Next Article in Journal
Solid Fe Resources Separated from Rolling Oil Sludge for CO Oxidation
Next Article in Special Issue
Physiologically Based Pharmacokinetic Modeling of Nanoparticle Biodistribution: A Review of Existing Models, Simulation Software, and Data Analysis Tools
Previous Article in Journal
Functions of the Zinc-Sensing Receptor GPR39 in Regulating Intestinal Health in Animals
Previous Article in Special Issue
Ligand-Enhanced Negative Images Optimized for Docking Rescoring
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Poisonous Piperidine Plants and the Biodiversity of Norditerpenoid Alkaloids for Leads in Drug Discovery: Experimental Aspects

by
Ashraf M. A. Qasem
,
Michael G. Rowan
and
Ian S. Blagbrough
*
School of Pharmacy, University of Bath, Bath BA2 7AY, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12128; https://doi.org/10.3390/ijms232012128
Submission received: 15 September 2022 / Revised: 4 October 2022 / Accepted: 7 October 2022 / Published: 12 October 2022

Abstract

:
There are famous examples of simple (e.g., hemlock, Conium maculatum L.) and complex (e.g., opium poppy, Papaver somniferum L., Papaveraceae) piperidine-alkaloid-containing plants. Many of these are highly poisonous, whilst pepper is well-known gastronomically, and several substituted piperidine alkaloids are therapeutically beneficial as a function of dose and mode of action. This review covers the taxonomy of the genera Aconitum, Delphinium, and the controversial Consolida. As part of studying the biodiversity of norditerpenoid alkaloids (NDAS), the majority of which possess an N-ethyl group, we also quantified the fragment occurrence count in the SciFinder database for NDA skeletons. The wide range of NDA biodiversity is also captured in a review of over 100 recently reported isolated alkaloids. Ring A substitution at position 1 is important to determine the NDA skeleton conformation. In this overview of naturally occurring highly oxygenated NDAs from traditional Aconitum and Delphinium plants, consideration is given to functional effect and to real functional evidence. Their high potential biological activity makes them useful candidate molecules for further investigation as lead compounds in the development of selective drugs.

1. Piperidines: Poisonous, Tasty, and Beneficial

Nature is rich with examples of plants that can be described as medicinal or poisonous. The controversy in the description spotlights the fact, well known in pharmacy and the pharmaceutical sciences, that dose and mode of action are critical. Out of thousands of plants with various types of active principles, many examples can be found of natural sources of alkaloids containing a substituted piperidine nucleus.
There are well-known examples of simple piperidine-alkaloid-containing plants, such as the famous poison hemlock (Conium maculatum L.), in the family Apiaceae (formerly Umbelliferae). It is a lethal poison that was given to criminals in ancient Greece and that the Greek philosopher, Socrates, was forced to drink (399 B.C.) [1]. The principal component of poison hemlock is the piperidine alkaloid coniine 1 (Figure 1), which is a nicotinic acetylcholine receptor (nAChR) agonist [1] where the importance of the positive centre was highlighted in the Beers–Reich model [2]. Consumption of C. maculatum leads to various degrees of toxicity in animals, where it has been found that it is more poisonous to cattle than to other animals. Human toxicity signs were described by Socrates’ pupil as trembling, staggering, and rapid muscular weakness. Death resulting from hemlock poisoning is mainly due to respiratory failure [1,3]. Another nAChR agonist is anabasine 2, a natural nicotine-3-like isomer compound from Nicotiana spp. (tobacco) of the family Solanaceae. Two important species are N. glauca (wild tree tobacco) and N. tabacum L. Anabasine 2 is the major component in N. glauca, while nicotine 3 is the main constituent in N. tabacum L. [3,4,5]. Another source of piperidine alkaloids is Lobelia spp. (Campanulaceae). An important example is the Indian tobacco, L. inflata L., which is used traditionally in smoking cessation and in the treatment of respiratory conditions [6]. Lobeline 4 is the major and the most biologically active component of L. inflata L. It was found that lobeline 4 can be described as agonist, antagonist, or mixed agonist/antagonist at nAChR [7].
Lupinus spp. (Fabaceae) consumption leads to teratogenic crooked calf disease. The teratogenicity was initially suggested to be due to the quinolizidine alkaloid, anagyrine 5 [5,8]. As similar teratogenicity happens in livestock due to the consumption of C. maculatum and N. glauca, which contain mainly piperidine alkaloids [4,9,10], it is now suggested that the main piperidine alkaloid, ammodendrine 6, which is found in many Lupinus spp. such as L. formosus, is responsible for such deformations. Keeler and Panter [8] showed that anagyrine 5 exists as a minor component in L. formosus, while ammodendrine 6 is the major one, supporting that the piperidine alkaloid 6 is responsible for the teratogenicity.
Black pepper, Piper nigrum L. (Piperaceae), is known as the king of spices, and it is the natural source of piperine 7 which shows anti-inflammatory, antioxidant, anticancer, and antimicrobial activities [11,12,13]. 1-Deoxynojirimycin 8 is another example of a piperidine alkaloid which acts as one of the most potent α-glycosidase inhibitors and has relevant biological activity in the treatment of hyperglycaemia and obesity. It is naturally occurring in the leaves of white mulberry, Morus alba L. (Moraceae) [14,15].
In addition to simple piperidines, there are many famous examples of piperidine-containing plants where the piperidine ring is part of a complex skeleton. Papaver somniferum L. (Papaveraceae), also known as opium poppy, is a natural source of the opioids morphine 9 and codeine 10. These piperidine-containing alkaloids are used as strong analgesics, but are also abused in addiction [16,17]. The potency of morphine 9 is much higher than codeine 10, and the fact that codeine 10 exerts its analgesic effect after being metabolized into morphine 9 highlights the importance of the phenolic alcohol in the activity at μ opioid receptors [18]. Another example is cytisine 11, which is derived from Laburnum anagyroides Medik. (Fabaceae), and it acts as an nAChR agonist. The skeleton of cytisine 11 shows that the piperidine coexists with a quinolizidine ring [3,19] and fits the Beers–Reich model, as cytisine 11 contains a cationic centre and heteroatoms, where the model showed that the distance between them in nAChR ligands is 5.9 Å [2].
There are piperidine-containing alkaloids, which are C-18 and C-19 norditerpenoid alkaloids (NDAs) from Aconitum and Delphinium (Ranunculaceae), and especially aconitine 12, lappaconitine 13, lycoctonine 14, lycaconitine 15, and methyllycaconitine (MLA) 16. The C-19 NDA aconitine 12 was first discovered in 1833 by P. L. Geiger from A. napellus [20,21]. It is considered a potent lethal cardiotoxin that acts on voltage-gated sodium channels (VGSC) and keeps them in an open conformation. In contrast, lappaconitine 13, which was the first C-18 NDA to be discovered, is a VGSC blocker [22]. The hydrogen bromide (HBr) salt of lappaconitine 13 (allapinine) is used clinically in Russia as an anti-arrhythmic drug. Lappaconitine 13 was first discovered from A. septentrionale Koelle by H. V. Rosendahl in 1895 [23,24]. Lycoctonine 14 was first reported in 1865 from A. lycoctonum L. [25]. Lycaconitine 15 was first reported in 1884 from A. lycoctonum L. [26]. MLA 16, a C-19 NDA, was first discovered by Manske in 1938 in D. brownii Rydb [27]. Goodson (1943) later determined its exact formula [28]. MLA 16 acts as a potent competitive antagonist on α7-nAChR [29]. The importance of the piperidine nitrogen of NDAs in the interaction with nAChR was proven, as semi-synthetic analogues with quaternary nitrogen showed higher activity, and that suggests that the nitrogen is a main site of receptor interaction [30]. All the previous examples show alkaloids which contain piperidine alone or they coexist with other heterocycles. Scopolamine (hyoscine) 17, L-hyoscyamine 18, and atropine (dl-hyoscyamine) 19 (Figure 1) are examples of alkaloids that contain a piperidine ring fused with pyrrolidine (tropane alkaloids). These alkaloids are antagonists at the muscarinic acetylcholine receptors (mAChR) and are derived from many plants of the nightshades (Solanaceae). These compounds meet the Beers–Reich pharmacophore criteria, as they contain a cationic centre and a Van der Waals surface (heteroatoms). A member of the nightshades is Datura stramonium L., which is also known as thornapple and jimsonweed [31,32].

2. Taxonomy of Aconitum, Delphinium, and Consolida

Diterpenoid alkaloids are found mainly in Aconitum, Delphinium, and Consolida within the family Ranunculaceae, and Garrya (silk tassel) from the family Garryaceae. Apart from these genera, three diterpenoid alkaloids, lycoctonine 14, MLA 16, and inuline (which is the 2-aminobenzoate ester of lycoctonine 14) have been reported from Inula royleana (Asteraceae) [33]. The three families (Ranunculaceae, Garryaceae, and Asteraceae) are classified under the Angiospermae class (flowering plants) [34,35,36]. C-18 and C-19 NDAs are derived from only three genera within Ranunculaceae: Aconitum, Delphinium, and Consolida. The Ranunculaceae family contains around 43 genera and more than 2000 species [37]. Aconitum L. with around 330 species and Delphinium L. with around 450 species are considered the major genera in this family [38,39].
All three of these genera are classified within the tribe Delphinieae of the subfamily Ranunculoideae. [40]. A scientific classification of Aconitum, Delphinium, and Consolida is shown in Scheme 1 [36,40,41,42].
Aconitum L. has been divided into three subgenera (Aconitum, Lycoctonum (DC.) Peterm., and Gymnaconitum (Stapf) Rapes). The Aconitum subgenus Aconitum produces biennial tuberous roots, while subgenus Lycoctonum (DC.) Peterm. species have perennial rhizomes. The only annual species of the Aconitum genus can be found in subgenus Gymnaconitum (Stapf) Rapes. [43]. The Delphinium L. genus is also divided into two subgenera (Delphinastnim (DC.) Wang and Delphinium) [44], and the species within this genus are usually perennial (occasionally annual) [45]. Due to the shape of flowers of the Delphinium species which resemble dolphins, the Delphinium genus takes its name from the Greek word delphis [45].
The genus Consolida has proved to be more controversial. A. P. De Candolle separated a group of annual species from the genus Delphinium L. to form an independent section (Consolida DC.). S.F. Gray changed the section Consolida to the rank of a genus in 1821 (Consolida (DC.) S.F. Gray). Boisser gave the rank of genus to the Consolida section of Delphinium L. in 1867 and Huth gave it the rank subgenus in 1895 [46]. Huth was the last worker to include Consolida within Delphinium. Much more recently, Jabbour and Renner (2011) suggested using a DNA phylogenetic study that Consolida should be embedded in Delphinium [47]. Commonly, Delphinium and Consolida species are called larkspur, which is also a name derived from the shape of the flowers [45]. The colourful flowers of the Delphinium species gave rise to many cultivated species (cultivars) that are used as ornamental plants in the garden. These hybrid species come from crossing different parent plants and mainly from the tetraploid D. elatum L. [48,49]. Examples of the hybrid Delphinium varieties are the giant pacific court hybrids which originate from D. elatum and other species such as D. exaltatum and D. formosum [50].

3. NDA Chemical Toxicity

North Americans have divided the Delphinium (larkspur) plants into three categories depending on habit of growth and environment. First are the tall larkspurs (such as D. barbeyi, D. occidentale), which are 1–2 m tall and generally exist at altitudes above 2400 m in moist habitats. Second are the intermediate larkspurs (such as D. geyeri, plains larkspur), which are 0.6–1 m tall and grow on the short grass prairies of Nebraska, Wyoming, and Colorado. The third category is low larkspurs (such as D. andersonii), which are less than 0.6 m tall and generally grow in the desert/semidesert, foothills, or low mountain ranges [51,52]. Delphinium (larkspur) alkaloids cause economically important livestock toxicity across North American ranges [51,53]. Tall larkspurs contain higher amounts of toxic NDAs and are therefore considered a greater threat [54]. Intoxication happens due to the action of NDAs at the α1-nAChR expressed at neuromuscular junctions (NMJ) [55].
It was found that the livestock intoxication by larkspurs is controlled by different factors, for example, cattle breed and genetics affect the susceptibility to the intoxication. Age is another factor, where young heifers are more susceptible than mature cows. The cattle sex was reported to be an effective factor, where heifers are more prone to the toxicity than steers and bulls. Lastly, the plant factor plays an important role, where the alkaloid concentration and composition of methylsuccinimidoanthranoyl-lycoctonine (MSAL) and non-MSAL (Figure 2), which depend on the population, species, climate, and the year, affect the toxicity in cattle and the amount needed to develop clinical signs [56,57].
The toxicity of NDAs found in three tall larkspur species (D. barbeyi, D. occidentale, D. glaucescens) was tested in mice. The assay revealed that the 7,8-methylenedioxy-lycoctonine (MDL) alkaloids are the least toxic NDAs. The lycoctonine-type is twice as toxic as MDL, but it is considered to be a low toxic group, where the least toxic alkaloid of this category, brownine 20, has a toxicity which is comparable to the MDL NDA. The MSAL alkaloids MLA 16 and 14-deacetylnudicauline 21 were 10-times more toxic than any other tested NDAs (Figure 2) [54].
MSAL is much more toxic than MDL, and the MSAL level in the tall larkspurs mainly contributes to livestock poisoning. A report investigated the importance of the MDL alkaloids and found that MDL alkaloids exacerbate the toxicity of the MSAL alkaloids; as the ratio of MDL to MSAL increases, the amount of MSAL that is needed to develop clinical signs decreases. The exact mechanism of this MDL action is not known, but it was suggested that MDL may act as a co-agonist in an allosteric manner or at the orthosteric ligand binding site of the receptor to exacerbate the toxicity of MSAL-type alkaloids on nAChR and therefore increase their toxicity [58]. The observed action could also be due to an effect of MDL alkaloids on metabolic enzymes which results in prolonged exposure to the MSAL alkaloids, but further investigation is needed.

4. Norditerpenoid Alkaloid (NDA) Biodiversity

NDAs have complex highly oxygenated hexacyclic systems, and as many of them are of pharmacological importance, their structures and 3D configuration are significant factors in their actions at various biological targets [59]. The majority of NDAs possess an N-Et group, as shown in Table 1, which shows various NDA skeletons and their abundance in the SciFinder database. Substitution at position 1 is important to determine the NDA skeleton conformation, as ring A in 1-OMe NDA free bases exists in a twisted-chair conformation, and in 1-OH NDA ring A adopts a twisted-boat conformation [60,61]. Table 1 shows that 1-OMe NDA abundancy is 10-times higher than 1-OH NDA. The biological activity of NDAs attracts natural product chemists to investigate their sources, and that has resulted in the discovery of interesting NDA skeletons, some of them with pharmacological importance.
Recent phytochemical investigations reported on Aconitum and Delphinium species show the wide variety of structural motifs in such NDAs. Chen et al. reported the extraction of a new aconitine-type NDA with a 1-OH substitution, pubescensine 22 from A. soongaricum var. pubescens, and this showed a potent insect antifeedant activity (EC50 < 1 mg/cm2) [62]. Ding and co-workers discovered four new NDAs, vilmorines A–D 2326 from A. vilmorinianum (Figure 3) [63]. Vilmorine D 26 exhibited moderate to weak antioxidant activity (Fe2+ chelation activity) with IC50 = 33.6 ± 0.2 μg/mL, and it showed antibacterial activity against Staphylococcus aureus and Bacillus subtilis with MICs of 64 and 32 μg/mL, respectively. Vilmorine A 23 has an unusual spiro junction. Only three such compounds were isolated with that characteristic skeleton (Table 1). Vilmorine A 23 also has the really unusual 1-β-OMe group, whereas the vast majority of the position 1 substituents have an α-configuration. Vilmorines B–C 2425 have an unusual cyclopropyl moiety, and they are rare examples containing an imine (piperideine) (Table 1).
Qin et al. isolated five new NDAs from A. carmichaelii, carmichaenine A–E 2731 [64], with characteristic 1-OH substitutions. Majusine D 32 from D. majus W. T. Wang and stapfianine A 33 from A. stapfianu were discovered as new C-19 NDAs [65,66] with 1-OH substitutions. Sharwuphinine B 34 was discovered from D. shawurense as one of the few quaternary C-19 NDAs (Figure 4) [67].
Chen et al. isolated two imine (piperideine)-type NDAs, vilmorrianines F–G 3536, in addition to new N-desethyl-N-formyl-8-O-methyltalatisamine 37 from A. vilmorinianum Komarov [68]. Six new NDAs, 6-dehydroeladine 38, elapacidine 39, iminopaciline 40, iminoisodelpheline 41, iminodelpheline 42 (piperideines), and N-formyl-4,19-secopacinine 43, were extracted from D. elatum seeds (Figure 4) [69]. Shan et al. reported two new C-18 NDAs, anthriscifoltine A–B 4445 from D. anthriscifolium var. majus [70]. Ding and co-workers also isolated three new NDAs, vilmotenitines A–C 4648 from A. vilmorinianum var. patentipilum, (Figure 5) where vilmotenitines A and B 4647 had an unusual (spiro) rearranged six-membered B ring [71], as they had found in vilmorine A 23 with the inverted substituent stereochemistry at position 1 [63].
Two new C19 NDAs, iliensine A and B 4950, were isolated from D. iliense, where iliensine A 49 had a characteristic glycosidic linkage [72]. Wada et al. isolated four new C19 NDAs with a 7,8-methylenedioxy moiety from D. elatum [73], 19-oxoisodelpheline 51, N-deethyl-19-oxoisodelpheline 52, N-deethyl-19-oxodelpheline 53, and melpheline 54 (Figure 6).
Wang and co-workers discovered three new C19 NDAs, szechenyianine A, B, and C 5557, from A. szechenyianum [74]. All three compounds were tested against nitric oxide (NO) release inhibition, as they were considered potential anti-inflammatory agents. Szechenyianine A 55 showed activity with IC50 36.6 ± 7 μM, while szechenyianine B 56 had IC50 3.3 ± 0.1 μM, and that highlights the importance of the N-O moiety. Szechenyianine C 57 (Figure 6) which is a 7,17-secoaconitine-type NDA, also showed potent activity, with IC50 7.5 ± 0.9 μM.
Chao Zhan et al. reported caerudelphinine A 58, a new 1-OH C-19 lycoctonine-type NDA from D. caeruleum Jacq. ex Camp [75]. Grandiflorine B 59 was also reported as a new C-19 lycoctonine-type NDA from D. grandiflorum [76]. The unusual skeleton of grandiflorine B 59 shows cleavage of the 7–17 bond and N-C19 bond and the formation of an unusual N-C7 bond. Zhao et al. reported three new NDAs, nagaconitine A–C 6062 from A. nagarum var. heterotrichum [77]. Nagaconitine A 60 has a unique acyl group which was reported in only three NDAs. 8,14-Diacetate diester 62 showed antitumor activity against cancer cell line SK-OV-3. Two more new C-19 NDAs, 14-benzoylliljestrandisine 63 and 14-anisoylliljestrandisine 64, were isolated from A. tsaii (Figure 7) [78].
Extensive phytochemical studies on D. anthriscifolium var. majus led to the isolation of six new C-19 NDAs with the 7,8-methylenedioxy moiety, which were named anthrisciflorine A–F 6570 (Figure 8) [79].
Guo et al. isolated two new NDAs, 7,8-epoxy-franchetine 71 and N-(19)-en-austroconitine 72, from A. iochanicum [80]. Tested against NO production in macrophages (mouse cell line), they showed a weak anti-inflammatory effect. Liang et al. isolated sinchiangensine A 73 as a new NDA from A. sinchiangense W.T. Wang (Figure 9), and it showed significant antitumour activity against cancer cell lines A-549, SMCC-7721, MCF-7, and SW-480 [81]. The IC50 (μM) values of 73 against these cell lines were 12.8, 9.6, 11.8, and 18.8, respectively, and these values were comparable with cisplatin, the positive control, the IC50 (μM) values of which were 22.3, 18.6, 28.8, and 18.2. Sinchiangensine A 73 also showed potent antibacterial activity against Gram-positive S. aureus ATCC-25923, with an MIC value (μmol/mL) of 0.15 which is comparable to 0.67, the MIC of the positive control berberine HCl.
Ajacisines A–E 7478 (Figure 10) were isolated as new NDAs from D. ajacis [82]. Testing the in vitro antiviral activity against respiratory syncytial virus (RSV), compounds 7678 showed moderate to weak effects. The IC50 (μM) values of 7678 against RSV were 75.2 ± 1.1, 35.1 ± 0.6, and 10.1 ± 0.3, respectively, while the IC50 (μM) of ribavirin (the positive control) was 3.1 ± 0.8 [82].
Meng et al. reported the isolation of four new C-19 NDAs, aconicarmichoside A–D 7982 (Figure 11), from the aqueous extract of fuzi and the lateral roots of A. carmichaeli [83]. These four alkaloids are the first examples of glycosidic NDAs where the glycosides are directly attached to the alkaloid skeleton.
Fukuyama and co-workers [84] achieved the synthesis of cardiopetaline 83 through Wagner–Meerwein rearrangement of the denudatine skeleton into an aconitine skeleton without the need of pre-activation of the hydroxy group. This means that there is no need to differentiate the hydroxy groups in the poly-oxygenated system as was needed before. Sarpong and co-workers [85] developed a unifying strategy to synthesize C-18 NDA (weisaconitine D 84) and C-19 NDA (liljestrandinine 85) from a common intermediate. Liu and Qin [86] highlighted the importance of dearomatization of aromatic compounds that yield o-benzoquinones coupled with Diels–Alder cycloaddition in the synthesis of complex structures such as 83, 84, 85, and many others. Yang et al. constructed a unique tricyclo [6.2.1.0] BCD-system 86 of the NDA skeleton (Figure 12) [87].
Lian et al. reported five new C-18 NDAs, anthriscifoltines C–G 8791 from D. anthriscifolium var. majus (Figure 12) [88]. Song et al. reported three new C-19 NDAs, szechenyianine D–F 9294 from A. szechenyianum (Figure 13) [89].
Another four new C-19 NDAs, elapacigine 95, N-deethyl-N-formylpaciline 96, N-deethyl-N-formylpacinine 97, and N-formyl-4,19-secoyunnadelphinine 98 (Figure 14), were isolated from D. elatum cv. Pacific giant [90].
Yamashita et al. also reported four new C-19 NDAs, 14-anisoyllasianine 99, 14-anisoyl-N-deethylaconine 100, N-deethylaljesaconitine A 101, and N-deethylnevadensine 102, from A. japonicum subsp. subcuneatum (Nakai) Kadota (Figure 15) [91].
Li et al. isolated four new C-19 NDAs, carmichasines A–D 103106 from A. carmichaelii Debeaux (Figure 15) [92]. Extraction of the roots of A. taronense Fletcher et Lauener, which has been used in traditional Chinese medicine (TCM) to treat rheumatism and arthritis, yielded four new C-19 NDAs, taronenines A–D 107110 (Figure 16) [93]; NDAs 107, 108, and 110 exhibited anti-inflammatory activity when tested in mice cells.
A study conducted on the D. pseudoaemulans C. Y. Yang et B. Wang resulted in the isolation of eight new C-19 NDAs, pseudophnines A−D 111114, pseudorenines A−B 115116, and pseudonidines A−B 117118 (Figure 17) [94].
Ahmad and co-workers reported the isolation of two new C-19 NDAs, jadwarine A–B 119120 from D. denudatum [95]. They also reported a new lycoctonine-type C-19 NDA, swatinine C 121 (Figure 18) which showed competitive inhibitory activity on acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) [96].
Extraction of the roots of A. brevicalcaratum led to the isolation of three new C-19 NDAs, brochyponines A–C 122124 (Figure 18) [97]. Abjalan et al. discovered a new lycoctonine-type C-19 NDA, aemulansine 125 from D. aemulans Navaski, which showed in vitro cytotoxicity [98]. Two novel 8,15-seco C-19 NDAs, nagarine A 126 and B 127 (Figure 19), were isolated from A. nagarum [99].
The variation in pharmacological activities of the NDAs, despite their structural similarities, is an attractive aspect for synthetic chemists to work on to obtain a better understanding of the structure–activity relationships (SAR). Liu et al. reported a total synthesis of the ABCDE system of the C-19 NDAs [100]. Another study reported the construction of the AEF ring system attached to a phenyl group as an analogue to ring D [101]. The construction of the fused CD-bicycle of aconitine was also achieved [102]. Lv et al. built the hexacyclic ring system of franchetine 128, a 7,17-seco NDA [103]. The importance of such NDAs continues to attract chemists to attempt to make a total synthesis of them. Progress has been made in the total synthesis of aconitine 12, but the construction of a pentacyclic system of the aconitine skeleton failed [104]. On the other hand, a total synthesis of talatisamine 129 (Figure 20) has been completed in 33 steps [105]. In addition, the synthesis of a [6-6-6] ABE-tricyclic analogue of MLA 16 has been achieved [106]. A synthetic approach has also been established for the BCD-tricyclic system [107].

5. Conclusions

It is clear that nature is rich with many examples of plants that can be described as medicinal or poisonous. The poisonous piperidine plants show that dose and mode of pharmacological action are critical. The biodiversity of natural sources of NDAs, based upon a substituted piperidine nucleus, are important for continuing to provide leads in drug discovery. NDAs from Aconitum and Delphinium have complex, highly oxygenated hexacyclic systems, and as many of them are of pharmacological importance, their structures and 3D configuration are significant factors in their actions at various protein targets with respect to medicine and toxicology.
The majority of NDAs possess an N-Et group. We investigated the occurrence count in the SciFinder database for NDA skeletons, including many new NDAs. Substitution at position 1 is important to determine the NDA skeleton conformation, as ring A in 1-OMe NDA free bases exists in a twisted-chair conformation. In 1-OH NDA, ring A adopts a twisted-boat conformation. In conclusion, screening NDAs for their biological activity has resulted in the discovery of new sets of ligands. These are promising natural compounds that are pharmacologically active. These hits potentially eventually will become selective leads for the treatment of a wide variety of disease states. These NDAs are new natural products, and if they can be isolated from easy-to-grow Aconitum or Delphinium plants, then the future is bright for further NDA development based on experimental aspects, including phytochemistry leading to SAR studies and hopefully to new, selective, if not specific, drugs.

Funding

This research was funded by Zarqa University, Jordan, for the Studentship to A.M.A.Q.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

There are no conflict of interest to declare.

References

  1. Vetter, J. Poison hemlock (Conium maculatum L.). Food Chem. Toxicol. 2004, 42, 1373–1382. [Google Scholar] [CrossRef] [PubMed]
  2. Beers, W.H.; Reich, E. Structure and activity of acetylcholine. Nature 1970, 228, 917–922. [Google Scholar] [CrossRef] [PubMed]
  3. Schep, L.J.; Slaughter, R.J.; Beasley, D.M.G. Nicotinic plant poisoning. Clin. Toxicol. 2009, 47, 771–781. [Google Scholar] [CrossRef] [PubMed]
  4. Green, B.T.; Lee, S.T.; Welch, K.D.; Panter, K.E. Plant alkaloids that cause developmental defects through the disruption of cholinergic neurotransmission. Birth Defects Res. Part C Embryo Today Rev. 2013, 99, 235–246. [Google Scholar] [CrossRef]
  5. Green, B.T.; Welch, K.D.; Panter, K.E.; Lee, S.T. Plant toxins that affect nicotinic acetylcholine receptors: A review. Chem. Res. Toxicol. 2013, 26, 1129–1138. [Google Scholar] [CrossRef]
  6. Folquitto, D.G.; Swiech, J.N.D.; Pereira, C.B.; Bobek, V.B.; Halila Possagno, G.C.; Farago, P.V.; Miguel, M.D.; Duarte, J.L.; Miguel, O.G. Biological activity, phytochemistry and traditional uses of genus Lobelia (Campanulaceae): A systematic review. Fitoterapia 2019, 134, 23–38. [Google Scholar] [CrossRef]
  7. Felpin, F.-X.; Lebreton, J. History, chemistry and biology of alkaloids from Lobelia inflata. Tetrahedron 2004, 60, 10127–10153. [Google Scholar] [CrossRef]
  8. Keeler, R.F.; Panter, K.E. Piperidine alkaloid composition and relation to crooked calf disease-inducing potential of Lupinus formosus. Teratology 1989, 40, 423–432. [Google Scholar] [CrossRef]
  9. Green, B.T.; Lee, S.T.; Panter, K.E.; Welch, K.D.; Cook, D.; Pfister, J.A.; Kem, W.R. Actions of piperidine alkaloid teratogens at fetal nicotinic acetylcholine receptors. Neurotoxicol. Teratol. 2010, 32, 383–390. [Google Scholar] [CrossRef]
  10. James, L.F.; Panter, K.E.; Gaffield, W.; Molyneux, R.J. Biomedical applications of poisonous plant research. J. Agric. Food Chem. 2004, 52, 3211–3230. [Google Scholar] [CrossRef]
  11. Meghwal, M.; Goswami, T.K. Piper nigrum and piperine: An update. Phytother. Res. 2013, 27, 1121–1130. [Google Scholar] [CrossRef] [PubMed]
  12. Smilkov, K.; Ackova, D.G.; Cvetkovski, A.; Ruskovska, T.; Vidovic, B.; Atalay, M. Piperine: Old spice and new nutraceutical? Curr. Pharm. Des. 2019, 25, 1729–1739. [Google Scholar] [CrossRef] [PubMed]
  13. Shityakov, S.; Bigdelian, E.; Hussein, A.A.; Hussain, M.B.; Tripathi, Y.C.; Khan, M.U.; Shariati, M.A. Phytochemical and pharmacological attributes of piperine: A bioactive ingredient of black pepper. Eur. J. Med. Chem. 2019, 176, 149–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Butt, M.S.; Nazir, A.; Sultan, M.T.; Schroën, K. Morus alba L. Nature’s functional tonic. Trends Food Sci. Technol. 2008, 19, 505–512. [Google Scholar] [CrossRef]
  15. Zhang, W.; Mu, W.; Wu, H.; Liang, Z. An overview of the biological production of 1-deoxynojirimycin: Current status and future perspective. Appl. Microbiol. Biotechnol. 2019, 103, 9335–9344. [Google Scholar] [CrossRef]
  16. Kalant, H. Opium revisited: A brief review of its nature, composition, non-medical use and relative risks. Addiction 1997, 92, 267–277. [Google Scholar] [CrossRef]
  17. Frick, S.; Kramell, R.; Schmidt, J.; Fist, A.J.; Kutchan, T.M. Comparative qualitative and quantitative determination of alkaloids in narcotic and condiment Papaver somniferum cultivars. J. Nat. Prod. 2005, 68, 666–673. [Google Scholar] [CrossRef]
  18. Drewes, A.M.; Jensen, R.D.; Nielsen, L.M.; Droney, J.; Christrup, L.L.; Arendt-Nielsen, L.; Riley, J.; Dahan, A. Differences between opioids: Pharmacological, experimental, clinical and economical perspectives. Br. J. Clin. Pharmacol. 2013, 75, 60–78. [Google Scholar] [CrossRef] [Green Version]
  19. Daly, J.W. Nicotinic agonists, antagonists, and modulators from natural sources. Cell. Mol. Neurobiol. 2005, 25, 513–552. [Google Scholar] [CrossRef]
  20. Keith, L.H.; Pelletier, S.W. The C19-diterpene alkaloids. In Chemistry of The Alkaloids; Pelletier, S.W., Ed.; Van Nostrand Reinhold: New York, NY, USA, 1970; pp. 549–590. [Google Scholar]
  21. Dunstan, W.R.; Ince, W.H. Contributions to our knowledge of the aconite alkaloids. Part I. On the crystalline alkaloid of Aconitum napellus. J. Chem. Soc. Trans. 1891, 59, 271–287. [Google Scholar] [CrossRef]
  22. Ameri, A. The effects of Aconitum alkaloids on the central nervous system. Prog. Neurobiol. 1998, 56, 211–235. [Google Scholar] [CrossRef]
  23. Wang, F.P.; Chen, Q.H.; Liang, X.T. Chapter 1. The C18-Diterpenoid Alkaloids; Elsevier: Amsterdam, The Netherlands, 2009; Volume 67, ISBN 9780123747853. [Google Scholar]
  24. Sun, W.; Wang, H.; Zhang, S.; Wang, X. Crystal structure of lappaconitine-ium nitrate monohydrate, C32H47N3O12. Z. Krist.-New Cryst. Struct. 2014, 229, 485–487. [Google Scholar] [CrossRef]
  25. Hubschmann, V.F. Ueber Aconitum lycoctonum L. Schweiz. Wochenschr. Pharm. 1865, 3, 269–271. [Google Scholar]
  26. Dragendorff, V.G.; Spohn, H. Reviews and bibliographical notices. Am. J. Pharm. 1884, 8, 44. [Google Scholar] [CrossRef] [Green Version]
  27. Manske, R.H.F. An alkaloid from Delphinium brownii Rydb. Can. J. Res. 1938, 16, 57–60. [Google Scholar] [CrossRef]
  28. Goodson, J.A. The alkaloids of the seeds of Delphinium elatum L. J. Chem. Soc. 1943, 139–141. [Google Scholar] [CrossRef]
  29. Wonnacott, S.; Albuquerque, E.X.; Bertrand, D. MLA discriminates between nicotinic receptor subclasses. Methods Neurosci. 1993, 263–275. [Google Scholar]
  30. Manners, G.D.; Panter, K.E.; Pelletier, S.W. Structure-activity relationships of norditerpenoid alkaloids occurring in toxic larkspur (Delphinium) species. J. Nat. Prod. 1995, 58, 863–869. [Google Scholar] [CrossRef]
  31. Lakstygal, A.M.; Kolesnikova, T.O.; Khatsko, S.L.; Zabegalov, K.N.; Volgin, A.D.; Demin, K.A.; Shevyrin, V.A.; Wappler-Guzzetta, E.A.; Kalueff, A.V. Dark classics in chemical neuroscience: Atropine, scopolamine, and other anticholinergic deliriant hallucinogens. ACS Chem. Neurosci. 2019, 10, 2144–2159. [Google Scholar] [CrossRef]
  32. Krenzelok, E.P. Aspects of Datura poisoning and treatment. Clin. Toxicol. 2010, 48, 104–110. [Google Scholar] [CrossRef]
  33. Wang, F.-P.; Chen, Q.-H. The C19-diterpenoid alkaloids. In The Alkaloids. Chemistry and Biology; Elsevier: Amsterdam, The Netherlands, 2010; Volume 69, pp. 1–577. [Google Scholar]
  34. ITIS Report: Asteraceae. Available online: https://www.itis.gov/servlet/SingleRpt/SingleRpt?search_topic=TSN&search_value=35420#null (accessed on 14 September 2022).
  35. ITIS Report: Garrya. Available online: https://www.itis.gov/servlet/SingleRpt/SingleRpt?search_topic=TSN&search_value=27826#null (accessed on 14 September 2022).
  36. ITIS Report: Ranunculaceae. Available online: https://www.itis.gov/servlet/SingleRpt/SingleRpt?search_topic=TSN&search_value=18410#null (accessed on 14 September 2022).
  37. Christenhusz, M.J.M.; Byng, J.W. Phytotaxa. Phytotaxa 2016, 261, 201–217. [Google Scholar] [CrossRef] [Green Version]
  38. The Plant List: Delphinium. Available online: http://www.theplantlist.org/browse/A/Ranunculaceae/Delphinium/ (accessed on 14 September 2022).
  39. The Plant List: Aconitum. Available online: http://www.theplantlist.org/browse/A/Ranunculaceae/Aconitum/ (accessed on 14 September 2022).
  40. Hao, D.C.; Xiao, P.G.; Ma, H.Y.; Peng, Y.; He, C.N. Mining chemodiversity from biodiversity: Pharmacophylogeny of medicinal plants of Ranunculaceae. Chin. J. Nat. Med. 2015, 13, 507–520. [Google Scholar] [CrossRef]
  41. ITIS: Aconitum. Available online: https://www.itis.gov/servlet/SingleRpt/SingleRpt?search_topic=TSN&search_value=18411#null (accessed on 14 September 2022).
  42. ITIS Report: Delphinium. Available online: https://www.itis.gov/servlet/SingleRpt/SingleRpt?search_topic=TSN&search_value=18457#null (accessed on 14 September 2022).
  43. Bisset, N.G. Arrow poisons in China. Part II. Aconitum botany, chemistry, and pharmacology. J. Ethnopharmacol. 1981, 4, 247–336. [Google Scholar] [CrossRef]
  44. Blanché, C. Delphinium L. Subgen. Delphinium: Origin and evolutionary trends. Collect. Bot. 1990, 19, 75–96. [Google Scholar] [CrossRef] [Green Version]
  45. Kuder, R.C. Larkspurs, delphiniums, and chemistry. J. Chem. Educ. 1947, 24, 418–422. [Google Scholar] [CrossRef]
  46. Trifonova, V.I. Comparative biomorphological study of the taxonomy and phylogeny of the genera Consolida (DC.) S.F. Gray and Aconitella spach. Collect. Botánica 1990, 19, 97–110. [Google Scholar] [CrossRef] [Green Version]
  47. Jabbour, F.; Renner, S.S. A phylogeny of Delphinieae (Ranunculaceae) shows that Aconitum is nested within Delphinium and that Late Miocene transitions to long life cycles in the Himalayas and Southwest China coincide with bursts in diversification. Mol. Phylogenet. Evol. 2012, 62, 928–942. [Google Scholar] [CrossRef]
  48. Legro, R.A.H. Species hybrids in Delphinium. Euphytica 1961, 10, 1–23. [Google Scholar] [CrossRef]
  49. Lawrence, W.J.C. The origin of new forms in Delphinium. Genetica 1936, 18, 109–115. [Google Scholar] [CrossRef]
  50. Dalla Guda, C.; Cervelli, C.; Farina, C. Effects of lighting regimes on flowering of Delphinium hybrids ‘Pacific Giants’. In XXV International Horticultural Congress, Part 5: Culture Techniques with Special Emphasis on Environmental Implications; International Society for Horticultural Science: Leuven, Belgium, 1998; Volume 515, pp. 173–178. [Google Scholar]
  51. Nielsen, D.B.; Ralphs, M.H.; Evans, J.O.; Call, C.A. Economic feasibility of controlling tall Larkspur on rangelands. Rangel. Ecol. Manag. J. Range Manag. Arch. 1994, 47, 369–372. [Google Scholar] [CrossRef]
  52. 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]
  53. Cook, D.; Gardner, D.R.; Lee, S.T.; Stonecipher, C.A.; Pfister, J.A.; Welch, K.D.; Green, B.T. Two Delphinium ramosum chemotypes, their biogeographical distribution and potential toxicity. Biochem. Syst. Ecol. 2017, 75, 1–9. [Google Scholar] [CrossRef]
  54. Manners, G.D.; Panter, K.E.; Ralphs, M.H.; Pfister, J.A.; Olsen, J.D.; James, L.F. Toxicity and chemical phenology of norditerpenoid alkaloids in the Tall Larkspurs (Delphinium species). J. Agric. Food Chem. 1993, 41, 96–100. [Google Scholar] [CrossRef]
  55. Dobelis, P.; Madl, J.E.; Pfister, J.A.; Manners, G.D.; Walrond, J.P. Effects of Delphinium alkaloids on neuromuscular transmission. J. Pharmacol. Exp. Ther. 1999, 291, 538–546. [Google Scholar] [PubMed]
  56. Green, B.T.; Keele, J.W.; Gardner, D.R.; Welch, K.D.; Bennett, G.L.; Cook, D.; Pfister, J.A.; Davis, T.Z.; Stonecipher, C.A.; Lee, S.T.; et al. Sex-dependent differences for larkspur (Delphinium barbeyi) toxicosis in yearling Angus cattle. J. Anim. Sci. 2019, 97, 1424–1432. [Google Scholar] [CrossRef] [Green Version]
  57. Green, B.T.; Keele, J.W.; Bennett, G.L.; Gardner, D.R.; Stonecipher, C.A.; Cook, D.; Pfister, J.A. Animal and plant factors which affect larkspur toxicosis in cattle: Sex, age, breed, and plant chemotype. Toxicon 2019, 165, 31–39. [Google Scholar] [CrossRef]
  58. Welch, K.D.; Green, B.T.; Gardner, D.R.; Cook, D.; Pfister, J.A.; Panter, K.E. The effect of 7,8-methylenedioxylycoctonine-type diterpenoid alkaloids on the toxicity of tall larkspur (Delphinium spp.) in cattle. J. Anim. Sci. 2012, 90, 2394–2401. [Google Scholar] [CrossRef]
  59. Qasem, A.M.A.; Zeng, Z.; Rowan, M.G.; Blagbrough, I.S. Norditerpenoid alkaloids from Aconitum and Delphinium: Structural relevance in medicine, toxicology, and metabolism. Nat. Prod. Rep. 2022, 39, 460–473. [Google Scholar] [CrossRef]
  60. Zeng, Z.; Qasem, A.M.A.; Kociok-Köhn, G.; Rowan, M.G.; Blagbrough, I.S. The 1α-hydroxy-A-rings of norditerpenoid alkaloids are twisted-boat conformers. RSC Adv. 2020, 10, 18797–18805. [Google Scholar] [CrossRef]
  61. Zeng, Z.; Qasem, A.M.A.; Woodman, T.J.; Rowan, M.G.; Blagbrough, I.S. Impacts of steric compression, protonation, and intramolecular hydrogen bonding on the 15N NMR spectroscopy of norditerpenoid alkaloids and their piperidine-ring analogues. ACS Omega 2020, 5, 14116–14122. [Google Scholar] [CrossRef]
  62. Chen, L.; Shan, L.; Zhang, J.; Xu, W.; Wu, M.; Huang, S.; Zhou, X. Diterpenoid alkaloids from Aconitum soongaricum var. pubescens. Nat. Prod. Commun. 2015, 10, 2063–2065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yin, T.-P.; Cai, L.; Fang, H.-X.; Fang, Y.-S.; Li, Z.-J.; Ding, Z.-T. Diterpenoid alkaloids from Aconitum vilmorinianum. Phytochemistry 2015, 116, 314–319. [Google Scholar] [CrossRef] [PubMed]
  64. Qin, X.; Yang, S.; Zhao, Y.; Gao, Y.; Ren, F.; Zhang, D.; Wang, F. Five new C19-diterpenoid alkaloids from Aconitum carmichaeli. Phytochem. Lett. 2015, 13, 390–393. [Google Scholar] [CrossRef]
  65. Zhao, Q.; Gou, X.J.; Liu, W.; He, G.; Liang, L.; Chen, F.Z. Majusine D: A new C19-diterpenoid alkaloid from Delphinium majus. Nat. Prod. Commun. 2015, 10, 2069–2070. [Google Scholar] [CrossRef] [Green Version]
  66. Yin, T.P.; Cai, L.; Li, Y.; Fang, Y.S.; Peng, L.; Ding, Z.T. New alkaloids from Aconitum stapfianum. Nat. Prod. Bioprospecting 2015, 5, 271–275. [Google Scholar] [CrossRef] [Green Version]
  67. Zhao, B.; Usmanova, S.K.; Yili, A.; Kawuli, A.; Abdulla, R.; Aisa, H.A. New C19-norditerpenoid alkaloid from Delphinium shawurense. Chem. Nat. Compd. 2015, 51, 519–522. [Google Scholar] [CrossRef]
  68. Chen, C.L.; Tan, W.H.; Wang, Y.; Xue, Z.G.; Wan, C.P.; Yang, Z.Y.; Zhou, Z.H.; Ma, X.X. New norditerpenoid alkaloids from Aconitum vilmorinianum Komarov. J. Nat. Med. 2015, 69, 601–607. [Google Scholar] [CrossRef]
  69. Wada, K.; Chiba, R.; Kanazawa, R.; Matsuoka, K.; Suzuki, M.; Ikuta, M.; Goto, M.; Yamashita, H.; Lee, K.H. Six new norditerpenoid alkaloids from Delphinium elatum. Phytochem. Lett. 2015, 12, 79–83. [Google Scholar] [CrossRef]
  70. Shan, L.; Zhang, J.; Chen, L.; Wang, J.; Huang, S.; Zhou, X. Two new C18-diterpenoid alkaloids from Delphinium anthriscifolium. Nat. Prod. Commun. 2015, 10, 2067–2068. [Google Scholar] [CrossRef] [Green Version]
  71. Cai, L.; Fang, H.-X.; Yin, T.-P.; Yu, J.; Li, Z.-J.; Dong, J.-W.; Ding, Z.-T. Unusual C19-diterpenoid alkaloids from Aconitum vilmorinianum var. patentipilum. Phytochem. Lett. 2015, 14, 106–110. [Google Scholar] [CrossRef]
  72. Zhang, J.F.; Dai, R.Y.; Shan, L.H.; Chen, L.; Xu, L.; Wu, M.Y.; Wang, C.J.; Huang, S.; Zhou, X. Two new C19-diterpenoid alkaloids from Delphinium iliense. Phytochem. Lett. 2016, 17, 299–303. [Google Scholar] [CrossRef]
  73. Wada, K.; Asakawa, E.; Tosho, Y.; Nakata, A.; Hasegawa, Y.; Kaneda, K.; Goto, M.; Yamashita, H.; Lee, K.H. Four new diterpenoid alkaloids from Delphinium elatum. Phytochem. Lett. 2016, 17, 190–193. [Google Scholar] [CrossRef]
  74. Wang, F.; Yue, Z.; Xie, P.; Zhang, L.; Li, Z.; Song, B.; Tang, Z.; Song, X. C19-norditerpenoid alkaloids from Aconitum szechenyianum and their effects on LPS-activated NO production. Molecules 2016, 21, 4–11. [Google Scholar] [CrossRef] [Green Version]
  75. Lin, C.Z.; Liu, Z.J.; Bairi, Z.D.; Zhu, C.C. A new diterpenoid alkaloid isolated from Delphinium caeruleum. Chin. J. Nat. Med. 2017, 15, 45–48. [Google Scholar] [CrossRef]
  76. Chen, N.H.; Zhang, Y.B.; Li, W.; Li, P.; Chen, L.F.; Li, Y.L.; Li, G.Q.; Wang, G.C. Grandiflodines A and B, two novel diterpenoid alkaloids from Delphinium grandiflorum. RSC Adv. 2017, 7, 24129–24132. [Google Scholar] [CrossRef] [Green Version]
  77. 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]
  78. Li, G.Q.; Zhang, L.M.; Zhao, D.K.; Chen, Y.P.; Shen, Y. Two new C19-diterpenoid alkaloids from Aconitum tsaii. J. Asian Nat. Prod. Res. 2017, 19, 457–461. [Google Scholar] [CrossRef]
  79. Shan, L.H.; Zhang, J.F.; Gao, F.; Huang, S.; Zhou, X.L. Diterpenoid alkaloids from Delphinium anthriscifolium var. majus. Sci. Rep. 2017, 7, 6063. [Google Scholar] [CrossRef] [Green Version]
  80. Guo, R.; Guo, C.; He, D.; Zhao, D.; Shen, Y. Two new C19-diterpenoid alkaloids with anti-inflammatory activity from Aconitum iochanicum. Chin. J. Chem. 2017, 35, 1644–1647. [Google Scholar] [CrossRef]
  81. Liang, X.; Chen, L.; Song, L.; Fei, W.; He, M.; He, C.; Yin, Z. Diterpenoid alkaloids from the root of Aconitum sinchiangense W. T. Wang with their antitumor and antibacterial activities. Nat. Prod. Res. 2017, 31, 2016–2023. [Google Scholar] [CrossRef]
  82. Yang, L.; Zhang, Y.B.; Zhuang, L.; Li, T.; Chen, N.H.; Wu, Z.N.; Li, P.; Li, Y.L.; Wang, G.C. Diterpenoid alkaloids from Delphinium ajacis and their anti-RSV activities. Planta Med. 2017, 83, 111–116. [Google Scholar] [CrossRef] [PubMed]
  83. Meng, X.H.; Guo, Q.L.; Zhu, C.G.; Shi, J.G. Unprecedented C19-diterpenoid alkaloid glycosides from an aqueous extract of “FU ZI”: Neoline 14-O-L-arabinosides with four isomeric L-anabinosyls. Chin. Chem. Lett. 2017, 28, 1705–1710. [Google Scholar] [CrossRef]
  84. Nishiyama, Y.; Yokoshima, S.; Fukuyama, T. Synthesis of cardiopetaline via a Wagner—Meerwein rearrangement without preactivation of the pivotal hydroxy group. Org. Lett. 2017, 19, 5833–5835. [Google Scholar] [CrossRef] [PubMed]
  85. Kou, K.G.M.; Kulyk, S.; Marth, C.J.; Lee, J.C.; Doering, N.A.; Li, B.X.; Gallego, G.M.; Lebold, T.P.; Sarpong, R. A unifying synthesis approach to the C18-, C19-, and C20-diterpenoid alkaloids. J. Am. Chem. Soc. 2017, 139, 13882–13896. [Google Scholar] [CrossRef] [PubMed]
  86. Liu, X.-Y.; Qin, Y. Enabling syntheses of diterpenoid alkaloids and related diterpenes by an oxidative dearomatization/Diels-Alder cycloaddition strategy. Nat. Prod. Rep. 2017, 34, 1044–1050. [Google Scholar] [CrossRef]
  87. Yang, X.; Cheng, B.; Cheng, H.; Xu, L.; Wang, J.L. Rapid construction of the unique BCD ring system of tricyclo [6.2.1.0] undecane in the C19-diterpenoid alkaloid aconitine. Chin. Chem. Lett. 2017, 28, 1788–1792. [Google Scholar] [CrossRef]
  88. Shan, L.H.; Zhang, J.F.; Gao, F.; Huang, S.; Zhou, X.L. C18-Diterpenoid alkaloids from Delphinium anthriscifolium var. majus. J. Asian Nat. Prod. Res. 2018, 20, 423–430. [Google Scholar] [CrossRef]
  89. Song, B.; Jin, B.; Li, Y.; Wang, F.; Yang, Y.; Cui, Y.; Song, X.; Yue, Z.; Liu, J. C19-Norditerpenoid alkaloids from Aconitum szechenyianum. Molecules 2018, 23, 1108. [Google Scholar] [CrossRef] [Green Version]
  90. Yamashita, H.; Katoh, M.; Kokubun, A.; Uchimura, A.; Mikami, S.; Takeuchi, A.; Kaneda, K.; Suzuki, Y.; Mizukami, M.; Goto, M.; et al. Four new C19-diterpenoid alkaloids from Delphinium elatum. Phytochem. Lett. 2018, 24, 6–9. [Google Scholar] [CrossRef]
  91. Yamashita, H.; Takeda, K.; Haraguchi, M.; Abe, Y.; Kuwahara, N.; Suzuki, S.; Terui, A.; Masaka, T.; Munakata, N.; Uchida, M.; et al. Four new diterpenoid alkaloids from Aconitum japonicum subsp. subcuneatum. J. Nat. Med. 2018, 72, 230–237. [Google Scholar] [CrossRef]
  92. Li, Y.; Gao, F.; Zhang, J.F.; Zhou, X.L. Four new diterpenoid alkaloids from the roots of Aconitum carmichaelii. Chem. Biodivers. 2018, 15, e1800147. [Google Scholar] [CrossRef] [PubMed]
  93. Yin, T.; Hu, X.; Mei, R.; Shu, Y.; Gan, D.; Cai, L.; Ding, Z. Four new diterpenoid alkaloids with anti-inflammatory activities from Aconitum taronense Fletcher et Lauener. Phytochem. Lett. 2018, 25, 152–155. [Google Scholar] [CrossRef]
  94. Xue, W.J.; Zhao, B.; Ruzi, Z.; Zhao, J.Y.; Aisa, H.A. Norditerpenoid alkaloids from Delphinium pseudoaemulans C. Y. Yang et B. Wang. Phytochemistry 2018, 156, 234–240. [Google Scholar] [CrossRef] [PubMed]
  95. Ahmad, H.; Ahmad, S.; Ali, M.; Latif, A.; Shah, S.A.A.; Naz, H.; Ur Rahman, N.; Shaheen, F.; Wadood, A.; Khan, H.U.; et al. Norditerpenoid alkaloids of Delphinium denudatum as cholinesterase inhibitors. Bioorg. Chem. 2018, 78, 427–435. [Google Scholar] [CrossRef]
  96. 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]
  97. Shu, Y.; Yin, T.P.; Wang, J.P.; Gan, D.; Zhang, Q.Y.; Cai, L.; Ding, Z.T. Three new diterpenoid alkaloids isolated from Aconitum brevicalcaratum. Chin. J. Nat. Med. 2018, 16, 866–870. [Google Scholar] [CrossRef]
  98. Ablajan, N.; Zhao, B.; Zhao, J.Y.; Kodirova, D.R.; Sagdullaev, S.S.; Aisa, H.A. Alkaloids from Delphinium aemulans. Chem. Nat. Compd. 2020, 56, 836–839. [Google Scholar] [CrossRef]
  99. Yin, T.; Shu, Y.; Zhou, H.; Cai, L.; Ding, Z. Nagarines A and B, two novel 8,15-seco diterpenoid alkaloids from Aconitum nagarum. Fitoterapia 2019, 135, 1–4. [Google Scholar] [CrossRef]
  100. Liu, M.; Cheng, C.; Xiong, W.; Cheng, H.; Wang, J.L.; Xu, L. Total synthesis of C19-diterpenoid alkaloid: Construction of a functionalized ABCDE-ring system. Org. Chem. Front. 2018, 5, 1502–1505. [Google Scholar] [CrossRef]
  101. Doering, N.A.; Kou, K.G.M.; Norseeda, K.; Lee, J.C.; Marth, C.J.; Gallego, G.M.; Sarpong, R. A copper-mediated conjugate addition approach to analogues of aconitine-type diterpenoid alkaloids. J. Org. Chem. 2018, 83, 12911–12920. [Google Scholar] [CrossRef]
  102. Zhou, X.H.; Liu, Y.; Zhou, R.J.; Song, H.; Liu, X.Y.; Qin, Y. Construction of the highly oxidized bicyclo [3.2.1] octane CD ring system of aconitine via a late stage enyne cycloisomerization. Chem. Commun. 2018, 54, 12258–12261. [Google Scholar] [CrossRef] [PubMed]
  103. Lv, Z.; Gao, L.; Cheng, C.; Niu, W.; Wang, J.L.; Xu, L. Expedient construction of the hexacycle of franchetine. Chem. Asian J. 2018, 13, 955–958. [Google Scholar] [CrossRef] [PubMed]
  104. Zhou, R.J.; Dai, G.Y.; Zhou, X.H.; Zhang, M.J.; Wu, P.Z.; Zhang, D.; Song, H.; Liu, X.Y.; Qin, Y. Progress towards the synthesis of aconitine: Construction of the AE fragment and attempts to access the pentacyclic core. Org. Chem. Front. 2019, 6, 377–382. [Google Scholar] [CrossRef]
  105. Kamakura, D.; Todoroki, H.; Urabe, D.; Hagiwara, K.; Inoue, M. Total synthesis of talatisamine. Angew. Chem. Int. Ed. 2020, 59, 479–486. [Google Scholar] [CrossRef]
  106. Xiao, D.; Zhao, X.; Lei, J.; Zhu, M.; Xu, L. Synthesis of [6-6-6] ABE tricyclic ring analogues of methyllycaconitine. Chin. Chem. Lett. 2021, 32, 3031–3033. [Google Scholar] [CrossRef]
  107. Cao, S.Y.; Yue, H.J.; Zhu, M.Q.; Xu, L. Synthesis of tricyclo [7.2.1.09,10] dodecan-11-one core ring systems of norditerpenoid alkaloids and racemulosine. Org. Chem. Front. 2020, 7, 933–937. [Google Scholar] [CrossRef]
Figure 1. Piperidine alkaloids from plants.
Figure 1. Piperidine alkaloids from plants.
Ijms 23 12128 g001
Scheme 1. Scientific classification of Aconitum, Delphinium, and Consolida from the Plantae kingdom.
Scheme 1. Scientific classification of Aconitum, Delphinium, and Consolida from the Plantae kingdom.
Ijms 23 12128 sch001
Figure 2. NDAs of lycoctonine (20), MSAL (16), and MDL (21) types.
Figure 2. NDAs of lycoctonine (20), MSAL (16), and MDL (21) types.
Ijms 23 12128 g002
Figure 3. NDAs 2226.
Figure 3. NDAs 2226.
Ijms 23 12128 g003
Figure 4. NDAs 2743.
Figure 4. NDAs 2743.
Ijms 23 12128 g004
Figure 5. NDAs 4448.
Figure 5. NDAs 4448.
Ijms 23 12128 g005
Figure 6. NDAs 4957.
Figure 6. NDAs 4957.
Ijms 23 12128 g006
Figure 7. NDAs 5864.
Figure 7. NDAs 5864.
Ijms 23 12128 g007
Figure 8. NDAs 65–70.
Figure 8. NDAs 65–70.
Ijms 23 12128 g008
Figure 9. NDAs 7173.
Figure 9. NDAs 7173.
Ijms 23 12128 g009
Figure 10. NDAs 7478.
Figure 10. NDAs 7478.
Ijms 23 12128 g010
Figure 11. Glycosidic NDAs 7982.
Figure 11. Glycosidic NDAs 7982.
Ijms 23 12128 g011
Figure 12. NDAs and their construction 8391.
Figure 12. NDAs and their construction 8391.
Ijms 23 12128 g012
Figure 13. NDAs 9294.
Figure 13. NDAs 9294.
Ijms 23 12128 g013
Figure 14. NDAs 9598.
Figure 14. NDAs 9598.
Ijms 23 12128 g014
Figure 15. NDAs 99106.
Figure 15. NDAs 99106.
Ijms 23 12128 g015
Figure 16. NDAs 107110.
Figure 16. NDAs 107110.
Ijms 23 12128 g016
Figure 17. NDAs 111–118.
Figure 17. NDAs 111–118.
Ijms 23 12128 g017
Figure 18. NDAs 119124.
Figure 18. NDAs 119124.
Ijms 23 12128 g018
Figure 19. NDAs 125127.
Figure 19. NDAs 125127.
Ijms 23 12128 g019
Figure 20. NDAs 128 and 129.
Figure 20. NDAs 128 and 129.
Ijms 23 12128 g020
Table 1. NDA skeleton occurrence count in the SciFinder database.
Table 1. NDA skeleton occurrence count in the SciFinder database.
Skeleton +Number of Compounds ++
Ijms 23 12128 i0012728
Ijms 23 12128 i002436
Ijms 23 12128 i003160
Ijms 23 12128 i0042598
Ijms 23 12128 i005427
Ijms 23 12128 i006128
Ijms 23 12128 i00778
Ijms 23 12128 i0085
Ijms 23 12128 i00913
Ijms 23 12128 i01013
Ijms 23 12128 i0114
Ijms 23 12128 i01267
Ijms 23 12128 i0130
Ijms 23 12128 i0140
Ijms 23 12128 i0150
Ijms 23 12128 i0160
Ijms 23 12128 i0170
Ijms 23 12128 i01836
Ijms 23 12128 i0193002
Ijms 23 12128 i020392
Ijms 23 12128 i02110 (all are N-Et)
Ijms 23 12128 i0220
Ijms 23 12128 i02324 (3 N-Pr, 1 N-Me, and 20 N-Et)
Ijms 23 12128 i0241 (N-Et)
Ijms 23 12128 i0255 (N-Et)
Ijms 23 12128 i0260
Ijms 23 12128 i0270
Ijms 23 12128 i0282
Ijms 23 12128 i0293
Ijms 23 12128 i0302
Ijms 23 12128 i0312
+ R = methyl (Me), ethyl (Et), propyl (Pr); ++ number of compounds includes the free base form and the salt form.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Qasem, A.M.A.; Rowan, M.G.; Blagbrough, I.S. Poisonous Piperidine Plants and the Biodiversity of Norditerpenoid Alkaloids for Leads in Drug Discovery: Experimental Aspects. Int. J. Mol. Sci. 2022, 23, 12128. https://doi.org/10.3390/ijms232012128

AMA Style

Qasem AMA, Rowan MG, Blagbrough IS. Poisonous Piperidine Plants and the Biodiversity of Norditerpenoid Alkaloids for Leads in Drug Discovery: Experimental Aspects. International Journal of Molecular Sciences. 2022; 23(20):12128. https://doi.org/10.3390/ijms232012128

Chicago/Turabian Style

Qasem, Ashraf M. A., Michael G. Rowan, and Ian S. Blagbrough. 2022. "Poisonous Piperidine Plants and the Biodiversity of Norditerpenoid Alkaloids for Leads in Drug Discovery: Experimental Aspects" International Journal of Molecular Sciences 23, no. 20: 12128. https://doi.org/10.3390/ijms232012128

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