1.5H), 4.52–4.32 (m, 6.5H), 4.26–4.22 (dd, J = 4.0 Hz, 0.5H), 4.15–4.10 (m, 1.5H), 4.06–4.03 (dd, J = 8.7, 4.2 Hz, 0.5H), 3.87–3.72 (m, 2.5H), 3.64–3.56 (m, 1H), 3.52–3.44 (m, 3H), 2.24–1.91 (m, 5H), 1.78–1.58 (m, 4H), 1.52–1.48 (m, 1H), 1.35–1.11 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 154.7, 154.3, 138.6, 138.4, 138.0, 138.0, 137.7, 137.6, 136.7, 134.3, 134.2, 129.7, 129.6, 128.6, 128.50, 128.47, 128.4, 128.2, 128.10, 128.07, 127.74, 127.65, 127.6, 127.5, 126.1, 84.5, 83.4, 83.2, 82.0, 73.1, 73.0, 71.2, 71.1, 70.9, 70.6, 70.5, 68.8, 67.8, 66.9, 65.1, 64.8, 63.0, 62.7, 40.9, 33.9, 32.7, 31.5, 30.2, 29.5, 29.2, 29.1, 26.6, 26.3, 26.1; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46407.
Data for (2S,3S,4S,5S)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5-((S)-13-(benzyloxy)-10- hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (ent-12): yellow syrup; yield: 45%; [α]D 20 + 27.8 (c 1.39 in CH2Cl2); νmax/cm−1: 3462 (m), 3031 (w), 2925 (s), 1699 (vs), 1409 (m), 1096 (vs), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.41–7.16 (m, 25H), 5.54–5.33 (m, 2H), 5.22–5.14 (m, 1H), 5.04–5.01 (m, 1H), 4.65–4.55 (m, 1.5H), 4.51–4.32 (m, 6.5H), 4.27–4.23 (m, 0.5H), 4.15–4.12 (m, 1.5H), 4.06–4.03 (dd, J = 8.2, 3.9 Hz, 0.5H), 3.85–3.69 (m, 2.5H), 3.59–3.54(m, 1H), 3.51–3.47 (m, 3H), 2.22–1.90 (m, 5H), 1.77–1.52 (m, 5H), 1.38–1.12 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 154.7, 154.3, 138.6, 138.3, 138.00, 137.95, 137.7, 136.6, 134.34, 134.28, 128.5, 128.44, 128.41, 128.37, 128.2, 128.1, 127.73, 127.69, 127.65, 127.6, 126.1, 84.5, 83.4, 83.2, 82.03, 77.5, 77.2, 76.8, 73.1, 73.0, 71.2, 71.1, 70.9, 70.54, 70.45, 68.79, 67.84, 66.9, 65.1, 64.7, 62.973.0, 62.6, 40.8, 35.4, 33.9, 32.7, 32.2, 31.5, 30.1, 29.4, 29.3, 29.2, 29.1, 27.5, 26.6, 26.3, 26.0; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46418.
Data for (2S,3R,4S,5S)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5-((R)-13-(benzyloxy)-10- hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (ent-3,10’-di-epi-12): yellow syrup; yield: 40%; [α]D 20 + 5.1 (c 1.05 in CH2Cl2); νmax/cm−1: 3461 (m), 3030 (w), 2926 (s), 1700 (vs), 1408 (m), 1096 (vs), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.38–7.17 (m, 25H), 5.53–5.38 (s, 2H), 5.25–5.00 (m, 2H), 4.85–4.45 (m, 7H), 4.39–4.22 (m, 3H), 4.12–4.17 (m, 1H), 3.81–3.74 (m, 2H), 3.71–3.68 (m, 0.5H), 3.65–3.57 (m, 1.5H), 3.52–3.46 (t, J = 6.1 Hz, 2H), 2.26–1.88 (m, 5H), 1.79–1.46 (m, 5H), 1.32–1.01 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 154.7, 154.5, 138.7, 138.4, 136.7, 134.2, 128.5, 128.5, 128.4, 128.2, 128.0, 127.72, 127.65, 127.5, 127.4, 126.30, 126.2, 81.1, 79.9, 78., 77.5, 77.2, 76.9, 73.0, 72.7, 72.5, 72.2, 71.9, 70.9, 70.6, 69.1, 66.9, 62.7, 62.3, 57.9, 40.9, 37.3, 33.9, 33.3, 32.7, 31.9, 29.4, 29.1, 26.6, 26.3; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46412.
Data for (2S,3R,4S,5S)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5-((S)-13-(benzyloxy)-10- hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (ent-3-epi-12): yellow syrup; yield: 41%; [α]D 20+ 5.9 (c 3.3 in CH2Cl2); νmax/cm−1: 3461 (m), 3030 (w), 2926 (s), 1701 (vs), 1408 (m), 1096 (vs), 697 (m); 1H-NMR (500 MHz, CDCl3) δ 7.38–7.17 (m, 25H), 5.53–5.36 (m, 2H), 5.24–5.01 (m, 2H), 4.82–4.75 (m, 1H), 4.71–4.46 (m, 6H), 4.42–4.34 (m, 1H), 4.31–4.23 (m, 2H), 4.15–4.14 (d, J = 3.7 Hz, 1H), 3.81–3.77 (m, 2H), 3.70–3.68 (d, J = 8.1 Hz, 0.5H), 3.60–3.57 (m, 1.5H), 3.52–3.46 (m, 2H), 2.22–1.89 (m, 5H), 1.78–1.58 (m, 5H), 1.34–1.01 (m, 8H); 13C-NMR (125 MHz, CDCl3) δ 154.7, 154.5, 1390, 138.7, 138.4, 138.3, 136.7, 136.6, 134.24, 134.17, 132.9, 129.6, 128.50, 128.45, 128.4, 128.2, 128.1, 127.99, 127.95, 127.73, 127.65, 127.5, 127.4, 127.3, 127.2, 126.3, 126.1, 81.0, 79.8, 78.0, 77.6, 77.4, 77.2, 76.9, 73.0, 72.9, 72.7, 72.4, 72.2, 71.9, 71.3, 70.9, 70.5, 70.5, 69.1, 66.9, 62.6, 62.3, 57.9, 57.8, 40.8, 35.5, 34.1, 33.9, 33.3, 32.8, 32.7, 32.6, 32.2, 31.8, 29.4, 29.3, 29.1, 29.0, 27.4, 26.6, 26.5, 26.4, 26.3, 26.0; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46437.
Data for (2S,3R,4R,5R)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5-((R)-13-(benzyloxy)-10- hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (2-epi-12): yellow syrup; yield: 43%; [α]D 20 −3.7 (c 0.88 in CH2Cl2); νmax/cm−1: 3461 (m), 3030 (w), 2926 (s), 1700 (vs), 1409 (m), 1097 (vs), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.34–7.24 (m, 25H), 5.52–5.38 (m, 2H), 5.11 (s, 2H), 4.64–4.58 (m, 2H), 4.55–4.46 (m, 6H), 4.36–4.32 (m, 1H), 4.15–4.12 (dd, J = 6.8, 4.6 Hz, 1H), 3.91–3.89 (t, J = 4.0 Hz, 1H), 3.85–3.66 (m, 3H), 3.61–3.57 (m, 1H), 3.52–3.46 (t, J = 6.1 Hz, 2H), 2.25–1.92 (m, 4H), 1.81–1.60 (m, 4H), 1.52–1.42 (m, 1H), 1.35–1.12 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 155.6, 138.5, 138.4, 138.2, 138.0, 136.8, 134.4, 132.9, 128.5, 128.48, 128.45, 128.4, 128.3, 128.0, 127.8, 127.8, 127.7, 127.6, 127.5, 126.0, 89.6, 82.5, 80.7, 73.4, 73.0, 73.0, 72.7, 71.6, 71.3, 70.9, 70.6, 70.5, 67.0, 62.3, 59.0, 40.9, 35.5, 34.1, 33.9, 32.7, 29.5, 29.4, 29.1, 27.5, 26.4, 26.3, 26.0, 25.9; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46422.
Data for (2S,3R,4R,5R)-3,4-bis(benzyloxy)-1-benzyloxycarbonyl-5-((S)-13-(benzyloxy)-10- hydroxydec-7-en-1-yl)-2-((benzyloxy)methyl)pyrrolidine (2,10’-di-epi-12): yellow syrup; yield: 43%; [α]D 20 −3.8 (c 1.43 in CH2Cl2); νmax/cm−1: 3470 (m), 3030 (w), 2926 (s), 1700 (vs), 1409 (m), 1097 (vs), 697 (m); 1H-NMR (400 MHz, CDCl3) δ 7.36–7.24 (m, 25H), 5.52–5.34 (m, 2H), 5.13 (s, 2H), 4.65–4.48 (m, 8H), 4.36–4.32 (m, 1H), 4.15–4.12 (dd, J = 6.8, 4.7 Hz, 1H), 3.92–3.90 (t, J = 3.9 Hz, 1H), 3.83 –3.67 (m, 3H), 3.52–3.45 (m, 2H), 2.26–1.85 (m, 5H), 1.77–1.41 (m, 5H), 1.35–1.13 (m, 8H); 13C-NMR (100 MHz, CDCl3) δ 153.0, 138.51, 138.45, 138.2, 138.1, 136.8, 134.3, 130.0, 128.53, 128.51, 128.47, 128.46, 128.4, 128.0, 1279, 127.79, 127.76, 127.69, 127.66, 127.5, 126.1, 83.9, 79.9, 73.4, 73.1, 73.0, 72.9, 71.7, 71.3, 71.1, 70.9, 70.64, 70.57, 69.4, 68.1, 67.0, 62.3, 59.1, 40.9, 37.4, 34.8, 34.7, 33.9, 32.8, 29.5, 29.4, 29.2, 26.4, 26.3, 25.9, 25.8; HRMS(ESI) calcd for C54H65O7NNa+ [M + Na]+ 862.46532, found 862.46448.

3.3.7. General Procedure for Synthesis of Compounds 3, 10’-epi-3, ent-10’-epi-3, ent-3, ent-3,10’-di-epi-3, ent-3-epi-3, 2-epi-3, 2, and 10’-di-epi-3, with Broussonetine M (3) as an Axample

Pd/C (10 wt%) was added to a stirred solution of 9 (43 mg, 0.055 mmol) and 3 N HCl (1 mL) in MeOH (10 mL) under Ar atmosphere and the reaction mixture was stirred under H2 atmosphere for 12 h. Then, the catalyst was filtered and the solvent was removed under reduced pressure. Purification of the residue by flash chromatography on silica gel (CHCl3/MeOH/NH3·H2O (2 N) = 90/9/1) afforded (2R,3R,4R,5R)-2-hydroxymethyl-3,4-dihydroxyl-5-((10R)-10,13-(dihydroxyl)dec- 1-yl)pyrrolidine (3) as white solid (14.4 mg, 0.055 mmol, quantative yield). Data for broussonetine M (3): [α]D 20 + 4.0 (c 0.7 in CH3OH) [lit. [36] [α]D 20 + 5.9 (c 0.3 in CH3OH)]; νmax/cm−1: 3104 (vs), 2926 (s), 2857 (w),1402 (vs); 1H-NMR (500 MHz, C5D5N) δ 6.41 (br, 7.15–5.54, 4H), 4.92 (t, J = 6.7 Hz, 1H), 4.67 (t, J = 7.1 Hz, 1H), 4.49–4.42 (m, 2H), 4.31–4.28 (m, 1H), 4.09–4.05 (m, 1H), 3.96 (t, J = 6.4 Hz, 2H), 3.92–3.87 (m 1H), 2.34–2.29 (m, 2H), 2.15–2.09 (m, 1H), 2.03–1.97 (m, 1H), 1.90–1.79 (m, 3H), 1.74–1.56 (m, 4H), 1.48–1.45 (m, 1H), 1.33–1.07 (m, 10H); 13C-NMR (125 MHz, C5D5N) δ 80.3, 76.2, 70.8, 65.0, 62.7, 62.3, 59.2, 38.3, 35.1, 31.5, 30.2, 29.9, 29.7, 29.5, 29.4, 29.3, 26.6, 26.1; 1H-NMR (400 MHz, CD3OD) δ 4.00–3.97 (t, J = 6.1 Hz, 1H), 3.90–3.80 (m, 3H), 3.58–3.48 (m, 4H), 3.34 (m, 1H), 1.98–1.28 (m, 22H); 13C-NMR (100 MHz, CD3OD) δ 80.3, 76.6, 72.3, 65.5, 64.3, 63.1, 59.5, 38.4, 34.7, 32.0, 30.8, 30.7, 30.6, 30.39, 30.35, 29.9, 27.2, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27432.
Data for (2R,3R,4R,5R)-2-hydroxymethyl-3,4-dihydroxyl-5-((10S)-10,13-(dihydroxyl)dec-1-yl) pyrrolidine (10’-epi-3): white solid, quantative yield; [α]D 20 + 4.0 (c 0.4 in CH3OH); νmax/cm−1: 3104 (vs), 2927 (s), 2861 (w), 1402 (vs); 1H-NMR (400 MHz, CD3OD) δ 3.99–3.96 (t, J = 5.7 Hz, 1H), 3.85 (m, 3H), 3.62–3.51 (m, 3H), 3.49–3.45 (m, 1H), 3.37–3.35 (m, 1H), 1.96–1.84 (m, 1H), 1.80–1.64 (m, 2H), 1.63–1.28 (m, 19H); 13C-NMR (100 MHz, CD3OD) δ 80.3, 76.6, 72.3, 65.5, 64.3, 63.2, 59.6, 38.4, 34.8, 32.2, 30.8, 30.7, 30.6, 30.4, 29.9, 27.2, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27411.
Data for (2S,3S,4S,5S)-2-hydroxymethyl-3,4-dihydroxyl-5-((10R)-10,13-(dihydroxyl)dec-1-yl) pyrrolidine (ent-10’-epi-3): white solid, quantative yield; [α]D 20 −16.8 (c 1.5 in CH3OH); νmax/cm−1: 3134 (vs), 2928 (s), 2854 (w), 1402 (vs); 1H-NMR (400 MHz, CD3OD) δ 4.01–3.98 (t, J = 6.2 Hz, 1H), 3.91–3.80 (m, 3H), 3.59–3.47 (m, 4H), 3.36–3.33 (m, 1H), 1.95–1.30 (m, 22H); 13C-NMR (100 MHz, CD3OD) δ 80.3, 76.6, 72.3, 65.5, 64.3, 63.1, 59.5, 38.4, 34.8, 32.0, 30.8, 30.7, 30.6, 30.39, 30.35, 29.9, 27.2, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27430.
Data for (2S,3S,4S,5S)-2-hydroxymethyl-3,4-dihydroxyl-5-((10S)-10,13-(dihydroxyl)dec-1-yl) pyrrolidine (ent-3): white solid, quantative yield; [α]D 20 −20.4 (c 1.55 in CH3OH); νmax/cm−1: 3134 (vs), 2928 (s), 2854 (w), 1403 (s); 1H-NMR (400 MHz, CD3OD) δ 4.01–3.98 (t, J = 6.1 Hz, 1H), 3.90–3.80 (m, 3H), 3.58–3.45 (m, 4H), 3.36–3.33 (m, 1H), 1.94–1.29 (m, 22H); 13C-NMR (100 MHz, CD3OD) δ 80.3, 76.6, 72.3, 65.5, 64.3, 63.1, 59.5, 38.4, 34.8, 32.0, 30.8, 30.7, 30.6, 30.39, 30.35, 29.9, 27.2, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27416.
Data for (2S,3R,4S,5S)-2-hydroxymethyl-3,4-dihydroxyl-5-((10R)-10,13-(dihydroxyl)dec-1-yl) pyrrolidine (ent-3,10’-di-epi-3): white solid, quantative yield; [α]D 20 −19.4 (c 0.5 in CH3OH); νmax/cm−1: 3124 (vs), 2926 (s), 2853 (w), 1402 (s); 1H-NMR (500 MHz, CD3OD) δ 4.16 (s, 1H), 3.97–3.92 (m, 2H), 3.90–3.85 (m, 1H), 3.70–3.68 (dd, J = 8.0, 3.9 Hz, 1H), 3.59–3.54 (m, 3H), 3.46–3.41 (td, J = 9.1, 5.2 Hz, 1H), 1.89–1.29 (m, 22H); 13C-NMR (125 MHz, CD3OD) δ 77.4, 72.3, 71.7, 63.5, 63.1, 62.0, 59.4, 38.5, 34.8, 32.0, 30.8, 30.7, 30.6, 30.46, 30.38, 29.9, 27.6, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27424.
Data for (2S,3R,4S,5S)-2-hydroxymethyl-3,4-dihydroxyl-5-((10S)-10,13-(dihydroxyl)dec-1-yl) pyrrolidine (ent-3-epi-3): white solid, quantative yield; [α]D 20 −33.4 (c 0.65 in CH3OH); νmax/cm−1: 3364 (vs), 2925 (vs), 2852 (m), 1402 (s); 1H-NMR (400 MHz, CD3OD) δ 4.16 (t, J = 3.4 Hz, 1H), 3.97–3.85 (m, 3H), 3.68–3.63 (m, 1H), 3.56 (t, J = 6.4 Hz, 2H), 3.53 (m, 1H), 3.42 (td, J = 9.2, 5.2 Hz, 1H), 1.90–1.28 (m, 22H); 13C-NMR (100 MHz, CD3OD) δ 77.4, 72.3, 71.7, 63.5, 63.1, 62.0, 59.4, 38.5, 34.8, 32.08, 30.8, 30.7, 30.6, 30.5, 30.4, 29.9, 27.6, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27427.
Data for (2S,3R,4R,5R)-2-hydroxymethyl-3,4-dihydroxyl-5-((10R)-10,13-(dihydroxyl)dec-1-yl) pyrrolidine (2-epi-3): white solid, quantative yield; [α]D 20 + 3.1 (c 0.45 in CH3OH); νmax/cm−1: 3124 (s), 2923 (s), 2852 (w), 1402 (s); 1H-NMR (500 MHz, CD3OD) δ 4.07 (s, 1H), 3.98–3.89 (m, 3H), 3.72–3.68 (m, 1H), 3.56 (t, J = 6.3 Hz, 2H), 3.54–3.52 (m, 1H), 3.30–3.26 (m, 1H), 1.88–1.29 (m, 22H); 13C-NMR (125 MHz, CD3OD) δ 80.8, 76.5, 72.3, 68.7, 65.6, 63.1, 58.6, 38.5, 34.8, 33.1, 30.8, 30.7, 30.6, 30.5, 30.3, 29.9, 27.8, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27439.
Data for (2S,3R,4R,5R)-2-hydroxymethyl-3,4-dihydroxyl-5-((10S)-10,13-(dihydroxyl)dec-1-yl) pyrrolidine (2,10’-di-epi-3): white solid, quantative yield; [α]D 20 + 0.12 (c 1.2 in CH3OH); νmax/cm−1: 3127 (s), 2923 (s), 2853 (m), 1403 (s); 1H-NMR (400 MHz, CD3OD) δ 4.08 (d, J = 2.0 Hz, 1H), 3.99–3.88 (m, 3H), 3.71 (m, 1H), 3.60–3.50 (m, 3H), 3.31–3.27 (m, 1H), 1.93–1.76 (m, 2H), 1.71–1.28 (m, 20H); 13C-NMR (100 MHz, CD3OD) δ 80.8, 76.5, 72.3, 68.7, 65.5, 63.1, 58.6, 38.4, 34.8, 33.1, 30.8, 30.7, 30.6, 30.4, 30.3, 29.9, 27.8, 26.8; HRMS(ESI) calcd for C18H38O5N+ [M + H]+ 348.27445, found 348.27429.

4. Conclusions

In summary, a general and versatile synthetic strategy has been developed for the synthesis of broussonetine M (3), ent-broussonetine M (ent-3), and six other stereoisomers with d-arabino-nitrone (14), l-arabino-nitrone (ent-14), l-lyxo-nitrone (ent-3-epi-14), and l-xylo-nitrone (2-epi-14) as the starting materials in 26%–31% total yield for five linear steps. Glycosidase inhibition assays on a range of enzymes showed that natural product broussonetine M (3) and 10’-epi-3 showed potent inhibition of β-glucosidase from bovine liver, while ent-3 and ent-10’-epi-3 were potent and selective inhibitors of rice α-glucosidase and rat intestinal maltase. It was also found that the configuration at C-3 was essential for α-glucosidase inhibition. This work has further explored the spectrum of glycosidase inhibition by DAB- and LAB-related iminosugars and will be helpful for the future design of potent and selective glycosidase inhibitors.

Supplementary Materials

The following are available online: copies of 1H-NMR, 13C-NMR, and 2D NMR spectra, and HPLC analysis methods.

Author Contributions

Q.-K.W. performed the chemical syntheses, analyzed the data, and wrote the original draft; K.K. performed the glycosidase inhibition assay; Y.-X.L. and Y.-M.J. validated all the experimental data and revised the manuscript; A.K., G.W.J.F., and C.-Y.Y. designed the experiments and reviewed the manuscript. All authors approved the final manuscript.

Funding

Financial support from the National Natural Science Foundation of China (No. 21272240 and No. 21772206) and National Engineering Research Center for Carbohydrate Synthesis of Jiangxi Normal University is gratefully acknowledged. This work was supported in part by an Emeritus Leverhulme Research Fellowship (G.W.J.F.) and a Grant-in-Aid for Scientific Research (C) from the Japanese Society for the Promotion of Science (JSPS KAKENHI Grant Number JP17K08362) (A.K.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Compain, P.; Martin, O.R. Iminosugars: From Synthesis to Therapeutic Applications, 1st ed.; Wiley-VCH: Chichester, UK, 2007; pp. 328–446. [Google Scholar]
  2. Wrodnigg, T.M. From Lianas to Glycobiology Tools: Twenty-Five Years of 2,5-dideoxy-2,5-imino-d-mannitol. Mon. Chem. 2002, 133, 393–426. [Google Scholar] [CrossRef]
  3. Asano, N.; Nash, R.J.; Molyneux, R.J.; Fleet, G.W.J. Sugar-mimic glycosidase inhibitors: Natural occurrence, biological activity and prospects for therapeutic application. Tetrahedron Asymmetry 2000, 11, 1645–1680. [Google Scholar] [CrossRef]
  4. Watson, A.A.; Fleet, G.W.J.; Asano, N.; Molyneux, R.J.; Nash, R.J. Polyhydroxylated alkaloids—Natural occurrence and therapeutic applications. Phytochemistry 2001, 56, 265–295. [Google Scholar] [CrossRef]
  5. Horne, G.; Wilson, F.X.; Tinsley, J.; Williams, D.H.; Storer, R. Iminosugars past, present and future: Medicines for tomorrow. Drug Discov. Today 2010, 16, 107–118. [Google Scholar] [CrossRef]
  6. Nash, R.J.; Kato, A.; Yu, C.-Y.; Fleet, G.W.J. Iminosugars as Therapeutic Agents: Recent Advances and Promising Trends. Future Med. Chem. 2011, 3, 1513–1521. [Google Scholar]
  7. Davis, B.G. A silver-lined anniversary of Fleet iminosugars: 1984–2009, from DIM to DRAM to LABNAc. Tetrahedron Asymmetry 2009, 20, 652–671. [Google Scholar] [CrossRef]
  8. Butters, T.D.; Dwek, R.A.; Platt, F.M. Inhibition of Glycosphingolipid Biosynthesis:  Application to Lysosomal Storage Disorders. Chem. Rev. 2000, 100, 4683–4696. [Google Scholar] [CrossRef]
  9. Nash, R.J.; Bell, E.A.; Williams, J.M. 2-Hydroxymethyl-3,4-dihydroxypyrrolidine in fruits of angylocalyx boutiqueanus. Phytochemistry 1985, 24, 1620–1622. [Google Scholar] [CrossRef]
  10. Welter, A.; Jadot, J.; Dardenne, G.; Marlier, M.; Casimir, J. 2,5-Dihydroxymethyl 3,4-dihydroxypyrrolidine dans les feuilles de Derris elliptica. Phytochemistry 1976, 15, 747–749. [Google Scholar] [CrossRef]
  11. Jones, D.W.C.; Nash, R.J.; Bell, E.A.; Williams, J.M. Identification of the 2-hydroxymethyl-3,4-dihydroxypyrrolidine (or 1,4-dideoxy-1,4-iminopentitol) from angylocalyx boutiqueanus and from arachniodes standishii as the (2R, 3R, 4S)-isomer by the synthesis of its enantiomer. Tetrahedron Lett. 1985, 26, 3125–3126. [Google Scholar] [CrossRef]
  12. Fleet, G.W.J.; Nicholas, S.J.; Smith, P.W.; Evans, S.V.; Fellows, L.E.; Nash, R.J. Potent competitive inhibition of α-galactosidase and α-glucosidase activity by 1,4-dideoxy-1,4-iminopentitols: Syntheses of 1,4-dideoxy-1,4-imino-d-lyxitol and of both enantiomers of 1,4-dideoxy-1,4-iminoarabinitol. Tetrahedron Lett. 1985, 26, 3127–3130. [Google Scholar] [CrossRef]
  13. Asano, N.; Kizu, H.; Oseki, K.; Tomioka, E.; Matsui, K.; Okamoto, M.; Baba, M. N-Alkylated Nitrogen-in-the-Ring Sugars: Conformational Basis of Inhibition of Glycosidases and HIV-1 Replication. J. Med. Chem. 1995, 38, 2349–2356. [Google Scholar] [CrossRef] [PubMed]
  14. Merino, P.; Delso, I.; Tejero, T.; Cardona, F.; Marradi, M.; Faggi, E.; Parmeggiani, C.; Goti, A. Nucleophilic additions to cyclic nitrones en route to iminocyclitols - Total syntheses of DMDP, 6-deoxy-DMDP, DAB-1, CYB-3, nectrisine, and radicamine B. Eur. J. Org. Chem. 2008, 2929–2947. [Google Scholar] [CrossRef]
  15. Cruz, F.P.d.; Newberry, S.; Jenkinson, S.F.; Wormald, M.R.; Butters, T.D.; Alonzi, D.S.; Nakagawa, S.; Becq, F.; Norez, C.; Nash, R.J.; et al. 4-C-Me-DAB and 4-C-Me-LAB—enantiomeric alkyl-branched pyrrolidine iminosugars—are specific and potent α-glucosidase inhibitors; acetone as the sole protecting group. Tetrahedron Lett. 2011, 52, 219–223. [Google Scholar] [CrossRef] [PubMed]
  16. Ayers, B.J.; Ngo, N.; Jenkinson, S.F.; Martínez, R.F.; Shimada, Y.; Adachi, I.; Weymouth-Wilson, A.C.; Kato, A.; Fleet, G.W.J. Glycosidase Inhibition by All 10 Stereoisomeric 2,5-Dideoxy-2,5-iminohexitols Prepared from the Enantiomers of Glucuronolactone. J. Org. Chem. 2012, 77, 7777–7792. [Google Scholar] [CrossRef] [PubMed]
  17. Fosgerau, K.; Westergaard, N.; Quistorff, B.; Grunnet, N.; Kristiansen, M.; Lundgren, K. Kinetic and Functional Characterization of 1,4-Dideoxy-1,4-imino-d-arabinitol: A Potent Inhibitor of Glycogen Phosphorylase with Anti-hyperglyceamic Effect in ob/ob Mice. Arch. Biochem. Biophys. 2000, 380, 274–284. [Google Scholar] [CrossRef]
  18. Yu, C.-Y.; Asano, N.; Ikeda, K.; Wang, M.-X.; Butters, T.D.; Wormald, M.R.; Dwek, R.A.; Winters, A.L.; Nash, R.J.; Fleet, G.W.J. Looking glass inhibitors: l-DMDP, a more potent and specific inhibitor of alpha-glucosidases than the enantiomeric natural product DMDP. Chem. Commun. 2004, 1936–1937. [Google Scholar] [CrossRef]
  19. Wang, G.-N.; Yang, L.; Zhang, L.-H.; Ye, X.-S. A Versatile Approach to N-Alkylated 1,4-Dideoxy-1,4-imino-d-arabinitols and 1,4-Dideoxy-1,4-imino-l-xylitols. J. Org. Chem. 2011, 76, 2001–2009. [Google Scholar] [CrossRef]
  20. Bouillon, M.E.; Pyne, S.G. Diastereoselective concise syntheses of the polyhydroxylated alkaloids DMDP and DAB. Tetrahedron Lett. 2014, 55, 475–478. [Google Scholar] [CrossRef]
  21. Li, Y.-X.; Kinami, K.; Hirokami, Y.; Kato, A.; Su, J.-K.; Jia, Y.-M.; Fleet, G.W.J.; Yu, C.-Y. Gem-difluoromethylated and trifluoromethylated derivatives of DMDP-related iminosugars: Synthesis and glycosidase inhibition. Org. Biomol. Chem. 2016, 14, 2249–2263. [Google Scholar] [CrossRef]
  22. Jenkinson, S.F.; Best, D.; Saville, A.W.; Mui, J.; Martínez, R.F.; Nakagawa, S.; Kunimatsu, T.; Alonzi, D.S.; Butters, T.D.; Norez, C.; et al. C-Branched Iminosugars: α-Glucosidase Inhibition by Enantiomers of isoDMDP, isoDGDP, and isoDAB–l-isoDMDP Compared to Miglitol and Miglustat. J. Org. Chem. 2013, 78, 7380–7397. [Google Scholar] [CrossRef] [PubMed]
  23. Best, D.; Jenkinson, S.F.; Saville, A.W.; Alonzi, D.S.; Wormald, M.R.; Butters, T.D.; Norez, C.; Becq, F.; Blériot, Y.; Adachi, I.; et al. Cystic fibrosis and diabetes: isoLAB and isoDAB, enantiomeric carbon-branched pyrrolidine iminosugars. Tetrahedron Lett. 2010, 51, 4170–4174. [Google Scholar] [CrossRef]
  24. Matassini, C.; Vanni, C.; Goti, A.; Morrone, A.; Marradi, M.; Cardona, F. Multimerization of DAB-1 onto Au GNPs affords new potent and selective N-acetylgalactosamine-6-sulfatase (GALNS) inhibitors. Org. Biomol. Chem. 2018, 16, 8604–8612. [Google Scholar] [CrossRef] [PubMed]
  25. D’Adamio, G.; Forcella, M.; Fusi, P.; Parenti, P.; Matassini, C.; Ferhati, X.; Vanni, C.; Cardona, F. Probing the Influence of Linker Length and Flexibility in the Design and Synthesis of New Trehalase Inhibitors. Molecules 2018, 23, 436. [Google Scholar] [CrossRef] [PubMed]
  26. Liu, Z.; Ma, S. Recent Advances in Synthetic -Glucosidase Inhibitors. Chemmedchem 2017, 12, 819–829. [Google Scholar] [CrossRef]
  27. Li, Y.-X.; Shimada, Y.; Adachi, I.; Kato, A.; Jia, Y.-M.; Fleet, G.W.J.; Xiao, M.; Yu, C.-Y. Fluorinated and Conformationally Fixed Derivatives of l-HomoDMDP: Synthesis and Glycosidase Inhibition. J. Org. Chem. 2015, 80, 5151–5158. [Google Scholar] [CrossRef]
  28. Ghavami, A.; Johnston, B.D.; Jensen, M.T.; Svensson, B.; Pinto, B.M. Synthesis of Nitrogen Analogues of Salacinol and Their Evaluation as Glycosidase Inhibitors. J. Am. Chem. Soc. 2001, 123, 6268–6271. [Google Scholar] [CrossRef]
  29. Kato, A.; Nakagome, I.; Sato, K.; Yamamoto, A.; Adachi, I.; Nash, R.J.; Fleet, G.W.J.; Natori, Y.; Watanabe, Y.; Imahori, T.; et al. Docking study and biological evaluation of pyrrolidine-based iminosugars as pharmacological chaperones for Gaucher disease. Org. Biomol. Chem. 2016, 14, 1039–1048. [Google Scholar] [CrossRef]
  30. Shibano, M.; Kitagawa, S.; Kusano, G. Studies on the constituents of Broussonetia species. 1. Two new pyrrolidine alkaloids, broussonetines C and D, as beta-galactosidase and beta-mannosidase inhibitors from Broussonetia kazinoki SIEB. Chem. Pharm. Bull. 1997, 45, 505–508. [Google Scholar] [CrossRef]
  31. Shibano, M.; Kitagawa, S.; Nakamura, S.; Akazawa, N.; Kusano, G. Studies on the Constituents of Broussonetia Species. II. Six New Pyrrolidine Alkaloids, Broussonetine A, B, E, F and Broussonetinine A and B, as Inhibitors of Glycosidases from Broussonetia kazinoki SIEB. Chem. Pharm. Bull. 1997, 45, 700–705. [Google Scholar] [CrossRef]
  32. Shibano, M.; Nakamura, S.; Akazawa, N.; Kusano, G. Studies on the Constituents of Broussonetia Species. III. Two New Pyrrolidine Alkaloids, Broussonetines G and H, as Inhibitors of Glycosidase, from Broussonetia kazinoki SIEB. Chem. Pharm. Bull. 1998, 46, 1048–1050. [Google Scholar] [CrossRef] [PubMed]
  33. Shibano, M.; Nakamura, S.; Kubori, M.; Minoura, K.; Kusano, G. Studies on the constituents of Broussonetia species. IV. Two new pyrrolidinyl piperidine alkaloids, broussonetines I and J, from Broussonetia kazinoki SIEB. Chem. Pharm. Bull. 1998, 46, 1416–1420. [Google Scholar] [CrossRef]
  34. Shibano, M.; Nakamura, S.; Motoya, N.; Kusano, G. Studies on the constituents of Broussonetia species. V. Two new pyrrolidine alkaloids, broussonetines K and L, as inhibitors of glycosidase, from Broussonetia kazinoki SIEB. Chem. Pharm. Bull. 1999, 47, 472–476. [Google Scholar] [CrossRef]
  35. Shibano, M.; Tsukamoto, D.; Kusano, G. A new pyrrolizidine alkaloid, Broussonetine N, as an inhibitor of glycosidase, from Broussonetia kazinoki Sieb. and absolute stereostructures of Broussonetines A and B. Chem. Pharm. Bull. 1999, 47, 907–908. [Google Scholar] [CrossRef]
  36. Shibano, M.; Tsukamoto, D.; Fujimoto, R.; Masui, Y.; Sugimoto, H.; Kusano, G. Studies on the Constituents of Broussonetia Species. VII. Four New Pyrrolidine Alkaloids, Broussonetines M, O, P, and Q, as Inhibitors of Glycosidase, from Broussonetia kazinoki SIEB. Chem. Pharm. Bull. 2000, 48, 1281–1285. [Google Scholar] [CrossRef]
  37. Tsukamoto, D.; Shibano, M.; Okamoto, R.; Kusano, G. Studies on the constituents of Broussonetia species VIII. Four new pyrrolidine alkaloids, broussonetines R, S, T, and V and a new pyrroline alkaloid, broussonetine U, from Broussonetia kazinoki Sieb. Chem. Pharm. Bull. 2001, 49, 492–496. [Google Scholar] [CrossRef]
  38. Shibano, M.; Tsukamoto, D.; Kusano, G. Polyhydroxylated Alkaloids with Lipophilic Moieties as Glycosidase Inhibitors from Higher Plants. Heterocycles 2002, 57, 1539–1553. [Google Scholar]
  39. Zhao, H.; Kato, A.; Sato, K.; Jia, Y.-M.; Yu, C.-Y. Total synthesis and glycosidase inhibition of broussonetine I and J2. J. Org. Chem. 2013, 78, 7896–7902. [Google Scholar] [CrossRef]
  40. Song, Y.-Y.; Kinami, K.; Kato, A.; Jia, Y.-M.; Li, Y.-X.; Fleet, G.W.J.; Yu, C.-Y. First total synthesis of (+)-broussonetine W: Glycosidase inhibition of natural product & analogs. Org. Biomol. Chem. 2016, 14, 5157–5174. [Google Scholar]
  41. D’Alonzo, D.; Guaragna, A.; Palumbo, G. Glycomimetics at the mirror: Medicinal chemistry of l-iminosugars. Curr. Med. Chem. 2009, 16, 473–505. [Google Scholar] [CrossRef]
  42. Best, D.; Wang, C.; Weymouth-Wilson, A.C.; Clarkson, R.A.; Wilson, F.X.; Nash, R.J.; Miyauchi, S.; Kato, A.; Fleet, G.W.J. Looking glass inhibitors: Scalable syntheses of DNJ, DMDP, and (3R)-3-hydroxy- l-bulgecinine from d-glucuronolactone and of l-DNJ, l-DMDP, and (3S)-3-hydroxy-d-bulgecinine from l-glucuronolactone. DMDP inhibits β-glucosidases and β-galactosidases whereas l-DMDP is a potent and specific inhibitor of α-glucosidases. Tetrahedron Asymmetry 2010, 21, 311–319. [Google Scholar]
  43. Mohan, S.; Eskandari, R.; Pinto, B.M. Naturally Occurring Sulfonium-Ion Glucosidase Inhibitors and Their Derivatives: A Promising Class of Potential Antidiabetic Agents. Acc. Chem. Res. 2014, 47, 211–225. [Google Scholar] [CrossRef] [PubMed]
  44. Boisson, J.; Thomasset, A.; Racine, E.; Cividino, P.; Sainte-Luce, T.B.; Poisson, J.-F.; Behr, J.-B.; Py, S. Hydroxymethyl-Branched Polyhydroxylated Indolizidines: Novel Selective alpha-Glucosidase Inhibitors. Org. Lett. 2015, 17, 3662–3665. [Google Scholar] [CrossRef] [PubMed]
  45. Kato, A.; Zhang, Z.-L.; Wang, H.-Y.; Jia, Y.-M.; Yu, C.-Y.; Kinami, K.; Hirokami, Y.; Tsuji, Y.; Adachi, I.; Nash, R.J.; et al. Design and Synthesis of Labystegines, Hybrid Iminosugars from LAB and Calystegine, as Inhibitors of Intestinal α-Glucosidases: Binding Conformation and Interaction for ntSI. J. Org. Chem. 2015, 80, 4501–4515. [Google Scholar] [CrossRef] [PubMed]
  46. Jackova, D.; Martinkova, M.; Gonda, J.; Vilkova, M.; Pilatova, M.B.; Takac, P. Stereoselective synthesis and anticancer activity of broussonetine analogues. Tetrahedron Asymmetry 2017, 28, 1175–1182. [Google Scholar] [CrossRef]
  47. Ribes, C.; Falomir, E.; Murga, J.; Carda, M.; Alberto Marco, J. Convergent, stereoselective syntheses of the glycosidase inhibitors broussonetines D and M. Org. Biomol. Chem. 2009, 7, 1355–1360. [Google Scholar] [CrossRef] [Green Version]
  48. D’Adamio, G.; Parmeggiani, C.; Goti, A.; Moreno-Vargas, A.J.; Moreno-Clavijo, E.; Robina, I.; Cardona, F. 6-Azido hyacinthacine A(2) gives a straightforward access to the first multivalent pyrrolizidine architectures. Org. Biomol. Chem. 2014, 12, 6250–6266. [Google Scholar] [CrossRef]
  49. Lahiri, R.; Ansari, A.A.; Vankar, Y.D. Recent developments in design and synthesis of bicyclic azasugars, carbasugars and related molecules as glycosidase inhibitors. Chem. Soc. Rev. 2013, 42, 5102–5118. [Google Scholar] [CrossRef]
  50. Lahiri, R.; Palanivel, A.; Kulkarni, S.A.; Vankar, Y.D. Synthesis of Isofagomine-Pyrrolidine Hybrid Sugars and Analogues of (-)-Steviamine and (+)-Hyacinthacine C-5 Using 1,3-Dipolar Cycloaddition Reactions. J. Org. Chem. 2014, 79, 10786–10800. [Google Scholar] [CrossRef]
  51. Chatterjee, A.K.; Choi, T.L.; Sanders, D.P.; Grubbs, R.H. A general model for selectivity in olefin cross metathesis. J. Am. Chem. Soc. 2003, 125, 11360–11370. [Google Scholar] [CrossRef]
  52. Wennekes, T.; van den Berg, R.J.B.H.N.; Boltje, T.J.; Donker-Koopman, W.E.; Kuijper, B.; van der Marel, G.A.; Strijland, A.; Verhagen, C.P.; Aerts, J.M.F.G.; Overkleeft, H.S. Synthesis and Evaluation of Lipophilic Aza-C-glycosides as Inhibitors of Glucosylceramide Metabolism. Eur. J. Org. Chem. 2010, 1258–1283. [Google Scholar] [CrossRef]
  53. Wang, W.-B.; Huang, M.-H.; Li, Y.-X.; Rui, P.-X.; Hu, X.-G.; Zhang, W.; Su, J.-K.; Zhang, Z.-L.; Zhu, J.-S.; Xu, W.-H.; et al. A Practical Synthesis of Sugar-derived Cyclic Nitrones: Powerful Synthons for the Synthesis of Iminosugars. Synlett 2010, 3, 488–492. [Google Scholar]
  54. Cardona, F.; Faggi, E.; Liguori, F.; Cacciarini, M.; Goti, A. Total syntheses of hyacinthacine A2 and 7-deoxycasuarine by cycloaddition to a carbohydrate derived nitrone. Tetrahedron Lett. 2003, 44, 2315–2318. [Google Scholar] [CrossRef]
  55. Carmona, A.T.; Whightman, R.H.; Robina, I.; Vogel, P. Synthesis and glycosidase inhibitory activity of 7-deoxycasuarine. Helv. Chim. Acta. 2003, 86, 3066–3073. [Google Scholar] [CrossRef]
  56. Tsou, E.-L.; Chen, S.-Y.; Yang, M.-H.; Wang, S.-C.; Cheng, T.-R.-R.; Cheng, W.-C. Synthesis and biological evaluation of a 2-aryl polyhydroxylated pyrrolidine alkaloid-based library. Bioorg. Med. Chem. 2008, 16, 10198–10204. [Google Scholar] [CrossRef]
  57. Tsou, E.-L.; Yeh, Y.-T.; Liang, P.-H.; Cheng, W.-C. A convenient approach toward the synthesis of enantiopure isomers of DMDP and ADMDP. Tetrahedron 2009, 65, 93–100. [Google Scholar] [CrossRef]
  58. Keck, G.E.; Geraci, L.S. Catalytic asymmetric allylation (CAA) reactions. II. A new enantioselective allylation procedure. Tetrahedron Lett. 1993, 34, 7827–7828. [Google Scholar] [CrossRef]
  59. Bhoite, S.P.; Kamble, R.B.; Suryavanshi, G.M. An enantioselective synthesis of (+)-hygroline and (+)-pseudohygroline via Keck allylation and CBS reduction. Tetrahedron Lett. 2015, 56, 4704–4705. [Google Scholar] [CrossRef]
  60. Wormald, M.R.; Nash, R.J.; Hrnciar, P.; White, J.D.; Molyneux, R.J.; Fleet, G.W.J. Configurational and conformational analysis of highly oxygenated pyrrolizidines: Definitive identification of some naturally occurring 7a-epi-alexines. Tetrahedron Asymmetry 1998, 9, 2549–2558. [Google Scholar] [CrossRef]
  61. Donohoe, T.J.; Sintim, H.O.; Hollinshead, J. A Noncarbohydrate Based Approach to Polyhydroxylated Pyrrolidizines:  Total Syntheses of the Natural Products Hyacinthacine A1 and 1-Epiaustraline. J. Org. Chem. 2005, 70, 7297–7304. [Google Scholar] [CrossRef]
  62. Calveras, J.; Egido-Gabás, M.; Gómez, L.; Casas, J.; Parella, T.; Joglar, J.; Bujons, J.; Clapés, P. Dihydroxyacetone Phosphate Aldolase Catalyzed Synthesis of Structurally Diverse Polyhydroxylated Pyrrolidine Derivatives and Evaluation of their Glycosidase Inhibitory Properties. Chem. Eur. J. 2009, 15, 7310–7328. [Google Scholar] [CrossRef] [PubMed]
  63. Kessler, M.; Acuto, O.; Storelli, C.; Murer, H.; Semenza, G.A. A modified procedure for the rapid preparation of efficiently transporting vesicles from small intestinal brush border membranes. Their use in investigating some properties of d-glucose and choline transport systems. Biochim. Biophys. Acta 1978, 506, 136–154. [Google Scholar] [CrossRef]
Sample Availability: Not available.
Figure 1. Structures of DAB, DMDP, and their related derivatives.
Figure 1. Structures of DAB, DMDP, and their related derivatives.
Molecules 24 03712 g001
Figure 2. Some representative broussonetines.
Figure 2. Some representative broussonetines.
Molecules 24 03712 g002
Scheme 1. Retrosynthesis of broussonetine M (3) from d-arabino-nitrone (14).
Scheme 1. Retrosynthesis of broussonetine M (3) from d-arabino-nitrone (14).
Molecules 24 03712 sch001
Scheme 2. Synthesis of the pyrrolidine core 13.
Scheme 2. Synthesis of the pyrrolidine core 13.
Molecules 24 03712 sch002
Scheme 3. Synthesis of the side chain.
Scheme 3. Synthesis of the side chain.
Molecules 24 03712 sch003
Scheme 4. Completion of the synthesis of broussonetine M (3).
Scheme 4. Completion of the synthesis of broussonetine M (3).
Molecules 24 03712 sch004
Table 1. Broussonetine M (3) and analogues synthesized from different cyclic nitrones and alcohols.
Table 1. Broussonetine M (3) and analogues synthesized from different cyclic nitrones and alcohols.
EntryCyclic NitronePyrrolidineYield aAlcoholProductYield b
1 Molecules 24 03712 i001 Molecules 24 03712 i00264% Molecules 24 03712 i003 Molecules 24 03712 i00443%
2 Molecules 24 03712 i005 Molecules 24 03712 i00643%
3 Molecules 24 03712 i007 Molecules 24 03712 i00865% Molecules 24 03712 i009 Molecules 24 03712 i01041%
4 Molecules 24 03712 i011 Molecules 24 03712 i01245%
5 Molecules 24 03712 i013 Molecules 24 03712 i01464% Molecules 24 03712 i015 Molecules 24 03712 i01640%
6 Molecules 24 03712 i017 Molecules 24 03712 i01841%
7 Molecules 24 03712 i019 Molecules 24 03712 i02071% Molecules 24 03712 i021 Molecules 24 03712 i02243%
8 Molecules 24 03712 i023 Molecules 24 03712 i02443%
a Total yield in 3 steps starting from cyclic nitrones to the corresponding pyrrolidine. b Total yield in 2 steps starting from pyrrolidine cores to broussonetine M or its analogues.
Table 2. Concentrations of Broussonetine M (3) and its DMDP-related analogs giving 50% inhibition of various enzymes.
Table 2. Concentrations of Broussonetine M (3) and its DMDP-related analogs giving 50% inhibition of various enzymes.
EnzymeIC50 (μM)
Molecules 24 03712 i025 Molecules 24 03712 i026 Molecules 24 03712 i027 Molecules 24 03712 i028 Molecules 24 03712 i029 Molecules 24 03712 i030 Molecules 24 03712 i031 Molecules 24 03712 i032
DAB (1) [15]DMDP (2) [16]310’-epi-3LAB (ent-1) [15]l-DMDP (ent-2) [16]ent-10’-epi-3ent-3
α-Glucosidase
Yeast0.15NI a (15.6%) bNI (2.6%)NI (4.6%)70NI (34.9%)NI (1.9%)NI (1.9%)
Rice250214NI (0%)NI (0%)3.25.81.31.2
Rat intestinal maltase55290NI (0%)NI (26.4%)0.931.20.180.29
β-Glucosidase
Almond25010NI (10.0%)NI (29.1%)NINI (12.1%)NI (8.0%)NI (9.6%)
Bovine liver6389.76.30.8NI (13%)NI (4.6%)51NI (46.5%)
α-Galactosidase
Coffee beansNI (7%)NI (10.5%)NI (0%)NI (0%)NI (2%)NI (13.8%)NI (12.2%)NI (0.3%)
β-Galactosidase
Bovine liverNI (37%)3.32.30.2NI (16%)NI (0%)9.050
α-Mannosidase
Jack bean320NI (31.5%)NI (2.5%)NI (0%)NI (0%)NI (17.3%)NI (1.8%)NI (2.2%)
β-Mannosidase
SnailNI (10%)721NI (1.7%)NI (0%)NI (1%)NI (12.6%)NI (2.0%)NI (0%)
α-l-Fucosidase
Bovine kidneyNI (11%)NI (37.2%)NI (0%)NI (0%)NI (0%)NI (0%)NI (6.1%)NI (0%)
α,α-Trehalase
Porcine kidney61200NI (0%)NI (0%)7548NI (0%)NI (2.1%)
Amyloglucosidase
A. niger362NI (3.8%)NI (36.5%)NI (41%)NI (6.1%)NI (0%)
α-l-Rhamnosidase
P. decumbensNI (5%)NI (24.1%)NI (6.1%)NI (4.7%)803NI (45.6%)NI (1.8%)NI (13.3%)
β-Glucuronidase
E.colicNI (0.4%)8620NI (0.8%)NI (36.2%)NI (44.3%)
Bovine liverNI (18.4%)NI (17.2%)NI (14.9%)NI (2.3%)
a NI: No inhibition (less than 50% at 100 μM for broussonetine M and its analogs; less than 50% at 1000 μM for DAB, DMDP, LAB, and l-DMDP); b ( ): inhibition % at 100 μM for broussonetine M and its analogs; inhibition % at 1000 μM for DMDP and l-DMDP; c —: not determined.
Table 3. Concentrations of Broussonetine M’s analogs related with l-altro-DMDP and d-gluco-DMDP giving 50% inhibition of various enzymes.
Table 3. Concentrations of Broussonetine M’s analogs related with l-altro-DMDP and d-gluco-DMDP giving 50% inhibition of various enzymes.
EnzymeIC50 (μM)
Molecules 24 03712 i033 Molecules 24 03712 i034 Molecules 24 03712 i035 Molecules 24 03712 i036 Molecules 24 03712 i037 Molecules 24 03712 i038
l-altro-DMDP [16]ent-3,10-di-epi-3ent-3-epi-3d-gluco-DMDP [16]2-epi-32,10-di-epi-3
α-Glucosidase
YeastNI a (35.1%) bNI (6.6%)NI (0%)167NI (1.6%)NI (3.2%)
RiceNI (9.5%)NI (9.5%)NI (23.0%)131NI (5.6%)12
Rat intestinal maltase7548442138NI (16.8%)1.4
β-Glucosidase
AlmondNI (15.9%)NI (7.3%)NI (4.6%)256NI (8.6%)NI (10.9%)
Bovine liverNI (10.3%)68715232333.9
α-Galactosidase
Coffee beans120NI (0%)NI (0%)NI (38.4%)NI (0%)NI (0%)
β-Galactosidase
Bovine liverNI (5.9%)31413617.413
α-Mannosidase
Jack beanNI (37.6%)NI (0%)NI (1.3%)NI (13.3%)NI (0%)NI (0.32%)
β-Mannosidase
SnailNI (3.7%)NI (2.3%)NI (0%)NI (14.6%)NI (0%)NI (0%)
α- l-Fucosidase
Bovine kidney2059850NI (37.2%)NI (4.3%)NI (10.8%)
α,α-Trehalase
Porcine kidney1000NI (3.8%)NI (0%)379NI (0%)NI (0%)
Amyloglucosidase
A. nigercNI (0%)NI (0%)NI (3.8%)NI (6.8%)
α-l-Rhamnosidase
P. decumbens91NI (11.4%)NI (15.2%)NI (24.1%)NI (3.3%)NI (4.7%)
β-Glucuronidase
E.coliNI (2.0%)42NI (28.7%)NI (5.1%)7380
Bovine liverNI (14.9%)NI (0%)NI (5.2%)NI (1.7%)
a NI: No inhibition (less than 50% at 100 μM for broussonetine M’s analogs; less than 50% at 1000 μM for l-altro-DMDP and d-gluco-DMDP); b ( ): inhibition % at 100 μM for broussonetine M’s analogs; inhibition % at 1000 μM for l-altro-DMDP and d-gluco-DMDP; c —: not determined.

Share and Cite

MDPI and ACS Style

Wu, Q.-K.; Kinami, K.; Kato, A.; Li, Y.-X.; Jia, Y.-M.; Fleet, G.W.J.; Yu, C.-Y. Synthesis and Glycosidase Inhibition of Broussonetine M and Its Analogues. Molecules 2019, 24, 3712. https://doi.org/10.3390/molecules24203712

AMA Style

Wu Q-K, Kinami K, Kato A, Li Y-X, Jia Y-M, Fleet GWJ, Yu C-Y. Synthesis and Glycosidase Inhibition of Broussonetine M and Its Analogues. Molecules. 2019; 24(20):3712. https://doi.org/10.3390/molecules24203712

Chicago/Turabian Style

Wu, Qing-Kun, Kyoko Kinami, Atsushi Kato, Yi-Xian Li, Yue-Mei Jia, George W. J. Fleet, and Chu-Yi Yu. 2019. "Synthesis and Glycosidase Inhibition of Broussonetine M and Its Analogues" Molecules 24, no. 20: 3712. https://doi.org/10.3390/molecules24203712

Article Metrics

Back to TopTop