G Model
ARTICLE IN PRESS
CHROMB-19632; No. of Pages 11
Journal of Chromatography B, xxx (2015) xxx–xxx
Contents lists available at ScienceDirect
Journal of Chromatography B journal homepage: www.elsevier.com/locate/chromb
Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines Tengfei Xu a,b , Shu Liu a , Lulu Meng c , Zifeng Pi a,∗ , Fengrui Song a , Zhiqiang Liu a,∗ a State Key Laboratory of Electroanalytical Chemistry, National Center for Mass Spectrometry in Changchun, Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China b University of the Chinese Academy of Sciences, Beijing 100039, China c Jilin Provincial Science & Technology Department, Changchun 130041, China
a r t i c l e
i n f o
Article history: Received 13 March 2015 Received in revised form 21 August 2015 Accepted 17 September 2015 Available online xxx Keywords: Diterpendoid alkaloids Analysis methods Biological activity Metabolism Biotransformation
a b s t r a c t The diterpenoid alkaloids as one type of heterocyclic alkaloids have been found in many traditional herbal medicines, such as genus Consolida, Aconitum, and Delphinium (Ranunculaceae). Pharmacological researches have indicated that many diterpenoid alkaloids are the main bioactive components which have analgesic, anti-inflammatory, anti-microbial, anti-tumor, cardiotonic, and anti-arrhythmic activities. Studies focused on the determination, quantitation and pharmacological properties of these alkaloids have dramatically increased during the past few years. Up to now, newly discovered diterpenoid alkaloids with important biological activities have been isolated and synthesized. Considering their significant role and diffusely used in many disease treatments, we summarized the information of their analysis methods, bioactivity, metabolism and biotransformation in vivo as well as the pharmacological mechanisms. Based on above review, the further researches are suggested. © 2015 Elsevier B.V. All rights reserved.
1. Introduction Since diterpenoid alkaloids (DAs) show a variety of important physiological activities, thus as potential drugs these compounds are used to the treatment of many diseases. At present, the investigation of these alkaloids highlights on the analysis method, biological activity, metabolism in vivo and the mechanism of interaction way. Most of the DAs were isolated from the Ranunculaceae family such as Aconitum, Consolida and Delphinium species, which are widely used as important ingredient in Oriental Medicine for thousands of years owing to their excellent efficacy in clinical. From 1998 to 2014, more than 400 new DAs isolated from nature products were reported [1–9]. Accordingly, three categories of DAs are classified as the C18-, C19-, and C20-diterpenoid alkaloids that, respectively, contain 18, 19 and 20 carbon atoms (see Fig. 1). Different types of DAs show diverse biological activities, for example, C18-, C19-diterpenoid alkaloids, major acontine-type with heterozcycles, exhibit excellent therapeutic effect in the treatments of joint pain, gastroenteritis, diarrhea, and rheumatoid arthritis with the anti-inflammatory and analgesic activities [10]. C20diterpenoid alkaloids possess potent inhibitory on tumor cells, and
∗ Corresponding authors. Fax: +86 431 85262236. E-mail addresses:
[email protected] (Z. Pi),
[email protected] (Z. Liu).
thus as prototype drugs could be used to develop new anticancer drugs [2,11,12]. Many studies have devoted to the pharmacological properties of DAs with in vitro and in vivo assays such as metabolism, antiinflammatory, analgesic, anti-tumor, antibacterial, effects on the nervous and cardiovascular system, and so on [13–17]. The diterpene structure alkaloids with heterocycles provide a rich resource for developing new drugs to the treatments of some thorny diseases. Although some achievements have been obtained, it is necessary to further research on the mechanisms about absorption, biotransformation and interaction with the corresponding receptors of DAs. This review focuses on the analysis methods, biological activities, processed ways in vivo, in vitro and the corresponding mechanisms of DAs. 2. Separation and analytical technology of diterpenoid alkaloids 2.1. Physicochemical properties of diterpenoid alkaloids In 2006, Nakamura et al. did an evaluation for physicochemical quality of DAs, including description, stability in solvent, moisture content, solubility, melting point and UV absorbance. They proved that the alkaloids degraded faster in MeOH than in other solutions, and a mixture of phosphate buffer solution with CH3 CN (1:1, v/v)
http://dx.doi.org/10.1016/j.jchromb.2015.09.023 1570-0232/© 2015 Elsevier B.V. All rights reserved.
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
2
Fig. 1. Chemical structure of typical C18-, C19- and C20-diterpenoid alkaloids.
might be suitable for test [18]. Meanwhile, Yue et al. have studied the stability of DAs in different pH buffers, storage time and solvents. The results indicated that dichloromethane was a better solvent to keep the stabilities of aconitine (AC), mesaconitine (MA) and hypaconitine (HA) than other solvents like ether, methanol and water. They found that the different stabilities of diester–diterpene alkaloids (DDAs) mainly were depend on their structures, in which C-3 and N position are important sites for hydrolysis [3]. 2.2. Separation technology of diterpenoid alkaloids Recently, developments of separation and purification techniques for DAs are emerged. These techniques include silica gel column chromatography, microwave-assisted extraction (MAE), supercritical-fluid extraction (SFE), and high-speed countercurrent chromatography (HSCCC). Silica gel column chromatography is applied in the separation of DAs to be a classical and conventional method being limited due to the multi-step, timeconsuming and potential hazard of losing compounds. MAE method has been applied to the extraction of total alkaloids from plants
[19], in which microwave was used for heating the extract solution resulting in a significant reduction of extraction time (usually <30 min) and quantity of organic solvent (usually <40 mL). SFE could be used for numerous commercial applications and could give out a high yield of alkaloids from aqueous extracts [20]. In the separation techniques, the purification efficiency of cation-exchange SPE is relative poor, but the cost is low, and it could purify crude extracts without much process [21,22]. Recently, HSCCC, a supportfree liquid–liquid partition chromatography, has been successful used in the isolation and purification of DAs as an efficient method. Tang et al. obtained six DAs with HSCCC, including Guanfu base O, Guanfu base Q, Guanfu base Z, hetisinone, hetisine and Guanfu base AA from A. coreanum with one-step separation [23]. Wang et al. and Liu et al. also developed an efficient method to separate DAs from A. coreanum, in which a pH-zone-refining counter-current chromatography was applied to purify seven alkaloid compounds. The purities of these alkaloid compounds were higher than 96 percent, which indicated that pH-zone-refining CCC was an efficient method for preparative separation of DAs from herbs even with low contents [24,25]. For the mass production of DAs, it is
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
3
suggested that at first, a coarse separation is performed, and followed by a higher efficient purification step. According to the separation methods mentioned above, SFE followed with HSCCC is meaningful to mass production of DAs. 2.3. Analysis methods of diterpenoid alkaloids Recently, the diterpenoid alkaloids are widely used in traditional Chinese medicine (TCM) and Chinese patent medicines, whereas these medicines have a narrow therapeutic index at the same time. To safely utilize herbal medicine, it is highly needed to develop a rapid and sensitive analysis method for quality control of the medicines. To date, a number of researches focused on the separation, identification and quantitation of DAs as well as DA metabolites. The analysis methods mainly include high-performance liquid chromatography–diode array detection (HPLC–DAD), high-performance liquid chromatographyelectrospray ionization–mass spectrometry (HPLC-ESI–MS), ultraperformance liquid chromatography-electrospray ionization-mass spectrometry (UPLC-ESI–MS) [26], matrix assisted laser desorption ionization-mass spectrometry (MALDI–MS) and so on. HPLC–DAD was widely used for quality control and finger-print for Aconitum species and related prescriptions. This technique is usually needs standard compounds to compare the retention time. It is not sufficient because this technique could not provide more information to character the structures of alkaloids in Aconitum species. However, by using mass spectrometry (MS), it is enable to determine DAs with different ionizations and detection modes, thus MS has been diffusely used to determinate structures of unknown alkaloids along with separations [27]. Ding et al. determined 13 aminoalcohol-diterpenoid alkaloids in Aconitum carmichaeli with solid phase extraction coupled with LC–MS/MS. Multiple reaction monitor (MRM) was used in their study, and the limit of detection (LOD) and the limit of quantitation (LOQ) were lower than 0.5 ng mL−1 and 2.0 ng mL−1 , respectively [28]. Wang et al. found that the fragmentation mechanisms of aconitinetype alkaloids were mainly due to the elimination of substituent at C-8, of which an acetic acid or a fatty acid lost in MS2 spectra and CH3 COOH, CH3 OH, H2 O, BzOH and CO lost in MS3 spectra, respectively. At the same time, they found 70 diterpenoid alkaloids in the flowers of Aconitum kusnezoffii, including a series of lipoaconitines [4]. This is the first time to be found in that plant. Wu et al. carried out a direct determination of alkaloids in A. carmichaeli Debx and its processed products using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDITOF-MS). The results showed that this method is valuable for rapidly analyzing component profiles of plants with less sample preparation, and that the new compounds in medicinal herbs might be discovered [29]. Eight C20-diterpenoid alkaloids were firstly identified, and thirty C19-diterpenoid alkaloids were isolated from A. carmichaeli by Hu et al. in 2009. The fragmentation mechanisms of DDAs, monoester-diterpene alkaloids (MDAs), amine-diterpene alkaloids (ADAs) and C20-diterpenoid alkaloids were elucidated in detail, and the results showed that different substituents in different sites may produce different fragment ions, which could be used to identify various alkaloids. The major differences of fragment ions between C19 and C20-diterpenoid alkaloids are the loss of CH3 OH for C19-diterpenoid alkaloids but not for C20-diterpenoid alkaloids [30]. Recently, Xu group developed a rapid and effective method for separation and identification DDAs in A. carmichaeli based on ultra-high-pressure liquid chromatography coupled with high resolution LTQ-Orbitrap tandem mass spectrometry (UHPLC-LTQOrbitrap-MSn ) [31,32]. In their study, a systematic strategy was successfully conducted by monitoring characteristic neutral loss of DDAs. A typical fragmentation of DDAs different from MDAs or deesterified DAs is that a molecule of AcOH is eliminated at C8
Fig. 2. The DART spectra of the preparatas of Radix Aconiti Radix (top) and Aconiti Kusnezoffii (bottom). Reproduced from [34] with permission from the author.
position in MS2 spectra. By calculating the C1, C6, C16 and C18 binding energies, they concluded that the losses of 32(MeOH) + 28(CO), 32(MeOH) + 18(H2 O), 122(BzOH) in MSn spectra were respectively corresponding to the structures with 15-OH, 16-OMe; 3-OH, 16OMe and 14-BzOH in DDAs. They separated and identified 42 DDAs including 16 new types of short chain lipo-DDAs and 4 N-dealkyltype DDAs in A. carmichaeli. [31]. Moreover, they detected and characterized more than 120 alkaloids including DDAs, MDAs, lipoditerpenoid alkaloids (LDAs) and ADAs in A. carmichaeli based on the procedures used in this research [32]. It has been proved that the analysis method with neutral loss is an effective tool to determine not only for DAs but also for compounds with similar structures in complex plant extracts. New ionization method like direct analysis in real time (DART) has already been applied to quality control and identification of herbal medicine. DART ionization is a solvent-free method that a steam of hot gas flowing onto a sample surface to adsorb and ionize the analytes from the surface, which could avoid multiple sample preparation procedures. Song et al. [33–35] accessed Radix Aconiti, in which the preparata samples were processed by two different procedures (boiling and steaming) by DART analysis method. The results indicated that the main chemical marker discrimination amongst the qualified and unqualified samples mainly were on some MDAs and DDAs, and the results by using the two processing methods have no obvious difference. After processing, the relative intensities of DDAs reduced while MDAs increased, and both the two processing procedures could reduce the toxicity of Radix Aconiti effectively. An in situ analysis of Aconitum herbal pieces using DART-MS has developed recently, which showed that almost all the aconitine-type alkaloids could be desorbed and ionized from the pieces of Aconitum products. From Fig. 2, it can be seen that the major differences of Radix Aconiti Kusnezoffii and Radix Aconiti were some MDAs (m/z from 573 to 603) [34]. From above we can conclude that DART-MS is an excellent method with advantages such as fast, high-throughput, and less pollution in rapid quality control of DAs. Apart from mass spectrometry, other detection methods were also developed for qualitative or quantitative determination of DAs. Capillary zone electrophoresis (CZE) is a reliable method with good precision, accuracy and recovery for simultaneous determination of DAs, which has been used in quality control of herbal medicines
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
4
[36]. Yang et al. successfully simultaneously determined AC, MA and HA in crude sample with CE-electrochemiluminescence (CEECL) detection system. They used ionic liquids to enhance the sensitivity and selectivity, and the detection limits were close to 10−9 mol L−1 of the alkaloids. The CE-ECL detection system can be used for analysis of trace DAs that especially with tertiary amine group in biological samples [37]. 2.4. Extraction, clean-up and quantitation of diterpenoid alkaloids from bio-matrices Xie et al. developed an analysis method for quantitative determinations of AC, MA, HA, benzoylaconine (BAC), benzoylmesaconine (BMC) and benzoylhypaconine (BHA) in Chinese medicinal herbs, respectively and Chinese patent medicines by HPLC–DAD [38]. But the application of this method is subjected to its higher limit of determination, say >800 ng mL−1 , i.e., it is not suitable for trace samples of DAs. Recently, Zhang et al. simultaneous quantified the six aconitum alkaloids mentioned above in human plasma with a LC–MS method within 4.5 min. The pharmacokinetic evaluations of the six aconitum alkaloids were preformed all with a good linearity (r2 > 0.995) ranged from 0.1 to 1000 ng mL−1 [39]. It is known that extraction procedure just like many other sample cases is a key step in the quantitative analysis of DAs in bio-matrices. A study conducted several years ago revealed that liquid–liquid extraction (LLE) showed a low recoveries for six Aconitum alkaloids (≈45%), but protein precipitation procedure (acetonitrile with 0.1% acetic acid or methanol with 0.2% formic acid) could give satisfactory recovery of the alkaloids with less matrix effect in rat plasma [26]. He et al. quantified the major metabolites of AC in rat urine by protein precipitation of acetonitrile/methanol (3:1, v/v), and good precisions and accuracies of the metabolites were obtained [40]. The method that using solid-phase extraction followed by LC–MS in quantified AC in biological samples (blood samples and autopsy samples) to be proved was a highly sensitive and no matrix peaks interfered with the detection of AC [41]. Hattori et al. used a new unique polymer column (Shodex ODP2HP-4B) coupled with mass spectrometry to separate and quantify four aconitine-type analogues alkaloids in whole blood simultaneously. The column with functions as an extraction medium for impurities and a LC stationary phase could exclude proteins and nucleic acid, thus it is not necessary to have complicated pretreatments of the blood in that analysis method [42]. Wu et al. extracted AC as well as other 16 poisonous alkaloids in human blood and gastric juice. A multi-step liquid extraction was preformed, firstly using chloroform and followed by chloroform-ether (2:1, v/v). The linearity (r > 0.9952) and the intra-day and inter-day relative standard deviation (RSD) (less than 10% and 15%, respectively) results indicated that the processing method was simple, rapid and stable to be enough to examine toxic alkaloids in bio-samples [43]. 3. Biological activities of diterpenoid alkaloids It seems that the synthesis of a new bioactive compound is more and more difficult, thus a great deal of bioactive substances and drugs has been derived from natural materials. In which, many DAs possess good treatment effects and favorable performance to interact with receptors in the neurotransmitter and gastrointestinal systems. 3.1. Anti-inflammatory and analgesic activity of diterpenoid alkaloids Clinically, non-steroidal anti-inflammatory drugs (NSAIDS) and opioids are two main type drugs for analgesc and antiinflammatory. But NSAIDS might cause gastrointestinal tract
damage due to suppress the production of prostaglandins, while opioids are harmful to the nervous system and strong potential drug addiction. Due to these reasons, developing a new type of antiinflammatory analgesic pharmaceutical is necessary. DAs isolated from Ranunculaceae plants such as Aconitum species, Delphinium species and Consolida species possess interaction with receptors in the neurotransmitter systems and electrophysiological properties, which are the most likely candidates for yielding compounds with spasmolytic, anti-inflammatory and analgesic activity, respectively. The five diterpene alkaloids (napelline, songorine, HA, MA, 12-epinapelline N-oxide) isolated from Aconitum baikalense are confirmed that they possess significant effect in inhibit edema and reduce acute inflammatory responseble for histamine or carrageenan. The activities of inhibt edema of napelline, HA, 12epinapelline N-oxide, MA and songorine were 12%, 34%, 39% 34% and 23%, respectively [44]. Another aconitine-type alkaloids extracted from Radix Aconiti Carmichaeli were observed effective on carrageenan-induced paw edema and acetic acid-induced abdominal constriction. The results showed that this plant extracts and the contained alkaloids have significant antinociceptive activities and anti-inflammatory effects as well as MA and AC are effective in decreasing the pain sensation [45]. Meanwhile, guiwuline, a new franchetine type of C19-diterpenoid alkaloid isolated from A. carmichaeli Debx, could be used as a lead molecule to develop novel analgesic agent, because it possesses high analgesic activity (ED50 , 15 mg/kg) and low toxicity (LD50 , 500 mg/kg) [46]. 3.2. Cardiovascular action of diterpenoid alkaloids As containing various noxious DAs, aconite roots exhibit cardiotoxicity and neurotoxicity. It was reported that aconite roots could cause serious ventricular tachycardia and arrhythmia [4]. The two diterpenoid alkaloids, methyllycaconitine and lycaconitine, separated from Delphinium species exhibited neuronal nicotinic acetylcholine receptor affinity [47]. In addition, methyllycaconitine has been approved in clinical with an favorable effect on muscle relaxants [48]. Lappaconitine hydrobromide has been used as an antiarrhythmic drug for the treatments of supraventricular and ventricular extrasystoly, atrial fibrillation and flutter syndromes [49,50]. From the electrocardiogram, it can be found that the occurrence of arrhythmia often follows an increase in the plasma concentration of aconitine after oral [51]. Interestingly, DAs revealed spasmolytic or spasmogenic activity as the structures are different. Futhermore, Tursunkhodzhaeva et al. reported the spasmolytic or spasmogenic activities of 82 diterpenoids alkaloids and gave out the structure–activity relationship of those componds. It was found that an aromatic ester in the 1- or 14position of aconitine-type, an aromatic ester in the C-14 and C-6 positions of lycoctonine-type alkaloids, and a C-4 anthranilic acid of lappaconitine-type alkaloids showed the higher spasmolytic activities. Aconitine-like alkaloids showed spasmogenic activities at the concentrations of 0.035–1.4 mol L−1 , in which the compound possessing the most activity was mesaconitine. With the experiments of in vitro and in vivo, 1-O-benzoylkaracoline and 1-Obenzoylisotalatisidine exhibited the higher spasmolytic activities (EC50 , 0.39 and 0.18 mol L−1 ) and less toxicity, indicating that the DAs might be a new source for discovering and developing highly efficacious, low-toxic, and long-acting drugs with myotropic spasmolytic activities [52]. 3.3. Anti-microbial activity of diterpenoid alkaloids In order to protect themselves from microbial infection, plants always produce many secondary metabolites such as alkaloids possessing antiviral, antibacterial and antifungal activities. Aconitum and Delphinium species containing DAs are often used for
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
the treatments of itches and other skin eruptions in traditional medicine. Different diterpenoid alkaloids possess different microbial inhibition activity. Lycoctonine, 18-O-methyllycoctonine, delcosine, 14-acetyldelcosine and 14-acetylbrowniine show significant antibacterial activities against K. pneumoniae and A. baumannii at 8 g mL−1 concentration, a high antifungal activity against C. albicans at 4 g mL−1 , and have a selective inhibition against PI-3 virus [53]. Lycoctonine also has an inhibity for Salmonella typhi and Pseudomonas aeruginosa. Meanwhile, 6dehydroacetylsepaconitine and 13-hydroxylappaconitine possess significant antibacterial activities against Staphylococcus aureus. Compared to imipenem, a broad-spectrum -lactam antibiotic drug, delphatine and lappaconitine prior inhibit Salmonella typhi and P. aeruginosa [54]. It has been reported that BMA combined with IL-12 could regulate severe herpes simplex virus type 1(HSV-1) infection in TI-mice [55]. 3.4. Anti-tumor activity of diterpenoid alkaloids Alkaloids like vincristine or paclitaxel possess significantly anti-tumor properties, and diterpenoid alkaloids isolated from extensive plants also possess anti-tumor activity. It was reported that several C19-norditerpenoids like neoline, pubescenine, 14-deacetylajadine, lycoctonine, dehydrotakaosamine and ajadelphinine had irreversible effects on SW480, HeLa and SkMel25 cell lines [12]. Lycaconitine, a C19-diterpenoid alkaloid, had potent P-glycoprotein multidrug resistance (P-gp MDR) inhibition activity, and could reduce multidrug resistant population into half with only 74 g mL−1 [56]. 8-O-azeloyl-14-benzoylaconine, isolated from the roots of Aconitum karacolicum Rapcs, showed prominent anti-tumor properties against three human tumor cell lines, HCT-15, A549 and MCF-7 [57]. Laterly, three bis-[O(14-benzoylaconine-8-yl)]esters, (i.e., bis-[O-(14-benzoylaconine8-yl)]-pimelate, bis-[O-(14-benzoylaconine-8-yl)]-suberate and bis-[O-(14-benzoylaconine-8-yl)]-azelate) were found that they possessed noticeable cytotoxities in vitro and IC50 vaules against tumor cells were ranging from 4 to 28 mmol L−1 [58]. Hazawa et al. had observed 39 aconitum alkaloids against tumor cells (A172, A549, HeLa and Raji), in which six C20-Diterpenoid alkaloids showed anti-proliferative activities with IC50 values ranged from 1 to 5 mol L−1 against A549. The results showed that the compounds derived from C20-diterpenoid alkaloids possessed a significantly suppressive effect of tumor cells while C19-diterpenoid alkaloids possessed no or only a slight effect [59]. Two Aconitum C20-diterpenoid alkaloid derivatives, 11-m-Trifluorometylbenzoyl-pseudokobuisne and 11Anisoyl-pseudokobusine, showed significant suppressive effects against Raji cells and their IC50 values were 2.2 g mL−1 and 2.4 g mL−1 , respectively. The structure-activity relationships of C20-diterpenoid alkaloids showed that atisine type alkaloids, i.e., the C-11 residues were the important components for the antiproliferative properties with a lower toxicity to hematopoiesis [60]. Further studies by Hazawa et al. suggested that C6 -derivatives with the identical bone structure of C20-diterpenoid alkaloids promoted the proliferation of hematopoietic stem/progenitor cells, while C11-derivatives had an opposing effects depending on the different derivatization sites [11]. The diterpenoid alkaloids inhibitory effects on tumur cells are shown in Table 1. 3.5. Toxicity of diterpenoid alkaloids Severe aconite poisoning would occur after accidental ingestion of the wild plant or consumption herbal decoction extracted from aconite roots [61]. The toxic activity of aconitum alkaloids mainly affect the central nervous system, heart and muscle tissues [17]. The oral median lethal doses (LD50 ) of the toxins in mice respec-
5
tively are 1.8 mg/kg for AC [62], 1.9 mg/kg for MA [63], 5.80 mg/kg for HA, 810 mg/kg for BMA, 830 mg/kg for BHA and 1500 mg/kg for BAC [64]. Long-term administration of AC in rice can increase the activity of acontine metabolish thus will decrease the effectiveness of AC [62]. It was reported that the high toxicity aconitum alkaloids are related to the acetyl group at C8, the hydroxyl group at C13, the four methoxyl groups individually at C1, C6, C16, and C18, and the benzoyl-ester group at C14 [65]. The investigation on the toxicity of Aconitum alkaloids to pregnant women showed that AC had direct embryotoxic effects during the rat organogenetic period, and severe dysmorphogenesis effect was induced when the concentration of AC was increased up to 5 mg mL−1 [66]. A quantitative structureactivity relationship (QSAR) analysis performed by Bello-Ramirez using a number of diterpene alkaloids from Aconitum sp. deduced that with an aroyl/aroyloxy group at R14 position had a significantly lower LD50 than with an aroyloxy group at R4 position [67]. Furthermore studies by QSAR analysis suggested that the toxicities of aconitum and delphinium alkaloids are caused by the interaction with sodium ion channel, in which hydrogen bonds act a vital role in the binding processes [68,69]. In addition to the traditional analysis method, metabolomics research concerning the small biochemicals presenting in biological sample could not be ignored. Metabolomics becomes more and more important in the toxicity arena for understanding the toxicity mechanism of traditional Chinese medicine [70,71]. Radix Aconiti, the mother root of A. carmichaelii Debx., contains many diterpenoid alkaloids [33,72], Wang et al. using High-Definition Mass Spectrometry combined with pattern recognition methods discovered that toxicities of the diterpenoid alkaloids were related to tryptophan metabolism, aminoacyl-tRNA biosynthesis and sphingolipid metabolism pathways. Moreover, it was found that Radix Aconiti could lead to heart and liver to serious poisoning, which is depent on the time and dose of given drugs [73,74].
4. Metabolism and biotransformation of diterpenoid alkaloids It was reported that the pharmacokinetic behaviors were similar among DDAs, but different with MDAs after oral administration. The concentrations of DDAs in plasma after oral administration increased very rapidly, and then decreased also very rapidly (time for maximum concentration (Tmax ,), 1.75 ± 0.27 h; half-life (T1/2 ), 2.95 ± 1.36–4.92 ± 5.17 h). In addition, a double-absorption peak was observed in all the concentration—time curves of six aconitine-type alkaloids. The double-peak phenomenon of Aconitum alkaloids might be as a low gastric pH which could delay the drug absorption, and the cyclical fluctuations in stomach and intestine [26]. A study demonstrated that the mechanisms of their pharmacokinetic behaviors could be explained based on pharmacokinetic parameters of three MDAs (BMC, BAC and BHA) after oral administration to rats. The three MDAs were absorbed into blood rapidly and reached the maximum plasma concentrations within about 0.2 h, during the time, the maximum concentration (Cmax ) were less than 40 ng mL−1 . The results indicated that each of the three MDAs had a low bioavailability, and the absorption was blocked. A metabolic stability in rat live microsomes showed that almost no metabolism of the three alkaloids was observed, meaning that rats had un-enough enzymes in vivo to metabolism MDAs [75]. A poor protein bounding (23.9 – 31.9%) illuminated that AC had a low bioavailability (8.24 ± 2.52%) with T1/2 (i.v., 80.98 ± 6.40 min) and Tmax (30.08 ± 9.73 min) in vivo. It was reported that the DDAs firstly dissolved out in gastrointestinal tract and then were absorbed in blood after oral administration [14], and further the DDAs were transformed into detoxication or amine compounds. The pharmacokinetic experiments showed that
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
6
Table 1 Inhibitory Effects of C19-, C20-diterpenoid alkaloids on tumor cell growth. Compound
Anti-cell lines
IC50
Structure
Reference
Lycaconitine Bis-[O-(14-benzoylaconine-8-yl)]-pimelate
KB V20C A-549 MCF-7 HCT-15 A-549 MCF-7 HCT-15 A-549 MCF-7 HCT-15 Raji Raji HCT-15 A549 MCF-7 A172 A172 A172 A172 A172 A172 A172 A172 A172 A172 A172 A172 A172 A172 No effect or only slight effect A549 A549 A549 A549 A549 A549
74 g/mL 9.50 ± 3.21 mol/L 7.56 ± 0.84 mol/L 4.64 ± 1.53 mol/L 7.53 ± 3.08 mol/L 6.90 ± 1.62 mol/L 4.01 ± 0.51 mol/L 19.5 mol/L 16.9 mol/L 28.0 mol/L 2.2 g/mL 2.4 g/mL 16.8 mol/L 19.4 mol/L 10.3 mol/L >5.0 g/mL >5.0 g/mL >5.0 g/mL 5.6 ± 0.15 g/mL >5.0 g/mL >5.0 g/mL >5.0 g/mL >5.0 g/mL 1.3 ± 0.72 g/mL 3.7 ± 0.57 g/mL 1.2 ± 0.10 g/mL 0.89 ± 0.16 g/mL 1.3 ± 0.11 g/mL 1.5 ± 0.01 g/mL C19 4.4 mol/L 3.2 mol/L 1.7 mol/L 3.5 mol/L 3.5 mol/L 5.1 mol/L
C19 C19-derivative
[56] [58]
Bis-[O-(14-benzoylaconine-8-yl)]-suberate
Bis-[O-(14-benzoylaconine-8-yl)]-azelate
11-m-Trifluorometylbenzoyl (Mb)-pseudokobuisne 11-Anisoyl (As)-pseudokobusine 8-O-Azeloyl-14-benzoylaconine
Yesoxine Dehydrolucidusculine 1,12,15-Triacetylluciculine 12-Acetylluciculine Kobusine Pseudokobusine 15-Veratroylpseudokobusine N,15-dibenzyl-N,6-seco-6-dehydropseudokobusine 6,11-Dibenzoylpseudokobusine 6-Veratroylpseudokobusine 11-Veratroylpseudokobusine 11-Cinnamoylpseudokobusine 11-Anisoylpseudokobusine 11-p-Nitrobenzoylpseudokobusine 1–7a 8a 9a 10a 11a 12a 13a a
C19-derivative
C19-derivative
C20-derivative C20-derivative C19
[60]
C20 C20 C20-derivative C20-derivative C20 C20 C20 C20-derivative C20-derivative C20-derivative C20-derivative C20-derivative C20-derivative C20-derivative [59] C20 C20 C20 C20 C20 C20
[61]
[57]
The number of the compounds are shown in Fig. 1.
DDAs disappeared readily in rat plasma after oral administration, however MDAs could be detected in plasma, indicating that there was a strong first-pass effect of DDAs thus DDAs might be transform into MDAs in vivo [76,77]. Enzyme immunoassay results also showed that aconitum alkaloids such as AC, MA and HA hydrolyzed to the metabolites with less toxicity in rats, thus suggesting that these metabolic processes in vivo are detoxication [78]. The alkaloid detoxicity metabolic process might be due to the efflux transporters and metabolic enzymes in the intestine and liver to protect the body be not suffered from the invasion xenobiotics, especial toxicants. P-glycoprotein (P-gp), intestinal bacteria and cytochrome P450 are responsible to impede intestinal absorptions of diterpenoid alkaloids, which may be the detoxicity mechanisms of DAs after oral. Hence, the three main interact ways of DAs metabolization are summarized as follows. 4.1. Drug–drug interactions In traditional Chinese medicine formula contains Radix aconiti and Aconitum Carmichael, other herbs are always added for reducing their toxicity. It was found that in the combined extracts of Radix Glycyrrhizae and A. carmichaelii Debeaux, the contents of DDAs and LDAs increased while the MDAs decreased [79]. In an everted gut sac permeability experiment, three DDAs in co-decoction extracts of Radix Glycyrrhizae and A. carmichaelii Debeaux firstly dissolved in gastrointestinal tract and then was absorbed in blood after oral administration, that is to say, in this way it can avoid dose dumping thus achieve the purpose of detoxification [14]. Liu et al. determined the contents of DDAs in Radix aconiti before and after co-decoction with 9 kinds of Chinese medicines by a semi-quantitative ESI–MS
method. The results revealed that the DDAs increased a lot in extracts when Radix aconiti co-decoction with Rhizoma pinelliae, Semen trichosanthis, Fructus trichosanthis, Pericarpium trichosanthis, Bulbus fritillariae thunbergli and Rhizoma bletillae, which mean that it should be careful to use several herbs in combined way [80]. Zhang et al. studied the pharmacokinetic parameters of HA in Sini decoction (SND), a formula contains A. carmichaelii, Glycyrrhiza uralensis and Zingiber. They found that alkaloids through HA could be absorbed rapidly in SND; the Tmax , Cmax , k, area under the plasma concentration (AUC) were decreased; T1/2 and mean residence time (MRT) had been lengthened, which indicated Glycyrrhiza/uralensis and Zingiber inhibited the absorptions of HA in SND, and maintained the concentration of HA in a relatively moderate range in vivo [81]. A similar pharmacokinetic behavior of BMA was observed when Aconiti Kusnezoffii Radix Cocta decocting together with Piperis Longi Fructus and Chebulae Fructu, wherein a decreased absorption rate of BMA was appeared [82]. However, as Chinese medicine has the multi-compound characteristics, therefore many of the mechanisms such as synergistic effect, drug–drug interactions as well as incompatibilities are still unclear, consequently further researches should be performed. 4.2. Diterpenoid alkaloids impacted by P-glycoprotein P-gp as an efflux transporter plays a key role for reducing toxicity of DAs. The efflux transport ratio of DAs could be adjusted based on the two factors, safety and therapeutic effect, therefore, the performance of DAs affected by P-gp is important. DAs absorption increased in the presence of P-gp inhibitors in vivo. It has reported that HA was not only an inhibiter but also a substrate
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
7
Table 2 The effects of P-gp inbititors on the efflux ratios of the Aconitium alkaloids. Drug
Inhibitor
Efflux ratio (Pb-a/Pa-b)
Aconitine
— Cyclosporin A Verapamil — Cyclosporin A Verapamil — Cyclosporin A Verapamil — Cyclosporin A Verapamil — Cyclosporin A Verapamil — Cyclosporin A Verapamil — Cyclosporin A Verapamil — Cyclosporin A Verapamil
34.6 1.0 1.1 29.7 1.0 1.1 15.6 1.0 0.9 5.0 1.3 2.7 4.0 1.0 1.9 4.3 1.3 3.3 1.1 0.6 0.9 1.2 0.7 0.8
Mesaconitine
Hypaconitine
Benzoylaconitine
Benzoylmesaconine
Benzoylhypacoitine
Aconine
Mesaconine
of P-gp [83]. The human colonic adenocarcinoma Caco-2 cell line has been widely used for simulating drug absorption to understand transport mechanisms in intestinal environment. The efflux ratios (Er) of AC, MA, and HA in Caco-2 cells were 34.6 ± 4.2, 29.7 ± 2.1, and 15.6 ± 2.1, respectively, while all their hydrolysates (BAC, BMA, BHA) were close to 4 (See Table 2) [84,85]. With the presence of Pgp inhibiters (i.e., verapamil and cyclosporinA), all the Er values of AC, MA, HA, BAC and BHA significantly decreased (nearly to 1.0) [86].The results proved that P-gp was involved respectively in the transports of AC, MA, HA, BAC and BHA. Not only DDAs but also MDAs were substrates of P-gp [75]. Additionally, the toxicity of AC was decreased in rats after co-administration with paeoniflorin that is a substrate of P-gp [87], and an in silico docking model of the interaction between P-gp and AC constructed strongly suggested that AC is a specific substrate of P-gp [88]. 4.3. Diterpenoid alkaloids metabolized by cytochrome P450 Cytochromes P450 (CYP) enzymes are the most important phase-I drug metabolizing enzymes. They are responsible for the oxidation of drug metabolism and other xenobiotics [89]. A great deal of studies shows that C19-diterpenoid alkaloids are primarily metabolized by CYP enzymes. Many isoforms of CYP inhibitors, such as CYP3A, on AC metabolism depended to concentration have been observed in rat liver microsomes. Another one in rat liver microsomes affecting AC metabolism was CYP1A1/2 isoform. Among the isoforms of CYP, CYP3A is the most important, which is highly expressed in critical tissues to be responsible for drug metabolism in vivo [90]. While CYP2B1/2, 2D and 2E1 isoforms had no obvious inhibitory effects on aconitine metabolism [91]. A similar result was obtained to HA which was mainly metabolized by CYP3A, while CYP2C, CYP1A2, CYP2D, CYP2E1 had less responsible to the metabolism reaction of HA [92]. Continuing their investigation on the aconitum alkaloids metabolism, Tang et al. discovered that HA was metabolized primarily by CYP3A4 and 3A5, next by CYP2C19, 2D6, and CYP2E1. MA might be metabolized by CYP3A4, CYP3A5, CYP2C8, CYP2C9, and CYP2D6 [93]. Interestingly, a research conducted by Tang et al. showed that both CYP3A activity and protein levels were not significantly affected by aconitine after oral administration, indicating that aconitine neither inhibiting nor inducing CYP3A activity thus
± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ± ±
4.2 0.02* 0.01* 2.1 0.05* 0.03* 2.1 0.01* 0.05* 1.4 0.3* 1.3 2.4 0.3 0.6 0.9 0.5* 1.3 0.4 0.2 0.6 0.5 0.3 0.6
does not cause CYP3A-related drug–drug interaction in the live of rats [94]. Hu et al. used three probe drugs to test the activity of CYP enzymes after multiple oral administrations of aconitine in rats (phenacetin for CYP1A2, omeprazole for CYP2C19 and midazolam for CYP3A4). Compared with pre-administration of probe drugs, T1/2 and Tmax of phenacetin decreased, however, T1/2 and Tmax of omeprazole and midazolam increased, which implied that aconitine could increase the activity of CYP1A2 [95]. From above, it can be concluded that aconitine has a significant inhibition effect on the activities of CYP2C19. As for MDAs, previous research proved that BAC, BMA, and BHAC could be metabolized by CYP3A4 and CYP3A5 [95]. Bi et al. found that the components of MDAs had strong inhibitory effects on the activities of CYP2C and CYP 2D. The IC50 values were 7.44 and 6.74 mol L−1 , respectively. The components of DDAs had weaker inhibitory effects on the activities of CYP1A2, 3A, 2C and 2D, and their IC50 values were 39.48, 70.44, 17.136 and 86.04 mol L−1 , respectively [96]. Briefly, DDAs and MDAs can be transformed into less toxic metabolites by CYP3A4 and CYP3A5, which could greatly reduce the toxicity of Aconitum plants, prevent Aconitum alkaloids to be absorbed excessively into bloodstream [97]. An in silico docking study showed that verapamil and aconitine had similar residues and similar interaction ways with P-gp. Verapamil was mainly metabolized by CYP3A, which may be a corroborative evidence indicating that aconitine was also metabolized by CYP3A [88]. Recently, with the application of LCMS, metabolites can be detected and identified in a low LOD. After analyzing the metabolites, the metabolism pathways for HA were demethylation, dehydrogenation, hydroxylation, and didemethylation conducted by those CYP isoforms, and a generally similar pathway for AC, an additional dehydrogenation pathway for MA compared with HA. [93,98,99]. 4.4. Diterpenoid alkaloids decomposed by intestinal bacteria Human intestinal is rich with enzymes, acting as a metabolic system like human liver. The bioavailabilities of some prototype drugs are low, but their metabolites can be absorbed with a benign pharmacological activity. The small intestine provides alkalescent drugs an optimal pH environment for absorption. With an everted gut sac permeability experiment, the results showed that DDAs such as AC, HA and MA had steady effective absorption rates (Ka)
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
8
and apparent permeabilities (Papp) in ileum [14]. So the study on the metabolites about DAs metabolized by intestinal bacteria is of great significance. In our group, some techniques, such as mass spectrometer, have been applied to study the Aconitum alkaloids metabolized by intestinal bacteria in vitro. It was found that diester deoxyaconitine was transformed into monoester alkaloids, diester- and lipo-alkaloids under the influence of human intestinal bacteria, in which the toxicities were reduced. Deoxyaconitine was converted into more than ten kinds of new metabolites by deacetylation, debenzoylation, dehydroxylation, demethylation and esterification [100]. With use of ion trap and Fourier transform ion cyclotron resonance electrospray ionization tandem mass spectrometry, Liu and co-workers found that 16-Odemethylaconitine from biotransformation of aconitine could be further transformed into mone-ester alkaloids and lipo-alkaloids by human intestinal bacteria [101,102]. Aconitum alkaloids were metabolized by intestines bacteria through not only decomposition but also addition reactions to lead to that a new group might be added at the C-8 position to prolong the branch chain. The rat intestinal flora can further reduce the toxicity of DDAs and increase the contents of lipo-alkaloids. Aconitine was converted into other DDAs or MDAs by the bacteria metablism [103]. Through comparative analysis of different pH levels, aconitine had the highest metabolic activity at pH 7.0, which is close to the pH condition of rat intestinal. However, mesaconitine was metabolized more easily than aconitine because of different groups linked at N heterocyclic position of aconitine [104,105]. 5. The pharmacological mechanisms of diterpenoid alkaloids 5.1. Mechanism of cardiovascular action The electrophysiological mechanism of arrhythmia is due to delayed after-depolarization and early after-depolarization. Aconitine [106] and lappaconitine [107] bind with high affinity to the open gate of Na+ channels at receptor site 2 of the cell membranes, and prolong the sodium influx during the action potential, in which lappaconitine could block sodium channel in cloned human heart (hH1) irreversibly. The sustained Na+ influx delays the repolarization phase of the action potential and initiates premature excitation to cause persistent activation of these channels [108–110]. Seventeen diterpene alkaloids obtained from Aconitum toxicum and Consolida orientalis were investigated by Judit Hohmann on G protein gated inwardly rectififying K channels (GIRK), of which aconitine exerted 45 ± 9% GIRK inhibition at 10 mol L−1 , which showed significant blocking activity compared with control measurements [111]. These results suggest that AC possess a more complex cardiac action, with which multiple ion channels might be affected. 5.2. Mechanisms of anti-inflammatory and analgesic The mechanism of anti-inflammatory activity of alkaloids is related to the ability of alkaloids inhibiting the generation of inflammatory mediators. The inhibition action suppresses the secretion of proinflammatory cytokines, such as histamine, serotonin, arachidonic acid, TNF-␣ and IL-6 [16]. Alkaloids had less effect on prostaglandin synthase, thus few ulcerogenic effects were observed after the alkaloids used. C19-diterpenoid alkaloids such as MA, AC and HA decreased TNF-␣ level and revealed a potent analgesic effect, which played an important role in anti-inflammatory [112,113]. The DAs together with their metabolites exhibited analgesic or neurotoxic effects through the central nervous system as these
compounds distribute in the spinal cord [78]. Neurotoxic effects and the corresponding mechanisms of aconitine were conducted by Peng et al. on cerebral cortex neuron cells. The results showed that aconitine increased the release of substance P and opioid peptide, which inhibited Na+ -K+ -ATPase activity to lead increase of the concentrations of Na+ as well as the related Ca2+ , but decreases of the concentrations of K+ as well as the related Mg2+ . The increased opioid peptide stated above could raise the pain threshold, while substance P possesses analgesia function. The increased Ca2+ , say it is overload, would lead to the damage of the cellular form and change the release of correlative neurotransmitters, resulting in neurological symptoms after oral aconitine [114]. However, it has been reported that some DAs, such as bullatine A, AC, MA and HA, possessed both central and peripheral antinociceptive effects and had potential anti-inflammatory activities, indicating that the anti-inflammatory of DAs were not via PGE2 pathway with Na+ channel interference [115]. Recently, Dong group reported that the DAs screened from Fuzilizhong Pills could suppress NF-B activity, which demonstrate that the Aconitum alkaloid derivatives can be regarded as new diester-diterpenoid-type NF-B inhibitors [116]. 5.3. Mechanisms of anti-tumor Usually, various tumor treatment agents like vincristine possess anti-tumor properties through inhibiting the growth of deregulating lesions and promoting apoptosis in tumor cells. For these reasons, the anti-tumor drugs often have damage to normal cells with their cytotoxicity that interfere the DNA synthesis during the cell divisions. Two C20-diterpenoid alkaloids, 11-m-trifluoromethylbenzoyl–pseudokobusine and 11anisoyl-pseudokobusine, caused no remarkable G2/M arrest, which was different from the disruption of microtubule function or DNA damage. 11-m-trifluoromethylbenzoyl–pseudokobusine acted as a G1/S mediator through the Erk and PI3K pathway in Raji cells, with down-regulate Erk-activation and inhibit PI3K-activation. The results above revealed that the DAs have lower toxicity to hematopoiesis through regulating the tumor cells enzymes systems [60]. It was reported that neoline, pubescenine, 14-de-acetylajadine, lycoctonine and dehydrotakaosamine had an irreversible effect on SW480. In order to gain insights into the mechanism of irreversible cytotoxic action of these compounds, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and acid phosphatase (AP) methods were compared to evaluate the cell viability. The results suggested that the mode of action of these compounds might relate to the inhibition of ATP production. This will explain SW480 cells that whose resistance mechanism are related to higher energy demand, are the most sensitive to these compounds [117]. 5. Future prospects Natural products have been drawn attentions in recent years, thus lots of compounds isolated from natural products have already been confirmed with excellent pharmacological efficacies. Meanwhile, heterocyclic alkaloids with diterpene structure owning varieties of pharmacological activities including analgesic, anti-inflammatory, anti-microbial, anti-tumor, cardiotonic and anti-arrhythmic activities, as well as bioactivities like antiparasite, antioxidant ability [12] were also discovered. Further investigations should be pursued on the basic mechanisms of actions of diterpenoid alkaloids, and based on the mechanisms to synthesize “drug-like” alkaloids. In addition, new analysis methods like multivariate statistical analysis opens a new perspective in drug metabolism, which offer a comprehensive strategy to find out chemical markers without much process and has applied in
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
xenobiotic and endogenic metabolome [33,118,119]. QSAR technique together with quantum chemistry calculation [67–69,120], as a supplementary method, provided the binding energies and structures of the diterpenoid alkaloids to verify experimental data should be used to couple with MS and enzyme assay, and so on.
Acknowledgements This research work was supported by the National Natural Science Foundation of China (No. 81274046, 81073040) and National Basic Research Program of China (973 Program) (No. 2011CB505300, 2011CB505305).
[19]
[20]
[21]
[22]
[23]
[24]
References [1] H. Yue, Z.F. Pi, F.R. Song, Z.Q. Liu, Z.W. Cai, S.Y. Liu, Studies on the aconitine-type alkaloids in the roots of Aconitum carmichaeli Debx. by HPLC/ESIMS/MSn, Talanta 77 (2009) 1800–1807. [2] F.P. Wang, Q.H. Chen, X.Y. Liu, Diterpenoid alkaloids, Prod. Rep. 27 (2010) 529–570. [3] H. Yue, Z.F. Pi, H.L. Li, F.R. Song, Z.Q. Liu, S.Y. Liu, Studies on the stability of diester-diterpenoid alkaloids from the genus Aconitum L. by high performance liquid chromatography combined with electrospray ionisation tandem mass spectrometry (HPLC/ESI/MSn ), Phytochem. Anal. 19 (2008) 141–147. [4] Y. Wang, F.R. Song, Q.X. Xu, Z.Q. Liu, S.Y. Liu, Characterization of aconitine-type alkaloids in the flowers of Aconitum kusnezoffii by electrospray ionization tandem mass spectrometry, J. Mass Spectrom. 38 (2003) 962–970. [5] Y. Wang, F.R. Song, D.M. Jin, Z.Q. Liu, S.Y. Liu, Studies on the aconitum alkaloids in the Sini concoction by electrospray ionization mass spectrometry, Chem. J. Chin. Univ. 25 (2004) 85–89. [6] Y. Wang, Z.Q. Liu, F.R. Song, S.Y. Liu, Electrospray ionization tandem mass spectrometric study of the aconitines in the roots of aconite, Rapid Commun. Mass Spectrom. 16 (2002) 2075–2082. [7] Y. Wang, S.Y. Liu, Z.Q. Liu, F.R. Song, D.M. Jin, C.M. Liu, Analysis of aconitum alkaloids in the roots of Aconitum brachypudum by electrospray ionization mass spectrometry, Chin. J. Anal. Chem. 31 (2003) 139–142. [8] W.X. Sun, S.Y. Liu, Z.Q. Liu, F.R. Song, S.P. Fang, A study of Aconitum alkaloids from aconite roots in Aconitum carmichaeli Debx using matrix-assisted laser desorption ionization mass spectrometry, Rapid Commun. Mass Spectrom. 12 (1998) 821–824. [9] G.L. Yan, H. Sun, W.J. Sun, L. Zhao, X.C. Meng, X.J. Wang, Rapid and global detection and characterization of aconitum alkaloids in Yin Chen Si Ni Tang, a traditional Chinese medical formula, by ultra performance liquid chromatography–high resolution mass spectrometry and automated data analysis, J. Pharm. Biomed. Anal. 53 (2010) 421–431. [10] H. Sato, C. Yamada, C. Konno, Y. Ohizumi, K. Endo, H. Hikino, Pharmacological actions of aconitine alkaloids, Tohoku J. Exp. Med. 128 (1979) 175–187. [11] M. Hazawa, K. Wada, I. Kashiwakura, Aconitum C20-diterpenoid alkaloid derivatives regulate the growth and differentiation of cord blood derived human hematopoietic stem/progenitor cells, Phytother. Res. 26 (2012) 1262–1264. [12] M. Reina, A. González-Coloma, Structural diversity and defensive properties of diterpenoid alkaloids, Phytochem. Rev. 6 (2007) 81–95. [13] F. Moritz, P. Compagnon, I.G. Kaliszczak, Y. Kaliszczak, V. Caliskan, C. Girault, Severe acute poisoning with homemade Aconitum napellus capsules: toxicokinetic and clinical data, Clin. Toxicol. 43 (2005) 873–876. [14] J.M. Zhang, W. Liao, Y.X. He, Y. He, D. Yan, C.M. Fu, Study on intestinal absorption and pharmacokinetic characterization of diester diterpenoid alkaloids in precipitation derived from Fuzi-Gancao herb-pair decoction for its potential interaction mechanism investigation, J. Ethnopharmacol. 147 (2013) 128–135. [15] L. Tang, Y. Gong, C. Lv, L. Ye, L. Liu, Z.Q. Liu, Pharmacokinetics of aconitine as the targeted marker of Fuzi (Aconitum carmichaeli) following single and multiple oral administrations of Fuzi extracts in rat by UPLC/MS/MS, J. Ethnopharmacol. 141 (2012) 736–741. [16] F. Zhao, Z.T. Gao, W.H. Jiao, L.P. Chen, L. Chen, X.S. Yao, In vitro anti-inflammatory effects of beta-carboline alkaloids, isolated from Picrasma quassioides, through Inhibition of the iNOS Pathway, Planta Med. 78 (2012) 1906–1911. [17] M. Fu, M. Wu, J.F. Wang, Y.J. Qiao, Z. Wang, Disruption of the intracellular Ca2+ homeostasis in the cardiac excitation-contraction coupling is a crucial mechanism of arrhythmic toxicity in aconitine-induced cardiomyocytes, Biochem. Biophys. Res. Commun. 354 (2007) 929–936. [18] Y. Nakamura, K. Yomura, T. Kammoto, M. Ishimatsu, Y. Kikuchi, K. Niitsu, S. Terabayashi, S. Takeda, H. Sasaki, K. Arimoto, M. Okada, S. Sekita, M. Satake, Y. Goda, Physicochemical quality evaluation of natural compounds isolated from crude drugs—standard compounds for the official specification and
[25]
[26]
[27]
[28]
[29]
[30]
[31]
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
9
testing method of processed aconite root and powdered processed aconite root in the Japanese pharmacopoeia, J. Nat. Med. Tokyo 60 (2006) 285–294. Z. Jian-guo, L. Fei, X. Xiao-long, L. Yan-yan, P. Hai-long, W. Hai-peng, L. Ping, Optimization of microwave-assisted extraction of total alkaloids from semen strychni, Chin. Food Sci. 31 (2010) 116–119. M. Then, K. Szentmihalyi, A. Sarkozi, V. Illes, E. Forgacs, Effect of sample handling on alkaloid and mineral content of aqueous extracts of greater celandine (Chelidonium majus L.), J. Chromatogr. A 889 (2000) 69–74. T. Mroczek, K. Glowniak, J. Kowalska, Solid–liquid extraction and cation-exchange solid-phase extraction using a mixed-mode polymeric sorbent of Datura and related alkaloids, J. Chromatogr. A 1107 (2006) 9–18. C.H. Deng, N. Liu, M.X. Gao, X.M. Zhang, Recent developments in sample preparation techniques for chromatography analysis of traditional Chinese medicines, J. Chromatogr. A 1153 (2007) 90–96. Q.F. Tang, C.H. Yang, W.C. Ye, J.H. Liu, S.X. Zhao, Preparative isolation and purification of chemical components from Aconitum coreanum by high-speed counter-current chromatography coupled with evaporative light scattering detection, Phyt. Anal. 19 (2008) 155–159. X.Y. Wang, X.K. Shu, X. Wang, J.Q. Yu, F. Jing, Preparative isolation of seven diterpenoid alkaloids from Aconitum coreanum by pH-zone-refining counter-current chromatography, Molecules 19 (2014) 12619–12629. D.H. Liu, X.K. Shu, X. Wang, L. Fang, L.Q. Huang, X.J. Xi, Z.J. Zheng, Preparative separation of C-19-diterpenoid alkaloids from Aconitum carmichaeliii debx by pH-zone-refining counter-current chromatography, Quim. Nova 36 (2013), 1366–U332. J.J. Liu, Q. Li, Y.D. Yin, R. Liu, H.R. Xu, K.S. Bi, Ultra-fast LC-ESI–MS/MS method for the simultaneous determination of six highly toxic Aconitum alkaloids from Aconiti kusnezoffii radix in rat plasma and its application to a pharmacokinetic study, J. Sep. Sci. 37 (2014) 171–178. X. Li, Q.Q. Wang, C. Xu, F.R. Song, Z.Q. Liu, The method and application on mass spectrometry analysis of Chinese medicine research, Sci. Sin. Chim. 44 (2014) 701–709. J.Y. Ding, X.X. Liu, D.M. Xiong, L.M. Ye, R.B. Chao, Simultaneous determination of thirteen aminoalcohol-diterpenoid alkaloids in the lateral roots of Aconitum carmichaeli by solid-phase extraction-liquid chromatography–tandem mass spectrometry, Planta Med. 80 (2014) 723–731. W. Wu, Z.T. Liang, Z.Z. Zha, Z.W. Cai, Direct analysis of alkaloid profiling in plant tissue by using matrix-assisted laser desorption/ionization mass spectrometry, J. Mass Spectrom. 42 (2007) 58–69. R. Hu, J. Zhao, L.W. Qi, P. Li, S.L. Jing, H.J. Li, Structural characterization and identification of C-19- and C-20-diterpenoid alkaloids in roots of Aconitum carmichaeli by rapid-resolution liquid chromatography coupled with time-of-flight mass spectrometry, Rapid Commun. Mass Spectrom. 23 (2009) 1619–1635. J. Zhang, Z.H. Huang, X.H. Qiu, Y.M. Yang, D.Y. Zhu, W. Xu, Neutral fragment filtering for rapid identification of new diester-diterpenoid alkaloids in roots of Aconitum carmichaeli by ultra-high-pressure liquid chromatography coupled with linear ion trap-orbitrap mass spectrometry, PloS One (2012). W. Xu, J. Zhang, D.Y. Zhu, J. Huang, Z.H. Huang, J.Q. Bai, X.H. Qiu, Rapid separation and characterization of diterpenoid alkaloids in processed roots of Aconitum carmichaeli using ultra high performance liquid chromatography coupled with hybrid linear ion trap-Orbitrap tandem mass spectrometry, J. Sep. Sci. 37 (2014) 2864–2873. H.B. Zhu, C.Y. Wang, Y. Qi, F.R. Song, Z.G. Liu, S.Y. Liu, Rapid quality assessment of Radix Aconiti Preparata using direct analysis in real time mass spectrometry, Anal. Chim. Acta 752 (2012) 69–77. F. Zhou, H.B. Zhu, S. Liu, K. Ma, F.R. Song, In situ analysis for herbal pieces of Aconitum plants by using direct analysis in real time mass spectrometry, Chin. J. Chem. 33 (2015) 241–246. X. Li, G.Y. Hou, J.P. Xing, F.R. Song, Z.Q. Liu, S.Y. Liu, Direct analysis in real time-mass spectrometry for the rapid detection of metabolites of aconite alkaloids in intestinal bacteria, J. Am. Soc. Mass Spectrom. 25 (2014) 2181–2184. J.Z. Song, Q.B. Han, C.F. Qiao, P.P.H. But, H.X. Xu, Development and validation of a rapid capillary zone electrophoresis method for the determination of aconite alkaloids in aconite roots, Phyt. Anal. 21 (2010) 137–143. Y. Bao, F. Yang, X.R. Yang, CE-electrochemiluminescence with ionic liquid for the facile separation and determination of diester-diterpenoid aconitum alkaloids in traditional Chinese herbal medicine, Electrophoresis 32 (2011) 1515–1521. Y. Xie, Z.H. Jiang, H. Zhou, H.X. Xu, L. Liu, Simultaneous determination of six Aconitum alkaloids in proprietary Chinese medicines by high-performance liquid chromatography, J. Chromatogr. A 1093 (2005) 195–203. F. Zhang, M.H. Tang, L.J. Chen, R. Li, X.H. Wang, J.G. Duan, X. Zhao, Y.Q. Wei, Simultaneous quantitation of aconitine, mesaconitine, hypaconitine, benzoylaconine, benzoylmesaconine and benzoylhypaconine in human plasma by liquid chromatography–tandem mass spectrometry and pharmacokinetics evaluation of SHEN-FU injectable powder, J. Chromatogr. B 873 (2008) 173–179. H.W. He, F. Yan, Relative quantification of the metabolite of aconitine in rat urine by LC-ESI–MS/MS and its application to pharmacokinetics, Anal. Sci. 28 (2012) 1203–1205. J. Beike, L. Frommherz, M. Wood, B. Brinkmann, H. Kohler, Determination of aconitine in body fluids by LC–MS–MS, Int. J. Legal. Med. 118 (2004) 289–293.
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11 10
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
[42] H. Hattori, Y. Hirata, M. Hamajima, R. Kaneko, K. Ito, A. Ishii, O. Suzuki, H. Seno, Simultaneous analysis of aconitine, mesaconitine, hypaconitine, and jesaconitine in whole blood by LC–MS–MS using a new polymer column, Forensic Toxicol. 27 (2009) 7–11. [43] H.Q. Wu, X.T. Xiong, X.L. Huang, Z.X. Zhu, F. Huang, X.S. Lin, Simultaneous determination of 17 toxic alkaloids in human fluids by liquid chromatography coupled with electrospray ionization tandem mass spectrometry, J. Liq. Chromatogr. Rel. Techn. 36 (2013) 1149–1162. [44] Y.V. Nesterova, T.N. Povetieva, N.I. Suslov, G.N. Zyuz’kov, S.G. Aksinenko, S.V. Pushkarskii, A.V. Krapivin, Anti-inflammatory activity of diterpene alkaloids from Aconitum baikalense, Bull. Exp. Biol. Med. 156 (2014) 665–668. [45] M.C. Lai, I.M. Liu, S.S. Liou, Y.S. Chang, Mesaconitine plays the major role in the antinociceptive and anti-inflammatory activities of radix Aconiti Carmichaeli (Chuan Wu), J. Food Drug Anal. 19 (2011) 362–368. [46] H.Y. D.P. Wang, L. Lou, X.J. Huang, G.Y. Hao, Z.C. Liang, W.D. Pan Yang, A novel franchetine type norditerpenoid isolated from the roots of Aconitum Carmichaeli Debx. with potential analgesic activity and less toxicity, Bioorg. Med. Chem. Lett. 22 (2012) 4444–4446. [47] J.M. Jacyno, J.S. Harwood, N.H. Lin, J.E. Campbell, J.P. Sullivan, M.W. Holladay, Lycaconitine revisited: partial synthesis and neuronal nicotinic acetylcholine receptor affinities, J. Nat. Prod. 59 (1996) 707–709. [48] M.S. Yunusov, Antiarrhythmic agents based on diterpenoid alkaloids, Russ. Chem. B+ 60 (2011) 633–638. [49] T.G. Tolstikova, A.O. Bryzgalov, I.V. Sorokina, S.A. Osadchii, E.E. Shults, M.P. Dolgikh, M.V. Khvostov, Solification with hydrobromic acid as a factor defining the antiarrhythmic effect of lappaconitine derivatives, Lett. Drug Des. Discov. 6 (2009) 475–477. [50] Y.V. Vakhitova, E.I. Farafontova, R.Y. Khisamutdinova, V.M. Yunusov, I.P. Tsypysheva, M.S. Yunusov, A study of the mechanism of the antiarrhythmic action of Allapinin, Russ. J. Bioorg. Chem. 39 (2013) 92–101. [51] Q.L. Zhang, J.H. Hu, Z.P. Jia, D. Wang, Q.G. Zhu, Pharmacokinetics of aconitine in rat skin after oral and transdermal gel administrations, Biomed. Chromatogr. 26 (2012) 622–626. [52] F.N. Dzhakhangirov, F.M. Tursunkhodzhaev, M.N. Sultankhodzhaev, B.T. Salimov, Spasmlytic activity of diterpenoid alkaloids and their derivatives structure-activity relationship, Vet. Hum. Toxicol. 49 (2013) 702–706. [53] B. Sener, I. Orhan, B. Ozcelik, Diterpenoid alkaloids from some Turkish Consolida species and their antiviral activities, Arkivoc (2007) 265–272. [54] M. Ahmad, W. Ahmad, M. Ahmad, M. Zeeshan, F. Obaidullah, F. Shahee, Norditerpenoid alkaloids from the roots of Aconitum heterophyllum wall with antibacterial activity, J. Enzym. Inhib. Med. Chem. 23 (2008) 1018–1022. [55] M. Kobayashi, H. Takahashi, D.N. Herndon, R.B. Pollard, F. Suzuki, Therapeutic effects of IL-12 combined with benzoylmesaconine, a non-toxic aconitine-hydrolysate, against herpes simplex virus type 1 infection in mice following thermal injury, Burns 29 (2003) 37–42. [56] D.K. Kim, H.Y. Kwon, K.R. Lee, D.K. Rhee, O.P. Zee, Isolation of a multidrug resistance inhibitor from Aconitum pseudo-laeve var. erectum, Arch. Pharmacal. Res. 21 (1998) 344–347. [57] A. Chodoeva, J.J. Bosc, J. Guillon, A. Decendit, M. Petraud, C. Absalon, C. Vitry, C. Jarry, J. Robert, 8-O-Azeloyl-14-benzoylaconine: a new alkaloid from the roots of Aconitum karacolicum Rapcs and its antiproliferative activities, Bioorgan. Med. Chem. 13 (2005) 6493–6501. [58] A. Chodoeva, J.J. Bosc, J. Guillon, P. Costet, A. Decendit, J.M. Merillon, J.M. Leger, C. Jarry, J. Robert, Hemisynthesis and antiproliferative properties of mono-[O-(14-benzoylaconine-8-yl)] esters and bis-[O-(14-benzoylaconine-8-yl)] esters, Eur. J. Med. Chem. 54 (2012) 343–351. [59] M. Hazawa, K. Wada, K. Takahashi, T. Mori, N. Kawahara, I. Kashiwakura, Suppressive effects of novel derivatives prepared from Aconitum alkaloids on tumor growth, Invest. New Drug 27 (2009) 111–119. [60] M. Hazawa, K. Takahashi, K. Wada, T. Mori, N. Kawahara, I. Kashiwakura, Structure-activity relationships between the Aconitum C-20-diterpenoid alkaloid derivatives and the growth suppressive activities of Non-Hodgkin’s lymphoma Raji cells and human hematopoietic stem/progenitor cells, Invest. New Drug 29 (2011) 1–8. [61] R. Pullela, L. Young, B. Gallagher, S.P. Avis, E.W. Randell, A case of fatal aconitine poisoning by Monkshood ingestion, J. Forensic Sci. 53 (2008) 491–494. [62] K. Wada, M. Nihira, H. Hayakawa, Y. Tomita, M. Hayashida, Y. Ohno, Effects of long-term administrations of aconitine on electrocardiogram and tissue concentrations of aconitine and its metabolites in mice, Forensic Sci. Int. 148 (2005) 21–29. [63] M. Taki, K. Niitu, Y. Omiya, M. Noguchi, M. Fukuchi, M. Aburada, M. Okada, 8-O-cinnamoylneoline, a new alkaloid from the flower buds of Aconitum carmichaeli and its toxic and analgesic activities, Planta. Med. 69 (2003) 800–803. [64] N.G. Bisset, Arrow poisons in China. part ii. aconitum-botany, chemistry, and pharmacology, J. Ethnopharmacol. 4 (1981) 247–336. [65] A. Ameri, The effects of Aconitum alkaloids on the central nervous system, Prog. Neurobiol. 56 (1998) 211–235. [66] K. Xiao, L. Wang, Y. Liu, C. Peng, G. Yan, J. Zhang, Y. Zhuo, H. Li, Study of aconitine toxicity in rat embryos in vitro, Birth Defects Res. 80 (2007) 208–212. [67] A.M. Bello-Ramirez, A.A. Nava-Ocampo, A QSAR analysis of toxicity of Aconitum alkaloids, Fundam. Clin. Pharmacol. 18 (2004) 699–704.
[68] M.A. Turabekova, B.F. Rasulev, A QSAR toxicity study of a series of alkaloids with the lycoctonine skeleton, Molecules (2004) 1194–1207. [69] M.A. Turabekova, B.F. Rasulev, F.N. Dzhakhangirov, S.I. Salikhov, Aconitum and Delphinium alkaloids drug-likeness descriptors related to toxic mode of action, Environ. Toxicol. Pharmacol. 25 (2008) 310–320. [70] J.K. Nicholson, J.C. Lindon, E. Holmes, Metabonomics’: understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data, Xenobiotica 29 (1999) 1181–1189. [71] Y. Qi, S.Z. Li, Z.F. Pi, F.R. Song, N. Lin, S. Li, Z.Q. Liu, Metabonomic study of Wu-tou decoction in adjuvant-induced arthritis rat using ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry, J. Chromatogr. B 953 (2014) 11–19. [72] H.B. Zhu, C.Y. Wang, Y. Qi, F.R. Song, Z.Q. Liu, S.Y. Liu, Fingerprint analysis of Radix Aconiti using ultra-performance liquid chromatography-electrospray ionization/tandem mass spectrometry (UPLC-ESI/MSn) combined with stoichiometry, Talanta 103 (2013) 56–65. [73] H. Dong, A. Zhang, H. Sun, H. Wang, X. Lu, M. Wang, B. Ni, X. Wang, Ingenuity pathways analysis of urine metabolomics phenotypes toxicity of Chuanwu in Wistar rats by UPLC-Q-TOF-HDMS coupled with pattern recognition methods, Mol. Biosyst. 8 (2011) 1206–1221. [74] X. Wang, H. Wang, A. Zhang, X. Lu, H. Sun, H. Dong, P. Wang, Metabolomics study on the toxicity of aconite root and its processed products using UPLC-ESI synapt HDMS coupled with pattern recognition approach and ingenuity pathways, J. Proteome Res. 11 (2011) 284–1301. [75] H. Zhang, Q. Wu, W.H. Li, S. Sun, W. Zhang, Z.Y. Zhu, G.Q. Zhang, Y.F. Chai, Absorption and metabolism of three monoester-diterpenoid alkaloids in Aconitum carmichaeli after oral administration to rats by HPLC–MS, J. Ethnopharmacol. 154 (2014) 645–652. [76] Q. Zhang, Y.M. Ma, Z.T. Wang, C.H. Wang, Pharmacokinetics difference of multiple active constituents from decoction and maceration of Fuzi Xiexin Tang after oral administration in rat by UPLC-MS/MS, J. Pharm. Biomed. Anal. 92 (2014) 35–46. [77] W.L. Liu, Z.Q. Liu, F.R. Song, S.Y. Liu, Specific conversion of diester-diterpenoid Aconitum alkaloids components into hydrolysis monoester-diterpenoid alkaloids components and lipo-alkaloids components, Chem. J. Chin. Univ. 32 (2011) 717–720. [78] F. Zuo, J. Zhao, N. Nakamura, J.J. Gao, T. Akao, M. Hattori, Y. Oomiga, Y. Kikuchi, Pharmacokinetic study of benzoylmesaconine in rats using an enzyme immunoassay system, J. Nat. Med. Tokyo 60 (2006) 313–321. [79] Y. Yang, X.J. Yin, H.M. Guo, R.L. Wang, R. Song, Y. Tian, Z.J. Zhang, Identification and comparative analysis of the major chemical constituents in the extracts of single Fuzi herb and Fuzi-Gancao herb-pair by UFLC-IT-TOF/MS, Chin. J. Nat. Med. 12 (2014) 542–553. [80] W.L. Liu, F.R. Song, Z.Q. Liu, S.Y. Liu, D.F. Zhang, Chemical study on combination taboo of Radix aconiti with Rhizoma pinelliae, Fructus trichosanthis Bulbus fritillariae thunbergli Radix ampelopsis and Rhizoma bletillae, Acta Chim. Sin. 68 (2010) 889–896. [81] W. Zhang, H. Zhang, S. Sun, F.F. Sun, J. Chen, L. Zhao, G.Q. Zhang, Comparative pharmacokinetics of hypaconitine after oral administration of pure hypaconitine, Aconitum carmichaelii extract and sini decoction to rats, Molecules 20 (2015) 1560–1570. [82] Y.D. Yin, J.J. Liu, H.R. Xu, C.X. Lv, Y.Y. Du, Q. Li, K.S. Bi, Simultaneous determination of benzoylmesaconine and piperine in rat plasma after oral administration of Naru-3 by an ultra fast liquid chromatography-tandem mass spectrometry method and its application in a comparative pharmacokinetic study, Anal. Method 10 (2014) 3420–3428. [83] T.J. Han, Y. Liu, Z.F. Pi, Z.Y. Liu, F.R. Song, Z.Q. Liu, Absorption of hypaconitine and p-glycoprotein-mediated drug-hypaconitine interactions by Caco-2 human intestinal cell monolayers, J. Liq. Chromatogr. R. T. 36 (2013) 1207–1220. [84] C. Yang, Z. Li, T. Zhang, F. Liu, J. Ruan, Z. Zhang, Transcellular transport of aconitine across human intestinal Caco-2 cells, Food Chem. Toxicol. 57 (2013) 195–200. [85] T.J. Han, F.R. Song, Z.Y. Liu, Z.F. Pi, Z.Q. Liu, S.Y. Liu, Studies of the intestinal transport of the diterpenoid alkaloids in the Aconitum carmichaeli combined with different medicinal herbs in a Caco-2 cell culture system with UPLC/MS, Acta Chim. Sinica 69 (2011) 1795–1802. [86] L. Ye, X.S. Yang, Z. Yang, S. Gao, T.J. Yin, W. Liu, F. Wang, M. Hu, The role of efflux transporters on the transport of highly toxic aconitine, mesaconitine, hypaconitine, and their hydrolysates, as determined in cultured Caco-2 and transfected MDCKII cells, Toxicol. Lett. 216 (2013) 86–99. [87] Y.F. Fan, Y. Xie, L. Liu, H.M. Ho, Y.F. Wong, Z.Q. Liu, H. Zhou, Paeoniflorin reduced acute toxicity of aconitine in rats is associated with the pharmacokinetic alteration of aconitine, J. Ethnopharmacol. 141 (2012) 701–708. [88] C. Yang, T. Zhang, Z. Li, L. Xu, F. Liu, J. Ruan, K. Liu, Z. Zhang, P-glycoprotein is responsible for the poor intestinal absorption and low toxicity of oral aconitine: in vitro, in situ, in vivo and in silico studies, Toxicol. Appl. Pharmacol. 273 (2013) 561–568. [89] S.K. Kim, R.F. Novak, The role of intracellular signaling in insulin-mediated regulation of drug metabolizing enzyme gene and protein expression, Pharmacol. Therapeut. 113 (2007) 88–120. [90] O. Burk, L. Wojnowski, Cytochrome P450 3A and their regulation, N.-S. Arch. Pharmacol. 369 (2004) 105–124.
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023
G Model CHROMB-19632; No. of Pages 11
ARTICLE IN PRESS T. Xu et al. / J. Chromatogr. B xxx (2015) xxx–xxx
[91] L. Ye, L. Tang, Y. Gong, C. Lv, Z. Zheng, Z. Jiang, Z. Liu, Characterization of metabolites and cytochrome P450 isoforms involved in the microsomal metabolism of aconitine, Xenobiotica 41 (2006) 46–58. [92] Y. Bi, X. Li, Z. Pi, F. Song, Z. Liu, Analysis of hypaconitine’s metabolites and related metabolic CYP isoforms in rat liver microsomal by UPLC-MS/MS, J. Chin. Mass Spectrom. Soc. 34 (2013) 330–337. [93] L. Tang, L. Ye, C. Lv, Z. Zheng, Y. Gong, Z. Liu, Involvement of CYP3A4/5 and CYP2D6 in the metabolism of aconitine using human liver microsomes and recombinant CYP450 enzymes, Toxicol. Lett. 202 (2011) 47–54. [94] L. Zhu, X. Yang, J. Zhou, L. Tang, B. Xia, M. Hu, F. Zhou, Z. Liu, The exposure of highly toxic aconitine does not significantly impact the activity and expression of cytochrome P450 3A in rats determined by a novel ultra performance liquid chromatography-tandem mass spectrometric method of a specific probe buspirone, Food Chem. Toxicol. 51 (2013) 396–403. [95] M. Hu, S. Zheng, Q. Ye, Effect of aconitine on three cytochrome P450 isoforms in rats, Lat. Am. J. Pharm. 33 (2014) 65–70. [96] Y.F. Bi, S. Liu, X. Li, Z.Q. Liu, F.R. Song, Metabolic fingerprint and effects of aconite alkaloid components on the activities of CYP450 isozymes in rat liver microsomes, Chem. J. Chin. Univ. 34 (2013) 2084–2089. [97] L. Ye, X.S. Yang, L.L. Lu, W.Y. Chen, S. Zeng, T.M. Yan, L.N. Dong, X.J. Peng, J. Shi, Z.Q. Liu, Monoester-diterpene Aconitum alkaloid metabolism in human liver microsomes: predominant role of CYP3A4 and CYP3A5, Evid. Based Compl. Alt. 10 (2013) 1–24. [98] L. Ye, T. Wang, C.H. Yang, L. Tang, J. Zhou, C. Lv, Y. Gong, Z.H. Jiang, Microsomal cytochrome P450-mediated metabolism of hypaconitine, an active and highly toxic constituent derived from Aconitum species, Toxicol. Lett. 204 (2011) 81–91. [99] L. Ye, L. Tang, Y. Gong, C. Lv, Z.J. Zheng, Z.H. Jiang, Z.Q. Liu, Characterization of metabolites and human P450 isoforms involved in the microsomal metabolism of mesaconitine, Xenobiotica 41 (2011) 46–58. [100] Y.F. Zhao, F.R. Song, H. Yue, X.H. Guo, H.L. Li, Z.Q. Liu, S.Y. Liu, Biotransformation of deoxyaconitine of metabolite of aconitine by human intestinal bacteria and electrospray ionization tandem mass spectrometry, Chem. J. Chin. Univ. 28 (2007) 2051–2055. [101] Y.F. Zhao, F.R. Song, H. Yue, X.H. Guo, H.L. Li, Z.Q. Liu, S.Y. Liu, Studies on the biotransformation of 16-O-demethyldeoxyaconitine of the metabolite of aconitine in human intestinal bacteria, Chin. J. Anal. Chem. 35 (2007) 1711–1715. [102] Y.F. Zhao, F.R. Song, X.Y. Wang, X.H. Guo, Z.Q. Liu, S.Y. Liu, Studies on the biotransformation of 16-O-demethylaconitine and electrospray ionization tandem mass spectrometry, Acta Chin. Sinica 66 (2008) 525–530. [103] X.Y. Wang, Z.F. Pi, W.L. Liu, F.R. Song, Z.Q. Liu, S.Y. Liu, Biotransformation of Aconitum alkaloids before and after the combination of Radix Aconiti Preparata by rat intestinal flora using semiquantitative analysis method of electrospray ionization mass spectrometry, Chem. J. Chin. Univ. 32 (2011) 1526–1531. [104] X.Y. Wang, Z.F. Pi, W.L. Liu, Y.F. Zhao, S.Y. Liu, Effect of pH on the metabolism of aconitine under rat intestinal bacteria and analysis of metabolites using HPLC/MS-MSn technique, Chin. J. Chem. 28 (2010) 2494–2500. [105] Y. Xin, Z.F. Pi, F.R. Song, Z.Q. Liu, S.Y. Liu, Study on the metabolic characteristics of aconite alkaloids in the extract of Radix aconiti under intestinal bacteria of rat by UPLC/MSn technique, Chin. J. Chem. 30 (2012) 656–664.
11
[106] T.Y.K. Chan, Aconite poisoning, Clin. Toxicol. 47 (2009) 279–285. [107] S.N. Wright, Irreversible block of human heart (hH1) sodium channels by the plant alkaloid lappaconitine, Mol. Pharmacol. 59 (2001) 183–192. [108] C.C. Lin, T.Y.K. Chan, J.F. Deng, Clinical features and management of herb-induced aconitine poisoning, Ann. Emerg. Med. 43 (2004) 574–579. [109] P. Honerjager, A. Meissner, The positive inotropic effect of aconitine, N.-S. Arch. Pharmacol. 322 (1983) 49–58. [110] L.J. Voss, J.M. Voss, L. McLeay, J.W. Sleigh, Aconitine induces prolonged seizure-like events in rat neocortical brain slices, Eur. J. Pharmacol. 584 (2008) 291–296. [111] T. Kiss, P. Orvos, S. Bansaghi, P. Forgo, N. Jedlinszki, L. Talosi, J. Hohmann, D. Csupor, Identification of diterpene alkaloids from Aconitum napellus subsp firmum and GIRK channel activities of some Aconitum alkaloids, Fitoterapia 90 (2013) 85–93. [112] P.J. Tong, C.L. Wu, X.F. Wang, H.Z. Hu, H.T. Jin, C.Y. Li, Y. Zhu, L.T. Shan, L.W. Xiao, Development and assessment of a complete-detoxication strategy for Fuzi (lateral root of Aconitum carmichaeli) and its application in rheumatoid arthritis therapy, J. Ethnopharmacol. 146 (2013) 562–571. [113] M. Zhang, Y. Long, Y. Sun, Y.K. Wang, Q.A. Li, H.J. Wu, Z.J. Guo, Y.H. Li, Y.B. Niu, C. Li, L. Liu, Q.B. Mei, Evidence for the complementary and synergistic effects of the three-alkaloid combination regimen containing berberine, hypaconitine and skimmianine on the ulcerative colitis rats induced by trinitrobenzene-sulfonic acid, Eur. J. Pharmacol. 651 (2011) 187–196. [114] C. Peng, T. Zheng, F. Yang, Y.X. Li, D.K. Zhang, Study of neurotoxic effects and underlying mechanisms of aconitine on cerebral cortex neuron cells, Arch. Pharmacal. Res. 32 (2009) 1533–1543. [115] W. Ren, L. Yuan, J. Li, X.J. Huang, S. Chen, D.J. Zou, X.M. Liu, X.Z. Yang, Ethanolic extract of Aconiti Brachypodi Radix attenuates nociceptive pain probably via inhibition of voltage-dependent Na(+) channel, Afr. J. Tradit. Complem. 9 (2012) 574–583. [116] L.Y. Dong, B.F. Cheng, Y. Luo, N. Zhang, H.Q. Duan, M. Jiang, Y.M. Wang, G. Bai, G.A. Luo, Identification of nuclear factor-kappa B inhibitors and beta(2) adrenergic receptor agonists in Chinese medicinal preparation Fuzilizhong pills using UPLC with quadrupole time-of-flight MS, Phytochem. Anal. 25 (2014) 113–121. [117] C. de Ines, M. Reina, J.A. Gavin, A. Gonzalez-Coloma, In vitro cytotoxicity of norditerpenoid alkaloids, Z. Naturforsch. C 61 (2006) 11–18. [118] P. Cui, H. Han, R. Wang, L. Yang, Identification and determination of Aconitum alkaloids in aconitum herbs and Xiaohuoluo pill using UPLC-ESI–MS, Molecules 17 (2012) 10242–10257. [119] G.G. Tan, M. Liu, X. Dong, S. Wu, L. Fan, Y.B. Qiao, Y.F. Chai, H. Wu, A strategy for rapid analysis of xenobiotic metabolome of Sini decoction in vivo using ultra-performance liquid chromatography-electrospray ionization quadrupole-time-of-flight mass spectrometry combined with pattern recognition approach, J. Pharm. Biomed. Anal. 96 (2014) 187–196. [120] L.H. Chen, L.J. Jin, Z.M. Su, Y.Q. Qiu, Y. Wang, S.Y. Liu, ESI–MSn behavior and quantum chemistry calculation of stability of fragment ions of diester-diterpenoid alkaloids(DDA), Chem. J. Chin. Univ. 26 (2005) 2340–2344.
Please cite this article in press as: T. Xu, et al., Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines, J. Chromatogr. B (2015), http://dx.doi.org/10.1016/j.jchromb.2015.09.023