Skip to main content
Evidence-based Complementary and Alternative Medicine : eCAM logoLink to Evidence-based Complementary and Alternative Medicine : eCAM
. 2015 Feb 3;2015:252434. doi: 10.1155/2015/252434

In Vivo and In Vitro Metabolites from the Main Diester and Monoester Diterpenoid Alkaloids in a Traditional Chinese Herb, the Aconitum Species

Min Zhang 1, Chong-sheng Peng 1, Xiao-bo Li 1,*
PMCID: PMC4332761  PMID: 25705235

Abstract

Diester diterpenoid alkaloids (DDAs), such as aconitine (AC), mesaconitine (MA), and hypaconitine (HA), are both pharmacologically active compounds and toxic ingredients in a traditional Chinese herb, the Aconitum species. Many DDA metabolism studies have been performed to explore mechanisms for reducing toxicity in these compounds and in Aconitum species extracts for safe clinical administration. In this review, we summarize recent progress on the metabolism of toxic AC, MA, and HA and corresponding monoester diterpenoid alkaloids (MDAs) in the gastrointestinal tract and liver in different animal species and humans in vivo and/or in vitro, where these alkaloids are primarily metabolized by cytochrome P450 enzymes, carboxylesterases, and intestinal bacteria, which produces phase I metabolites, ester hydrolysed products, and lipoalkaloids. Furthermore, we classify metabolites detected in the blood and urine, where the aforementioned metabolites are absorbed and excreted. Less toxic MDAs and nontoxic alcohol amines are the primary DDA metabolites detected in the blood. Most other DDAs metabolites produced in the intestine and liver detected in the urine have not been reported in the blood. We propose an explanation for this nonconformity. Finally, taking AC, for instance, we generalize a process of toxicity reduction in the body after oral AC administration for the first time.

1. Introduction

Diester diterpenoid alkaloids (DDAs, Table 1), such as aconitine (AC), mesaconitine (MA), and hypaconitine (HA), are a family of highly toxic alkaloids from the root of a traditional Chinese herb, the Aconitum species (sp.), which has been used clinically for years. Monoester diterpenoid alkaloids (MDAs, Table 1) are the ester hydrolysis products of DDAs at the C-8 position, which are also components of this herb. Both DDAs and MDAs exhibit excellent pharmacological effects, including anti-inflammatory, analgesic, and cardiotonic activities [1, 2].

Table 1.

DDA, MDA, and alcohol amine chemical structures.

graphic file with name ECAM2015-252434.tab1.i001.jpg

Compounds R1 R2 R3 R4 Formula Mass

DDAs
 Aconitine (AC) Ethyl (Et) Hydroxy (OH) Acetyl (Ac) Benzoyl (Bz) C34H47NO11 645.3149
 Mesaconitine (MA) Methyl (Me) OH Ac Bz C33H45NO11 631.2992
 Hypaconitine (HA) Me Hydrogen (H) Ac Bz C33H45NO10 615.3043
MDAs
 Benzoylaconine (BAC) Et OH H Bz C32H45NO10 603.3043
 Benzoylmesaconine (BMA) Me OH H Bz C31H43NO10 589.2887
 Benzoylhypaconine (BHA) Me H H Bz C31H43NO9 573.2938
Alcohol amines
 Aconine Et OH H H C25H41NO9 499.2781
 Mesaconine Me OH H H C24H39NO9 485.2625
 Hypaconine Me H H H C24H39NO8 469.2676

However, these compounds, especially DDAs, have narrow therapeutic windows. For example, a single lethal AC dose for humans is estimated at 2–6 mg [3, 4] with poisoning symptoms, such as hypotension, palpitations, ventricular tachyarrhythmias, asystole, and numbness of the face and limbs [1]. Severe poisoning may occur after improper ingestion of DDA-containing drugs or prescriptions, such as Chuanwu [5], Caowu [6], and Fuzi [7]. Therefore, Aconitum herbs are traditionally boiled or steamed before oral administration to ensure safety [8]. During this process, DDAs are mainly hydrolysed to less toxic MDAs. Further MDA hydrolysis yields almost nontoxic alcohol amines (Table 1), such as aconine, mesaconine, and hypaconine [3, 9, 10]. In contrast with AC, the half-maximal lethal dose (LD50, mg/kg, i.v. mice) of 14-benzoylaconine (BAC) and aconine increases by approximately 38- and 430-fold, respectively [11].

On the other hand, many valuable studies have recently been performed on DDA and MDA metabolism to explore the toxicity reduction mechanisms and obtain information for clinical guidance. In this paper, we review for the first time the metabolites biotransformed in the gastrointestinal tract and liver from toxic AC, MA, and HA of DDAs as well as their corresponding ester hydrolysed products, BAC, 14-benzoylmesaconine (BMA), and 14-benzoylhypaconine (BHA) of MDAs, in different animal species and humans in vivo and in vitro. Furthermore, we classify the metabolites detected in the blood and urine, in which these metabolites are absorbed and excreted. Our study will be fundamental and helpful for further studies on reducing the toxicity of DDA-containing drugs compatible with other medicine based on DDAs absorption and metabolism [12, 13].

2. Metabolism in the Gastrointestinal Tract and Liver

Traditional Chinese prescriptions are commonly prepared through decoction and ingested orally. The active compounds are unavoidably converted in the gastrointestinal tract.

2.1. Metabolism in the Stomach

The stomach provides an acidic environment for drug dissolution and absorption; however, studies on stomach metabolism are typically ignored. Only one study has focused on AC metabolism in the stomach.

In this study, 14 metabolites and 2 ester hydrolysis products are identified in gastric content in rabbits after oral AC administration [14]. Metabolism includes hydroxylation, deoxylation, demethylation, didemethylation/deethylation, and ester exchange at the C-8 position with long chain fatty acids (Table 2). The enzymes responsible for metabolism have not been reported. The aforementioned metabolic process may be catalysed by CYP2C9 and CYP2C8 that are expressed in parietal gastric cells [15] and by bacteria that are located in the human stomach [16].

Table 2.

AC metabolites produced in rabbit stomachs.

DDAs m/z
(ESI+)
Formula Identification Neutral loss (Da), identification of fatty acid Metabolic procedure MS detection References
AC 662 C34H47NO12 2′-Hydroxy AC or
3′-AC (M1)a
NAb Rabbits and rats; ig, in vivo. IT, FT-ICR [14]
3′-Hydroxy AC or
2′-hydroxy AC (M3)a
4′-Hydroxy AC (M6)a
632 C33H45NO11 Demethyl AC (M4) NA
630 C34H47NO10 Indaconitine (15-deoxy AC, M5)c NA
Deoxyaconitine (3-deoxy AC, M7)
618 C32H43NO11 Didemethyl AC or
N-deethyl AC (M2)
NA
604 C32H45NO10 BAC (hydrolysis product 2) NA Rabbits and rats; ig, in vivo. IT, FT-ICR
542 C27H43NO10 14-O-Debenzoyl AC (hydrolysis product 1) NA Rabbits and rats; ig, in vivo. IT, FT-ICR
828 C47H73NO11 8-O-Pentadecanoyl BAC (M10) 242, pentadecanoic acid Rabbits and rats; ig, in vivo. IT, FT-ICR
842 C48H75NO11 8-O-Palmitoyl BAC (M12) 256, palmitic acid
864 C50H73NO11 8-O-Linolenoyl BAC (M9) 278, linolenic acid
866 C50H75NO11 8-O-Linoleoyl BAC (M11) 280, linoleic acid
868 C50H77NO11 8-O-Oleoyl BAC (M13) 282, oleic acid
870 C50H79NO11 8-O-Stearoyl BAC (M14) 284, stearic acid
978 C58H91NO11 8-O-Hexacosandienoyl BAC (M8) 392, hexacosandienoic acid

a2′, 3′, and 4′, the position in benzoyl group.

bNot available.

cDeoxy may also be referred to as dehydroxy in the literature.

The ester hydrolysis products at the C-8 and C-14 positions are not only observed in rabbit stomachs but also in acid solutions (negative control). Ester hydrolysis in the stomach may be catalysed by carboxylesterases (CEs) in the gastric mucosa [17] because CE expression has also been reported in the stomach, although CEs are predominantly distributed in the liver, plasma, and intestine [18]. However, this finding also implies that DDAs can be nonenzymatically ester hydrolysed under acidic conditions, which is discussed in Section 5.

In addition, AC, MA, HA, and their hydrolysis products (MDAs and alcohol amines) are detected in gastric contents in a dead female, who was suspected of dying from acute drug poisoning involving Aconitum alkaloids [19]. However, the reference did not indicate whether the hydrolysis products were metabolized from DDAs in the stomach or were originally in the toxicant.

2.2. Metabolism in the Intestine

A large number of bacteria populate the gastrointestinal tract; the bacterial concentration increases distally. The majority of bacteria reside in the colon, where the density approaches 1011-1012 cells/mL, and anaerobic species dominate. This microbiota secretes a diverse array of enzymes that participate in various metabolic processes, such as reduction, hydrolysis, deoxylation, acetylation, deacetylation, and N-demethylation; thus, the intestinal microbiota is important to orally ingested drug metabolism [20, 21]. Notably, hydrolysis catalysed by bacteria is common in glycosides. Based on DDA and MDA structures, ester hydrolysis is likely driven by CEs, which also dominate the intestine [18].

The intestinal bacteria DDA metabolism reviewed herein was mainly performed in vitro through anaerobic incubation in a feces suspension, which included high levels of intestinal bacteria. The intestinal bacteria DDA metabolism is similar to metabolism in the stomach and included hydroxylation, deoxylation, demethylation, demethylation with deoxylation, ester hydrolysis at the C-8 and/or C-14 position, and ester exchange at the C-8 position with short and long chain fatty acids (Table 3). AC metabolites, such as 16-O-demethyl AC, 3-deoxy AC, and 16-O-demethyl-3-deoxy AC, were further converted to deoxylation, demethylation, ester hydrolysis, and ester exchange products (Table 4). These results imply that MDAs, which are DDA ester hydrolysed products, may be metabolized through the same pathway; however, no studies have reported on intestinal MDA metabolism.

Table 3.

Metabolites of AC, MA, and HA converted in intestine.

DDAs m/z (ESI+) Formula Identification Neutral loss (Da), identification of fatty acid Metabolic procedure MS detection References
AC 662 C34H47NO12 10-Hydroxy AC NAa Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P4)
632 C33H45NO11 16-O-Demethyl AC* NA Rabbits; contents from small intestine and caecum and feces; ig, in vivo. IT [23] (M3)
Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24] (M1)
630 C34H47NO10 Indaconitine (15-deoxy AC)b Rabbits; contents from small intestine and caecum and feces; ig, in vivo. IT [23] (M6)
Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P5)
Deoxy AC* NA Rabbits; contents from small intestine and caecum and feces; ig, in vivo. IT [23] (M5)
Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24] (M2)
Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P10)
616 C33H45NO10 16-O-Demethyl-deoxy AC* NA Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24] (M3)
604 C32H45NO10 BAC NA Rabbits; contents from small intestine and caecum and feces; ig, in vivo. IT [23] (M2)
Rats; intestinal bacteria; anaerobic incubation, in vitro.c IT [25]
Rats; intestinal bacteria; anaerobic incubation, in vitro.d IT [26]
Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P1)
590 C31H43NO10 16-O-Demethyl BAC NA Rabbits; contents from small intestine and caecum and feces; ig, in vivo. IT [23] (M1)
588 C32H45NO9 15-Deoxy BAC NA Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P2)
586 C32H43NO9 Deacetoxy AC NA Rats; intestinal bacteria; anaerobic incubation, in vitro.c,d IT [25, 26]
660 C35H49NO11 8-O-Propionyl BAC 74, propionic acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
NA Rats; intestinal bacteria; anaerobic incubation, in vitro.e IT, MALDI source-FT-ICR [27]
NA Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P8)
674 C36H51NO11 8-O-Butyryl BAC 88, butyric acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
NA Rats; intestinal bacteria; anaerobic incubation, in vitro.e IT, MALDI source-FT-ICR [27]
NA Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P9)
688 C37H53NO11 8-O-Valeryl BAC 102, valeric acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
NA Rats; intestinal bacteria; anaerobic incubation, in vitro.e IT, MALDI source-FT-ICR [27]
700 C38H53NO11 8-O-Hexenoyl BAC 114, hexenoic acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
NA Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P7)
690 C36H51NO12 8-O-(3-Hydroxy)-butyryl BAC NA Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P11)
702 C38H55NO11 8-O-Hexanoyl BAC 116, hexanoic acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
716 C39H57NO11 8-O-Heptanoyl BAC 130, heptanoic acid Ibid. Ibid. Ibid.
722 C40H51NO11 8-O-Phenylacetyl BAC 136, phenylacetic acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
NA Rats; intestinal bacteria; anaerobic incubation, in vitro.e IT, MALDI source-FT-ICR [27]
728 C40H57NO11 8-O-Octenoyl BAC NA Rats; intestinal bacteria; anaerobic incubation at pH 7.0, in vitro. IT [22] (P3)
736 C41H53NO11 8-O-Phenylpropionyl BAC 150, phenylpropionic acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
800 C45H69NO11 8-O-Tridecanoyl BAC 214, tridecanoic acid Ibid. Ibid. Ibid.
814 C46H71NO11 8-O-Tetradecanoyl BAC 228, tetradecanoic acid Ibid. Ibid. Ibid.
828 C47H73NO11 8-O-Pentadecanoyl BAC 242, pentadecanoic acid Ibid. Ibid. Ibid.
842 C48H75NO11 8-O-Palmitoyl BAC 256, palmitic acid Ibid. Ibid. Ibid.
854 C49H75NO11 8-O-Heptadecenoyl BAC 268, heptadecenoic acid Ibid. Ibid. Ibid.
856 C49H77NO11 8-O-(Methyl)-palmitoyl BAC 270, methyl palmitic acid Ibid. Ibid. Ibid.
866 C50H75NO11 8-O-Linoleyl BAC 280, linoleic acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
NA Rats; intestinal bacteria; anaerobic incubation, in vitro.c,d IT [25, 26]
868 C50H77NO11 8-O-Oleoyl BAC 282, oleic acid Human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [24]
870 C50H79NO11 8-O-Stearoyl BAC 284, stearic acid Ibid. Ibid. Ibid.
882 C51H79NO11 8-O-(9)-Nonadecenoyl BAC 296, nonadecene Ibid. Ibid. Ibid.
886 C50H79NO12 8-O-(3-Hydroxy)-stearoyl BAC 300, 3-hydroxy stearic acid Ibid. Ibid. Ibid.
954 C56H91NO11 8-O-Tetracosanoyl BAC 368, tetracosanoic acid Ibid. Ibid. Ibid.
962 C57H87NO11 8-O-Pentacosatrienoyl BAC 376, pentacosatrienoic acid Ibid. Ibid. Ibid.

MA 590 C31H43NO10 BMA NA Rats; intestinal bacteria; anaerobic incubation, in vitro.c,d IT [25, 26]
572 C31H41NO9 Deacetoxy MA NA Ibid. Ibid. Ibid.
660 C35H49NO11 8-O-Butyryl BMA NA Rats; intestinal bacteria; anaerobic incubation, in vitro.e IT, MALDI source-FT-ICR [27]
674 C36H51NO11 8-O-Valeryl BMA NA Ibid. Ibid. Ibid.
852 C49H73NO11 8-O-Linoleyl BMA NA Rats; intestinal bacteria; anaerobic incubation, in vitro.c,d IT [25, 26]

HA 574 C31H43NO9 BHA NA Rats; intestinal bacteria; anaerobic incubation, in vitro.e IT, MALDI source-FT-ICR [27]
Rats; intestinal bacteria; anaerobic incubation, in vitro.c,d IT [25, 26]
556 C31H41NO8 Deacetoxy HA NA Rats; intestinal bacteria; anaerobic incubation, in vitro.c,d IT [25, 26]
630 C34H47NO10 8-O-Propionyl BHA NA Rats; intestinal bacteria; anaerobic incubation, in vitro.e IT, MALDI source-FT-ICR [27]
644 C35H49NO10 8-O-Butyryl BHA NA Ibid. Ibid. Ibid.
658 C36H51NO10 8-O-Valeryl BHA NA Ibid. Ibid. Ibid.
692 C39H49NO10 8-O-Phenylacetyl BHA NA Ibid. Ibid. Ibid.
836 C49H73NO10 8-O-Linoleyl BHA NA Rats; intestinal bacteria; anaerobic incubation, in vitro.c,d IT [25, 26]

aNot available.

bDeoxy may also be referred to as dehydroxy in the literature.

cDDA was produced through decoction of Aconiti Radix Cocta with Fritillariae Thunbergii Bulbus, Pinelliae Rhizoma Preparatum, and Ampelopsis Radix.

It is not clear whether these compounds were directly metabolized from DDAs or were originally ingested.

dDDA was produced through decoction of Aconiti Lateralis Radix Praeparata with Glycyrrhizae Radix and Rhizome as well as with Atractylodis Macrocephalae Rhizoma.

It is not clear whether these compounds were directly metabolized from DDAs or were originally ingested.

eIn addition to AC and HA monomers, DDAs were also generated from ethyl alcohol extraction ofRadix Aconiti.

It is not clear whether these compounds were directly metabolized from DDAs or were originally ingested.

*These metabolites were further biotransformed in the intestine. Metabolites of these intermediate products are listed in Table 4.

Table 4.

Further biotransformation of intestinal AC metabolites in the intestine.

m/z (ESI+) Formula Identification Neutral loss (Da),
identification of fatty acid
Metabolic procedure MS detection References
618 C32H43NO11 1,16-Didemethyl AC (M1) NAa 16-O-Demethyl AC (C33H45NO11, 632) from AC; human; intestinal bacteria; anaerobic incubation, in vitro. IT, FT-ICR [28]
616 C33H45NO10 16-O-Demethyl-3-deoxy AC (M2)b NA
602 C32H43NO10 1,16-Didemethyl-3-deoxy AC (M3) NA
590 C31H43NO10 16-O-Demethyl BAC (M4) NA
486 C24H39NO9 16-O-Demethyl aconine (M5) NA
646 C34H47NO11 16-O-Demethyl-8-O-propionyl BAC 74, propionic acid
660 C35H49NO11 16-O-Demethyl-8-O-butyryl BAC 88, butyric acid
674 C36H51NO11 16-O-Demethyl-8-O-valeryl BAC 102, valeric acid
16-O-Demethyl-8-O-(methyl)-butyryl BAC 102, methyl butyric acid
696 C38H49NO11 16-O-Demethyl-8-O-heptatrienoyl BAC 124, heptatrienoic acid
698 C38H51NO11 16-O-Demethyl-8-O-heptadienoyl BAC 126, heptadienoic acid
700 C38H53NO11 16-O-Demethyl-8-O-heptenoyl BAC 128, heptenoic acid
702 C38H55NO11 16-O-Demethyl-8-O-heptanoyl BAC 130, heptanoic acid
710 C39H51NO11 16-O-Demethyl-8-O-octatrienoyl BAC 138, octatrienoic acid
716 C39H57NO11 16-O-Demethyl-8-O-octanoyl BAC 144, octanoic acid
730 C40H59NO11 16-O-Demethyl-8-O-nonanoyl BAC 158, nonanoic acid
736 C41H53NO11 16-O-Demethyl-8-O-decatetraenoyl BAC 164, decatetraenoic acid
762 C43H55NO11 16-O-Demethyl-8-O-dodecapentaenoyl BAC 190, dodecapentaenoic acid
764 C43H57NO11 16-O-Demethyl-8-O-dodecatetraenoyl BAC 192, dodecatetraenoic acid
766 C43H59NO11 16-O-Demethyl-8-O-dodecatrienoyl BAC 194, dodecatrienoic acid
778 C44H59NO11 16-O-Demethyl-8-O-tridecatetraenoyl BAC 206, tridecatetraenoic acid
786 C44H67NO11 16-O-Demethyl-8-O-(methyl)-dodecanoyl BAC 214, methyl dodecanoic acid
800 C45H69NO11 16-O-Demethyl-8-O-retradecanoyl BAC 228, tetradecanoic acid
854 C49H75NO11 16-O-Demethyl-8-O-oleoyl BAC 282, oleic acid
856 C49H77NO11 16-O-Demethyl-8-O-stearoyl BAC 284, stearic acid
870 C50H79NO11 16-O-Demethyl-8-O-(methyl)-stearoyl BAC 298, methyl stearic acid
884 C51H81NO11 16-O-Demethyl-8-O-arachidyl BAC 312, arachidic acid
898 C52H83NO11 16-O-Demethyl-8-O-heneicosanoyl BAC 326, heneicosanoic acid
926 C54H87NO11 16-O-Demethyl-8-O-tricosanoyl BAC 354, tricosanoic acid

616 C33H45NO10 16-O-Demethyl-3-deoxy AC (M1) NA 3-Deoxy AC (C34H47NO10, 630) from AC;
human; intestinal bacteria;
anaerobic incubation, in vitro.
IT, FT-ICR [29]
614 C34H47NO9 1,13-Dideoxy AC (M2) NA
588 C32H45NO9 3-Deoxy BAC (M3) NA
484 C25H41NO8 3-Deoxy aconine (M4) NA
644 C35H49NO10 3-Deoxy-8-O-propionyl BAC 74, propionic acid
658 C36H51NO10 3-Deoxy-8-O-butyryl BAC 88, butyric acid
700 C39H57NO10 3-Deoxy-8-O-heptanoyl BAC 130, heptanoic acid
702 C38H55NO11 3-Deoxy-8-O-(2-methyl-3-hydroxy)-valeryl BAC 132,
2-methyl-3-hydroxy valeric acid
714 C40H59NO10 3-Deoxy-8-O-octanoyl BAC 144, octanoic acid
730 C40H59NO11 3-Deoxy-8-O-(3-hydroxy)-octanoyl BAC 160, 3-hydroxy octanoic acid
746 C43H55NO10 3-Deoxy-8-O-undecapentaenoyl BAC 176, undecapentaenoic acid
762 C44H59NO10 3-Deoxy-8-O-dodecatetraenoyl BAC 192, dodecatetraenoic acid
786 C44H67NO11 3-Deoxy-8-O-(hydroxy)-dodecanoyl BAC 216, hydroxy dodecanoic acid
800 C45H69NO11 3-Deoxy-8-O-(hydroxy)-tridecanoyl BAC 230, hydroxy tridecanoic acid
814 C46H71NO11 3-Deoxy-8-O-(3-hydroxy)-tetradecanoyl BAC 244, hydroxy tetradecanoic acid
828 C47H73NO11 3-Deoxy-8-O-(hydroxy)-pentadecanoyl BAC 258, hydroxy pentadecanoic acid
854 C50H79NO10 3-Deoxy-8-O-propionyl BAC 284, stearic acid

602 C32H43NO10 1,16-O-Didemethyl-3-deoxy AC (M1) NA 16-O-Demethyl-3-deoxy AC (C33H45NO10, 616) from AC;
human; intestinal bacteria;
anaerobic incubation, in vitro.
IT, FT-ICR [30]
600 C33H45NO9 16-O-Demethyl-3-deoxy-deoxy AC (M2) NA
574 C31H43NO9 16-O-Demethyl-3-deoxy BAC (M3) NA
470 C24H39NO8 16-O-Demethyl-3-deoxy aconine (M4) NA
630 C34H47NO10 16-O-Demethyl-3-deoxy-8-O-propionyl BAC 74, propionic acid
644 C35H49NO10 16-O-Demethyl-3-deoxy-8-O-butyryl BAC 88, butyric acid
696 C39H53NO10 16-O-Demethyl-3-deoxy-8-O-octadienoyl BAC 140, octadienoic acid
700 C39H57NO10 16-O-Demethyl-3-deoxy-8-O-octanoyl BAC 144, octanoic acid
702 C38H55NO11 16-O-Demethyl-3-deoxy-8-O-(hydroxy)-heptanoyl BAC 146, hydroxy heptanoic acid
730 C40H59NO11 16-O-Demethyl-3-deoxy-8-O-(hydroxy)-nonanoyl BAC 174, hydroxy nonanoic acid
746 C43H55NO10 16-O-Demethyl-3-deoxy-8-O-dodecapentaenoyl BAC 190, dodecapentaenoic acid
762 C44H59NO10 16-O-Demethyl-3-deoxy-8-O-tridecatetraenoyl BAC 206, tridecatetraenoic acid
778 C45H63NO10 16-O-Demethyl-3-deoxy-8-O-tetradecatrienoyl BAC 222, tetradecatrienoic acid

aNot available.

bDeoxy may also be referred to as dehydroxy in the literature.

Ester exchange metabolites are classified as lipoalkaloids or lipoaconitines with an acetyl group at the C-8 position of DDAs replaced by other fatty acid acyl groups [24, 31]. Presumably, the short chain fatty acids (such as propionic, butyric, hexanoic, phenylacetic, and phenylpropionic acids) for ester exchange are generated from xenobiotics, such as food decomposed by intestinal bacteria, while certain long chain fatty acids (such as palmitic, oleic, and stearic acids) are generated from bacterial cell walls [24]. DDA toxicity is reduced after ester exchange. For example, the LD50 of 8-O-butyryl- (from short chain fatty acid) benzoylmesaconine is 15.78 mg/kg, which is 5.5-fold greater than MA (8-O-acetyl-benzoylmesaconine) [22]. The LD50 for mice with lipomesaconitines (from long chain fatty acids) are from 10 to 40 mg/kg, which are 20-fold greater than MA [32].

2.3. Metabolism in the Liver

The liver is an important organ for drug metabolism, and it expresses many drug-metabolising enzymes. After oral administration, drugs are typically subjected to hepatic metabolism, including CEs that catalyse ester hydrolysis [18], phase I drug metabolic enzymes that catalyse oxidation, and phase II metabolic enzymes that catalyse conjugation [21]. The metabolites are hydrophilic and are more rapidly excreted from the body than parent drugs. Cytochrome P450 enzymes (CYP450s) and uridine 5′-diphosphate (UDP)-glucuronosyltransferases (UGTs) are the most common phase I and phase II metabolic enzymes, respectively [33].

The hepatic metabolism studies reviewed herein were mainly performed in vitro through incubation with liver microsomes. CYP450- or UGT-catalysed metabolism in microsomes can be selectively performed in different reaction systems with auxiliary enzymes and exclusive substrates [34, 35].

The DDA and MDA phase I metabolic pathways are similar and include hydroxylation, deoxylation, demethylation, didemethylation/deethylation, dehydrogenation, and demethylation with dehydrogenation (Table 5). The individual CYP450s responsible for specific metabolites were further determined via individual inhibitors or recombinant isoenzymes. CYP3A4 and CYP3A5 are the most common isoenzymes that catalyse both DDAs and MDAs. In addition, CYP2D6, CYP1A1/2, CYP2C9, CYP2C8, CYP2C19, and CYP2E1 also partially catalyse DDAs.

Table 5.

Metabolites of DDAs and MDAs converted in the liver.

Alkaloids m/z (ESI+) Formula Identification Involved CYP450s Metabolic procedure MS detection References
AC 662 C34H47NO12 Hydroxy AC CYP3A5, CYP2D6 Human; liver microsomes and recombinant CYP450s; incubation, in vitro. Q-TOF [35] (M6)
NAa Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M5)
Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M6)
644 C34H45NO11 3-Dehydrogen AC CYP3A4, CYP3A5 Human; liver microsomes and recombinant CYP450s; incubation, in vitro. Q-TOF [35] (M5)
NA Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M5)
Dehydrogen AC CYP3A, CYP1A1/2 Rats; liver microsomes; incubation, in vitro. IT [4] (M6)
NA Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M7)
632 C33H45NO11 16-O-Demethyl AC CYP3A, CYP1A1/2 Rats; liver microsomes; incubation, in vitro. IT [4] (M2)
CYP3A4, CYP3A5, CYP2D6, CYP2C9 Human; liver microsomes and recombinant CYP450s; incubation, in vitro. Q-TOF [35] (M2)
NA Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M6)
NA Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M2)
O-Demethyl AC CYP3A, CYP1A1/2 Rats; liver microsomes; incubation, in vitro. IT [4] (M1)
CYP3A4, CYP3A5, CYP2C8, CYP2D6 Human; liver microsomes and recombinant CYP450s; incubation, in vitro. Q-TOF [35] (M1)
NA Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M1)
630 C34H47NO10 Deoxyaconitine (3-deoxy AC) NA Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M7)
Deoxy AC NA Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M8)
618 C32H43NO11 O-Didemethyl AC CYP3A, CYP1A1/2 Rats; liver microsomes; incubation, in vitro. IT [4] (M3)
CYP2D6, CYP3A5 Human; liver microsomes and recombinant CYP450s; incubation, in vitro. Q-TOF [35] (M4)
NA Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M4)
NA Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M3)
N-Deethyl AC CYP3A, CYP1A1/2 Rats; liver microsomes; incubation, in vitro. IT [4] (M4)
CYP3A4, CYP3A5, CYP2D6, CYP2C9 Human; liver microsomes and recombinant CYP450s; incubation, in vitro. Q-TOF [35] (M3)
NA Rats; liver microsomes; incubation, in vitro. Q-TOF [38] (M4)
NA Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M2)
NA Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M4)
604 C32H45NO10 BAC CYP3A, CYP1A1/2 Rats; liver microsomes; incubation, in vitro. IT [4] (M5)
NA Rats; liver microsome and S9 fraction; incubation, in vitro. Q-Trap [39]
NA Rats; liver microsomes; incubation, in vitro. Q-TOF [38] (M2)
NA Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M1)
NA Guinea pigs and mice; liver microsomes; incubation, in vitro. HRMS, MS2 [37] (M8)
586 C32H43NO9 Deacetoxy ACb NA Rats; liver microsome S9 fraction; incubation, in vitro. IT [36] (M3)
482 C25H39NO8 Dehydrated aconine NA Rabbits; liver; ig, in vivo. IT [40]

MA 648 C33H45NO12 Hydroxy MA CYP3A4, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M5)
2-Hydroxy MA NA Rats; liver microsomes; incubation, in vitro. Q-TOF, QQQ [38] (M5)
CYP3A, CYP2C, CYP2D Rats; liver microsomes; incubation, in vitro. QQQ; IM [42] (M5)
630 C33H43NO11 Dehydrogen MA CYP3A4, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M4)
NA Rats; liver microsomes; incubation, in vitro. Q-TOF, QQQ [38] (M6)
3-Dehydrogen MA CYP3A, CYP2D Rats; liver microsomes; incubation, in vitro. QQQ; IM [42] (M2)
618 C32H43NO11 16-O-Demethyl MA CYP2C8, CYP3A4, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M2)
CYP3A Rats; liver microsomes; incubation, in vitro. QQQ; IM [42] (M4)
1-O-Demethyl MA CYP3A, CYP2C Rats; liver microsomes; incubation, in vitro. QQQ; IM [42] (M3)
18-O-Demethyl MA CYP3A, CYP2C Rats; liver microsomes; incubation, in vitro. QQQ; IM [42] (M6)
Demethyl MA CYP2C8, CYP2D6, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M1)
Demethyl MA CYP3A4, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M3)
616 C32H41NO11 Demethyl-dehydrogen MA CYP3A4, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M6)
Demethyl-dehydrogen MA CYP2C8, CYP3A4, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M7, M8)
Demethyl-dehydrogen MA CYP2C8, CYP2C9, CYP2D6, CYP3A4, CYP3A5 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [41] (M9)
590 C31H44NO10 BMA NA Rats; liver microsome and S9 fraction; incubation, in vitro. Q-Trap [39]
NA Rats; liver microsomes; incubation, in vitro. Q-TOF, QQQ [38] (M1)

HA 632 C33H45NO11 MA CYP3A4, CYP3A5, CYP2C19, CYP2D6, CYP2E1 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43] (M8)
CYP3A, CYP2D, CYP2C, CYP2E1 Rats; liver microsomes; incubation, in vitro. QQQ [44] (M6)
2-Hydroxy HA CYP3A, CYP2C, CYP2D, CYP1A2 Rats; liver microsomes; incubation, in vitro. QQQ [44] (M4)
Hydroxy HA CYP3A4, CYP3A5, CYP2C19, CYP2D6, CYP2E1 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43] (M7)
614 C33H43NO10 15-Dehydrogen HA CYP3A, CYP2D, CYP2E1 Rats; liver microsomes; incubation, in vitro. QQQ [44] (M2)
602 C32H43NO10 16-O-Demethyl HA CYP3A4, CYP3A5, CYP2C19, CYP2D6, CYP2E1 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43] (M2)
1-O-Demethyl HA CYP3A, CYP2D, CYP2C Rats; liver microsomes; incubation, in vitro. QQQ [44] (M5)
18-O-Demethyl HA CYP3A, CYP2C Rats; liver microsomes; incubation, in vitro. QQQ [44] (M7)
Demethyl HA CYP3A4, CYP3A5, CYP2C8, CYP2C19, CYP2D6, CYP2E1 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43] (M1)
Demethyl HA CYP3A4, CYP3A5, CYP1A2, CYP2C8, CYP2C19, CYP2D6, CYP2E1 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43] (M3)
600 C32H41NO10 Demethyl-dehydrogen HA CYP3A4, CYP3A5, CYP2C19, CYP2D6, CYP2E1 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43] (M4–M6)
590 C31H43NO10 2-Hydroxy BHA CYP3A, CYP2C Rats; liver microsomes; incubation, in vitro. QQQ [44] (M1)
588 C31H41NO10 Didemethyl HA CYP3A4, CYP3A5, CYP2C19, CYP2D6, CYP2E1 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43]
(M9, M10)
Didemethyl HA CYP3A4, CYP3A5, CYP2C19 Human (male); liver microsomes and recombinant CYP450s;
incubation, in vitro.
Q-TOF [43] (M11)
574 C31H43NO9 BHA CYP3A, CYP2D Rats; liver microsomes; incubation, in vitro. QQQ [44] (M3)
NA Rats; liver microsomes; incubation, in vitro. Q-TOF, QQQ [38] (M3)
NA Rats; liver microsome and S9 fraction; incubation, in vitro. Q-Trap [39]

BAC 602 C32H43NO10 Dehydrogen BAC (M1, M2) CYP3A4, CYP3A5 Human; liver microsomes;
incubation, in vitro.
Q-TOF [45]
590 C31H43NO10 Demethyl BAC (M5) CYP3A4, CYP3A5, CYP2D6
Demethyl BAC (M6) CYP3A4, CYP3A5
588 C31H41NO10 Demethyl-dehydrogen BAC (M3) CYP3A4, CYP3A5
576 C30H41NO10 Deethyl BAC or
didemethyl BAC (M7)
CYP3A4, CYP3A5
574 C30H39NO10 Didemethyl-dehydrogen BAC or
deethyl-dehydrogen BAC (M4)
CYP3A4, CYP3A5

BMA 606 C31H43NO11 Hydroxy BMA (M8) CYP3A4, CYP3A5 Human; liver microsomes;
incubation, in vitro.
Q-TOF [45]
588 C31H41NO10 Dehydrogen BMA (M1, M2) CYP3A4, CYP3A5
576 C30H41NO10 Demethyl BMA (M5) CYP3A4, CYP3A5, CYP2D6, CYP2C8
Demethyl BMA (M6, M7) CYP3A4, CYP3A5
574 C30H39NO10 Demethyl-dehydrogen BMA (M3, M4) CYP3A4, CYP3A5

BHA 590 C31H43NO10 Hydroxy BHA (M7) CYP3A4, CYP3A5 Human; liver microsomes;
incubation, in vitro.
Q-TOF [45]
BMA (M8) CYP3A4, CYP3A5
572 C31H41NO9 Dehydrogen BHA (M1, M2) CYP3A4, CYP3A5
560 C30H41NO9 Demethyl BHA (M5) CYP3A4
Demethyl BHA (M4, M6) CYP3A4, CYP3A5
558 C30H39NO9 Demethyl-dehydrogen BHA (M3) CYP3A4, CYP3A5
556 C30H37NO9 Demethyl-didehydrogen BHA (M9) CYP3A4, CYP3A5

aNot available.

bDeacetoxy aconitine may also be referred to as pyroaconitine in the literature.

Hydrophobic drug biotransformation commonly occurs first through phase I metabolism in which functional groups, such as hydroxy, sulfhydryl, carboxyl, and amino group, are formed and provide reaction sites for the subsequent phase II conjugation [46, 47]. For lipophilic DDAs and MDAs, hydroxy groups are initially present and are formed after hydroxylation during the phase I metabolism. However, phase II metabolites of either DDAs or MDAs were not detected in hepatic metabolism in vitro and in vivo, which demonstrates that phase II metabolism is not dominant compared with phase I metabolism in the liver. DDA ester hydrolysis should be catalysed by CEs. However, CYP3A, CYP1A1, and CYP1A2 are also involved in ester hydrolysis of AC, which reflects the complexity of metabolism.

2.4. A Comparison of DDA and MDA Metabolism in the Gastrointestinal Tract and Liver

The metabolites generated in the stomach, intestine, and liver are compared in Table 6. The polarity of most metabolites increased after DDA gastrointestinal and hepatic metabolism, except lipoalkaloids. Metabolites of AC from dehydrogenation and demethylation with dehydrogenation were only observed in the liver. The AC metabolites from demethylation with deoxylation observed from intestinal bacteria incubation [24] were also detected in the urine after oral AC administration in rabbits. However, these metabolites were not found in the urine after intravenous injection [48]. This observation suggests that the gastrointestinal tract may participate in biotransformation. The characteristic metabolites in the gastrointestinal tract were lipoalkaloids, which might be converted by enzymes that are only produced by intestinal bacteria. In addition, more lipoalkaloid varieties were detected in the intestine than in the stomach, which is consistent with abundant bacterial distribution in the gastrointestinal tract [16]. More studies have focused on DDAs than MDAs. However, it is speculated that MDAs may share similar metabolic pathways (except for ester hydrolysis at the C-8 position) with DDAs in the gastrointestinal tract based on the similarity in their hepatic metabolism and chemical structures.

Table 6.

A comparison of DDA and MDA metabolites in different metabolic procedures.

Alkaloids Stomach Intestine Liver (CYP450s, phase I metabolism)
DDAs Ester hydrolysis Ester hydrolysis commonly occurs at C-8 Ester hydrolysis commonly occurs at C-8
Hydroxylation at 2′/3′/4′ of the benzoyl group Hydroxylation at C-10 Hydroxylation at C-2
Deoxylation at C-3/15 Deoxylation at C-3/15 Deoxylation at C-3/15
Demethylation at the methoxy group Demethylation at the methoxy group,
often at C-1/6/16 or the N-methyl group
Demethylation at the methoxy group,
often at C-1/6/16 or the N-methyl group
Didemethylation at the methoxy group or deethylation at the N-ethyl group NAa Didemethylation at the methoxy group or deethylation at the N-ethyl group
NA Deacetoxylation (pyrolysis) Deacetoxylation (pyrolysis)
NA NA Dehydrogenation at C-3/15
NA NA Demethylation at C-1/6/16 or the N-methyl group with dehydrogenation at C-3/15;
demethylation with dehydrogenation at the same methoxyl group, O remained as a carbonyl group.
NA Demethylation and deoxylation NA
Lipoalkaloids via ester exchange at C-8 with long chain fatty acids. Lipoalkaloids via ester exchange at C-8 with short/long chain fatty acids. NA

MDAs NA NA Hydroxylation
Demethylation
Didemethylation or deethylation
Dehydrogenation
Demethylation and (di)dehydrogenation

aNot available.

Interestingly, phase I metabolites of hydroxylation, deoxylation, demethylation, and didemethylation/deethylation were detected not only in the liver but also in the gastrointestinal tract. As mentioned above in Section 2.2, intestinal bacteria participate in metabolism, such as through deoxylation, reduction, and deacetylation. However, it has also been reported that human small intestinal epithelial cells express a range of P450s, which include CYP3A, the isoenzyme that dominates in the liver [49]. Intestinal metabolism was performed in vitro through anaerobic incubation in a feces suspension, despite the symbiotic intestinal bacteria, which should also contain apoptosis-undergoing intestinal epithelial cells that release phase I and phase II metabolic enzymes into the suspension. Thus, intestinal metabolites are likely converted by both bacteria and phase I metabolic enzymes.

Metabolic isoenzyme expression is not identical among different species [50] that lead to metabolic differences in different species. Based on references in this review, we find that DDAs were ester hydrolysed to MDAs in rat intestine and liver, but not in humans. On the other hand, the same metabolites converted in different species have been reported. For example, 16-O-demethyl BAC, the ester hydrolysed products from 16-O-demethyl AC in intestinal metabolism, was detected not only in rats but also in humans. Hydroxy aconitine from AC was detected through incubation in liver microsomes or S9 from humans, rats, guinea pigs, and mice. It is notable that the AC demethylation at the C-16 position is catalysed by CYP3A and CYP1A1/2 in rats while it is catalysed by CYP3A, CYP2D6, and CYP2C9 in humans. However, no studies have specifically compared metabolites from DDAs or MDAs among humans and different experimental animals. Briefly, the metabolic differences in different species yield certain risks in predicting human drug metabolism based on data from experimental animals.

The metabolic pathways proposed for DDAs are generalized in Figure 1.

Figure 1.

Figure 1

Proposed DDA metabolic pathways. The organ/tissue metabolic processes are partially indicated. The wavy bonds indicate the potential metabolic positions. Me, Et, Ac, and Bz indicate methyl, ethyl, acetyl, and benzoyl groups, respectively.

The organ/tissue metabolic processes are partially indicated. The wavy bonds indicate the potential metabolic positions. Me, Et, Ac, and Bz indicate methyl, ethyl, acetyl, and benzoyl groups, respectively.

3. Metabolites Detected in the Blood

MDAs and alcohol amines are the main DDA metabolites in the blood (Table 7). It has been suggested that AC and related alkaloids can be rapidly absorbed by the upper gastrointestinal tract for the short latent period between the ingestion of aconite roots and the onset of poisoning features [3]. Therefore, the absorbed DDAs may be partially and gradually ester hydrolysed to less toxic MDAs and nontoxic alcohol amines by CEs distributed in the blood. Furthermore, the blood provides a suitable pH environment for ester hydrolysis. This hypothesis is supported by an analysis of rat plasma after DDA administration via a tail vein, wherein MDAs and alcohol amines were detected [39].

Table 7.

DDA metabolites detected in the plasma.

DDAs m/z (ESI+) Formula Identification Metabolic procedure MS detection References
AC 604 C32H45NO10 BAC Mouse; plasma; ig, in vivo. GC/MS [51]
Rabbit; plasma; ig, in vivo. IT [52] (M2)
590 C31H43NO10 16-O-Demethyl BAC Rabbit; plasma; ig, in vivo. IT [52] (M3)
500 C25H41NO9 Aconine Rats; plasma; iv, in vivo.a Q-Trap [39]
Mouse; plasma; ig, in vivo. GC/MS [51]
Rabbit; plasma; ig, in vivo. IT [52] (M4)

MA 590 C31H43NO10 BMA Rats; plasma; iv, in vivo.a Q-Trap [39]
486 C24H40NO9 Mesaconine

HA 574 C31H44NO9 BHA Rats; plasma; iv, in vivo.a Q-Trap [39]

aA mixture of AC, MA, and HA was administered via the tail vein.

MDAs and alcohol amines are commonly considered markers in forensic and clinical evaluations of aconitine poisoning because their half-lives are longer than DDAs [19], which might lead to the neglect of other metabolites in the blood. Additionally, many efflux/influx transporters, such as P-glycoprotein (P-gp), multidrug resistance-associated protein 2 (MRP2), and MRP3 expressed in intestinal epithelial and hepatic cells, are involved in drug absorption [53]. It is difficult to determine whether the various metabolites produced in the gastrointestinal tract and liver are transported into the blood from the few studies on their transport mechanism.

4. Metabolites Detected in the Urine

The metabolites found in the urine are shown in Table 8. Compared with intestinal and hepatic metabolites, most metabolites from hydroxylation, deoxylation, demethylation, deethylation/didemethylation, dehydrogenation, ester hydrolysis, deacetoxylation (pyrolysis), and demethylation with deoxylation have been found in the urine. Further, a few phase II metabolites as glucuronide and sulfate conjugates have been found in the urine but have not been reported in hepatic or intestinal metabolism in vitro. Glucuronidation catalysed by UGTs occurs in human and rat kidneys [63, 64]; glucuronidation might be responsible for phase II biotransformation processes in addition to hepatic and intestinal metabolism.

Table 8.

Metabolites of AC, MA, and HA (DDAs) detected in the urine.

DDAs m/z (ESI+) Formula Identification Metabolic procedure MS detection References

AC 780 C38H53NO16 BAC glucuronide conjugate Rats; ig, in vivo. IT [54]
726 C34H47NO14S AC sulfate conjugate
662 C34H47NO12 10-Hydroxy AC Rats; ig, in vivo. IT [54]
Rats; ig, in vivo. IT [36] (M5)
644 C34H45NO11 3-Dehydrogen AC Rats; ig, in vivo. IT [36] (M7)
632 C33H45NO11 16-O-Demethyl AC Rats; ig, in vivo. IT [54]
Rats; ig, in vivo. IT [55] (M2)
Rabbits; ig, in vivo. IT [56] (M1)
Rabbits; iv and ig, in vivo. IT [48] (M1, found in both iv and ig)
Rabbits (male and female); ig, in vivo. IT [57] (M5)
Human (female); po, in vivo.a IT [58] (M4)
Rats; ig, in vivo. IT [36] (M6)
Rabbits; ig, in vivo. IT [59] (M1)
Human (female); po, in vivo.b IT [60] (M7)
1-O-Demethyl AC Rats; ig, in vivo. IT [54]
6-O-Demethyl AC
MA Rats; ig, in vivo. IT [55] (M1)
630 C34H47NO10 Deoxy AC Rats; ig, in vivo. IT [54]
Rats; ig, in vivo. IT [36] (M8)
618 C32H43NO11 16-O-Demethyl MA Rats; ig, in vivo. IT [55] (M3)
8-Methoxy BAC Rats; ig, in vivo. IT [54]
1-O-Demethyl MA Rats; ig, in vivo. IT [54]
N-Deethyl AC (M2) Rats; ig, in vivo. IT [36]
O-Didemethyl AC (M4)
616 C33H45NO10 1-O-Demethyl-13-deoxy AC Rats; ig, in vivo. IT [54]
Demethyl-deoxy AC Rabbits; iv and ig, in vivo. IT [48] (M2, found in ig only)
606 C31H43NO11 10-Hydroxy BMA Rats; ig, in vivo. IT [54]
604 C32H45NO10 BAC Rabbits; ig, in vivo. IT [56] (M2)
Rats; ig, in vivo. IT [55] (M4)
Rabbits (male and female); ig, in vivo. IT [57] (M2)
Rabbits; ig, in vivo. IT [59] (M2)
Rats; ig, in vivo. IT [54]
Human (female); po, in vivo.a IT [58] (M1)
Human (female); po, in vivo.b IT [60] (M4)
Rats; ig, in vivo. IT [36] (M1)
590 C31H43NO10 16-O-Demethyl BAC Rabbits; ig, in vivo. IT [56] (M3)
Rabbits (male and female); ig, in vivo. IT [57] (M3)
Rabbits; ig, in vivo. IT [59] (M3)
588 C32H45NO9 3-Deoxy BAC Rats; ig, in vivo. IT [54]
586 C32H43NO9 Pyroaconitine (deacetoxy AC) Rabbits (male and female); ig, in vivo. IT [57] (M6, found in male only)
Rats; ig, in vivo. IT [54]
Rats; ig, in vivo. IT [36] (M3)
500 C25H41NO9 Aconine Rabbits; ig, in vivo. IT [56] (M4)
Rabbits (male and female); ig, in vivo. IT [57] (M4)
Rabbits; ig, in vivo. IT [59] (M4)
Rats; ig, in vivo. IT [54]
482 C25H39NO8 Dehydrated aconine Human; po, in vivo.c IT [40]

Alkaloids m/z (ESI+) Formula Identification Metabolic procedure MS detection References

MA 766 C37H51NO16 BMA glucuronide conjugate Rats; ig, in vivo. IT [61] (M1)
648 C33H45NO12 10-Hydroxy MA Rats; ig, in vivo. IT [61] (M2)
618 C32H43NO11 1-O-Demethyl MA Rats; ig, in vivo. IT [61] (M3)
Demethyl MA Rats; ig, in vivo.d TOF [62] (M10)
616 C33H45NO10 Deoxy MA Rats; ig, in vivo. IT [61] (M4)
590 C31H43NO10 BMA Rats; ig, in vivo. IT [61] (M5)
Human (female); po, in vivo.a IT [58] (M2)
Human (female); po, in vivo.b IT [60] (M5)
468 C24H37NO8 Dehydrated mesaconine Human; po, in vivo.c IT [40]

HA 602 C32H43NO10 16-O-Demethyl HA Human (female); po, in vivo.a IT [58] (M5)
Human (female); po, in vivo.b IT [60] (M8)
574 C31H43NO9 BHA Human (female); po, in vivo.a IT [58] (M3)
Human (female); po, in vivo.b IT [60] (M6)

a,bDDA was produced through decoction containing Aconiti and Aconiti Kusnezoffii Radix.

It is not clear whether these compounds were directly metabolized from DDAs or originally ingested.

cDDA was produced from a medical liquor containing Aconiti Kusnezoffii Radix.

It is not clear whether these compounds were directly metabolized from DDAs or originally ingested.

dDDA was produced from a liquid of crude aconite root decoction via ethanol precipitation.

It is not clear whether these compounds were directly metabolized from DDAs or originally ingested.

Additionally, mRNA for CYP3A4 and CYP3A5, which are the major isoforms that catalyse DDA metabolism, is also expressed in human kidneys, but the expression levels are much lower than in the liver and intestine [65]. Based on the data in Section 3, metabolites from DDAs in the blood are fewer than in the urine. Further, the urine is converted from the blood in the kidney. Perhaps, the various metabolites in the urine are converted from DDAs and their ester hydrolysed products in the blood by metabolic enzymes expressed at low levels in the kidney. Is it possible that various metabolites from DDAs produced in the intestine and liver are absorbed in the blood and excreted in the urine? However, as noted in Section 3, the data on metabolites in the blood is insufficient.

No studies have reported on metabolites of lipoalkaloids in the urine, which are the metabolites characteristically produced in the gastrointestinal tract. DDA lipophilicity may be reasonably increased through ester exchange with long chain fatty acids at the C-8 position, which results in easier absorption of lipoalkaloids into the blood. Are the ester groups then hydrolysed by CEs in the blood and liver, producing MDAs and alcohol amines, or are they directly excreted through the feces? Such conjecture requires further investigation.

5. Original Compound Stability

All of the in vivo and in vitro metabolism reactions occur in fluid. Therefore, the stability of DDAs and MDAs in different pH aqueous solutions should be considered. One study reported that AC and MA were decomposed dramatically after incubation in water for 24 h at 25°C (degrees Celsius), and the products of AC were BAC, aconine, deacetoxy AC, and deoxy AC. In addition, almost half of the AC and MA were depleted in phosphate buffer at pH 2.0 and 6.8 over 12 h at 25°C (degrees Celsius); these pH values are similar to gastric acid and intestinal juice, respectively [66]. These results imply that metabolites, such as BAC and aconine, may be partially converted from DDAs in body fluid without enzyme catalysis. On the other hand, the rate of MDA formation from DDAs was much higher in phosphate buffer (pH 7.4) with hepatic microsomes than in the negative control without hepatic microsomes [39]. The facts imply that the enzymes did affect bioconversion of instable DDAs.

6. Metabolite Detection and Identification

Metabolites are typically varied at trace levels with endogenous interference from biological matrices, such as tissue, the blood, or urine. Liquid chromatography multiple-stage tandem mass spectrum (LC/MSn) has been widely applied for drug metabolite detection due to its high sensitivity and selectively.

For DDAs and MDAs, positive electrospray ionization (ESI+) is suitable for alkaloid ionization. Quadrupole time of flight (Q-TOF) and Fourier transform ion cyclotron resonance (FT-ICR) MS techniques are applied to metabolite identification due to their high resolution of pseudomolecular ions. Fragment ions are obtained step-by-step through ion trap (IT) MS, which is helpful for deducing the chemical structures. The acyl groups from fatty acids are confirmed by GC-MS, and neutral fatty acid losses are observed in LC-MS [24].

The fragmentation pathways of different types of Aconitum alkaloids include diagnostic ions. For the AC-type of alkaloid, the diagnostic ions are [M+H-18 (water)]+, [M+H-60 (acetate from C-8 and C-15)]+, [M+H-60-32 (methanol)-28 (carbonyl group)]+, and [M+H-60-32-28-122 (benzoic acid at C-14)]+[14, 22]. For the BAC-type, the diagnostic ions are [M+H-50 (methanol and water)]+, [M+H-50-32]+, and [M+H-50-32-18]+ [60]. For lipoaconitine, the diagnostic ions are 586 ([Mass of AC+H-60]+) with neutral fatty acid losses that correspond to acyl groups at the C-8 position [24].

However, MSn analyses only provide a possible fragmentation pattern based on the mass difference between pseudomolecular and fragment ions, and the metabolite confirmations are not necessarily accurate. Considering HA, the demethylation reaction position is ambiguous due to the five methyl groups at the C-1, C-6, C-16, C-18, and nitro positions. Demethylation with dehydrogenation was inferred to occur at the methoxy and hydroxy groups that attach to different skeleton carbons in MA [41] (see Figure 1), while it occurs at the same methoxy group in HA, forming a carbonyl group [43] (see Figure 1). However, detailed structure determination for these two types of metabolites was not provided.

7. Conclusions

In this review, we classify and summarize metabolites of highly toxic DDAs and less toxic MDAs from the gastric and intestinal content, intestinal bacterial juice, hepatic microsomes, blood, and urine from different animal species and humans in vivo and in vitro. For example, considering AC, which is the most researched toxic DDA, we generalize a process of toxicity reduction in body after oral AC administration for the first time (Figure 2).

Figure 2.

Figure 2

The proposed process of toxicity reduction after oral AC administration in humans and experimental animals. The metabolites from ester exchange are lipo-alkaloids. Ester hydrolysis occurs at the C-8 or/and C-14 position, producing benzoylaconine (BAC) and aconine. Phase I metabolism refers to hydroxylation, deoxylation, dehydrogenation, demethylation, and didemethylation/deethylation. A few phase II metabolites were detected in the urine, including BAC glucuronide and AC sulfate conjugates. Cytochrome P450 enzymes (CYP450s), carboxylesterases (CEs), and enzymes produced by intestinal bacteria are involved in gastrointestinal and hepatic metabolism of aconitine (AC).

The metabolites from ester exchange are lipoalkaloids. Ester hydrolysis occurs at the C-8 or/and C-14 position, producing benzoylaconine (BAC) and aconine. Phase I metabolism refers to hydroxylation, deoxylation, dehydrogenation, demethylation, and didemethylation/deethylation. A few phase II metabolites were detected in the urine, including BAC glucuronide and AC sulfate conjugates. Cytochrome P450 enzymes (CYP450s), carboxylesterases (CEs), and enzymes produced by intestinal bacteria are involved in gastrointestinal and hepatic metabolism of aconitine (AC).

In conclusion, CYP450s, CEs, and enzymes produced by intestinal bacteria are mainly involved in DDA metabolism in both the gastrointestinal tract and liver after oral administration, including hydroxylation, deoxylation, demethylation, dehydrogen, pyrolysis, ester hydrolysis, and ester exchange. Phase II conjugation of DDAs is not the dominant metabolic process and only a few conjugated DDAs are found in the urine. DDA metabolites in the blood are not as various as those in the urine.

Thus far, reports of less toxic MDA metabolism have only been related to hepatic metabolism. Nevertheless, MDAs may share similar metabolic pathways (except ester hydrolysis at the C-8 position) with DDAs in the gastrointestinal tract based on the same DDA and MDA diterpenoid skeletons and similar hepatic metabolism between DDAs and MDAs.

As summarized above, toxic DDAs and MDAs are converted to metabolites that are less toxic or easier to excrete in the gastrointestinal tract and liver after oral administration. However, for drug excretion, few phase II metabolism conjugations are formed, which are the most hydrosoluble metabolites. Further, this detoxification effect is likely restricted due to rapid DDA absorption by the upper gastrointestinal tract.

Although the many available studies on metabolism and toxicity of DDAs and MDAs are helpful, they are insufficient for safe clinical administration of Aconitum herbs. Several issues must be further studied and verified. More attention should be paid to metabolism of MDAs because they are not sufficiently safe for clinical use. Due to metabolic interspecific differences, it is more reasonable to apply human recombinant metabolic isozymes or humanized animal models [67] to a human metabolism study. Studies have not confirmed whether the various metabolites detected in the urine are from gastrointestinal and hepatic metabolism via absorption into the blood or from biotransformation in the kidney. Because the metabolites are detected at trace levels, it is difficult to accumulate such metabolites for identification, bioassays, or toxicity studies. However, the changes in bioactivity or toxicity after metabolism are unambiguous.

Based on our conclusions, it is worthwhile to perform an in-depth investigation of the Aconitum herbs compatible with other medicines, such as prescription licorice, which is featured in and crucial to clinical application of Aconitum herbs in traditional Chinese medicine. To a certain extent, drug-drug interactions are the essence of a drug-drug combination, in which drug metabolism and/or absorption is changed by affecting (inducing or inhibiting) another with respect to metabolic enzymes or/and transporters; thus, drug pharmacological activity or toxicity is consequently affected [12, 13, 67].

Acknowledgment

This work was mainly supported by the National Natural Science Foundation of China (no. 81274062).

Abbreviations

AC:

Aconitine

BAC:

14-Benzoylaconine or 8-O-deacetyl aconitine

BHA:

14-Benzoylhypaconine or 8-O-deacetyl hypaconitine

BMA:

14-Benzoylmesaconine or 8-O-deacetyl mesaconitine

CEs:

Carboxylesterases

CYP450s:

Cytochrome P450 enzymes

DDAs:

Diester diterpenoid alkaloids

FT-ICR:

Fourier transform ion cyclotron resonance

HA:

Hypaconitine

HLM:

Human liver microsomes

IM:

Ion mobility

IT:

Ion trap

LD50:

Half-maximally lethal dose

MA:

Mesaconitine

MDAs:

Monoester diterpenoid alkaloids

MRP:

Multidrug resistance-associated protein

MS:

Mass spectrometry

NA:

Not available

P-gp:

P-glycoprotein

QQQ:

Triple quadrupole

Q-trap:

Quadrupole trap

Q-TOF:

Quadrupole time of flight

UGTs:

Uridine 5-diphosphate- (UDP-) glucuronosyltransferases.

Conflict of Interests

There is no financial conflict of interests with the authors of this review.

References

  • 1.Chan T. Y. K. Aconite poisoning presenting as hypotension and bradycardia. Human and Experimental Toxicology. 2009;28(12):795–797. doi: 10.1177/0960327109353056. [DOI] [PubMed] [Google Scholar]
  • 2.Singhuber J., Zhu M., Prinz S., Kopp B. Aconitum in traditional Chinese medicine—a valuable drug or an unpredictable risk? Journal of Ethnopharmacology. 2009;126(1):18–30. doi: 10.1016/j.jep.2009.07.031. [DOI] [PubMed] [Google Scholar]
  • 3.Chan T. Y. K. Aconite poisoning. Clinical Toxicology. 2009;47(4):279–285. doi: 10.1080/15563650902904407. [DOI] [PubMed] [Google Scholar]
  • 4.Wang Y.-G., Wang S.-Q., Liu Y.-X., Yan L.-P., Dou G.-F., Gao Y. Characterization of metabolites and cytochrome P450 isoforms involved in the microsomal metabolism of aconitine. Journal of Chromatography B. 2006;844(2):292–300. doi: 10.1016/j.jchromb.2006.07.059. [DOI] [PubMed] [Google Scholar]
  • 5.Chinese Pharmacopoeia Commission. Chinese Pharmacopoeia, Part I. Beijing, China: China Medical Science Press; 2010. Aconiti Radix; p. p. 36. [Google Scholar]
  • 6.Chinese Pharmacopoeia Commission. Chinese Pharmacopoeia Part I. Beijing, China: China Medical Science Press; 2010. Aconiti Kusnezoffii Radix Cocta; p. p. 220. [Google Scholar]
  • 7.Chinese Pharmacopoeia Commission. Chinese Pharmacopoeia. 2010. part 1. Beijing, China: China Medical Science Press; 2010. Aconiti lateralis radix praeparata; p. p. 177. [Google Scholar]
  • 8.Chinese Pharmacopoeia Commission. Chinese Pharmacopoeia, Part I. Beijing, China: China Medical Science Press; 2010. Aconiti Radix Cocta; p. p. 37. [Google Scholar]
  • 9.Huang Q.-A., Zhang Y.-M., He Y., Lu J., Lin R.-C. Studies on hydrolysis of aconitine. China Journal of Chinese Materia Medica. 2007;32(20):2143–2145. [PubMed] [Google Scholar]
  • 10.Zheng Q., Lu H.-W., Hao W.-W., Liu J.-Y., Wang S.-J., Yang M. Study on hydrolysis of Aconitum alkaloids and quantitative analysis method of their hydrolysates. Chinese Pharmaceutical Journal. 2011;46(9):652–656. [Google Scholar]
  • 11.Zhou Y.-P., Liu W.-H., Zeng G.-Y., Chen D.-H., Li H.-Y., Song W.-L. The toxicity of aconitine and its analogs and their effects on cardiac contractile function. Acta Pharmaceutica Sinica. 1984;19(9):641–646. [PubMed] [Google Scholar]
  • 12.Rahimi R., Abdollahi M. An update on the ability of St. John's wort to affect the metabolism of other drugs. Expert Opinion on Drug Metabolism & Toxicology. 2012;8(6):691–708. doi: 10.1517/17425255.2012.680886. [DOI] [PubMed] [Google Scholar]
  • 13.König J., Müller F., Fromm M. F. Transporters and drug-drug interactions: important determinants of drug disposition and effects. Pharmacological Reviews. 2013;65(3):944–966. doi: 10.1124/pr.113.007518. [DOI] [PubMed] [Google Scholar]
  • 14.Sui Z.-G., Li N., Liu Z.-Q., Yan J., Liu Z.-Y. Metabolite profile analysis of aconitine in rabbit stomach after oral administration by liquid chromatography/electrospray ionization/multiple-stage tandem mass spectrometry. Xenobiotica. 2013;43(7):628–635. doi: 10.3109/00498254.2012.753490. [DOI] [PubMed] [Google Scholar]
  • 15.Enayetallah A. E., French R. A., Thibodeau M. S., Grant D. F. Distribution of soluble epoxide hydrolase and of cytochrome P450 2C8, 2C9, and 2J2 in human tissues. Journal of Histochemistry and Cytochemistry. 2004;52(4):447–454. doi: 10.1177/002215540405200403. [DOI] [PubMed] [Google Scholar]
  • 16.DiBaise J. K., Zhang H., Crowell M. D., Krajmalnik-Brown R., Decker G. A., Rittmann B. E. Gut microbiota and its possible relationship with obesity. Mayo Clinic Proceedings. 2008;83(4):460–469. doi: 10.4065/83.4.460. [DOI] [PubMed] [Google Scholar]
  • 17.Fukuhara A., Imai T., Otagiri M. Stereoselective disposition of flurbinprofen from a mutual prodrug with a histamine H-2-antagonist to reduce gasrointestinal lesions in the rat. Chirality. 1996;8:494–502. doi: 10.1002/(SICI)1520-636X(1996)8:7<494::AID-CHIR6>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  • 18.Satoh T., Hosokawa M. The mammalian carboxylesterases: from molecules to functions. Annual Review of Pharmacology and Toxicology. 1998;38:257–288. doi: 10.1146/annurev.pharmtox.38.1.257. [DOI] [PubMed] [Google Scholar]
  • 19.Usui K., Hayashizaki Y., Hashiyada M., Nakano A., Funayama M. Simultaneous determination of 11 aconitum alkaloids in human serum and urine using liquid chromatography-tandem mass spectrometry. Legal Medicine. 2012;14(3):126–133. doi: 10.1016/j.legalmed.2012.01.006. [DOI] [PubMed] [Google Scholar]
  • 20.Ley R. E., Peterson D. A., Gordon J. I. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124(4):837–848. doi: 10.1016/j.cell.2006.02.017. [DOI] [PubMed] [Google Scholar]
  • 21.Sousa T., Paterson R., Moore V., Carlsson A., Abrahamsson B., Basit A.-W. The gastrointestinal microbiota as a site for the biotransformation of drugs. International Journal of Pharmaceutics. 2008;363(1-2):1–25. doi: 10.1016/j.ijpharm.2008.07.009. [DOI] [PubMed] [Google Scholar]
  • 22.Wang X.-Y., Pi Z.-F., Liu W.-L., Zhao Y.-F., Liu S.-Y. Effect of pH on the metabolism of aconitine under rat intestinal bacteria and analysis of metabolites using HPLC/MS-MSn technique. Chinese Journal of Chemistry. 2010;28(12):2494–2500. doi: 10.1002/cjoc.201190028. [DOI] [Google Scholar]
  • 23.Sui Z.-G., Jiang Y.-Q., Liu Z.-Q., Liang F., Yan J., Liu Z.-Y. Study on metabolites of aconitine in rabbit intestines by using LC/ESI-MSn. Acta Chimica Sinica. 2009;67(21):2439–2444. [Google Scholar]
  • 24.Zhao Y.-F., Song F.-R., Guo X.-H., Liu S.-Y. Studies on the biotransformation of aconitine in human intestinal bacteria using soft-ionization mass spectrometry. Chemical Journal of Chinese Universities. 2008;29(1):55–59. [Google Scholar]
  • 25.Wang X.-Y., Pi Z.-F., Liu W.-L., Song F.-R., Liu Z.-Q., Liu S.-Y. 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. Chemical Journal of Chinese Universities. 2011;32(7):1526–1531. [Google Scholar]
  • 26.Wang X.-Y., Pi Z.-F., Song F.-R., Liu Z.-Q., Liu S.-Y. Studies on the biotransformation of Licorice and Aconite accessory root decoction and Atractylodes Macrocephala and Aconite accessory root decoction under rat intestinal bacteria. Acta Chimica Sinica. 2011;69(11):1368–1374. [Google Scholar]
  • 27.Xin Y., Pi Z.-F., Song F.-R., Liu Z.-Q., Liu S.-Y. Study on the metabolic characteristics of aconite alkaloids in the extract of Radix aconiti under intestinal bacteria of rat by UPLC/MSn technique. Chinese Journal of Chemistry. 2012;30(3):656–664. doi: 10.1002/cjoc.201100228. [DOI] [Google Scholar]
  • 28.Zhao Y.-F., Song F.-R., Wang X.-Y., Guo X.-H., Liu Z.-Q., Liu S.-Y. Studies on the biotransformation of 16-O-demethylaconitine and electrospray ionization tandem mass spectrometry. Acta Chimica Sinica. 2008;66(5):525–530. [Google Scholar]
  • 29.Zhao Y.-F., Song F.-R., Yue H., et al. Biotransformation of deoxyaconitine of metabolite of aconitine by human intestinal bacteria and electrospray ionization tandem mass spectrometry. Chemical Journal of Chinese Universities. 2007;11:2051–2055. [Google Scholar]
  • 30.Zhao Y.-F., Song F.-R., Yue H., et al. Studies on the biotransformation of 16-O-edmethyldeoxyaconitine of the metabolite of aconitine in human intestinal bacteria. Chinese Journal of Analytical Chemistry. 2007;35(12):1711–1715. [Google Scholar]
  • 31.Liu W.-L., Liu Z.-Q., Song F.-R., Liu S.-Y. Specific conversion of diester-diterpenoid aconitum alkaloids components into hydrolysis monoester-diterpenoid alkaloids components and lipo-alkaloids components. Chemical Journal of Chinese Universities. 2011;32(3):717–720. [Google Scholar]
  • 32.Qi M.-F. Another explanation of processing mechanism of Aconiti radix and Aconiti lateralis radix praeparata. Journal of Chinese Medicinal Materials. 1986;(6):37–38. [Google Scholar]
  • 33.Bock K. W. Functions and transcriptional regulation of adult human hepatic UDP-glucuronosyl-transferases (UGTs): mechanisms responsible for interindividual variation of UGT levels. Biochemical Pharmacology. 2010;80(6):771–777. doi: 10.1016/j.bcp.2010.04.034. [DOI] [PubMed] [Google Scholar]
  • 34.Zhu L., Ge G., Liu Y., et al. Characterization of UDP-glucuronosyltransferases involved in glucuronidation of diethylstilbestrol in human liver and intestine. Chemical Research in Toxicology. 2012;25(12):2663–2669. doi: 10.1021/tx300310k. [DOI] [PubMed] [Google Scholar]
  • 35.Tang L., Ye L., Lv C., Zheng Z.-J., Gong Y., Liu Z.-Q. Involvement of CYP3A4/5 and CYP2D6 in the metabolism of aconitine using human liver microsomes and recombinant CYP450 enzymes. Toxicology Letters. 2011;202(1):47–54. doi: 10.1016/j.toxlet.2011.01.019. [DOI] [PubMed] [Google Scholar]
  • 36.Chen X.-G., Lai Y.-Q., Cai Z.-W. Simultaneous analysis of aconitine and its metabolites by liquid chromatography-electrospray ion trap mass spectrometry. Journal of Chinese Mass Spectrometry Society. 2012;33:65–73. doi: 10.1093/jat/bks004. [DOI] [PubMed] [Google Scholar]
  • 37.Xie L.-X., Lv C., Ye L., Tang L. Study on metabolism of aconitine in liver microsomes of guinea pig and mice. China Pharmacy. 2012;23:590–593. [Google Scholar]
  • 38.Bi Y.-F., Liu S., Li X., Liu Z.-Q., Song F.-R. Metabolic fingerprint and effects of aconite alkaloid components on the activities of CYP450 isozymes in rat liver microsomes. Chemical Journal of Chinese Universities. 2013;34(9):2084–2089. doi: 10.7503/cjcu20130492. [DOI] [Google Scholar]
  • 39.Ye L., Gao S., Feng Q., et al. Development and validation of a highly sensitive UPLC-MS/MS method for simultaneous determination of aconitine, mesaconitine, hypaconitine, and five of their metabolites in rat blood and its application to a pharmacokinetics study of aconitine, mesaconitine, and hypaconitine. Xenobiotica. 2012;42(6):518–525. doi: 10.3109/00498254.2011.641608. [DOI] [PubMed] [Google Scholar]
  • 40.Liu Y.-G., Sun M.-Q., Zhang H.-G. Studies on the metabolic pathway of aconitine in rabbit and human using electrospray ionization-mass spectrometry. Journal of Liquid Chromatography and Related Technologies. 2013;36(12):1686–1696. doi: 10.1080/10826076.2012.695315. [DOI] [Google Scholar]
  • 41.Ye L., Tang L., Gong Y., et al. Characterization of metabolites and human P450 isoforms involved in the microsomal metabolism of mesaconitine. Xenobiotica. 2011;41(1):46–58. doi: 10.3109/00498254.2010.524950. [DOI] [PubMed] [Google Scholar]
  • 42.Bi Y.-F., Liu S., Zhang R.-X., Song F.-R., Liu Z.-Q. Metabolites and metabolic pathways of mesaconitine in rat liver microsomal investigated by using UPLC-MS/MS method in vitro. Yaoxue Xuebao. 2013;48(12):1823–1828. [PubMed] [Google Scholar]
  • 43.Ye L., Wang T., Yang C.-H., et al. Microsomal cytochrome P450-mediated metabolism of hypaconitine, an active and highly toxic constituent derived from Aconitum species. Toxicology Letters. 2011;204(1):81–91. doi: 10.1016/j.toxlet.2011.04.015. [DOI] [PubMed] [Google Scholar]
  • 44.Bi Y.-F., Li X., Pi Z.-F., Song F.-R., Liu Z.-Q. Analysis of hypaconitine's metabolites and related metabolic CYP isoforms in rat liver microsomal by UPLC-MS/MS. Journal of Chinese Mass Spectrometry Society. 2013;34(6):330–337. doi: 10.7538/zpxb.2013.34.06.0330. [DOI] [Google Scholar]
  • 45.Ye L., Yang X.-S., Lu L.-L., et al. Monoester-diterpene Aconitum alkaloid metabolism in human liver microsomes: predominant role of CYP3A4 and CYP3A5. Evidence-Based Complementary and Alternative Medicine. 2013;2013:24. doi: 10.1155/2013/941093.941093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li X., Yu Y.-N. Advance in research of drug phase II metabolisms and conjugatio enzymes. Chinese Journal of Clinical Pharmacology. 2000;16:458–465. [Google Scholar]
  • 47.Silva M. J., Barr D. B., Reidy J. A., et al. Glucuronidation patterns of common urinary and serum monoester phthalate metabolites. Archives of Toxicology. 2003;77:561–567. doi: 10.1007/s00204-003-0486-3. [DOI] [PubMed] [Google Scholar]
  • 48.Liang F., Sui Z.-G., Yan J., Liu Z.-Y. Comparison of metabolites of aconitine in rabbit urine under different routes of administration. Journal of Jilin University. 2010;36(3):443–445. [Google Scholar]
  • 49.Galetin A., Gertz M., Houston J.-B. Potential role of intestinal first-pass metabolism in the prediction of drug-drug interactions. Expert Opinion on Drug Metabolism and Toxicology. 2008;4(7):909–922. doi: 10.1517/17425255.4.7.909. [DOI] [PubMed] [Google Scholar]
  • 50.Komura H., Iwaki M. In vitro and in vivo small intestinal metabolism of CYP3A and UGT substrates in preclinical animals species and humans: species differences. Drug Metabolism Reviews. 2011;43(4):476–498. doi: 10.3109/03602532.2011.597401. [DOI] [PubMed] [Google Scholar]
  • 51.Wada K., Nihira M., Hayakawa H., Tomita Y., Hayashida M., Ohno Y. Effects of long-term administrations of aconitine on electrocardiogram and tissue concentrations of aconitine and its metabolites in mice. Forensic Science International. 2005;148(1):21–29. doi: 10.1016/j.forsciint.2004.04.016. [DOI] [PubMed] [Google Scholar]
  • 52.Zhang H.-G., Shi X.-G., Sun Y., Zhong D.-F., Zhang H.-Q. Study on the metabolites of aconitine in rabbit blood. Journal of Jilin University (Science edition) 2006;44:284–286. [Google Scholar]
  • 53.Zamek-Gliszczynski M. J., Chu X.-Y., Polli J. W., Paine M. F., Galetin A. Understanding the transport properties of metabolites: case studies and considerations for drug development. Drug Metabolism and Disposition. 2014;42:650–664. doi: 10.1124/dmd.113.055558. [DOI] [PubMed] [Google Scholar]
  • 54.Ye X.-L., He X.-W., Song Q.-Q., et al. Analysis on the metabolites of aconitine in Sini decoction (SND) in the rat urine by liquid chromatography and electrospray ionization mass spectrometry. Proceedings of the Annual Conference Symposium of Innovation and Development Forum on Chinese Medicine Preparations; 2011; Kunming, China. pp. 556–562. [Google Scholar]
  • 55.Wang C.-H., Wen J., Chen Y.-H., He Y. Study on determination of metabolites of aconitine in rat urine by HPLC/MS. Chinese Journal of Forensic Medicine. 2006;21(2):88–90. [Google Scholar]
  • 56.Sun Y., Zhang H.-G., Shi X.-G., Duan M.-Y., Zhong D.-F. Study on metabolites on aconitine in rabbit urine. Acta pharmaceutica Sinica. 2002;37(10):781–783. [PubMed] [Google Scholar]
  • 57.Sun Y., Zhang Q.-S., Dong L.-D., Chen Y.-J. Metabolites of major alkaloids of Aconitum Chinese herbal medicine in different gender rabbit urine. Journal of Jilin University (Science edition) 2007;45:1032–1034. [Google Scholar]
  • 58.Ai L., Sun Y., Zhang H.-G. Metabolites of aconitum alkaloids from compound formula of Chinese medicine in human body. Journal of Beijing University of Traditional Chinese Medicine. 2007;30:417–422. [Google Scholar]
  • 59.Zhang H.-G., Shi X.-G., Sun Y., Duan M.-Y., Zhong D.-F. New metabolites of aconitine in rabbit urine. Chinese Chemical Letters. 2002;13(8):758–760. [Google Scholar]
  • 60.Zhang H.-G., Sun Y., Duan M.-Y., Chen Y.-J., Zhong D.-F., Zhang H.-Q. Separation and identification of Aconitum alkaloids and their metabolites in human urine. Toxicon. 2005;46(5):500–506. doi: 10.1016/j.toxicon.2005.06.014. [DOI] [PubMed] [Google Scholar]
  • 61.Chen P.-P., Zhao N., Xu X.-L., Ruan Y.-P., Wei Y.-H., Li F.-Z. Analysis on the metabolites of mesaconitine in the rat urine by liquid chromatography and electrospray ionization mass spectrometry. Acta Pharmaceutica Sinica. 2010;45(8):1043–1047. [PubMed] [Google Scholar]
  • 62.Tan G.-G., Lou Z.-Y., Jing J., et al. Screening and analysis of aconitum alkaloids and their metabolites in rat urine after oral administration of aconite roots extract using LC-TOFMS-based metabolomics. Biomedical Chromatography. 2011;25(12):1343–1351. doi: 10.1002/bmc.1607. [DOI] [PubMed] [Google Scholar]
  • 63.Nakamura A., Nakajima M., Yamanaka H., Fujiwara R., Yokoi T. Expression of UGT1A and UGT2B mRNA in human normal tissues and various cell lines. Drug Metabolism and Disposition. 2008;36(8):1461–1464. doi: 10.1124/dmd.108.021428. [DOI] [PubMed] [Google Scholar]
  • 64.Shelby M. K., Cherrington N. J., Vansell N. R., Klaassen C. D. Tissue mRNA expression of the rat UDP-glucuronosyltransferase gene family. Drug Metabolism and Disposition. 2003;31(3):326–333. doi: 10.1124/dmd.31.3.326. [DOI] [PubMed] [Google Scholar]
  • 65.Nishimura M., Yaguti H., Yoshitsugu H., Naito S., Satoh T. Tissue distribution of mRNA expression of human cytochrome P450 isoforms assessed by high-sensitivity real-time reverse transcription PCR. Yakugaku Zasshi. 2003;123(5):369–375. doi: 10.1248/yakushi.123.369. [DOI] [PubMed] [Google Scholar]
  • 66.Yue H., Pi Z.-F., Li H.-L., Song F.-R., Liu Z.-Q., Liu S.-Y. 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) Phytochemical Analysis. 2008;19(2):141–147. doi: 10.1002/pca.1027. [DOI] [PubMed] [Google Scholar]
  • 67.Scheer N., Wolf C. R. Genetically humanized mouse models of drug metabolizing enzymes and transporters and their applications. Xenobiotica. 2014;44(2):96–108. doi: 10.3109/00498254.2013.815831. [DOI] [PubMed] [Google Scholar]

Articles from Evidence-based Complementary and Alternative Medicine : eCAM are provided here courtesy of Wiley

RESOURCES