Next Article in Journal
S-72, a Novel Orally Available Tubulin Inhibitor, Overcomes Paclitaxel Resistance via Inactivation of the STING Pathway in Breast Cancer
Previous Article in Journal
The Potential of Epigallocatechin-3-gallate (EGCG) as Complementary Medicine for the Treatment of Inflammatory Bowel Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neuropharmacological Potential of Diterpenoid Alkaloids

1
Department of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
2
Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
3
Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
4
Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81745-33871, Iran
5
Institute of Clinical Pharmacy, Faculty of Pharmacy, University of Szeged, 6720 Szeged, Hungary
6
National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(5), 747; https://doi.org/10.3390/ph16050747
Submission received: 15 March 2023 / Revised: 28 April 2023 / Accepted: 11 May 2023 / Published: 14 May 2023
(This article belongs to the Section Natural Products)

Abstract

:
This study provides a narrative review of diterpenoid alkaloids (DAs), a family of extremely important natural products found predominantly in some species of Aconitum and Delphinium (Ranunculaceae). DAs have long been a focus of research attention due to their numerous intricate structures and diverse biological activities, especially in the central nervous system (CNS). These alkaloids originate through the amination reaction of tetra or pentacyclic diterpenoids, which are classified into three categories and 46 types based on the number of carbon atoms in the backbone structure and structural differences. The main chemical characteristics of DAs are their heterocyclic systems containing β-aminoethanol, methylamine, or ethylamine functionality. Although the role of tertiary nitrogen in ring A and the polycyclic complex structure are of great importance in drug-receptor affinity, in silico studies have emphasized the role of certain sidechains in C13, C14, and C8. DAs showed antiepileptic effects in preclinical studies mostly through Na+ channels. Aconitine (1) and 3-acetyl aconitine (2) can desensitize Na+ channels after persistent activation. Lappaconitine (3), N-deacetyllapaconitine (4), 6-benzoylheteratisine (5), and 1-benzoylnapelline (6) deactivate these channels. Methyllycaconitine (16), mainly found in Delphinium species, possesses an extreme affinity for the binding sites of α7 nicotinic acetylcholine receptors (nAChR) and contributes to a wide range of neurologic functions and the release of neurotransmitters. Several DAs such as bulleyaconitine A (17), (3), and mesaconitine (8) from Aconitum species have a drastic analgesic effect. Among them, compound 17 has been used in China for decades. Their effect is explained by increasing the release of dynorphin A, activating the inhibitory noradrenergic neurons in the β-adrenergic system, and preventing the transmission of pain messages by inactivating the Na+ channels that have been stressed. Acetylcholinesterase inhibitory, neuroprotective, antidepressant, and anxiolytic activities are other CNS effects that have been investigated for certain DAs. However, despite various CNS effects, recent advances in developing new drugs from DAs were insignificant due to their neurotoxicity.

1. Introduction

Diterpenoid alkaloids (DAs) are characteristic components of some genera of the Ranunculaceae family, the occurrence of which is extraordinarily high in the genera Aconitum, Delphinium, and Consolida. Almost half of these natural compounds have been isolated from Aconitum sp. [1,2,3,4,5]. Despite being highly toxic, DAs possess interesting complex structures and noteworthy pharmacological and physiological effects which have attracted an exponentially increasing interest worldwide. Special emphasis has recently been placed on the research activities concerning the pharmacological and toxicological evaluations of these alkaloids. DAs are derivatives of the two nitrogen-free diterpenoids, ent-kaurene and ent-atisane, and therefore may be considered pseudoalkaloids. These compounds receive their nitrogen atoms from amino acids through an enzymatic transamination by aminotransferases [6]. Aconitum species commonly known as monkshood or wolfsbane were regarded as extremely toxic plants anciently used as arrow poisons, whereas Delphinium species have been considered to be less toxic [7]. Although DAs are known for their neurotoxicity, they exhibit a broad range of promising biological effects including analgesic, anticonvulsant, neuroprotective antiarrhythmic, antitumor, antimicrobial, and local anesthetic activities [8,9,10,11]. Before 1821, the Consolida species were classified in the Delphinium genus as a phylogroup. Since these two genera are morphologically very similar, they are repeatedly confused [12]. Regarding the similar chemical composition, their medicinal application has overlapped. Consolida sp. has been used for hundreds of years in China and the west of Asia as a medicinal plant for several nociceptive conditions, e.g., rheumatism, sciatica, and also as an anthelmintic [13,14]. Apart from the Ranunculaceae plants, nine species from Spiraea (Rosaceae) were found to have DAs [15]. The medical application of Spiraea sp. refers to traditional Chinese medicine which is used in some inflammatory and painful conditions such as toothache and headache. In addition, some sporadic studies reported the presence of DAs in Thalictrum (Ranunculaceae), Inula (Asteraceae), Garrya (Garryaceae), Erythrophleum (Fabaceae), Anopterus (Escalloniaceae), Rumex Pictus (Polygonaceae), Caesalpinia Sappan (Fabaceae), Artemisia korshinskyi (Asteraceae), and Isodon rubescens (Lamiaceae) species [6,15]. In traditional Chinese and Japanese medicine, for centuries, plants containing DAs have conventionally been used after being processed into less toxic products [16]. The unique pharmacological impacts of DAs on the central nervous system (CNS) encouraged scientists to seek new lead compounds (Scheme 1). Several authors have previously reviewed the recent evidence associated with DAs’ diversity, toxicity, biosynthesis, pharmacokinetics, structure–activity (or toxicity) relationship, and their analgesic, cytotoxicity, anti-inflammatory, and antimicrobial activities. However, the anticonvulsant, antagonizing α7 nAChR, antidementia, and neuroprotective effects as well as the biomolecular mode of action have not been concluded [6,8,17]. This narrative review aimed to summarize the CNS effects and biomolecular mechanism of action of DAs and plants containing DAs, along with classification, structural diversity, biosynthetic pathway, and the aspects restricting their widespread utilization mainly from the Aconitum and Delphinium species. A systematic search was conducted through the databases PubMed, Scopus, and Web of Science in December 2021.

2. Chemistry and Biosynthetic Pathway

2.1. Chemical Classification

Diterpenoid alkaloids originate through the amination reaction of tetra or pentacyclic diterpenoids. Based on the number of carbon atoms in the backbone structure, the 46 identified types are classified into three categories: C18, C19, and C20 diterpenoid alkaloids. These alkaloids possess a heterocyclic system containing β-aminoethanol, methylamine, or ethylamine functionality [4].
The small category of C18-DAs formed by demethylation at the C4 position of C19-DAs. C18-DAs are divided into five types: ranaconitines, lappaconitines, puberudines, puberunines, and sinomontadines (Figure 1). Lappaconitines possess a methine at C7 while ranaconitines are characterized by an oxygen-containing group at this position [8,18]. The second difference is in the C4 position, where the ranaconitines have a methine unit, while lappaconitines contain a methine unit or an oxygenated quaternary carbon (several compounds have been reported to possess chloro-substituents at C4). The three remaining types have undergone rearrangements as puberudines (open A ring), puberunines (unusual E ring), and sinomontadines (seven-membered A ring) [6].
It was reported that napeline and denudatine type of C20-DAs could lose a carbon unit to originate C19-DAs by the action of specific cyclases [6]. C19-DAs are structurally classified into six main types, aconitine, lactone, lycoctonine, 7,17-seco, franchetine, and glycosides, to gather with six rearranged subtypes which are chemically biosynthesized from aconitine type: acoseptines, vilmoraconitines, hemsleyaconitines, vilmotenitines, grandiflodines, and N-formyl-4, 19-secopacinine (Figure 2). Glycosides are characterized by a sugar unit (L-Arap and L-Araf) at the C1 or C14 positions of the aconitine structure [6]. C19-DAs are the largest category and hundreds of them were isolated from Aconitum and Delphinium species [6,8].
C20-DAs have a more complex structure which is characterized by a tetracyclic diterpenic backbone. Delphinium species are the major source of this category. C20-DAs are divided into 23 types: atisine, denudatine, hetisine, hetidine, napelline, vacognavine, veatchine, and anopterine, besides fifteen rearranged types. Atisine is assumed as the original group with a pentacyclic core structure and nitrogen which is located between C19 and C20. Denudatine is hexacyclic by an additional bond between C7 and C20. Hetisine is characterized by two excess bonds (N-C6 and C14-C2) in atisine. Hetidine structure is achieved by a connection between C14 and C20 in hetisines. The hexacyclic backbone of vokognavine is achieved by breaking the hetisine bond at N-C19 (forming seco). The pentacyclic backbone of veatchine is similar to kaurane diterpenes. Napelline and anopterine are similar to veatchines with the additional C7-C20 and C14-C20 bonds, respectively. There are also fifteen rearranged types in C20-DAs: spireine, cardionidine, albovionitine, arcutine, aconicarmisulfonine, kaurine A, kaurine B, delnudine, kusnesoline, grandiflodine, tricalysiamide, caesanine, anthriscifolsine, actaline, and racemulosine (Figure 3) [6,8].

2.2. Biosynthetic Pathway

The five-carbon unit, isoprenoid (IPP), which is formed through methylerythritol (MEP) or mevalonate (MVA) pathways, initiates the biosynthesis of terpenoids [19]. Four IPP units bind (catalyzed by geranylgeranyl pyrophosphate synthase) to form a 20-carbon geranylgeranyl pyrophosphate (GGPP) which further cyclizes by an enzymatic reaction to produce ent-copalyl diphosphate (ent-CPP) [20,21]. Among all the identified diterpenoid skeletons, only the two ent-kaurene and ent-atisane types can contribute to DA biosynthesis. These two precursors of DAs originate from ent-CPP by the actions of ent-kaurene/ent-atisane synthase [22]. The ent-kaurene transforms to provide the ent-kaurenoic aldehyde through two repeated oxidations by the ent-kaurene oxidase (a cytochrome P450 mono-oxygenases) [23]. On the other hand, L-serine is a plausible amino acid that could provide nitrogen for alkaloid biosynthesis [22]. Transferring nitrogen from L-serine to ent-kaurenoic aldehyde (an intermediate product) takes place through the enzymatic transamination by aminotransferases (serine-pyruvate transaminase and serine-glyoxylate transaminase are considered to have the major roles) (Scheme 2).
Expressing ent-kaurene oxidase genes in plants is the reason for the presence of these alkaloids. Since various aldehydes may be produced in the plants, the position of nitrogen in DAs might be different [6]. It has been shown that C18 and C19-DAs biochemically derive from C20-DAs and the involved biochemical reactions are thought to be Wagner–Meerwein rearrangement, Prins cyclization (ajaconine subtype into hetidine class), Schiff base cyclization (atisine class to lycoctoine class), and Mannich reaction, which supports the hypothesis that cyclases have a huge impact on DAs diversity [24,25]. Monooxygenases of cytochrome P450 could prompt oxidative rearrangements and oxygenize various carbon positions on DAs’ structure [6,26]. Thus, the created hydroxyls are the site of action for methyltransferases and acyltransferases to produce more toxic methyl, benzoyl (Bz), acetyl (Ac), anisoyl (As), Cinnamoyl (Cn), Veratroyl (Vr), and O-lipo derivatives [27].

3. Pharmacological Activities

To affect the central nervous system, primarily, the drug should pass the blood–brain barrier (BBB). Transmembrane diffusion is the most common route of drugs to pass the BBB and, in contrast to the transport system, shows a non-saturable kinetic. Physiochemical features of the drug mainly determine the amounts of this passage. Molecular weight (400–600 Da is optimum), lipid solubility, molecular charge, and tertiary structure are the most important factors necessary for transmembrane diffusion through BBB [28]. The diterpenic backbone of DAs provides their suitable lipid solubility, but the presence of tertiary nitrogen makes them different from normal diterpenoids. The tertiary nitrogen with the highest proton affinity in the molecule (in water) rearranges the electronic structure of DA by its protonation. Moreover, computational modeling showed that the function of nitrogen besides ester sidechains causes DAs to interact with the active sites as well as their toxicity [29,30].

3.1. Anticonvulsant Effects

An abnormal and transient neuronal synchronization in the brain causes epilepsy. This phenomenon alters the correct pattern of neuronal connections and is characterized by the peaks and troughs of electric discharge in the electroencephalograph (EEG) and may lead to various mental and physical symptoms associated with the origin of the disruption [31]. Voltage-gated sodium channels (VGSCs) play an essential role in the generation and coordination of action potential in the central nervous system, which participates fundamentally in the pathophysiology of seizures; thus, these channels could be an important aim for pharmaco-therapy [32]. The Fuzi total alkaloids (Aconiti Lateralis Radix Praeparata), an essential medicine in traditional Chinese medicine, significantly prolong the seizure latency time and decrease the mortality in pentylenetetrazole-induced mice [33]. Additionally, anticonvulsant effects of several alkaloidal-rich fractions and also isolated DAs from Delphinium and Aconitum species have been investigated (Table 1; Figure 4). Ameri’s findings have greatly enhanced our knowledge of the anticonvulsant effects of DAs [9]. Five distinct toxin binding sites were explored on voltage-dependent Na+ channels. Aconitine (1), batrachotoxin, and veratridine are tightly bound to site 2 in the α subunit of Na+ channels which are localized in the transmembrane region [34,35,36]. The voltage-dependent Na+ channels are activated by 1, resulting in the depolarization of the presynaptic membrane [37]. Compound 1 activates Na+ channels at resting membrane potential permanently by blocking Na+ channel inactivation and causing hyperpolarization of threshold activation due to sustained Na+ influx; thereby, inexcitability occurs [9]. The structurally related 3-acetylaconitine (2) was shown to activate voltage-dependent Na+ channels by a similar mechanism of action [38]. In contrast to compounds 1 and 2, some other Aconitum DAs exist, revealing inhibitory effects on the Na+ channels; lappaconitine (3), N-desacetyllappaconitine (4), 6-benzoylheteratisine (5), and 1-benzoylnapelline (6) reduce the peak amplitude of the current and show an inhibitory effect on the Na+ channel [9]. While 3 could antagonize the suppressive effect of 1 on population spike in rat hippocampal slices which are attributed to their opposite mode of action; both 1 and 3 possess an anticonvulsant potential [34,39,40]. Compounds 1 and 2 indirectly suppressed the uptake of [3H] noradrenaline via increasing sodium concentration in synaptosomes, while 3 and 4 do not indicate such an impact on [3H] noradrenaline uptake [9,41]. Despite Ameri’s investigations, Voss et al. showed acute seizure-like activity induced by 1 [42]. Songorine (7) might antagonize the GABA receptor in the rat brain via a different site of action different from the GABA recognition site [43]. However, in an in vivo experiment, 7 acted as an agonist on D2 and GABA A receptors, which might be translated as a potential antiepileptic effect [44]. The involvement of α-adrenoceptors and modulation of GABA receptors were reported for the antiepileptic activity of mesaconitine (8) and Fuzi total alkaloid, respectively [33,45].

3.2. Antagonizing α7 Nicotinic Acetylcholine Receptor

Nicotinic acetylcholine receptors (nAChRs) are cation-conducting ligand-gated receptors broadly expressed in the peripheral and central nervous systems and also in muscular cells. Seventeen nAChR subunits, comprising ten α, four β, γ, δ, and ε subunits have so far been identified in mammals where α7 homomeric nAChRs and heteromeric α4β2* nAChRs are predominant and expressed functionally in the brain. These receptors contribute to a range of neuronal functions from cognitive performance to neurotransmitter release (including glutamate, GABA, and dopamine) through altering intracellular Ca2+ concentration. It has been clarified that the pathogenesis of several neurological disorders containing Alzheimer’s disease, schizophrenia, Parkinson’s disease, and depression as well as nicotine addiction are related to α7 and α4β2* receptor functions. Although many efforts have been devoted to developing a (partial) agonist of α7 nAChR targeting neurologic and psychiatric conditions, none has already been approved as medicine due to their undesirable side effects [62,63,64]. Moreover, results from a meta-analysis indicated that curing the cognitive dysfunction in schizophrenia and Alzheimer’s disease with agonists of α7 nAChR cannot be considered a robust treatment [65]. In 1986, Jennings et al. found that the insecticidal effect of Delphinium plants is mainly due to the presence of methyllycaconitine (16) (Figure 5a), showing a high affinity to cholinergic receptors of insects [66]. Indeed, 16 is a potent and highly selective α7 nAChR antagonist which acts similarly to the α-bungarotoxin protein of snake venom (Figure 5b) [67]. Despite former research, sporadic studies have recently focused on the beneficial role which low doses of 16 play in neurologic disorders (Table 2); equivalent results were also reported for other nAChR antagonists, e.g., mecamylamine [68]. Several hypotheses have been proposed to explain their mechanism of action.
Based on the allosteric theory of ion channel functioning, nAChRs might have three different conformational states, resting, active, and desensitized, having the capability of altering their conformational states through spontaneous transitions. Ligand affinity highly depends on conformational states. On the other hand, α7 nAChRs possess five binding sites for interacting with agonists, where binding to two of them is necessary for activating the receptor and opening the channel. Binding an antagonist, e.g., 16, to remaining binding sites may prevent or delay the desensitization of the receptor and affect the affinity of the receptor to other ligands. As a further explanation, the stoichiometry of the binding site with which the acetylcholine molecules (AChs) interact determines the degree of channel activation. The binding of two AChs to two consecutive binding sites leads to slow channel activation, while binding to two non-consecutive sites results in rapid activation and receptor desensitization. The binding of 16 impacts the binding pattern of ACh to sites. In case one, two, or three molecules of 16 connect to the binding sites, the probability of non-consecutive binding of ACh increases, whereas binding four and five molecules of 16 eliminates the possibility of simultaneous binding of two ACh molecules, which is essential for the activation of the receptor. The hypothesis explains the contradictory responses of α7 nAChR to lower and higher doses of 16 (Figure 6) [64].

3.3. Analgesic Activity

Nociceptive pain develops in response to specific noxious stimulation of normal tissue in a normal somatosensory nervous system; in contrast, neuropathic pain comes from damage to the nerves or nervous system; there is also a third kind of pain called inflammatory pain, which may be described as increased sensitivity due to the inflammatory response associated with tissue damage [73]. Herbal preparations containing Aconitum have been widely used in traditional Chinese medicine to relieve rheumatoid arthritis [74]. The effectiveness of Aconitum-containing preparations for diabetic peripheral neuropathic pain was shown in a small clinical trial [75]. Pharmacological in vivo studies confirmed the antinociceptive activity of various DAs and extracts from Aconitum species (Table 3; Figure 7). Bulleyaconitine A (17), a diterpenoid alkaloid isolated from the rhizomes of A. bulleyanum, is structurally related to 1 and was approved in 1985 in China for the alleviation of chronic pain and is available as tablets, intramuscular injections, and soft gel capsules [76]. Compound 17 showed more significant analgesic potency than morphine. The clinical utility of 17 in the three past decades in China has demonstrated the advantages of its application for relieving chronic pain without any serious side effects, contrary to what is observed in the case of the application of morphine or non-steroidal analgesics [77]. Several studies have emphasized the positive central effect of 17 in the treatment of nociceptive pains [63]. The analgesic effect of 17 could be antagonized by reserpine but not naloxone, which suggests an involvement of catecholamine in the 17 mechanisms of action [78]. Compound 7 revealed a visceral antinociceptive effect by stimulating the release of dynorphin A, leading to the activation of presynaptic κ-opioid receptors in afferent neurons [66]. Consistently, it was indicated that 17 can directly stimulate the dynorphin A expression in spinal microglia [79]. On the other hand, Xie et al. suggested a preferable blocking effect on tetrodotoxin-sensitive Nav1.7 and Nav1.3 in dorsal root ganglion neurons as the mode of action of 17 [80]. Compound 17 could also inhibit Nav1.7 and Nav1.8 Na+ currents in a use-dependent manner [81]. Depressing of long-term potentiation at C-fiber synapses in the spinal dorsal horn and inhibition of transmitter release in paclitaxel-induced neuropathic pain were accounted for as well [82].
Another diterpenoid alkaloid, lappaconitine (3) from Aconitum species revealed antinociceptive effects in nociceptive test models. Compound 3 has been used for its analgesic properties in both traditional Chinese and Kampo medicine [83]. Its analgesic potency is comparable to that of tramadol and morphine [84,85]. Pretreatment with antagonists of adrenergic and serotoninergic systems could decrease the antinociceptive effects of 3, which might be mediated by β-adrenoceptors and 5-HT2 receptors in the brain and α-adrenoceptors and 5-HT receptors in the spinal cord [86]. Moreover, when 3 was administered via subcutaneous or intracerebroventricular routes, it could inhibit behavioral response induced by substance P or somatostatin, while the intrathecal injection showed no effect. These observations provide some evidence to support the supra-spinal descending mechanism of action of 3 and suppressing the transmission of the nociceptive neuronal message [87]. Inhibition of P2X(4) expression in microglia in the dorsal horn and P2X3 receptors in dorsal root ganglia neurons in the spinal cord were demonstrated as further plausible mechanisms of action [88,89,90]. Li et al. indicated that inhibition of the Nav1.7 channel is involved as well [91]. Similar to 17 stimulation of the expression of spinal microglial dynorphin A was suggested [92]. Ono et al. indicated that the antinociceptive effect of 3 was not antagonized by naloxone in the hot plate and acetic acid-induced writing tests, contrary to what was observed in the tail pinch test. It was suggested that the action of 3 was only partially antagonized by naloxone, which explains the naloxone-resistant antinociceptive activity for 3 at the spinal sites [93].
Mesaconitine (8), one of the most important ingredients of Aconitum species, also exhibits antinociceptive activity in animal models. Subcutaneous administration of 8 has shown much greater analgesic activity compared to that of morphine in animal models [94]. Friese et al. categorized Aconitum alkaloids into two groups based on their binding affinity to Na+ channel epitope site 2. Alkaloids of the high-affinity group (1 and 8) are supposed to act as Na+ activators due to their increasing synaptosomal [Na+]i and [Ca2+]i effect. A higher antinociceptive action was shown in the case of the high-affinity group than the other, indicating the importance of Na+ channels in the activity of 8 [95]. The analgesic action of 8 was not inhibited by levallorphan (an opiate antagonist) in an animal model. On the other hand, 8 could increase the release of endogenous norepinephrine from sympathetic nerve fibers [94]. Application cyclic AMP and isoproterenol (a β receptor agonist) in combination with 8 enhanced its analgesic activity, while propranolol (a β receptor antagonist) suppressed it, which magnifies the role of the adrenergic system in the mode of action [96]. Involvement of the nucleus reticularis gigantocellularis (NRGC), the nucleus reticularis paragigantocellularis (NRPG), the periaqueductal gray (PAG), and the lumbar enlargement in the antinociceptive action of 8 was shown in a rat model. Indeed, 8 might activate the inhibitory noradrenergic system in descending neurons from the NRPG, especially through β-receptor-mediated effects of noradrenaline [86,87]. Moreover, it was also revealed that the nucleus raphe magnus (NRM) has a key role via the serotoninergic system in the mechanism of action [97].
Isotalatizidine (18) attenuates allodynia in neuropathic pain in a mice model. An increase in the expression of dynorphin A was shown, and its release from microglia was observed, which might be triggered by the activation of the ERK1/2 pathway and phosphorylation of CREB (cAMP response element-binding) [98].
Bullatine A (19) was shown to have a synergistic effect with morphine as it increases morphine analgesic action while counteracting morphine tolerance. Stimulation of spinal microglia and subsequent enhancing of the expression of dynorphin A were thought to have a role [99,100].
Table 3. Antinociceptive effects of fractions/constituents from Aconitum and Delphinium species.
Table 3. Antinociceptive effects of fractions/constituents from Aconitum and Delphinium species.
PlantUsed Part/ConstituentsBiological ActivityRef.
Aconitum episcopaleEpiscopaline B (20)Antinociceptive effect 2-fold lower than aspirin and
acetaminophen
[101]
Aconitum pseudostapfianumPseudostapine C (21)2-fold more potent antinociceptive effect than
aspirin and acetaminophen
[102]
Aconitum episcopaleEpiscopine A (22)2-fold more potent antinociceptive effect than aspirin and acetaminophen[103]
Aconitum
carmichaelii
18Antinociceptive effect[98]
Aconitum carmichaeliPlant extract and neoline (23)Attenuated the mechanical hyperalgesia[104]
Aconitum carmichaeliPlant extract and 23Attenuated cold and mechanical hyperalgesia[105]
Aconitum carmichaeliAqueous extractsAntinociceptive activity[106]
Aconitum carmichaeliProcessed aconitum tuberHigh doses of processed Aconiti tuber inhibit the acute but potentiate the chronic antinociception of morphine[107]
Delphinium denudatumAqueous root extractAntinociceptive activity[108]
Aconitum carmichaeliProcessed aconitum tuber and 8Antinociceptive activity of 8 was more potent than morphine[109]
Aconitum sp.1Significant analgesic effects[110]
Aconitum carmichaeliiAconicatisulfonines A (24) and B (25)Analgesic activities[111]
Delphinium denudatumEthanolic extract and methanol fractionAnalgesic activity[112]
Aconitum kusnezoffii23Analgesic activity[113]
Aconitum carmichaeliiAconicarmichosides E (26), F (27), H (28), I (29), and J (30)Analgesic activity[114]
Aconitum sp.3Relieves the pain [82]
Aconitum baikalensisNapelline (31), hypaconitine (32), 7, 8, 12-epinapelline N-oxide (33).Analgesic activity comparable to that of sodium metamizole[115]
Aconitum
weixiense
Weisaconitines D (34)Analgesic activity[116]
Aconitum
carmichaeli
Guiwuline (35)Potential analgesic activity[117]
Aconitum
carmichaeli
8-O-cinnamoylneoline (36)Analgesic activity[118]

3.4. Antidementia Effect

A decrease in acetylcholine (ACh) in the central nervous system is associated with dementia. Several DAs possess an acetylcholinesterase (AChE) inhibitory effect (Table 4; Figure 8), and therefore, these compounds may theoretically act as antidementia agents. Some evidence has emphasized the role of K+ channel blockers as drug candidates for the treatment of neurodegenerative disorders [119]. Virtual screening from the Chinese natural product database resulted in the identification of four Aconitum alkaloids, namely, songorine (7), pyrochasmaconitine (37), 14-benzoyl talatisamine (12), and talatisamine (13), as candidates for this effect; an electrophysiological assay of rat dissociated hippocampal neurons verified this assumption [120]. All of the four alkaloids potently inhibited the delayed rectifier K+ current, while 13 was slightly effective on Na+ and Ca2+ channels in rat hippocampal neurons [119].
Macroautophagy was previously considered one of the plausible mechanisms of the pathogenesis of Alzheimer’s disease (AD). Compound 16 protects the human neuroblastoma SH-SY5Y cell line against amyloid-β peptide-induced cytotoxicity. Compound 16 might prevent autophagy induced by amyloid-β through the mammalian target of the rapamycin (m-tor) pathway [121]. Apetalrine B (38), the semisynthetic DAs from its parent compound, aconorine (A. apetalum), exhibits a neuroprotective effect in H2O2-treated SH-SY5Y cells by inhibiting cell apoptosis [122]. Firstly, 7 was superior to piracetam (a nootropic agent) in the correction of the cholinergic abnormalities induced by scopolamine, which indicates its antiamnestic activity. Secondly, 7 might act as a cholinomimetic, antihypoxic, and cerebro-protective agent [123,124]. Among the DAs from A. anthoroideum, rotundifosine F (39) possesses AChE inhibitory activity. Moreover, nominine (40) represented a neuroprotective effect due to its protection against 1-methyl-4-phenylpyridinium (MPP+)-induced apoptosis in SH-SY5Y cells [10]. Fuzi considerably induced growth of cell projection and cell proliferation in 0.4–0.8 mg/mL and 1.6–100 mg/mL, respectively, in the AD cell model. In addition, the involvement of GRIN1 and MAPK1 genes contributed to the antiAD mechanism of action [125].
Table 4. Antidementia effect of fractions/constituents from Aconitum and Delphinium species.
Table 4. Antidementia effect of fractions/constituents from Aconitum and Delphinium species.
PlantUsed Part/ConstituentsBiological ActivityRef.
Aconitum hemsleyanumHemsleyaline (41)Mild AChE inhibitory effect[126]
Aconitum kirinenseDiterpenoid alkaloidsModerate AChE inhibitory effect[127]
Aconitum laeveDiterpenoid alkaloidsSwatinine-C (42) and hohenackerine (43)
competitively inhibited AChE and BChE
Aconorine (44) and 3 noncompetitively inhibited AChE
[128]
Delphinium denudatumDiterpenoid alkaloidsJadwarine-A (45), 18, and dihydropentagynine (46) competitively inhibited AChE and BChE, while 1β-hydroxy,14β-acetyl condelphine (47) and jadwarine-B (48) showed non-competitive inhibition[129]
Aconitum heterophyllumDiterpenoid alkaloidsCompounds 6b-methoxy, 9b-dihydroxylheteratisine (49), 6,15b-dihydroxylhetisine (50), iso-atisine (51), heteratisine, 19-epi-isoatisine (52), and atidine (53) non-competitively inhibited AChE and BChE, while compounds 1a,11,13b-trihydroxylhetisine (54) and hetisinone (55) were determined as competitive inhibitors[130]
Delphinium denudatumIsotalatizidine (18) hydratePotent dual cholinesterase inhibitor[131]
Aconitum heterophyllumHeterophyllinine A(56) and B (57)56 and 57 inhibited AChE and BChE enzymes[132]
Aconitum falconeriFaleoconitine (58)
and Pseudaconitine (59)
Moderate inhibitory activity on AChE[133]

3.5. Antidepressant Effects

Two antagonists of nicotinic acetylcholine receptor (nAChR) were demonstrated to induce antidepressant effects in the forced swim and tail suspension tests in mice, while agonists of nAChR did exhibit no similar activity. The observed effect was more potent in the case of the non-selective nAChR antagonist mecamylamine compared to that of 16 (an α7 nAChR selective antagonist) [134]. Modulating serotonin sensitivity is considered a promising mechanism of action of DAs’ antidepressant activity in Aconitum baicalense. Besides their positive results in the tail suspension test, these compounds showed an antiexudative effect in edema induced by serotonin in mice. These alkaloids were 7, 8, 32, and 31 in order from most to least potent [135]. Antidepressant actions were also reported for chronic administration of Fuzi total alkaloids in normal and ovariectomized mice. An study designed to elucidate the mode of action discovered the potential roles of cAMP response element-binding (CREB) and brain-derived neurotrophic factor (BDNF) protein pathways [136]. Moreover, the sex-dependent antidepressant effect of 16 was consistently reported in male mice, while female mice did not exhibit the same. Local administration of 16 in male mice hippocampus could reverse the depression-like effect which was induced by physostigmine [137].

3.6. Miscellaneous Effects

Compound 7 has exhibited a more potent anxiolytic activity compared to phenazepam applying the Vogel conflict test in mice without a sedative effect. Anxiolytic activity of 7 was noted in a dose of 0.25 mg/kg, while a 10 times higher dose (2.5 mg/kg) showed an inferior activity [138].
An ethanolic extract of Delphinium denudatum has protected the brain against injury in a rat model of Parkinson’s disease. The expression of ipsilateral tyrosine hydroxylase was increased. The extract also prevented lipid peroxidation, depleted reduced glutathione in the substantia nigra, and attenuated the activity of superoxide dismutase and catalase in the striatum. Reduction in dopamine concentration was also reported in the striatum after the injection of 6-hydroxydopamine [139].

4. Clinical Trials

A double-blind, randomized, placebo-controlled, parallel-group, multi-center study was conducted on seventy-five patients with postherpetic neuralgia to evaluate the analgesic activity of bulleyaconitine A. The intervention group received tablets of bulleyaconitine A besides the first-line therapy (gabapentin), while the control group took gabapentin and placebo tablets. Treatment with bulleyaconitine A significantly improved the effective rate and the time taken to reach the primary outcomes in comparison with the control group [140]. In a clinical study, post-operative analgesia in one hundred and twenty participants was assessed for lappaconitine in four treatment groups: A: lappaconitine (12 mg) + bupivacaine (22.5 mg); B: lappaconitine (12 mg); C: bupivacaine (22 mg); D: morphine (2 mg) by epidural injection. The order of efficacy was D > A > C > B, while the onset of action was lower in the case of groups A and C [141]. Intravenous injection of lappaconitine was explored in a controlled clinical study for relief of pain after rectum carcinoma surgery. Sixty patients were divided into three groups to receive intravenous A: lappaconitine (8 mg), B: tramadol (100 mg), and C: Pethidine (50 mg) + droperidol (2.5 mg). Pain assessment was carried out using a visual analog scale (VAS) at different intervals. Lappaconitine significantly decreased the pain in patients who had undergone surgery for rectum carcinoma. Its VAS score was less than group D, while there was no significant difference between intravenous injection of 8 mg lappaconitine and 100 mg tramadol for pain relief after surgery [85]. In another study, of fifty-six patients after cholecystecystectomy, 40 mg lappaconitine showed an equal analgesic effect to 0.4 mg fentanyl, but with less nausea, vomiting, and itching [142].

5. Discussion

Although Aconitum and Delphinium species have been used for millennia in traditional medicine in Eastern societies, to date, there have been insufficient clinical studies evidencing their safety [74,83]. These plants contain DAs with characteristic bioactivities, and the use of these compounds, particularly in pure form, lends a fresh perspective to medicine. Despite the noticeable bioactivities of DAs that might be beneficial in various diseases, these compounds have played no major role in drug development in recent years. This gap is justified by the fact that there is a narrow therapeutic window between effective and toxic doses of Aconitum plants and their constituents.
Four active pharmacophores have been described previously for 1: a tertiary nitrogen atom, a hydroxyl at C13, a benzoyl ester at C14, and acetyl at C8, which contribute to the Na+ channel-activating effect. DAs with Na+ channel-blocking effects have a similar backbone structure to 1 but it was explained that lacking each of the four active pharmacophores causes them to exhibit contrasting effects. Accordingly, it was also indicated experimentally that 3 could reverse the arrhythmogenic effects of 1 [30,143,144]. A more recent in silico study of an electronic structure investigation of 19 DAs revealed that what makes the difference is not different structures but a wide distance between their efficiency, i.e., the blockers ban the channel conductance, while the activators permit only a very slow ion flow. In addition, the computational analysis demonstrated that both activators and blockers could interact with the binding site of Na+ channels via the benzoyl ester residue [30].
The presence of a tertiary N-ethyl substituted in ring A and a saturated ring D, in addition to an OAc or OC2H5 moiety at C8 and an aromatic ester (O-Bz or O-As) at C14, which is necessary for analgesic activity. A 3α or 5-hydroxyl group may support the analgesic effects but methylation of C1 hydroxyl will decrease the potency [6,145].
The lycoctonine backbone is the base for interfering cholinergic receptors which are responsible for Delphinium intoxication. A methylsuccinylanthranoyl ester at C18 (e.g., compound 16) is necessary due to providing proper orientation and improving binding affinity. The C14 functionality and the tertiary nitrogen also affect the activity [146].
The compounds responsible for the toxic effects are diester alkaloids which contain an acetyl group in C-8 besides a benzoyl or anisoyl functionality in C-14 [6,74,147]. A QSAR analysis investigated the structural toxicity relationship of diterpenoid alkaloids explaining that the number of C=O groups, lipophilicity, the atomic packing density in the molecular volume, surface polarity, and lipophilicity contribute to DA toxicity [29]. The most toxic compound in aconite is 1 with a lethal dose of 2 mg for an adult [148]. The main poisoning mechanism of action of 1 is the deactivation of the voltage-dependent sodium channels which leads to the disruption of neuromuscular release [9]. The cardiovascular system is another site where aconite derivatives can cause lethal ventricular arrhythmia [6]. Compound 16 exerts its toxicity through a different mechanism, i.e., by reversibly blocking the action of nicotinic acetylcholine receptors (nAChR), thereby paralyzing the respiratory muscles [149].
Several methods were emphasized to reduce the toxic effects of aconites in traditional Chinese medicine substantially based on heating processes (boiling, cooking, baking, steaming, and autoclaving), resulting in the hydrolysis of diester DAs to the less toxic monoesters and lipoalkaloids [148,150]. However, the medical effects do not necessarily decrease during the processing, as was demonstrated in the example of the anti-inflammatory activities of native DAs and their lipoalkaloid derivatives [151]. Additionally, the concomitant use of plants such as ginseng, licorice, dried ginger, and dahuang with aconite is believed to reduce its toxic effects and enhance its efficacy in traditional Chinese medicine [148].

6. Conclusions

As a conclusion to the above literature review, DAs originate from ent-atisane and ent-kaurene diterpenes through an enzymatic amination reaction. To date, 46 types of DA have been identified from three main categories: C18, C19, and C20-diterpenoid alkaloids. One of their most obvious effects is the anticonvulsant activity caused by interfering with the voltage-gated Na+ channels in the central nervous system through desensitization after persistent activation (e.g., 1 and 2) or deactivation (e.g., 36) of Na+ channels. The most effective chemical moieties involved in the binding of DAs to Na+ channels are four pharmacophores: the tertiary nitrogen atom, hydroxyl at C13, benzoyl ester at C14, and acetyl at C8. Some of these alkaloids showed promising analgesic effects. For example, 3, 8, 17, and 18 exert potent activity by increasing the release of dynorphin A, activating the inhibitory noradrenergic neurons in the β-adrenergic system, or preventing the transmission of pain messages by inactivating Na+ channels. On the other hand, 16 has mixed effects. It is marked as a potent antagonist of α7 nACh receptors while reversely activating it in very low concentrations. The tertiary N-ethyl substituted in ring A, a saturated ring D, an OAc or OC2H5 moiety at C8, and an aromatic ester (OBz or OAs) at C14 are likely to be necessary for analgesic activity. Although the role of tertiary nitrogen in ring A and polycyclic structure is of great importance in drug-receptor affinity, in silico studies have emphasized the role of sidechains in C13, C14, C8, etc. Moreover, some of the DAs have potent acetylcholinesterase inhibitory activity, which indicates their potential against neurodegenerative disorders such as Alzheimer’s disease. Antidepressant and anxiolytic effects have also been investigated in certain surveys; nevertheless, more experiments should be conducted to provide clearer evidence. Altogether, due to the undesirable profile of toxicity, no DA has been considered as medicine in the treatment of central nervous system disorders to date. A less favorable benefit-to-risk ratio might be justified in certain situations, such as in the treatment of epilepsy or dementia. Nevertheless, the necessity for more clinical trials is felt to clarify the effectiveness and eliminate the safety concerns, resulting in the development of new drugs based on DAs with lower toxicity and higher functional activities.

7. Future Perspectives

Some of the DAs showed extreme affinity for α7 nAChR, and are potential candidates for developing new research drugs in the treatment of cognitive disorders. Several DAs have prospecting potential in the treatment of Alzheimer’s disease. It is due to inhibiting the AChE and BChE enzymes in similar potency as standard drugs. They are also considered in developing the anticonvulsant drugs by blocking voltage-gated Na+ channels, or as analgesic in alleviation of chronic pain.

Author Contributions

A.S.: Methodology, Investigation, Writing—Original draft. M.G.: Supervision, Conceptualization. B.Z.: Supervision. A.R.J.: Visualization, Conceptualization. P.R.: Visualization, Conceptualization. M.F.: Writing—Reviewing and Editing. D.C., I.A.K. and Z.A.: Reviewing and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

The Ethics Committee of Isfahan University of Medical Sciences approved this research with the code IR.MUI.RESEARCH.REC.1399.796.

Data Availability Statement

The data presented in this study are available in the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kiss, T.; Cank, K.; Orbán-Gyapai, O.; Liktor-Busa, E.; Rutkovska, S.; Pučka, I.; Hohmann, J.; Csupor, D. Screening for diterpene alkaloids in the Spiraea genus. Planta Med. 2015, 81, PM_19. [Google Scholar] [CrossRef]
  2. Wang, F.P.; Chen, Q.H.; Liang, X.T. The C18-diterpenoid alkaloids. Alkaloids Chem. Biol. 2009, 67, 1–78. [Google Scholar]
  3. Wang, F.-P.; Chen, Q.-H.; Liu, X.-Y. Diterpenoid alkaloids. Nat. Prod. Rep. 2010, 27, 529–570. [Google Scholar] [CrossRef]
  4. Wang, F.-P.; Liang, X.-T. C20-diterpenoid alkaloids. Alkaloids Chem. Biol. 2002, 59, 1–280. [Google Scholar]
  5. Wang, F.-P.; Chen, Q.-H. The C19-diterpenoid alkaloids. Alkaloids Chem. Biol. 2010, 69, 1–577. [Google Scholar]
  6. Shen, Y.; Liang, W.J.; Shi, Y.N.; Kennelly, E.J.; Zhao, D.K. Structural diversity, bioactivities, and biosynthesis of natural diterpenoid alkaloids†. Nat. Prod. Rep. 2020, 37, 763–796. [Google Scholar] [CrossRef]
  7. Dewick, P.M. Medicinal Natural Products: A Biosynthetic Approach; John Wiley & Sons: West Sussex, UK, 2002; pp. 385–388. [Google Scholar]
  8. Thawabteh, A.M.; Thawabteh, A.; Lelario, F.; Bufo, S.A.; Scrano, L. Classification, Toxicity and Bioactivity of Natural Diterpenoid Alkaloids. Molecules 2021, 26, 4103. [Google Scholar] [CrossRef]
  9. Ameri, A. The effects of Aconitum alkaloids on the central nervous system. Prog. Neurobiol. 1998, 56, 211–235. [Google Scholar] [CrossRef]
  10. Huang, S.; Zhang, J.-F.; Chen, L.; Gao, F.; Zhou, X.-L. Diterpenoid alkaloids from Aconitum anthoroideum that offer protection against MPP+–Induced apoptosis of SH–SY5Y cells and acetylcholinesterase inhibitory activity. Phytochemistry 2020, 178, 112459. [Google Scholar] [CrossRef]
  11. Bisset, N.G. Arrow poisons in China. part ii. Aconitum—Botany, chemistry, and pharmacology. J. Ethnopharmacol. 1981, 4, 247–336. [Google Scholar] [CrossRef]
  12. Wang, W.C. A revision of the genus Delphinium (Ranunculaceae) of China (I). Guihaia 2019, 39, 1425–1469. [Google Scholar]
  13. Yin, T.; Cai, L.; Ding, Z. A systematic review on the chemical constituents of the genus Consolida (Ranunculaceae) and their biological activities. RSC Adv. 2020, 10, 35072–35089. [Google Scholar] [CrossRef]
  14. Bitiş, L.; Süzgeç, S.; Mericli, F.; Özçelik, H.; Zapp, J.; Becker, H.; Meriçli, A.H. Alkaloids from Consolida olopetala. Pharm. Biol. 2006, 44, 244–246. [Google Scholar] [CrossRef]
  15. Kiss, T.; Cank, K.B.; Orban-Gyapai, O.; Liktor-Busa, E.; Zomborszki, Z.P.; Rutkovska, S.; Pučka, I.; Németh, A.; Csupor, D. Phytochemical and pharmacological investigation of Spiraea chamaedryfolia: A contribution to the chemotaxonomy of Spiraea genus. BMC Res. Notes 2017, 10, 762. [Google Scholar] [CrossRef]
  16. Chung, J.Y.; Lee, S.J.; Lee, H.J.; Bin Bong, J.; Lee, C.-H.; Shin, B.-S.; Kang, H.G. Aconitine Neurotoxicity According to Administration Methods. J. Clin. Med. 2021, 10, 2149. [Google Scholar] [CrossRef]
  17. Liang, X.; Gao, Y.; Luan, S. Two decades of advances in diterpenoid alkaloids with cytotoxicity activities. RSC Adv. 2018, 8, 23937–23946. [Google Scholar] [CrossRef]
  18. Yin, T.; Cai, L.; Ding, Z. An overview of the chemical constituents from the genus Delphinium reported in the last four decades. RSC Adv. 2020, 10, 13669–13686. [Google Scholar] [CrossRef]
  19. Pal, T.; Malhotra, N.; Chanumolu, S.K.; Chauhan, R.S. Next-generation sequencing (NGS) transcriptomes reveal association of multiple genes and pathways contributing to secondary metabolites accumulation in tuberous roots of Aconitum heterophyllum Wall. Planta 2015, 242, 239–258. [Google Scholar] [CrossRef]
  20. Vranová, E.; Coman, D.; Gruissem, W. Structure and Dynamics of the Isoprenoid Pathway Network. Mol. Plant 2012, 5, 318–333. [Google Scholar] [CrossRef]
  21. Devkota, K.P.; Sewald, N. Terpenoid alkaloids derived by amination reaction. In Natural Products: Phytochemistry, Botany and Metabolism of Alkaloids, Phenolics and Terpenes; Ramawat, K.G., Mérillon, J.-M., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 923–951. [Google Scholar] [CrossRef]
  22. Zhao, P.-J.; Gao, S.; Fan, L.-M.; Nie, J.-L.; He, H.-P.; Zeng, Y.; Shen, Y.-M.; Hao, X.-J. Approach to the Biosynthesis of Atisine-Type Diterpenoid Alkaloids. J. Nat. Prod. 2009, 72, 645–649. [Google Scholar] [CrossRef]
  23. Morrone, D.; Chen, X.; Coates, R.M.; Peters, R.J. Characterization of the kaurene oxidase CYP701A3, a multifunctional cytochrome P450 from gibberellin biosynthesis. Biochem. J. 2010, 431, 337–347. [Google Scholar] [CrossRef]
  24. Li, X.; Zhu, M.; Wang, Z.; Liu, X.; Song, H.; Zhang, D.; Wang, F.; Qin, Y. Synthesis of Atisine, Ajaconine, Denudatine, and Hetidine Diterpenoid Alkaloids by a Bioinspired Approach. Angew. Chem. 2016, 128, 15896–15900. [Google Scholar] [CrossRef]
  25. Zhao, D.; Shen, Y.; Shi, Y.; Shi, X.; Qiao, Q.; Zi, S.; Zhao, E.; Yu, D.; Kennelly, E.J. Probing the transcriptome of Aconitum carmichaelii reveals the candidate genes associated with the biosynthesis of the toxic aconitine-type C19-diterpenoid alkaloids. Phytochemistry 2018, 152, 113–124. [Google Scholar] [CrossRef]
  26. Mizutani, M.; Sato, F. Unusual P450 reactions in plant secondary metabolism. Arch. Biochem. Biophys. 2011, 507, 194–203. [Google Scholar] [CrossRef]
  27. Zhou, G.; Tang, L.; Zhou, X.; Wang, T.; Kou, Z.; Wang, Z. A review on phytochemistry and pharmacological activities of the processed lateral root of Aconitum carmichaelii Debeaux. J. Ethnopharmacol. 2015, 160, 173–193. [Google Scholar] [CrossRef]
  28. Banks, W.A. Characteristics of compounds that cross the blood-brain barrier. BMC Neurol. 2009, 9, S3. [Google Scholar] [CrossRef]
  29. Turabekova, M.A.; Rasulev, B.F. QSAR Analysis of the Structure—Toxicity Relationship of Aconitum and Delphinium Diterpene Alkaloids. Chem. Nat. Compd. 2005, 41, 213–219. [Google Scholar] [CrossRef]
  30. Turabekova, M.A.; Rasulev, B.F.; Levkovich, M.G.; Abdullaev, N.D.; Leszczynski, J. Aconitum and Delphinium sp. alkaloids as antagonist modulators of voltage-gated Na+ channels: AM1/DFT electronic structure investigations and QSAR studies. Comput. Biol. Chem. 2008, 32, 88–101. [Google Scholar] [CrossRef]
  31. Moshé, S.L.; Perucca, E.; Ryvlin, P.; Tomson, T. Epilepsy: New advances. Lancet 2015, 385, 884–898. [Google Scholar] [CrossRef]
  32. Kaplan, D.I.; Isom, L.L.; Petrou, S. Role of Sodium Channels in Epilepsy. Cold Spring Harb. Perspect. Med. 2016, 6, a022814. [Google Scholar] [CrossRef]
  33. Li, B.; Tang, F.; Wang, L.; Liu, L.; Zhao, J.; Zhou, Y.; Wang, Y.; Song, Y.; Li, Y.; Cui, R. Anticonvulsant Effects of Fuzi Total Alkaloid on Pentylenetetrazole-Induced Seizure in Mice. J. Pharmacol. Sci. 2013, 123, 195–198. [Google Scholar] [CrossRef]
  34. Catterall, W.A. Structure and Function of Voltage-Sensitive Ion Channels. Science 1988, 242, 50–61. [Google Scholar] [CrossRef]
  35. Catterall, W.A. Cellular and molecular biology of voltage-gated sodium channels. Physiol. Rev. 1992, 72, S15–S48. [Google Scholar] [CrossRef]
  36. Adams, M.E.; Olivera, B.M. Neurotoxins: Overview of an emerging research technology. Trends Neurosci. 1994, 17, 151–155. [Google Scholar] [CrossRef]
  37. Yamanaka, H.; Doi, A.; Ishibashi, H.; Akaike, N. Aconitine facilitates spontaneous transmitter release at rat ventromedial hypothalamic neurons. Br. J. Pharmacol. 2002, 135, 816–822. [Google Scholar] [CrossRef]
  38. Ameri, A. Inhibition of rat hippocampal excitability by the plant alkaloid 3-acetylaconitine mediated by interaction with voltage-dependent sodium channels. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1997, 355, 273–280. [Google Scholar] [CrossRef]
  39. Ameri, A.; Simmet, T. Antagonism of the aconitine-induced inexcitability by the structurally related Aconitum alkaloids, lappaconitine and ajacine. Brain Res. 1999, 842, 332–341. [Google Scholar] [CrossRef]
  40. Ameri, A.; Shi, Q.; Aschoff, J.; Peters, T. Electrophysiological effects of aconitine in rat hippocampal slices. Neuropharmacology 1996, 35, 13–22. [Google Scholar] [CrossRef]
  41. Seitz, U.; Ameri, A. Different Effects on [3H]Noradrenaline Uptake of the Aconitum Alkaloids Aconitine, 3-Acetylaconitine, Lappaconitine, and N-Desacetyllappaconitine in Rat Hippocampus. Biochem. Pharmacol. 1998, 55, 883–888. [Google Scholar] [CrossRef]
  42. Voss, L.J.; Voss, J.M.; McLeay, L.; Sleigh, J.W. Aconitine induces prolonged seizure-like events in rat neocortical brain slices. Eur. J. Pharmacol. 2008, 584, 291–296. [Google Scholar] [CrossRef]
  43. Zhao, X.-Y.; Wang, Y.; Li, Y.; Chen, X.-Q.; Yang, H.-H.; Yue, J.-M.; Hu, G.-Y. Songorine, a diterpenoid alkaloid of the genus Aconitum, is a novel GABAA receptor antagonist in rat brain. Neurosci. Lett. 2003, 337, 33–36. [Google Scholar] [CrossRef]
  44. Koszegi, Z.; Atlasz, T.; Csupor, D.; Hohmann, J.; Hernadi, I. Aconitum alkaloid songorine acts as a potent GABAA receptor agonist in the rat brain in vivo. Acta Neurobiol. Exp. Wars. 2007, 94, 367–368. [Google Scholar]
  45. Ameri, A. Effects of the Aconitum alkaloid mesaconitine in rat hippocampal slices and the involvement of α-and β-adrenoceptors. Br. J. Pharmacol. 1998, 123, 243–250. [Google Scholar] [CrossRef]
  46. Raza, M.L.; Zeeshan, M.; Ahmad, M.; Shaheen, F.; Simjee, S.U. Anticonvulsant activity of DNS II fraction in the acute seizure models. J. Ethnopharmacol. 2010, 128, 600–605. [Google Scholar] [CrossRef]
  47. Raza, M.; Zeeshan, M.; Ahmad, M.; Shaheen, F.; Simjee, S. Anticonvulsant activity of Aconitum violaceum against maximal electroshock induced seizure model. Behv Pharm. 2008, 19, 658–659. [Google Scholar]
  48. Raza, M.; Shaheen, F.; Choudhary, M.I.; Sombati, S.; Rahman, A.-U.; DeLorenzo, R.J. Inhibition of sustained repetitive firing in cultured hippocampal neurons by an aqueous fraction isolated from Delphinium denudatum. J. Ethnopharmacol. 2004, 90, 367–374. [Google Scholar] [CrossRef]
  49. Raza, M.; Shaheen, F.; Choudhary, M.I.; Rahman, A.-U.; Sombati, S.; DeLorenzo, R.J. In vitro inhibition of pentylenetetrazole and bicuculline-induced epileptiform activity in rat hippocampal pyramidal neurons by aqueous fraction isolated from Delphinium denudatum. Neurosci. Lett. 2002, 333, 103–106. [Google Scholar] [CrossRef]
  50. Raza, M.; Shaheen, F.; Choudhary, M.I.; Sombati, S.; Rafiq, A.; Suria, A.; Rahman, A.-U.; DeLorenzo, R.J. Anticonvulsant activities of ethanolic extract and aqueous fraction isolated from Delphinium denudatum. J. Ethnopharmacol. 2001, 78, 73–78. [Google Scholar] [CrossRef]
  51. Ameri, A.; Zimmermann, T.; Simmet, T. Frequency- and structure-dependent inhibition of normal and epileptiform activity by 6-benzoyldeltamine in rat hippocampal slices. Eur. J. Pharmacol. 1999, 369, 279–288. [Google Scholar] [CrossRef]
  52. Ameri, A.; Simmet, T. Interaction of the structurally related Aconitum alkaloids, aconitine and 6-benzyolheteratisine, in the rat hippocampus. Eur. J. Pharmacol. 1999, 386, 187–194. [Google Scholar] [CrossRef]
  53. Ameri, A. Effects of the Aconitum alkaloid songorine on synaptic transmission and paired-pulse facilitation of CA1 pyramidal cells in rat hippocampal slices. Br. J. Pharmacol. 1998, 125, 461–468. [Google Scholar] [CrossRef]
  54. Ameri, A. Structure-dependent inhibitory action of the Aconitum alkaloids 14-benzoyltalitasamine and talitasamine in rat hippocampal slices. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1998, 357, 585–592. [Google Scholar] [CrossRef]
  55. Ameri, A. Inhibition of stimulus-triggered and spontaneous epileptiform activity in rat hippocampal slices by the Aconitum alkaloid mesaconitine. Eur. J. Pharmacol. 1998, 342, 183–191. [Google Scholar] [CrossRef]
  56. Ameri, A.; Gleitz, J.; Peters, T. Inhibition of neuronal activity in rat hippocampal slices by Aconitum alkaloids. Brain Res. 1996, 738, 154–157. [Google Scholar] [CrossRef]
  57. Ameri, A. Structure-dependent differences in the effects of the Aconitum alkaloids lappaconitine, N-desacetyllappaconitine and lappaconidine in rat hippocampal slices. Brain Res. 1997, 769, 36–43. [Google Scholar] [CrossRef]
  58. Ameri, A. Inhibition of rat hippocampal excitability by the Aconitum alkaloid, 1-benzoylnapelline, but not by napelline. Eur. J. Pharmacol. 1997, 335, 145–152. [Google Scholar] [CrossRef]
  59. Ameri, A. Electrophysiological actions of the plant alkaloid 6-benzoylheteratisine in rat hippocampal slices. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1997, 355, 538–544. [Google Scholar] [CrossRef]
  60. Ameri, A. Effects of the alkaloids 6-benzoylheteratisine and heteratisine on neuronal activity in rat hippocampal slices. Neuropharmacology 1997, 36, 1039–1046. [Google Scholar] [CrossRef]
  61. Ameri, A.; Gleitz, J.; Peters, T. Aconitine inhibits epileptiform activity in rat hippocampal slices. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1996, 354, 80–85. [Google Scholar] [CrossRef]
  62. Ho, T.N.T.; Abraham, N.; Lewis, R.J. Structure-Function of Neuronal Nicotinic Acetylcholine Receptor Inhibitors Derived from Natural Toxins. Front. Neurosci. 2020, 14, 609005. [Google Scholar] [CrossRef]
  63. Dineley, K.T.; Pandya, A.A.; Yakel, J.L. Nicotinic ACh receptors as therapeutic targets in CNS disorders. Trends Pharmacol. Sci. 2015, 36, 96–108. [Google Scholar] [CrossRef] [PubMed]
  64. Van Goethem, N.P.; Paes, D.; Puzzo, D.; Fedele, E.; Rebosio, C.; Gulisano, W.; Palmeri, A.; Wennogle, L.P.; Peng, Y.; Bertrand, D.; et al. Antagonizing α7 nicotinic receptors with methyllycaconitine (MLA) potentiates receptor activity and memory acquisition. Cell. Signal. 2019, 62, 109338. [Google Scholar] [CrossRef] [PubMed]
  65. Lewis, A.S.; van Schalkwyk, G.I.; Bloch, M.H. Alpha-7 nicotinic agonists for cognitive deficits in neuropsychiatric disorders: A translational meta-analysis of rodent and human studies. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2017, 75, 45–53. [Google Scholar] [CrossRef] [PubMed]
  66. Jennings, K.R.; Brown, D.G.; Wright, D.P. Methyllycaconitine, a naturally occurring insecticide with a high affinity for the insect cholinergic receptor. Experientia 1986, 42, 611–613. [Google Scholar] [CrossRef]
  67. Daly, J.W. Nicotinic Agonists, Antagonists, and Modulators from Natural Sources. Cell. Mol. Neurobiol. 2005, 25, 513–552. [Google Scholar] [CrossRef]
  68. Levin, E.D.; Caldwell, D.P. Low-dose mecamylamine improves learning of rats in the radial-arm maze repeated acquisition procedure. Neurobiol. Learn. Mem. 2006, 86, 117–122. [Google Scholar] [CrossRef]
  69. Green, B.T.; Welch, K.D.; Cook, D.; Gardner, D.R. Potentiation of the actions of acetylcholine, epibatidine, and nicotine by methyllycaconitine at fetal muscle-type nicotinic acetylcholine receptors. Eur. J. Pharmacol. 2011, 662, 15–21. [Google Scholar] [CrossRef]
  70. Hahn, B.; Shoaib, M.; Stolerman, I.P. Selective nicotinic receptor antagonists: Effects on attention and nicotine-induced attentional enhancement. Psychopharmacology 2011, 217, 75–82. [Google Scholar] [CrossRef]
  71. Burke, D.A.; Heshmati, P.; Kholdebarin, E.; Levin, E.D. Decreasing nicotinic receptor activity and the spatial learning impairment caused by the NMDA glutamate antagonist dizocilpine in rats. Eur. J. Pharmacol. 2014, 741, 132–139. [Google Scholar] [CrossRef]
  72. Levin, E.D.; Cauley, M.; Rezvani, A.H. Improvement of attentional function with antagonism of nicotinic receptors in female rats. Eur. J. Pharmacol. 2013, 702, 269–274. [Google Scholar] [CrossRef]
  73. Xie, M.-X.; Zhu, H.-Q.; Pang, R.-P.; Wen, B.-T.; Liu, X.-G. Mechanisms for therapeutic effect of bulleyaconitine A on chronic pain. Mol. Pain 2018, 14, 1744806918797243. [Google Scholar] [CrossRef] [PubMed]
  74. Singhuber, J.; Zhu, M.; Prinz, S.; Kopp, B. Aconitum in Traditional Chinese Medicine-A valuable drug or an unpredictable risk? J. Ethnopharmacol. 2009, 126, 18–30. [Google Scholar] [CrossRef] [PubMed]
  75. Feng, L.; Liu, W.-K.; Deng, L.; Tian, J.-X.; Tong, X.-L. Clinical Efficacy of Aconitum-Containing Traditional Chinese Medicine for Diabetic Peripheral Neuropathic Pain. Am. J. Chin. Med. 2014, 42, 109–117. [Google Scholar] [CrossRef]
  76. Huang, S.-N.; Wei, J.; Huang, L.-T.; Ju, P.-J.; Chen, J.; Wang, Y.-X. Bulleyaconitine A Inhibits Visceral Nociception and Spinal Synaptic Plasticity through Stimulation of Microglial Release of Dynorphin A. Neural Plast. 2020, 2020, 1484087. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, Y.Q.; Ding, X.N.; Wang, Y.D. The clinical studies of BLA tablets to treat common chronic pain. Chin. J. Pain. Med. 2011, 17, 314–315. [Google Scholar]
  78. Tang, X.C.; Liu, X.J.; Lu, W.H.; Wang, M.D.; Li, A.L. Studies on the analgesic action and physical dependence of bulleyaconitine A. Yao Xue Xue Bao Acta Pharm. Sin. 1986, 21, 886–891. [Google Scholar]
  79. Li, T.-F.; Fan, H.; Wang, Y.-X. Aconitum-Derived Bulleyaconitine A Exhibits Antihypersensitivity through Direct Stimulating Dynorphin A Expression in Spinal Microglia. J. Pain 2016, 17, 530–548. [Google Scholar] [CrossRef]
  80. Xie, M.-X.; Yang, J.; Pang, R.-P.; Zeng, W.-A.; Ouyang, H.-D.; Liu, Y.Q.; Liu, X.-G. Bulleyaconitine A attenuates hyperexcitability of dorsal root ganglion neurons induced by spared nerve injury: The role of preferably blocking Nav1.7 and Nav1.3 channels. Mol. Pain 2018, 14, 1744806918778491. [Google Scholar] [CrossRef]
  81. Wang, C.-F.; Gerner, P.; Schmidt, B.; Xu, Z.Z.; Nau, C.; Wang, S.-Y.; Ji, R.-R.; Wang, G.K. Use of Bulleyaconitine A as an Adjuvant for Prolonged Cutaneous Analgesia in the Rat. Anesth. Analg. 2008, 107, 1397–1405. [Google Scholar] [CrossRef]
  82. Zhu, H.-Q.; Xu, J.; Shen, K.-F.; Pang, R.-P.; Wei, X.-H.; Liu, X.-G. Bulleyaconitine A depresses neuropathic pain and potentiation at C-fiber synapses in spinal dorsal horn induced by paclitaxel in rats. Exp. Neurol. 2015, 273, 263–272. [Google Scholar] [CrossRef]
  83. Nyirimigabo, E.; Xu, Y.; Li, Y.; Wang, Y.; Agyemang, K.; Zhang, Y. A review on phytochemistry, pharmacology and toxicology studies of Aconitum. J. Pharm. Pharmacol. 2015, 67, 1–19. [Google Scholar] [CrossRef] [PubMed]
  84. Ono, M.; Satoh, T. Pharmacological studies of lappaconitine. Analgesic activities. Arzneimittelforschung 1988, 38, 892–895. [Google Scholar]
  85. Gong, Q.-A.; Li, M. Effect of Lappaconitine on Postoperative Pain and Serum Complement 3 and 4 Levels of Cancer Patients Undergoing Rectum Surgery. Zhongguo Zhong Xi Yi Jie He Za Zhi Zhongguo Zhongxiyi Jiehe Zazhi Chin. J. Integr. Tradit. West. Med. 2015, 35, 668–672. [Google Scholar]
  86. Ono, M.; Satoh, T. Pharmacological Studies on Lappaconitine: Possible Interaction with Endogenous Noradrenergic and Serotonergic Pathways to Induce Antinociception. Jpn. J. Pharmacol. 1992, 58, 251–257. [Google Scholar] [CrossRef] [PubMed]
  87. Ono, M.; Satoh, T. Pharmacological studies on lappaconitine: Antinociception and Inhibition of the Spinal Action of Substance P and Somatostatin. Jpn. J. Pharmacol. 1991, 55, 523–530. [Google Scholar] [CrossRef]
  88. Zhao, D.-K.; Shi, X.-Q.; Zhang, L.-M.; Yang, D.-Q.; Guo, H.-C.; Chen, Y.-P.; Shen, Y. Four new diterpenoid alkaloids with antitumor effect from Aconitum nagarum var. heterotrichum. Chin. Chem. Lett. 2017, 28, 358–361. [Google Scholar] [CrossRef]
  89. Yang, C.L.; Wei, Z.R.; Zhang, T.H.; Zeng, X.Q.; Wu, B.H. Effects of lappaconitine on pain and inflammatory response of severely burned rats and the mechanism. Zhonghua Shao Shang Za Zhi Zhonghua Shaoshang Zazhi Chin. J. Burn. 2017, 33, 374–380. [Google Scholar] [CrossRef]
  90. Ou, S.; Zhao, Y.-D.; Xiao, Z.; Wen, H.-Z.; Cui, J.; Ruan, H.-Z. Effect of lappaconitine on neuropathic pain mediated by P2X3 receptor in rat dorsal root ganglion. Neurochem. Int. 2011, 58, 564–573. [Google Scholar] [CrossRef]
  91. Li, Y.F.; Zheng, Y.M.; Yu, Y.; Gan, Y.; Gao, Z.B. Inhibitory effects of lappaconitine on the neuronal isoforms of voltage-gated sodium channels. Acta Pharmacol. Sin. 2019, 40, 451–459. [Google Scholar] [CrossRef]
  92. Sun, M.-L.; Ao, J.-P.; Wang, Y.-R.; Huang, Q.; Li, T.-F.; Li, X.-Y.; Wang, Y.-X. Lappaconitine, a C18-diterpenoid alkaloid, exhibits antihypersensitivity in chronic pain through stimulation of spinal dynorphin A expression. Psychopharmacology 2018, 235, 2559–2571. [Google Scholar] [CrossRef]
  93. Ono, M.; Satoh, T. Pharmacological studies of lappaconitine. Analgesia produced by intracerebroventricular, intracisternal and intrathecal injections. J. Pharm. Dyn. 1990, 13, 374–377. [Google Scholar] [CrossRef] [PubMed]
  94. Murayama, M.; Ito, T.; Konno, C.; Hikino, H. Mechanism of analgesic action of mesaconitine. I. Relationship between analgesic effect and central monoamines or opiate receptors. Eur. J. Pharmacol. 1984, 101, 29–36. [Google Scholar] [CrossRef] [PubMed]
  95. Friese, J.; Gleitz, J.; Gutser, U.T.; Heubach, J.F.; Matthiesen, T.; Wilffert, B.; Selve, N. Aconitum sp. alkaloids: The modulation of voltage-dependent Na+ channels, toxicity and antinociceptive properties. Eur. J. Pharmacol. 1997, 337, 165–174. [Google Scholar] [CrossRef]
  96. Hikino, H.; Murayama, M. Mechanism of the antinociceptive action of mesaconitine: Participation of brain stem and lumbar enlargement. Br. J. Pharmacol. 1985, 85, 575–580. [Google Scholar] [CrossRef]
  97. Suzuki, Y.; Oyama, T.; Ishige, A.; Isono, T.; Asami, A.; Ikeda, Y.; Noguchi, M.; Omiya, Y. Antinociceptive Mechanism of the Actonitine Alkaloids Mesaconitine and Benzoylmesaconine. Planta Med. 1994, 60, 391–394. [Google Scholar] [CrossRef] [PubMed]
  98. Shao, S.; Xia, H.; Hu, M.; Chen, C.; Fu, J.; Shi, G.; Guo, Q.; Zhou, Y.; Wang, W.; Shi, J.; et al. Isotalatizidine, a C19-diterpenoid alkaloid, attenuates chronic neuropathic pain through stimulating ERK/CREB signaling pathway-mediated microglial dynorphin A expression. J. Neuroinflammation 2020, 17, 13. [Google Scholar] [CrossRef]
  99. Huang, Q.; Mao, X.-F.; Wu, H.-Y.; Li, T.-F.; Sun, M.-L.; Liu, H.; Wang, Y.-X. Bullatine A stimulates spinal microglial dynorphin A expression to produce anti-hypersensitivity in a variety of rat pain models. J. Neuroinflammation 2016, 13, 214. [Google Scholar] [CrossRef]
  100. Huang, Q.; Sun, M.-L.; Chen, Y.; Li, X.-Y.; Wang, Y.-X. Concurrent bullatine A enhances morphine antinociception and inhibits morphine antinociceptive tolerance by indirect activation of spinal κ-opioid receptors. J. Ethnopharmacol. 2017, 196, 151–159. [Google Scholar] [CrossRef]
  101. Hu, J.; Li, J.X.; Li, Q.; Mao, X.; Peng, T.F.; Liu, H.Q.; Yin, S.; Yuan, H.J. Antinociceptive C19–diterpenoid alkaloids from the root of Aconitum episcopale. J. Asian Nat. Prod. Res. 2022, 24, 617–623. [Google Scholar] [CrossRef]
  102. Hu, J.; Li, J.X.; Li, Q.; Mao, X.; Peng, T.F.; Jin, N.H.; Yin, S.; Tang, Y. Antinociceptive C19–diterpenoid alkaloids isolated from Aconitum pseudostapfianum. J. Asian Nat. Prod. Res. 2021, 23, 637–643. [Google Scholar] [CrossRef]
  103. Hu, J.; Li, J.; Li, Q.; Mao, X.; Peng, T.; Jin, N.; Yin, S.; Shi, X.; Li, Y. Antinociceptive C19-Diterpenoid Alkaloids from Aconitum episcopale. Chem. Nat. Compd. 2021, 57, 503–506. [Google Scholar] [CrossRef]
  104. Tanimura, Y.; Yoshida, M.; Ishiuchi, K.; Ohsawa, M.; Makino, T. Neoline is the active ingredient of processed aconite root against murine peripheral neuropathic pain model, and its pharmacokinetics in rats. J. Ethnopharmacol. 2019, 241, 111859. [Google Scholar] [CrossRef]
  105. Suzuki, T.; Miyamoto, K.; Yokoyama, N.; Sugi, M.; Kagioka, A.; Kitao, Y.; Adachi, T.; Ohsawa, M.; Mizukami, H.; Makino, T. Processed aconite root and its active ingredient neoline may alleviate oxaliplatin-induced peripheral neuropathic pain. J. Ethnopharmacol. 2016, 186, 44–52. [Google Scholar] [CrossRef] [PubMed]
  106. Lai, M.C.; Liu, I.-M.; Liou, S.-S.; Chang, Y.-S. Mesaconitine plays the major role in the antinociceptive and anti-inflammatory activities of Radix Aconiti Carmichaeli (Chuan Wu). J. Food Drug Anal. 2011, 19, 362–368. [Google Scholar] [CrossRef]
  107. Shu, H.; Hayashida, M.; Arita, H.; Huang, W.; Xiao, L.; Chiba, S.; Sekiyama, H.; Hanaoka, K. High doses of processed Aconiti tuber inhibit the acute but potentiate the chronic antinociception of morphine. J. Ethnopharmacol. 2008, 119, 276–283. [Google Scholar] [CrossRef]
  108. Zafar, S.; Ahmad, M.A.; Siddiqui, T.A. Acute Toxicity and Antinociceptive Properties of Delphinium denudatum. Pharm. Biol. 2003, 41, 542–545. [Google Scholar] [CrossRef]
  109. Oyama, T.; Isono, T.; Suzuki, Y.; Hayakawa, Y. Anti-nociceptive Effects of Aconiti Tuber and its Alkaloids. Am. J. Chin. Med. 1994, 22, 175–182. [Google Scholar] [CrossRef] [PubMed]
  110. Deng, J.; Han, J.; Chen, J.; Zhang, Y.; Huang, Q.; Wang, Y.; Qi, X.; Liu, Z.; Leung, E.L.-H.; Wang, D. Comparison of analgesic activities of aconitine in different mice pain models. PLoS ONE 2021, 16, e0249276. [Google Scholar] [CrossRef]
  111. Wu, Y.; Shao, S.; Guo, Q.; Xu, C.; Xia, H.; Zhang, T.; Shi, J. Aconicatisulfonines A and B, Analgesic Zwitterionic C20-Diterpenoid Alkaloids with a Rearranged Atisane Skeleton from Aconitum carmichaelii. Org. Lett. 2019, 21, 6850–6854. [Google Scholar] [CrossRef]
  112. Zaheer, I.; Rahman, S.Z.; Khan, R.A.; Parveen, M.; Ahmad, M. Evaluation of analgesic activity of extracts of Delphinium denudatum in animal models: A dose dependent pre-clinical trial. J. Clin. Diagn. Res. 2018, 12, FC01–FC04. [Google Scholar] [CrossRef]
  113. Li, Q.; Sun, S.D.; Wang, M.Y.; Li, C.F.; Yuan, D.; Fu, H.Z. Chemical constituents and analgesic activity of Aconitum kusnezoffii Reichb. J. Chin. Pharm. Sci. 2018, 27, 855–863. [Google Scholar]
  114. Guo, Q.; Xia, H.; Meng, X.; Shi, G.; Xu, C.; Zhu, C.; Zhang, T.; Shi, J. C19-Diterpenoid alkaloid arabinosides from an aqueous extract of the lateral root of Aconitum carmichaelii and their analgesic activities. Acta Pharm. Sin. B 2018, 8, 409–419. [Google Scholar] [CrossRef] [PubMed]
  115. Nesterova, Y.V.; Povet’yeva, T.; Suslov, N.; Zyuz’kov, G.; Pushkarskii, S.; Aksinenko, S.; Schultz, E.; Kravtsova, S.; Krapivin, A. Analgesic Activity of Diterpene Alkaloids from Aconitum baikalensis. Bull. Exp. Biol. Med. 2014, 157, 488–491. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, D.-K.; Ai, H.-L.; Zi, S.-H.; Zhang, L.-M.; Yang, S.-C.; Guo, H.-C.; Shen, Y.; Chen, Y.-P.; Chen, J.-J. Four new C 18 -diterpenoid alkaloids with analgesic activity from Aconitum weixiense. Fitoterapia 2013, 91, 280–283. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, D.-P.; Lou, H.-Y.; Huang, L.; Hao, X.-J.; Liang, G.-Y.; Yang, Z.-C.; Pan, W.-D. A novel franchetine type norditerpenoid isolated from the roots of Aconitum carmichaeli Debx. with potential analgesic activity and less toxicity. Bioorganic Med. Chem. Lett. 2012, 22, 4444–4446. [Google Scholar] [CrossRef]
  118. Taki, M.; Niitu, K.; Omiya, Y.; Noguchi, M.; Fukuchi, M.; Aburada, M.; Okada, M. 8-O-Cinnamoylneoline, a New Alkaloid from the Flower Buds of Aconitum carmichaeli and its Toxic and Analgesic Activities. Planta Med. 2003, 69, 800–803. [Google Scholar]
  119. Song, M.-K.; Liu, H.; Jiang, H.-L.; Yue, J.-M.; Hu, G.-Y.; Chen, H.-Z. Discovery of talatisamine as a novel specific blocker for the delayed rectifier K+ channels in rat hippocampal neurons. Neuroscience 2008, 155, 469–475. [Google Scholar] [CrossRef]
  120. Liu, H.; Li, Y.; Song, M.; Tan, X.; Cheng, F.; Zheng, S.; Shen, J.; Luo, X.; Ji, R.; Yue, J. Structure-Based Discovery of Potassium Channel Blockers from Natural Products: Virtual Screening and Electrophysiological Assay Testing. Chem. Biol. 2003, 10, 1103–1113. [Google Scholar] [CrossRef]
  121. Zheng, X.L.; Xie, Z.H.; Zhu, Z.Y.; Liu, Z.; Wang, Y.; Wei, L.F.; Yang, H.; Yang, H.N.; Liu, Y.Q.; Bi, J.Z. Methyllycaconitine Alleviates Amyloid-β Peptides-Induced Cytotoxicity in SH-SY5Y Cells. PLoS ONE 2014, 9, e111536. [Google Scholar] [CrossRef]
  122. Wan, L.X.; Zhang, J.F.; Zhen, Y.Q.; Zhang, L.; Li, X.; Gao, F.; Zhou, X.L. Isolation, Structure Elucidation, Semi-Synthesis, and Structural Modification of C19-Diterpenoid Alkaloids from Aconitum apetalumand Their Neuroprotective Activities. J. Nat. Prod. 2021, 84, 1067–1077. [Google Scholar] [CrossRef]
  123. Nesterova, Y.V.; Povet’eva, T.N.; Suslov, N.I.; Zyuz’kov, G.N.; Zhdanov, V.V.; Fedorova, Y.S.; Kul’pin, P.V.; Shaposhnikov, K.V. Correction of Cholinergic Abnormalities in Mnestic Processes with Diterpene Alkaloid Songorine. Bull. Exp. Biol. Med. 2018, 165, 10–13. [Google Scholar] [CrossRef] [PubMed]
  124. Zyuz’kov, G.N.; Suslov, N.I.; Losev, E.A.; Ermolaeva, L.A.; Zhdanov, V.V.; Udut, E.V.; Miroshnichenko, L.A.; Simanina, E.V.; Demkin, V.P.; Povet’eva, T.N. Cerebroprotective and Regenerative Effects of Alkaloid Z77 under Conditions of Brain Ischemia. Bull. Exp. Biol. Med. 2015, 158, 352–355. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, Y.; Zhang, H.; Wang, J.; Yu, M.; Zhang, Q.; Yan, S.; You, D.; Shi, L.; Zhang, L.; Wang, L.; et al. Aconiti lateralis Radix Praeparata inhibits Alzheimer’s disease by regulating the complex regulation network with the core of GRIN1 and MAPK1. Pharm. Biol. 2021, 59, 311–320. [Google Scholar] [CrossRef] [PubMed]
  126. Luo, Z.-H.; Chen, Y.; Sun, X.-Y.; Fan, H.; Li, W.; Deng, L.; Yin, T.-P. A new diterpenoid alkaloid from Aconitum hemsleyanum. Nat. Prod. Res. 2020, 34, 1331–1336. [Google Scholar] [CrossRef] [PubMed]
  127. Jiang, G.-Y.; Qin, L.-L.; Gao, F.; Huang, S.; Zhou, X.-L. Fifteen new diterpenoid alkaloids from the roots of Aconitum kirinense Nakai. Fitoterapia 2020, 141, 104477. [Google Scholar] [CrossRef]
  128. Ahmad, H.; Ahmad, S.; Shah, S.A.A.; Khan, H.U.; Khan, F.A.; Ali, M.; Latif, A.; Shaheen, F.; Ahmad, M. Selective dual cholinesterase inhibitors from Aconitum laeve. J. Asian Nat. Prod. Res. 2018, 20, 172–181. [Google Scholar] [CrossRef]
  129. Ahmad, H.; Ahmad, S.; Ali, M.; Latif, A.; Shah, S.A.A.; Naz, H.; Rahman, N.U.; Shaheen, F.; Wadood, A.; Khan, H.U.; et al. Norditerpenoid alkaloids of Delphinium denudatum as cholinesterase inhibitors. Bioorganic Chem. 2018, 78, 427–435. [Google Scholar] [CrossRef]
  130. Ahmad, H.; Ahmad, S.; Shah, S.A.A.; Latif, A.; Ali, M.; Khan, F.A.; Tahir, M.N.; Shaheen, F.; Wadood, A.; Ahmad, M. Antioxidant and anticholinesterase potential of diterpenoid alkaloids from Aconitum heterophyllum. Bioorganic Med. Chem. 2017, 25, 3368–3376. [Google Scholar] [CrossRef]
  131. Ahmad, H.; Ahmad, S.; Khan, E.; Shahzad, A.; Ali, M.; Tahir, M.N.; Shaheen, F.; Ahmad, M. Isolation, crystal structure determination and cholinesterase inhibitory potential of isotalatizidine hydrate from Delphinium denudatum. Pharm. Biol. 2017, 55, 680–686. [Google Scholar] [CrossRef]
  132. Nisar, M.; Obaidullah; Ahmad, M.; Wadood, N.; Lodhi, M.A.; Shaheen, F.; Choudhary, M.I. New diterpenoid alkaloids from Aconitum heterophyllum Wall: Selective butyrylcholinestrase inhibitors. J. Enzym. Inhib. Med. Chem. 2009, 24, 47–51. [Google Scholar] [CrossRef]
  133. Atta-ur-Rahman; Fatima, N.; Akhtar, F.; Choudhary, M.I.; Khalid, A. New norditerpenoid alkaloids from Aconitum falconeri. J. Nat. Prod. 2000, 63, 1393–1395. [Google Scholar] [CrossRef]
  134. Andreasen, J.T.; Olsen, G.M.; Wiborg, O.; Redrobe, J.P. Antidepressant-like effects of nicotinic acetylcholine receptor antagonists, but not agonists, in the mouse forced swim and mouse tail suspension tests. J. Psychopharmacol. 2009, 23, 797–804. [Google Scholar] [CrossRef]
  135. Nesterova, Y.V.; Povetieva, T.N.; Suslov, N.I.; Semenov, A.A.; Pushkarskiy, S.V. Antidepressant Activity of Diterpene Alkaloids of Aconitum baicalense Turcz. Bull. Exp. Biol. Med. 2011, 151, 425–428. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, L.; Li, B.; Zhou, Y.; Wang, L.; Tang, F.; Shao, D.; Jiang, X.; Zhao, H.; Cui, R.; Li, Y. Antidepressant-like Effect of Fuzi Total Alkaloid on Ovariectomized Mice. J. Pharmacol. Sci. 2012, 120, 280–287. [Google Scholar] [CrossRef]
  137. Mineur, Y.S.; Mose, T.N.; Blakeman, S.; Picciotto, M.R. Hippocampal α7 nicotinic ACh receptors contribute to modulation of depression-like behaviour in C57BL/6J mice. Br. J. Pharmacol. 2018, 175, 1903–1914. [Google Scholar] [CrossRef]
  138. Nesterova, Y.V.; Povet’eva, T.N.; Suslov, N.I.; Shults, E.E.; Ziuz’kov, G.N.; Aksinenko, S.G.; Afanas’eva, O.G.; Krapivin, A.V.; Kharina, T.G. Anxiolytic Activity of Diterpene Alkaloid Songorine. Bull. Exp. Biol. Med. 2015, 159, 620–622. [Google Scholar] [CrossRef] [PubMed]
  139. Ahmad, M.; Yousuf, S.; Khan, M.B.; Ahmad, A.S.; Saleem, S.; Hoda, M.N.; Islam, F. Protective effects of ethanolic extract of Delphinium denudatum in a rat model of Parkinson’s disease. Hum. Exp. Toxicol. 2006, 25, 361–368. [Google Scholar] [CrossRef] [PubMed]
  140. Jin, Y.Y.; Qin, C.H.; Duan, B.L.; Huang, Y.Q.; Ma, K. Clinical efficacy of bulleyaconitine A combined with gabapentin on postherpetic neuralgia. Zhonghua Yi Xue Za Zhi 2021, 101, 3575–3580. [Google Scholar]
  141. Chen, M.G.; Wang, Q.H.; Lin, Y.B. Clinical study in epidural injection with lappaconitine for post-operative analgesia. Zhongguo Zhong Xi Yi Jie He Za Zhi Zhongguo Zhongxiyi Jiehe Zazhi Chin. J. Integr. Tradit. West. Med. 1995, 15, 274–276. [Google Scholar] [CrossRef]
  142. Wang, J. Clinical analgesic effect of lappaconitine in 56 patients after cholecystectomy. Chin. J. Clin. Pharmacol. Ther. 2010, 15, 551. [Google Scholar]
  143. Dzhakhangirov, F.N.; Sultankhodzhaev, M.N.; Tashkhodzhaev, B.; Salimov, B.T. Diterpenoid alkaloids as a new class of antiarrhythmic agents. Structure-activity relationship. Chem. Nat. Compd. 1997, 33, 190–202. [Google Scholar] [CrossRef]
  144. Valeev, A.E.; Verkhratskii, A.N.; Dzhakhangirov, F.N. Effects of allapinine on sodium currents in neurons isolated from the rat trigeminal ganglion and cardiomyocytes. Neurophysiology 1990, 22, 157–162. [Google Scholar] [CrossRef]
  145. Wang, J.-L.; Shen, X.-L.; Chen, Q.-H.; Qi, G.; Wang, W.; Wang, F.-P. Structure-Analgesic Activity Relationship Studies on the C18- and C19-Diterpenoid Alkaloids. Chem. Pharm. Bull. 2009, 57, 801–807. [Google Scholar] [CrossRef] [PubMed]
  146. Panter, K.E.; Manners, G.D.; Stegelmeier, B.L.; Lee, S.; Gardner, D.R.; Ralphs, M.H.; Pfister, J.A.; James, L.F. Larkspur poisoning: Toxicology and alkaloid structure–activity relationships. Biochem. Syst. Ecol. 2002, 30, 113–128. [Google Scholar] [CrossRef]
  147. Chan, T.Y.K. Aconite poisoning. Clin. Toxicol. 2009, 47, 279–285. [Google Scholar] [CrossRef] [PubMed]
  148. Chan, T.Y.K. Aconitum alkaloid content and the high toxicity of aconite tincture. Forensic Sci. Int. 2012, 222, 1–3. [Google Scholar] [CrossRef] [PubMed]
  149. Stegelmeier, B.L.; Hall, J.O.; Gardner, D.R.; Panter, K.E. The toxicity and kinetics of larkspur alkaloid, methyllycaconitine, in mice. J. Anim. Sci. 2003, 81, 1237–1241. [Google Scholar] [CrossRef] [PubMed]
  150. Chan, Y.-T.; Wang, N.; Feng, Y. The toxicology and detoxification of Aconitum: Traditional and modern views. Chin. Med. 2021, 16, 61. [Google Scholar] [CrossRef]
  151. Csupor, D.; Wenzig, E.-M.; Zupkó, I.; Wölkart, K.; Hohmann, J.; Bauer, R. Qualitative and quantitative analysis of aconitine-type and lipo-alkaloids of Aconitum carmichaelii roots. J. Chromatogr. A 2009, 1216, 2079–2086. [Google Scholar] [CrossRef]
Scheme 1. Biological effects of DAs on CNS.
Scheme 1. Biological effects of DAs on CNS.
Pharmaceuticals 16 00747 sch001
Figure 1. Backbone structures of different types of C18-diterpenoid alkaloids. Lappaconitines possess a methine at C7, while ranaconitines are characterized by an oxygen-containing group at this position. Ranaconitines have a methine unit at C4, while lappaconitines contain a methine unit or an oxygenated nonprotonated carbon, or chloro-substituents at C4.
Figure 1. Backbone structures of different types of C18-diterpenoid alkaloids. Lappaconitines possess a methine at C7, while ranaconitines are characterized by an oxygen-containing group at this position. Ranaconitines have a methine unit at C4, while lappaconitines contain a methine unit or an oxygenated nonprotonated carbon, or chloro-substituents at C4.
Pharmaceuticals 16 00747 g001
Figure 2. The backbone structures of different types of C19-diterpenoid alkaloids.
Figure 2. The backbone structures of different types of C19-diterpenoid alkaloids.
Pharmaceuticals 16 00747 g002
Figure 3. The backbone structures of different types of C20-diterpenoid alkaloids.
Figure 3. The backbone structures of different types of C20-diterpenoid alkaloids.
Pharmaceuticals 16 00747 g003
Scheme 2. Biosynthetic pathway of diterpene alkaloids in Ranunculaceae.
Scheme 2. Biosynthetic pathway of diterpene alkaloids in Ranunculaceae.
Pharmaceuticals 16 00747 sch002
Figure 4. The chemical structures of the anticonvulsant diterpene alkaloids.
Figure 4. The chemical structures of the anticonvulsant diterpene alkaloids.
Pharmaceuticals 16 00747 g004
Figure 5. (a) Chemical structure of methyllycaconitine; (b) Homomeric α7 nicotinic acetylcholine receptor.
Figure 5. (a) Chemical structure of methyllycaconitine; (b) Homomeric α7 nicotinic acetylcholine receptor.
Pharmaceuticals 16 00747 g005
Figure 6. Binding of methyllycaconitine could affect the binding pattern of ACh to binding sites and forms the three different conformational states. (a) The binding of two AChs to two consecutive binding sites leads to slow channel activation; (b) The binding of two AChs to two non-consecutive sites results in rapid activation and receptor desensitization; (c) Binding less than two AChs to binding sites causes a deactivation and the channel will be closed.
Figure 6. Binding of methyllycaconitine could affect the binding pattern of ACh to binding sites and forms the three different conformational states. (a) The binding of two AChs to two consecutive binding sites leads to slow channel activation; (b) The binding of two AChs to two non-consecutive sites results in rapid activation and receptor desensitization; (c) Binding less than two AChs to binding sites causes a deactivation and the channel will be closed.
Pharmaceuticals 16 00747 g006
Figure 7. Chemical structures of the analgesic DAs.
Figure 7. Chemical structures of the analgesic DAs.
Pharmaceuticals 16 00747 g007
Figure 8. Chemical structures of diterpene alkaloids with antidementia activity.
Figure 8. Chemical structures of diterpene alkaloids with antidementia activity.
Pharmaceuticals 16 00747 g008aPharmaceuticals 16 00747 g008b
Table 1. Anticonvulsant effects of fractions/constituents from Aconitum and Delphinium species.
Table 1. Anticonvulsant effects of fractions/constituents from Aconitum and Delphinium species.
PlantUsed Part/ConstituentsBiological ActivityAffected In Vitro/In Vivo ModelRef.
Delphinium nordhageniiAcetone fractionAnticonvulsant activityMice[46]
Aconitum violaceumVarious fractionsAnticonvulsant activityMice[47]
Delphinium denudatumAqueous fractionAntiepileptiform activityCultured rat hippocampal pyramidal neurons[48]
Delphinium denudatumAqueous fraction of rootsAntiepileptiform activityPrimary hippocampal neuronal cultures[49]
Delphinium denudatumEthanolic extract and the aqueous fraction of rootsAnticonvulsant activityMice[50]
Aconitum carmichaeliFuzi total alkaloidsIncrease the seizure latency and decrease the mortalityMice[33]
Aconitum sp.6-Benzoyl deltamine (9) and eldeline (10)Antiepileptiform activityRat hippocampal slices[51]
1, 3, and ajacine (11)Antiepileptiform activityRat hippocampal slices[39]
5Antiepileptiform activityRat hippocampal slices[52]
7Increasing excitabilityRat hippocampal slices[53]
14-Benzoyl talatisamine (12) and talatisamine (13)Antiepileptiform activityRat hippocampal slices[54]
8Antiepileptiform activityRat hippocampal slices[45]
810 nM evoked excitation
30–100 nM biphasic effect
above 100 nM suppressed the orthodromic population spike
Rat hippocampal slices[55]
1, 3, and 5Antiepileptiform activityRat hippocampal slices[56]
3 and 4Antiepileptiform activityRat hippocampal slices[57]
2Antiepileptiform activityRat hippocampal slices[38]
1-Benzoyl napelline (14)Antiepileptiform activityRat hippocampal slices[58]
5Antiepileptiform activityRat hippocampal slices[59]
5 and heteratisine (15)Antiepileptiform activityRat hippocampal slices[60]
1Antiepileptiform activityRat hippocampal slices[61]
1Prolonged seizure-like activityNeocortical slices from juvenile Sprague-Dawley rats[42]
Table 2. Beneficial effects of low doses of methyllycaconitine (16) on neurologic functions.
Table 2. Beneficial effects of low doses of methyllycaconitine (16) on neurologic functions.
Biological ActivitiesAffected In Vitro/In Vivo ModelRef.
Picomolar concentration of 16 potentiated α7 nAChR activity/improved memory acquisition processes.Oocytes from mature Xenopus laevis females/rat[64]
Nanomolar concentrations of 16 acted as a co-agonist to potentiate TE-671 cell responses to acetylcholine, epibatidine, nicotine, and neostigmine.Rhabdomyosarcoma cell line TE-671[69]
16 (0.4–1.3 mg/kg) improved response
accuracy at low doses.
Rat model[70]
16 (1–4 mg/kg) significantly counteracted the learning impairment caused by dizocilpine.Rat model[71]
16 (1 mg/kg) significantly improved attentional function.Rat model[72]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Salehi, A.; Ghanadian, M.; Zolfaghari, B.; Jassbi, A.R.; Fattahian, M.; Reisi, P.; Csupor, D.; Khan, I.A.; Ali, Z. Neuropharmacological Potential of Diterpenoid Alkaloids. Pharmaceuticals 2023, 16, 747. https://doi.org/10.3390/ph16050747

AMA Style

Salehi A, Ghanadian M, Zolfaghari B, Jassbi AR, Fattahian M, Reisi P, Csupor D, Khan IA, Ali Z. Neuropharmacological Potential of Diterpenoid Alkaloids. Pharmaceuticals. 2023; 16(5):747. https://doi.org/10.3390/ph16050747

Chicago/Turabian Style

Salehi, Arash, Mustafa Ghanadian, Behzad Zolfaghari, Amir Reza Jassbi, Maryam Fattahian, Parham Reisi, Dezső Csupor, Ikhlas A. Khan, and Zulfiqar Ali. 2023. "Neuropharmacological Potential of Diterpenoid Alkaloids" Pharmaceuticals 16, no. 5: 747. https://doi.org/10.3390/ph16050747

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop