Abstract
The genus Aconitum belongs to the family Ranunculaceae, is endowed with more than 350 species on the earth. Medicinally important aconitine type of diterpenoid alkaloids are the characteristic compounds in most of the Aconitum species. The present review endeavored the major research carried out in the field of genetic resource characterization, pharmacological properties, phytochemistry, major factors influencing quantity, biosynthetic pathways and processing methods for recovery of active ingredients, variety improvement, propagation methods, and important metabolite production through cell/organ culture of various Aconitum species. More than 450 derivatives of aconitine-type C19 and C20-diterpenoid alkaloids along with a few other non-alkaloidal compounds, such as phenylpropanoids, flavonoids, terpenoids, and fatty acids, have been identified in the genus. A few Aconitum species and their common diterpenoid alkaloid compounds are also well characterized for analgesic, inflammatory and cytotoxic properties. However, the different isolated compound needs to be validated for supporting other traditional therapeutical uses of the plant species. Aconitine alkaloids shared common biosynthesis pathway, but their diversification mechanism remains unexplored in the genus. Furthermore, the process needs to be developed on secondary metabolite recovery, mass-scale propagation methods, and agro-technologies for maintaining the quality of products. Many species are losing their existence in nature due to over-exploitation or anthropogenic factors; thus, temporal monitoring of the population status in its habitat, and suitable management programs for ascertaining conservation needs to be developed.
Keywords: Aconite, Monkshood, Diterpene alkaloids, Ethnopharmacology, Omics studies, Tissue culture
Introduction
The genus Aconitum (Ranunculaceae family) is one of the most recognized medicinal plants representing over 350 species distributed majorly in temperate regions of the Northern Hemisphere (Been 1992). Aconitum is a perennial (sometimes biennial) herb having stout leafy stems, bulbs, or rhizomes (Jabeen et al. 2013). East and South-East Asia and Central European represent a rich diversity of Aconitum species; however, a few species have also been recorded from Western and Eastern parts of North America. Among Asian countries, over 200 species have been recorded from China (Li and Kadota 2001) of which more than 76 species are used for medicinal purposes in various parts of the country (Xiao et al. 2006). The Hengduan Mountain region has been recognized as a primary center of diversity and speciation of the genus Aconitum, which is represented by a total of 166 recorded species (Yan and Qin‐Er 2005). A total of 33 species are found in the Himalayan region spanning India, Pakistan, Nepal, Bhutan, and Tibet (Dar and Nordenstam 2014), of which 26 species are reported from Kashmir Himalaya (Jabeen et al. 2013), and have been recognized in local and traditional system of medicine of the region. However, 94 taxa, including 22 native species and 28 nothospecies and a few intra-specific natural hybrids, have been recorded from Europe (Mitka et al. 2021). A total of 6 species of Aconitum (e.g., A. columbianum, A. delphiniifolium, A. maximum, A. noveboracense, A. reclinatum Gray., and A. uncinatum) have been reported from North America (Brink et al. 1994; Kuchenreuther 1996). Different Aconitum species found in Asian countries have diverse medical uses and therapeutical efficacy. Indian species, A. ferox and A. chasmanthum, were reported three-to-seven times more effective than European species A. napellus. Aconitum has been used as a powerful poison since ancient times and was used in arrows and bait poison (Been 1992). Aconitum species have an important place in the Asian herbal medicine system which is evident from its larger applications in the cardio and neurological disorders in India, China, Japan, and some other Asian countries; however, wrong species identification or improper processing may cause a high risk of severe toxicity (Chan 2014). Its major medicinal application has been recognized as anti-rheumatic, anesthetics, analgesic, diaphoretic, diuretic, anti-periodic anodyne, anti-diabetic, anti-phlogistic, and anti-pyretic agent in different traditional systems of medicine of Asia (Dar and Naqshi 2001; Shah 2005; Yue et al. 2009; Yan et al. 2010; Nyirimigabo et al. 2015).
Proper processing reduces the toxicity of Aconitum and sometimes enhanced its efficacy (Liu et al. 2017). Detoxified tubers and roots are only permitted for oral administration for clinical decoctions after proper processing by the Food and Drug Administration of China. Currently, more than 70 processing techniques are available for detoxification in the Chinese traditional system of medicine (Nyirimigabo et al. 2015). Thus, various species are used for the treatment of rheumatoid arthritis, various types of pain, inflammatory diseases, trauma and fractures, intoxication, plaque, immune-suppression-induced ailments, colds, and coughs (Hao et al. 2015). The characteristic aconite-type diterpenoid alkaloids found in Aconitum species have a strong medicinal application in the modern pharmaceutical market. An array of derivatives of diterpenoid alkaloids present in different species has similar or sometimes different biological activity with diverse biological efficiency (Bessonova and Saidkhodzhaeva 2000; Shen et al. 2020). Owing to their pharmacological values, diterpenoid alkaloids in Aconitum have attracted the attention of researchers. Besides, various other phytochemicals, e.g., flavonoids, phenylpropanoids, phenolic acids, terpenoids, and steroids, have been isolated from Aconitum. In different pharmacological activities, analgesic and anti-inflammatory activities have been well explored in extracts as well as isolated diterpenoid alkaloids.
Due to high demand in the international market, A. spicatum is among the top ten highly traded medicinal plants from the Nepal Himalaya (Olsen and Larsen 2003). A. heterophyllum, an endangered medicinal plant, has an average price of 50 USD per kg and market demand has been estimated at approximately 2000 tons per annum, while its availability is decreasing in the range of 26–50% annually due to a continuous decrease in its wild stock (Olsen and Larsen 2003; Gupta and Souravi 2020). Similarly, A. carmichaelii Debx. and A. kusnezoffii Reichb. have a huge market in China, as these species are used in medicinal and well as traditional culinary recipes (Brinckmann 2016). Increasing global trade of different Aconitum species has imposed a heavy demand for the supply of raw material leading to unscientific and indiscriminate harvesting of many species from the wild. Due to the rapid reduction of wild populations, a few species of Aconitum, (A. chasmanthum, A. heterophyllum, A. violaceum, and A. corsicum) have been placed in different threat categories of IUCN (IUCN 2020). Due to the availability of large diversity of genetic resources in nature, beneficial therapeutical effects, and the presence of a large array of compounds, Aconitum genus has attracted the attention of researchers in medical, pharmaceutical, agriculture, and environmental sciences. Being a factory of diverse potent diterpenoid alkaloids, the genus can be a model plant for the study of diterpenoid alkaloid biosynthesis.
Although various review papers have already been published on mass propagation methods, pharmacological, and phytochemical aspects in this genus (Srivastava et al. 2010; Nyirimigabo et al. 2015; Kumari et al. 2017; Ali et al. 2021; Wani et al. 2022), the present review highlighted recent research on these aspects along with a few additional research areas, including best growth influencing factors for quantity control, optimum recovery methods for secondary metabolite, and transcriptome studies for elucidation of major biosynthetic pathways of active ingredients. Also, available information on diversity in genetic resources (including genetic diversity studies and phylogenetics) and genomic resources along with full plastid genome sequencing studies has been systematically analyzed for improving the quality and quantity of active ingredients. Research gaps have also been highlighted for crop improvement programs and conservation and sustainable utilization of the genus Aconitum.
Medicinal uses and pharmacological properties
Medicinal uses
Aconitum has been traditionally used in different traditional medicine systems, including, Ayurveda, Unani, Chinese medicine system, and Homeopathy. The species has been used in treating different types of pains and inflammations, anxiety, neurological disorders, gastrointestinal disorders, edema, bronchial asthma, rheumatic fever, and some endocrinal disorders (Dar and Naqshi 2001; Nyirimigabo et al. 2015). In the traditional Indian medicine system Ayurveda, Aconitum species are used as anti-pyretic, anti-inflammatory, anti-hemorrhoid, antidotes, anti-tussive, and anti-diarrheal agents (Adams et al. 2013). Roots of Himalayan species, such as A. balfourii, A. ferox, and A. heterophyllum, are used in the treatment of vomiting, diarrhea, abdominal pain, body ache, and arthritis in India (Bisht et al. 2013). Roots of A. heterophyllum are also used as a remedy for the nervous system, digestive system, cough, cold, fever, and rheumatism (Shah 2005). However, the roots of A. spicatum have extensively been used to treat fever, headache, muscular pain, cuts, and wounds in Nepal and other parts of the Himalaya (Shyaula 2011). A. delavayi Franch. roots are used to treat rheumatism, traumatic injuries, pain, swelling, hematemesis, blood disease, hematochezia, and piles (Jiang et al. 2007). Roots of A. deinorrhizumare are effective against headache and rheumatic fever (Pullaiah 2006). However, a fermented preparation of A. violaceum flowers is useful in cough, fever, colds, stomach, and liver disorders, and preparations of root powder are effective in painful joints and boils, toothache, and preventing tooth cavities (Fig. 1) (Lone et al. 2014). Similarly, A. bulleyanumis traditionally used in influenza, rashes, and snake bite. A. orochryseum Stapf is used in bilious fever, dysentery, cough and cold, and as a febrifuge for malarial fever, stomach ulceration, and kidney dysfunction (Wangchuk et al. 2010). A. carmichaelii Debx, a species found in East Asia and eastern Russia, has been known for its anti-inflammatory, analgesic, diuretics, and cardio-tonic properties (Yan et al. 2010; Yue et al. 2009). A. brachypodum, one of the commonly used Chinese traditional herbs, has anti-rheumatic and analgesic properties. A. napellus L. a native and endemic species of Western and Central Europe is used in homeopathic preparations (Watad et al. 1995). A.kusnezoffii Rchb. found in Eastern China, Serbia, and Korean peninsula is used as an analgesic and anti-rheumatic agent for heart gout, neuralgia, failure congestion, and rheumatism (Zhao et al. 2009). A. coreanum has rheumatic arthralgia, anti-arrhythmia, analgesic, and anti-inflammatory properties to cure migraine headache, cardialgia, vertigo, epilepsy, and infantile convulsion (Park et al. 2017a, b). Similarly, A. taipeicum Hand-Mzt, A. finetianum Hand-Mazz, A. sungpanense hand. Mazz, A. vulparia Rchb, A. naviculare (Bru¨hl) Stapf, A. kirinense Nakai, and many other species have various health-beneficial effects and are used in different parts of the world (Wu et al. 1996; Fico et al. 2003; Wang et al. 2004; Jiang et al. 2007; Shrestha and Jha 2010; Zhang et al. 2013; Zhang et al. 2014).
Fig. 1.
Pharmacological properties of various species of Aconitum validated in various studies
Pharmacological properties
Anti-inflammatory potential
Different Aconitum species have been used as anti-inflammatory and analgesic agents in different systems of medicines since ancient times (Huang et al. 2011). The ethanolic root extract of A. heterophyllum has been reported to inhibit inflammation by reducing the weight of cotton pellet in cotton pellet-induced granuloma in rats and the activity was comparable to diclofenac sodium, which is a strong non-steroidal anti-inflammatory drug (Verma et al. 2010). Aconitum roots (Pendulous monkshood) displayed significant anti-inflammatory properties on lipopolysaccharides-induced mouse peritoneal macrophages at the dosage of 4–200 µg/ml body weight (Huang et al. 2013). Similarly, A. vilmorinianum Kom. exhibited improvement in swelling, hyperemia, allodynia, and vascular permeability in arthritic knee joints (Li et al. 2013). Butanol and ethanol extract of A. flavum possesses significant anti-inflammatory activity in dimethylbenzene-induced ear vasodilatation and acetic acid-induced capillary permeability enhancement in mice and carrageenan-induced paw edema in rats. The plant extracts showed significant anti-nociceptive activity using acetic acid-induced writhes, hot-plate test, and formalin test in mice, and it did not show any cytotoxicity at the tested dose (Zhang et al. 2015). Different plant parts of A. carmichaelii have shown analgesia and anti-inflammatory, and were found useful in the retreatment of rheumatoid arthritis and these activities showed no significant difference among leaves and stem parts used (Ya-Nan et al. 2018). Oral administration of the ethanolic extract of A. napellus L. (200 mg/kg) showed potent anti-inflammatory activity by inhibiting the acute as well as chronic inflammation adjuvant carrageenan-induced inflammation in rats (Gupta et al. 2019). Chloroform extract of A. sinomontanum showed significant anti-nociceptive and anti-inflammatory activities by improving the alleviated pain induced by acetic acid and significantly reducing the xylene-induced mouse ear edema (Zhang et al. 2020). Similarly, extracts of A. tanguticum (Maxim.) Stapf have displayed potential anti-inflammatory effects in rats (Wu et al. 2014).
Guiwuline, a franchetine type C19-diterpenoid alkaloid isolated from A. carmichaelii, showed potential analgesic activity in mouse hot-plate test and showed little toxicity to mice (Wang et al. 2012). Four new C19-diterpenoid alkaloids, taronenines A–D, isolated from the roots of A. taronense showed inhibition activities of interleukin-6 in LPS-activated RAW 264.7 cells with IC50 values between 18.87 and 29.60 μg/mL (Yin et al. 2018). Diterpene alkaloids isolated from A. baikalense (e.g., napelline, songorine, hypaconitine, mesaconitine, and 12-epinapelline N-oxide) improved histamine-induced acute inflammation condition by reducing swelling (more than 20%) of the injured limb. However, the reference drug diclofenac sodium salt exhibited 1.5 times higher activity than these isolated compounds (Nesterova et al. 2014). Two new diterpenoid alkaloids, 7,8-epoxy-franchetine and N-(19)-en-austroconitine A, isolated from A. iochanicum exhibited inhibitory activity against nitric oxide (NO) production in LPS-activated RAW264.7 macrophages. The anti-inflammatory activity of these compounds was ~ 25% weaker than diclofenac sodium drug (Guo et al. 2017). Also, paconitine and puberanine alkaloid isolated from the aerial parts of the leaves of Aconitum Royle. exhibited significant anti-inflammatory activity by decreasing the neutrophil accumulation in inflammatory regions (Shaheen et al. 2005). Aconitine, Songorine, 16,17‑dihydro‑12β, 16β‑epoxynapelline, and 12‑epi‑napelline application significantly improved the lipo-polysaccharide-induced cellular inflammatory responses improving the content of IL-6, IL-1β, TNF-α, and PGE-2 (Zhang et al. 2021a). A vakognavine-type alkaloid found in different Aconitum species inhibited the activity of cyclooxygenase-2, which was found comparable to aspirin (Liang et al. 2016). Lappaconitine also exhibited significant analgesic properties by inhibiting the voltage-dependent sodium channels, increasing epinephrine release in synaptic cleft, and inhibiting the release of P substance (Li et al. 2019).
Besides, diterpenoid alkaloids, four caffeoyl derivatives, caffeic acid, 4,5-dicaffeoylquinic acid, 3,5-dicaffeoylquinic acid, and 3,5-dicaffeoylquinic acid methyl ester isolated with activity guided fractionation process of A. koreanum root extract exhibited strong anti-inflammatory effects by inhibiting nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells and inhibited the expressions of iNOS and COX-2 related genes at mRNA levels in a dose-dependent manner (Park et al. 2009). A brief mechanism of the inflammatory response of aconitine alkaloid is summarized in Fig. 2.
Fig. 2.
The molecular mechanisms of anti-inflammatory response of aconitine alkaloid (IKKα IκB kinase-α, IKKβ IκB kinase-α, IκBα nuclear factor of kappa light polypeptide gene enhancer in B-cell inhibitor-α, NF-κB Nuclear factor kappa-light-chain-enhancer of activated B cells, P65, P65 protein of transcription factor, TNF-α tumor necrosis factor-α, IL-6 interleukin-6, IL-1 interleukin-1, COX-2 cyclooxygenase-2, PGE2 Prostaglandin E2, iNOS inducible nitric oxide synthase, NOS nitric oxide synthase). Red arrow (↓) indited inhibition (Lan et al. 2021; Li et al. 2022)
Anti‑cancer and anti‑proliferative properties
Aconitum plant extracts exhibit significant anticancer and cytotoxic properties by inhibiting the growth of different cancer cell lines. Lipopolysaccharide-rich aqueous fraction of dried tuberous roots of A. austrokoreense Koidz significantly inhibited nitric oxide production induced in RAW 264.7 cell lines and inhibited cytotoxic activity against human cancer cell lines (Ju et al. 2020). Ethanolic extract of Aconitum roots (Pendulous monkshood) inhibited the proliferation of HepG 2 cells, sP 2/0 cells, and Hela cells in a dose-dependent manner (Huang et al. 2013). The aqueous solution of aconitine-containing extract administered to mice having ascites or a solid form of Ehrlich’s carcinoma showed effectiveness against growth tumor growth and the average life span of experimental animals (Dasyukevich and Solyanik 2007). Similarly, aconitine-containing herbal extract significantly inhibits the growth of tumors and metastasis of Lewis lung carcoma (LLC-R9) cell lines (Solyanik et al. 2004). Oral administration of crude polysaccharides’ extraction of A. coreanum stems inhibits the growth of H22 tumor cells and prolonged the life span of H22 ascites tumor-bearing mice (Liang et al. 2012).
A new vakognavine-type alkaloid displayed strong inhibitory effects on HT-29, SGC-7901 and HepG2 cell lines with IC50 values between 0.948 and 3.645 μM (Liang et al. 2016). Taipeinine alkaloids found in various species of Aconitum blocked the cell cycle at G1/S phase and exhibited its usefulness in the inhibition of HepG2 cell proliferation (Zhang et al. 2014). Also, veatchine-type C20-diterpenoid alkaloids including 12-acetylluciculine and its derivatives isolated from A. japonicum inhibited the growth of human malignant A172 cells (Wada et al. 2007). Lappaconitine, a C19-diterpenoid alkaloid found in the roots of Aconitum pseudolaeve, inhibits the proliferation of the human lung cancer cells A549. At a higher concentration of lappaconitine, apoptosis rate increased with the down-regulated expression of Cyclin E1A in 549 cells along with an increase in G1 + G0 phase and a decrease in S and G2 + M phases (Sheng et al. 2014). Lycaconitine also has potent inhibitory effects on human fibro-carcinoma KB V20C, which was resistant to 20 nM vincristine, but it was found non-cytotoxic to NK cells (Kim et al. 1998). Five compounds (e.g., oxonitine, deoxyaconitine, hypaconitine, mesaconitine, and crassicauline A) were isolated from the roots of A. carmichaelii Debx. showed significant cytotoxic activities against various cancer cell lines belonging to leucocythemia, breast cancer, and liver cancer. Compounds having two ester groups contained in the structure possess a stronger effect than others (Gao et al. 2012). Also, 8-O-azeloyl-14-benzoylaconine isolated from the roots of A. karacolicum Rapcs. has two ester groups that exhibited significant anti-proliferative activities against cell lines of the colon (HCT-15), lung (A549), and breast (MCF-7) cancer (Chodoeva et al. 2005). Ten new acylated alkaloid derivatives prepared from the natural diterpenoid alkaloids of A. yesoense and A. japonicum inhibited the growth of A549 cells by arresting the cell cycle at G1 stage. Further, cytotoxic properties showed a significant increase after the addition of an acyl group at both C-11 and C-15 positions (Wada et al. 2011).
Effects on the central nervous system
Aconitum has been traditionally considered an analgesic agent and it significantly reduced neuropathic pain in the rat chronic constriction injury model (Xu et al. 2006). The diterpenoid alkaloids present in Aconitum exhibit potent N-cholinolytic activity and induced signals to neuronal nicotinic acetylcholine receptors (nAChRs) (Turabekova et al. 2010). Furthermore, Bullatine A, a diterpenoid alkaloid found in many species of Aconitum, can attenuate ATP-induced BV-2 microglia apoptosis through P2X receptor pathways (Li et al. 2013). Diterpene alkaloids often displayed anti-epileptic properties by guarding the Na+ channels as Na+ channels are involved at the beginning of epilepsy. Lappaconitine normalizes neuronal activity by blocking Na+ channels (Ameri 1998a). A few structure–relationship studies indicated that aromatic substitutes of different compounds have better anti-epileptic activity. Aromatic substitutes of aconitine-type diterpene alkaloids, such as 6-benzoylheteratisine, 1-benzoylnapelline, lappaconitine, and 14 benzoyl-talatisamine, exhibited better hippocampal excitability in rat model system than many commercially available compounds, such as heteratisine, napelline, lappaconidine, and talatisamine (Ameri 1997, 1998b). However, aconitine alkaloids can also suppress delayed rectifier K+ current in NG108-15 neuronal cells. However, alterations in action potentials caused by aconitine might be concerned with abnormal neuronal excitability (Lin et al. 2008).
Anti‑arrhythmic effect
Guan-Fu base S (a diterpenoid alkaloid) isolated from A. coreanum exhibited inhibitory effects on blocking the ventricular-specific sodium current using a whole-cell patch voltage-clamp technique (Xing et al. 2014). Alkaloids isolated from A. carmichaelii species exhibited strong anti-arrhythmogenic properties (Liu et al. 2012). Similarly, allapinin isolated from different Aconitum species prevented paroxysmal atrial fibrillations and hence exhibited strong application as an anti-arrhythmic drug (Mazur et al. 1986).
Anti‑diabetic and hypolipidemic effects
Different solvent extracts of A. pseudolaeve var erectum root showed over 90% of α-amylase inhibition and over 60% of α-glucosidase inhibition, which is an important mechanism to regulate glucose uptake from the diet (Kim et al. 2013). Ethanolic extract of A. ferox showed strong inhibitory activity against α-glucosidase and improved the blood level of Alloxan-induced diabetic rats (Shoaib et al. 2020). Aqueous extract of A. napellus exhibited promising hypoglycaemic activity comparable to reference standard glibenclamide to reduce blood glucose levels in alloxan-induced hyperglycaemic adult Wistar albino rats (Chhetree et al. 2010). Also, A. carmichaelii extract appeared more effective in insulin immune reactivity than the control and treated group after oral administration on blood sugar in Type II diabetic mice (Jung et al. 2010). Similarly, the methanolic extract of A. heterophyllum exhibited hypolipidemic potential (Subash and Augustine 2012). Also, polysaccharides found in A. carmichaelii inhibited high-cholesterol levels by up-regulating the mRNA expression of cholesterol 7α-hydroxylase (Zhou et al. 2015).
Other biological properties
Major Aconitum diterpene alkaloids are toxic due to their ability to interact with voltage-gated sodium channels, which increases the permeability of the smooth muscle membrane for sodium ions, followed by an increase the calcium ion availability (Fig. 3). Consequently, during muscular contraction, the release of neurotransmitters and changes in receptors and the promotion of lipid peroxidation create severe toxic effects followed by cell apoptosis in the heart, liver, and other tissues (Chan 2009). Aconitum species are also well known as immune-stimulant agents and many reports indicated that polysaccharides in these species have significant immune-stimulant properties (Zhao et al 2006; Gao et al. 2010; Fu et al. 2022). FPS-1, a water-soluble polysaccharide isolated from roots of A. carmichaelii Debx., showed potent stimulating effects on splenocyte antibody production and murine lymphocyte proliferation induced by concanavalin A or lipopolysaccharide (Zhao et al 2006). The water-soluble polysaccharides from A. kusnezoffii Reichb. (WKCP-A fraction) showed notable splenic lymphocyte proliferation activities and macrophage phagocytosis activities (Gao et al. 2011). Recently, some neutral polysaccharides isolated from A. carmichaelii leaves also exhibited promising immune modulatory properties (Fu et al. 2022), The roots of the plant A. heterophyllum traditionally used for curing dyspepsia, abdominal pain, and diarrhea showed antisecretory and antimotility effect mediated through the nitric oxide pathway (Prasad et al. 2014). Besides, antimicrobial, antivirus, anti-plasmodial, and antioxidant activities are reported from extracts, fractions, polysaccharides, diterpenoid alkaloids, and non-alkaloidal compounds obtained from different Aconitum species (Reviewed by Nyirimigabo et al. 2015; Zhou et al. 2015; Ali et al. 2021).
Fig. 3.
Mechanism action of aconitum alkaloid through the interaction with voltage-gated sodium ion pump of muscle and cardiac cells
Major active ingredients of Genus
Aconitum species are well known for the presence of diterpenoid alkaloid components, which have nitrogen-containing cyclic organic compounds with complex chemical structures. Aconitine was the first alkaloid isolated from the Aconitum in 1833 by Geiger (Nyirimigabo et al. 2015). Furthermore, other alkaloids, such as hypaconitine, mesaconitine, benzoylmesaconine, atidine, isotisine, hetidine, heteratisine, hetsinone, heterophyllisine, heterophylline, and heterophyllidine, were isolated from roots of different species of Aconitum (Fig. 4). Now, more than 1000 diterpene alkaloids, structurally classified into four categories, C18-, C19-, C20-, and bis-subtypes have been isolated from this genus. The diterpene alkaloids in Aconitum plants have captured the focus of modern research in recent years to develop therapeutical agents for anti-inflammatory, analgesic, or anti-pyretic drugs. The major active ingredients identified in some important species of Aconitum have been summarized in Table 1. Structure–activity relationship of aconitum alkaloid indicated that diterpenoid alkaloid having aroyl or aroyloxy group at R-14 position exhibited 30-times higher analgesic potential than the diterpenoid alkaloid with aroyloxy group at R-4 position (Bello-Ramírez et al. 2003). Also, the molecular weight of diterpenoid alkaloid inversely correlated with its analgesic properties. The analgesic effect is not only mediated by the central nervous system but by many other possible pathways, such as activation of voltage-dependent Na+ channels, inhibition of prostaglandin synthesis, and inhibition of noradrenaline uptake (Ameri 1998c). Toxicity of aconitine in neural, cardiac, and muscular tissues has been mediated by permanent activation of the voltage-dependent Na+ channel by shifting toward the hyperpolarized direction (Faber and Rudy 2000). Also, the presence of three groups [R(I)-C3’, R(I)-C5’, and R(I)-C2’] mainly the C5’ of the aromatic ring also determined the analgesic potency of these alkaloids. Furthermore, benzoyl ester and its position at aconitum alkaloid have been identified as an essential group that determined the binding efficacy of molecules at voltage-dependent Na + channels. High toxicity occurs if these ester groups are found at C8 and C14 positions and it also determines arrhythmia and analgesic properties (Isono et al. 1994). Besides alkaloidal constituents, various other active constituents, including, phenylpropanoids, flavonoids, terpenoids, steroids, free fatty acids, and polysaccharides, were isolated and identified from different Aconitum species. Major flavonoids present in Aconitum consist of glycosides of kaempferol and quercetin. To date, 55 known flavonoids have been isolated and identified from different species of the Aconitum; among them, 29 are new glycosides and have not been detected in other species (Yin et al. 2019). Similarly, 22 phenylpropionic acids, some steroids, and free fatty acids have been identified and quantified in different species of Aconitum (Yue et al. 2010; Weber et al. 2015; Liang et al. 2018; Yin et al. 2019). These non-alkaloidal constituents have significant antioxidant, antiparasitic, anti-phlogistic, antineoplastic, and immunoregulatory effects, and have also potential utility in discoveries of novel compound identification for drug discoveries and chemo-taxonomical significance.
Fig. 4.
Structure of some important pharmacologically potent diterpene alkaloids isolated various Aconitum species
Table 1.
Alkaloids identified in different Aconitum species and their content in root part (if quantified)
| Species | Alkaloids (quantitative value) | References |
|---|---|---|
| A. apetalum | Aconitine, aconorine, cammaconine, taurenine, songorine, songomine, 8-O-ethylcammaconine, 3-deoxyaconitine, apetaldines A−G, talassicumine A, acobretine E, | Hu et al. (2019) |
| A. balfourii | Pseudaconitine, aconitine, benzylaconitine, picroaconitine, andhaemonepellene | Khetwal and Pande (2004), Khetwal (2007) |
| A. barbatum | Puberunine and puberudine | Mu et al. (2012) |
| A. baikalense | Napelline, songorine, hypaconitine, mesaconitine, 12-epinapelline N-oxide | Nesterova et al. (2014) |
| A. chasmanthum | Aconitine, indaconitine, chasmanine, homochasmaninehomochasmaconitine, chasmoaconitine, chasmanthinine 14-o-benzoyl-8-ethoxybikhaconine, 14-o-benzoyl-8-methoxybikhaconine | Achmatowicz Jr and Marion (1964), Parvez et al. (1998) |
| A. carmichaeli | Benzoylmesaconine (0.017%); mesaconitine (0.027%); aconitine (0.003%); hypaconitine (0.179%); deoxyaconitine (0.013%); | Wang et al. (2006) |
| A. falconeri | Faleoconitine, mithaconitine, 3-methoxyacoforestinine, karakoline, 3-hydroxy-2-methyl-4 h-pyran-4-one; 3,4-dimethoxymethylbenzoate | Atta-ur-Rahman et al. (2000) |
| A. ferox | Bikhaconitine, pseudaconitine, veratroylbikhaconine, veratroylpseudaconine, diacetyl pseudaconitine | Hanuman and Katz (1993) |
| A. franchetii | chasmaconitine, chasmanthinine, talatisamine, indaconitine, leueandine | Csupor et al. (2006) |
| A. heterophyllum | Atisine, isoatisine, aconitic acid, hetisine, heteratisine, atidine, hetidine, hetisone, heterophyllisine, heterophylline, heterophyllidine, heterophyllinine-A, heterophyllinineB, lycoctonine, delphatine and lappaconitine | Wang et al. (2006), Ahmad et al. (2008), Nisar et al. (2009) |
| A. hemsleyanum | Benzoylmesaconine (0.004%); mesaconitine (0.013%); aconitine (0.004%); hypaconitine (0.014%); deoxyaconitine (0.002%); | Wang et al. (2006), Tang et al. (2009) |
| A. laciniatum | Pseudaconitine, 14-veratroylpseudaconine, 14-o-acetylneoline, neoline, senbusine a | Wangchuk et al. (2010) |
| A. laeve | Swatinine, delphatine, puberanine, n-acetylsepaconintine, swatinine a, swatinine b, foresticine, neoline, delvestine, chasmanine, 8-methylaconitine, 14-dimethylaconitine, n-deethyllyaconitine-naldehyde, lappaconitine, lycaconitine, lapaconidine, Lycoctonine | Ulubelen et al. (2002), Shaheen et al. (2005) |
| A. orientale | Demethylappaconitine; 7, 11, 14-trihydroxy-2,13-dioxohetisane; 6, 13, 15-trihydroxyhetisane; n-deethyldelphatine lappaconitine, lycoctonine, browniine | Ulubelen et al. (1996) |
| A. pendulum | Benzoylmesaconine (0.008%); mesaconitine (0.014%); aconitine (0.484%); hypaconitine (0.020%); deoxyaconitine (0.008%); | Wang et al. (2006) |
| A. palmuturn | Vakhmatine, vakhmadine, atisine, hetisine | Jiang and Pelletier (1991) |
| A. rotundifolium | Rotundifosines A–G, heterophyllidine, chellespontine | Frejat et al. (2017); Zhang et al. (2018) |
| A. spicatum | Indaconitine, chasmaconitine, ludaconitine, spicatine a, and spicatine b | Gao et al, (2006), Shyaula et al. (2016) |
| A. transsectum | Benzoylmesaconine (0.114%); mesaconitine (0.003%); aconitine (0.002%); hypaconitine (0.004%) | Wang et al. (2006) |
Major factors influencing the content of active ingredients
Being a sessile organism, toxic diterpene alkaloids acted as a plant defense system against environmental stresses and pathogenic attacks. Biosynthesis and accumulation of secondary metabolites are generally influenced by genetic architecture, developmental stage, environmental condition, metabolic expenditure of metabolites, and their other importance in plant tissue (Suyal et al. 2019; Wang et al. 2023). Besides the genetic makeup, epigenetic factors (e.g., DNA methylation and miRNA regulation) control the production of secondary metabolite with the influence of environmental stresses, such as drought, salinity, cold, pathogens, and heavy metals in pants by regulating stress-responsive genes (Wang et al. 2023). Thus, the accumulation of aconitine-type diterpene alkaloids exhibited great variation in different growing conditions within a species (Yu et al. 2017; Venkatasubramanian et al. 2018). Aconitine alkaloid is mainly accumulated in the hypogynous organs, while N-di-ethyl-aconitine accumulated in the epigeous organs. A significant variation across the seasons has been reported in total alkaloid content among the different Aconitum species. A high quantity of total alkaloids has been recorded in the leaf parts during the growth period. However, with the age, plant alkaloid content decreases (Colombo et al. 1988). Mesaconitine and aconitine in roots of A. japonicum have been reported in higher quantities in the samples harvested in May month, while hypaconitine has been reported in samples harvested in December month. Also, total alkaloid content was exhibited higher in the samples collected in May month, and lower in the samples collected in September month (Ito et al. 1996). The biosynthesis of total alkaloids and aconitine in the root of A. nagarum and A. elwesii was found to be highest in November month. However, in the leaf parts, the alkaloid content was highest in August month (pre-flowering season) in both these species (Sinam and Devi 2011). A considerable quantity of alkaloids was also reported in the stems and leaves of A. carmichaelii, and among the different tissues, total alkaloid contents were reported higher in the stem than in other parts. Among seasons, the total alkaloid of the stem part peaked in June, while in leaves, it was highest in July month (Li 2019).
Similarly, in the aerial parts of A. zeravschanicum, the highest total alkaloid content was observed at the initiation of the vegetation stage (Nigmatullaev and Salimov 2000). Under the conditions of adequate vegetative growth, the plant initiates sexual reproduction. A. kusnezoffii plant, inhibits its vegetative growth and reduces the metabolite accumulation in asexual organs to enhance the success of reproduction. Plant sacrifices their defense system by reducing the turnover of potentially active compounds to provide maximum resources for the important biological process during reproduction. Asexual organs (main storage root) acted as a sink of resources ensuring the storage, utilization, and allocation of resources from the storage root. However, lateral roots (i.e., initial asexual organ) accumulated higher biomass at the end of the growing season and become a mode of asexual reproductive method in the lateral stage (Tang et al 2021). Biomass of the aerial part (weight or the size) can be used as a marker for determining diester alkaloid content in A. carmichaelii, as a positive relationship was found between the weight of the aerial part and weight of the new tuberous root and the weight of the aerial part and diester alkaloid content in stem (Kawasaki et al. 2011).
Best recoveries of active ingredients
Diterpenoids alkaloids are the main active constituents in Aconitum with some toxicity. Quantification of these alkaloids and keeping the low level of toxic compounds important steps for the quality control of Aconitum-based drugs. Thus, various actions have been initiated to reduce its toxicity and increase the availability of non-toxic metabolites. Among the compound separation methods, a high-performance liquid chromatography (HPLC) method has been developed using the C18 column coupled with a photodiode array detector (DAD) for the simultaneous quantitative determination of six Aconitum alkaloids (i.e., aconitine, mesaconitine, hypaconitine, benzoylaconine, benzoylmesaconine, and benzoyl-hypaconine) from Aconitum roots. The best recovery (90–100%) was achieved in gradient elution using solvents of acetonitrile and ammonium bicarbonate buffer (pH 10.0) (Xie et al. 2005). Also, a capillary zone electrophoresis method has been developed for the simultaneous determination of six major alkaloids (i.e., aconitine, mesaconitine, hypaconitine, benzoylaconine, benzoylmesaconine, and benzoyl-hypaconine) from Aconitum roots. The best recovery was achieved in running buffer having a mixture of 200 mm Tris, 150 mm perchloric acid, and 40% 1,4-dioxane (pH 7.8), and the best operation temperature of the capillary was found 25 °C (Song et al. 2010).
Processing methods also significantly influenced the aconitine alkaloid in roots and herbal formulations. It has been observed that various processing steps cause the removal of the acetyl group at C8 position of diterpenoid alkaloid molecules and formed benzoylaconine with 100–400 times lesser toxicity than natural diterpenoid alkaloid molecules (Fig. 5). Toxicity is further reduced after the loss of benzoyl ester group at C14 to convert it into aconine molecules (Mizugaki and Ito 2005). In China, such processing of Aconitum is performed through soaking, heating, and decocting in water or alkaline solution (Liu et al. 2007a). After such processing, the pharmacological activity of plant extract or preparation did not exhibit any significant change (Liu et al. 2007b). However, during the steaming process, the quantity of diester–diterpenoid alkaloids decreased and the contents of monoester–diterpenoid alkaloids increased initially and reached to maximum level after 40 min, but then started to decrease gradually. However, the concentration of aconine alkaloids (e.g., mesaconine, aconine, and hypaconine) increased throughout the processing. On contrary, the concentrations of fuziline, songorine, karacoline, and salsolional remain constant or decreased slightly (Yang et al. 2014). During the baking process, the total and diterpenoids’ alkaloids changed significantly along with the total toxicity. However, a slight reduction in the concentration of diester–diterpenoid alkaloids has been obtained during the steaming process, and monoester–diterpenoid alkaloids, aconine alkaloids, and the total alkaloids have been reported to be degraded at different temperatures (Yang et al. 2014). Thus, optimization of processing parameters, such as temperature and time duration, appeared an important step for improving medicinal efficacy.
Fig. 5.
Change of toxic aconitine into benzaconine and acinine after hating treatment or boiling process (see Liu et al. 2017 for detail)
Biosynthetic pathways of diterpenoid alkaloids
The active ingredient diterpene alkaloids can be broadly classified into three types, viz., diester–diterpene alkaloids, monoester–diterpene alkaloids, and unesterified diterpene alkaloids (Zhou et al. 2015). Ester hydrolysis can transform diester–diterpene alkaloids into monoester–diterpene alkaloids or ester-free amine-diterpene alkaloids, which have relatively lower toxicity and better pharmacological effects. In rats, after hydrolysis, the toxicity of monoester–diterpene alkaloid remained up to less than 150 times that of toxicity of diester–diterpene alkaloid, while their pharmacology remained unchanged (Wen et al. 2013; Zhou et al. 2015). To date, over one thousand diterpene alkaloids have been isolated from different species of Aconitum, and after successful clinical testing, only a few compounds (lappaconitine, 3-acetylaconitine and bulleyaconitine) have been used as analgesics (Teng et al. 2021).
Diterpene alkaloids and their derivatives followed a common biosynthetic pathway and have been investigated in many species with modern technological advancements. Aconitine biosynthesis is initiated with MVA (Mevalonate) and MEP (methylerythritol) pathways (Fig. 2). The process is initiated with the condensation of three isopentenyl pyrophosphate (IPP) molecules for the formation of geranyl–geranyl pyrophosphate with the help of the enzyme geranyl–geranyl pyrophosphate synthase (GGPPS). Geranyl–geranyl pyrophosphate formed copalyl diphosphate through proton-induced cyclization in the presence of copalyl–diphosphate synthase (CDPS) (Zi et al. 2014). Furthermore, kaurene synthase (KS) formed kaurene through cyclization and rearrangement of copalyl diphosphate and atisane is formed through its alternate rearrangement reaction. After subsequent oxidation, hydroxylation, and addition of β-ethanolmaine, an atisine skeleton is formed.
Following further oxidation and modification of the atisine skeletal through various cytochrome P450s, different C20-type diterpene alkaloids are formed (Cherney et al. 2014; Devkota and Sewald 2013). The main toxic and pharmacological active constituents of Aconitum are aconitine-type C19-diterpene alkaloids (Jiang et al. 2022). The C19-type diterpene alkaloid skeletons were derived from various substitutions on the aconitine skeleton, leading to the formation of different alkaloids (Fig. 6).
Fig. 6.
Biosynthetic pathway for aconitine-type diterpenoid alkaloid (DAs) in aconitum genus. The biosynthesis pathway is initiated with the formation of mevalonate diphosphate formation from acetyl co-A in MVA pathway and dimethylallyl diphosphate from d-glyceraldehyde-3-phosphate and pyruvate in MEP pathway. After condensation of the end products of these two respective pathways, isopentenyl diphosphate is formed which is further condensed into geranyl–geranyl pyrophosphate as a precursor of diterpene alkaloids (see Rai et al. 2017; Yang et al. 2020 for detail)
Transcriptome studies for major biosynthetic pathways’ elucidation
In recent years, technological advancement created an opportunity to generate large information on genomics, proteomics, or transcriptomics in very less time in an inexpensive manner. Among these, transcriptomic studies aim at the genome-wide characterization of mRNA expression in cells, tissue, organs, or organisms as a function of a particular treatment, growth condition, ontological stage, environment, season, or pathological condition, and elucidated and/or characterized gene, transcription factors, and their epigenetic control of various biological processes (Druka et al. 2010; Wai et al. 2022; Yang et al. 2022). The technique has extensively been used to dissect the expression and genetic regulation of biosynthetic pathways of the pharmacologically important secondary metabolite. Rai et al. (2017) detected the candidate genes involved in the mevalonate (MVA) and methylerythritol (MEP) biosynthetic pathways for the synthesis of geranyl–geranyl pyrophosphate as a precursor of diterpene alkaloids. Also, a few unigenes involved in the biosynthesis of diterpene alkaloids were identified after geranyl–geranyl pyrophosphate formation (Rai et al. 2017). Among these, ent-CPP synthases (CPS), ent-kaurene synthases (KS), kaurene oxidases (KOX), cyclases, aminotransferases, monooxygenases, methyltransferase, and BAHD acyltransferases were found to be active during the biosynthesis of C19-diterpene alkaloids in Aconitum. Furthermore, regulating transcription factors (TFs) involved in the accumulation of diterpene alkaloids have been identified (Yang et al. 2020).
In a comparative transcriptome study of root and areal parts, genes like 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR), mevalonate kinase (MVK), mevalonate diphosphate decarboxylase (MVDD), and 1-hydroxy-2-methyl-2-(E)-butenyl 4-diphosphate synthase (HDS) were found to be involved in root part for the synthesis of aconitine-type diterpene alkaloids (Pal et al. 2015). Atisine biosynthesis is regulated by the simultaneous accumulation of two molecules, e.g., atisenol and steviol, in A. heterophyllum. network link between different metabolic pathways identified genes of glycolysis (e.g., G6PI, PFK, ALD, and ENO), serine biosynthesis (e.g., PGDH and PSAT), diterpene biosynthesis (e.g., KO and KH) and phosphorylated pathway as candidate genes for steviol and atisine biosynthesis (Kumar et al. 2016).
In A. carmichaelii, key genes encoding enzymes, such as ent-copalyl diphosphate synthases, ent-kaurene synthases, kaurene oxidases, cyclases, and aminotransferases, were found involved in the early steps of diterpenoid alkaloids’ biosynthetic pathway. Furthermore, candidate genes responsible for modification in diterpenoid alkaloids, such as monooxygenases, methyltransferase, and acyltransferases skeletons, were identified through transcriptome sequencing (Zhao et al. 2018a). Terpene synthase genes (CDPS4 and KS2) were correlated with 21 unigenes annotated as cytochrome P450 in A. carmichaelii (Rai et al. 2017).
In A. heterophyllum, higher expression of eight genes (out of 18 genes) encoding GDP-mannose pyrophosphorylase (GMPase), SHAGGY, Expansin, RING-box protein 1 (RBX1), SRF receptor kinase (SRF), b-amylase, ADP glucose pyrophosphorylase (AGPase), and auxin-responsive factor 2 (ARF2), was observed in roots than shoots which indicated their major contribution in accumulation and storage of primary metabolites in the tuberous root (Malhotra et al. 2016). Digital gene expression analysis in A. heterophyllum revealed that four genes (e.g., 3-hydroxy-3-methyl-glutaryl-CoA reductase, mevalonate kinase, mevalonate diphosphate decarboxylase, and 1-hydroxy-2-methyl-2-(E)-butenyl 4-diphosphate synthase) of aconite biosynthesis pathway and five genes of primary metabolic pathways (e.g., geranyl diphosphate mannose pyrophosphorylase, SHAGGY, RING-box protein 1, SRF receptor kinases, and b-amylase) over-expressed in root parts compared to shoots for the biosynthesis of aconitine and related alkaloids. Also, ABC transporters (399 nos.) involved in the biosynthesis and storage of aconitine-type diterpene alkaloids were identified (Pal et al. 2015).
In A. kusnezoffii, among the different plant parts, the mevalonate pathway was relatively more active than methylerythritol 4-phosphate pathway in the flower, while in the leaf and stem, the situation found the opposite. However, aconitine, mesaconitine, and hypaconitine contents in different tissues remain inconsistent with the expression of biosynthesis genes, indicating that there exists a sink–source relation in the plant (Bai et al. 2022). The transcriptome data also indicated the genes involved in alkaloid biosynthesis are differentially expressed in different varieties, consistent with differences in the accumulation of alkaloids (Yang et al. 2020). However, information on different CYP450s, methyl transferases, hydroxylases, and other enzymes involved in structural modification for the diversification of aconitine-type diterpene alkaloids remains largely unexplored.
Genomic resources and diversity studies
Representing over 350 species, the Aconitum genus provides a large genetic resource availability across the world (Nyirimigabo et al. 2015). With high species diversity, eastern Asia has been considered a center of diversification (Kadota 1987). The greatest species diversity has been found in the Hengduan Mountains region of southwestern China (Liang-Qian 1988). The subgenus Aconitum includes more than 250 species, mainly distributed in the Eastern Himalaya, southwestern China and Japan. Interestingly, a total of 22 native species are found in Europe and are regarded as a secondary center of Aconitum diversification (Mitka 2000). While, with more than 60 species, Aconitum subgenus Lycoctonum has a wide distribution range and well-known background of polyploidy (Kong et al. 2017a, b). Despite great medicinal value in traditional and modern medicines, fewer efforts have been made in Aconitum species in genomic resource creation for variety improvement.
Phylogenetic classification and genetic diversity
Within species, a large genetic diversity has been demonstrated in various studies. Aconitum species are well known for taxonomic complexity due to extensive inter-specific hybridization (Kita and Ito 2000). Nuclear ITS markers have been applied to resolve this issue in some recent studies. Many species from northern China exhibited close relationships with the species from Europe, North America, and eastern Asia. ITS-based categorization supports the sub-grouping of the subgenus Aconitum, which is based on the morphology of seeds and petals, suggesting that seed and petal morphology reflects the phylogenetic relationships within the subgenus in a better way. Other important morphological characters used for traditional taxonomical classification, such as the degree of leaf division, the shape of the upper sepal, and the attitude of the stem, might be unreliable in subdividing the subgenus (Luo et al. 2005). Similarly, based on phylogenetic analysis using multiple nuclear (ITS and ESTs) and chloroplast markers in Aconitum L. subgenus. Lycoctonum, the removal of many unique arrays of characters (the latter even having the aberrant base chromosome number of x = 6) was suggested to achieve monophyly in the section. Furthermore, the subgenus Lycoctonum was suggested to be redefined to include only two sections, the unspecific sect. Alatospermum and the relatively species-rich sect. Lycoctonum (Hong et al. 2017).
In different species of Aconitum, moderate-to-high level of genetic diversity was reported in various studies. RAPD and ISSR as preliminary marker systems used for genetic diversity characterization of Aconitum genus were successfully used in diversity characterization of A. noveboracense, A. columbianum Nutt, A. carmichaelii, and A. kongboense L. species and based on genetic diversity parameters, suitable conservation priorities were formulated (Cole and Kuchenreuther 2001; Zhao et al. 2015; Luo et al. 1994; Meng et al. 2015). However, these marker systems lack the issue of repeatability thus, AFPL emerged as another choice of technique for diversity characterization of A. brachypodum, A. kongboense, A. plicatum, and A. firmum (Mitka et al. 2015; Du et al. 2018; Meng et al. 2014).
More recently, sequence-specific markers, such as SSRs’ markers, have emerged as a new method for genetic characterization. With the development of sequencing technology, expression-based markers become helpful for the development of functional markers. The ubiquity and usefulness of SSRs or microsatellites have been well recognized in eukaryotes for constructing framework genetic maps, comparative mapping, marker-assisted selection, genetic diversity, and gene targeting due to high frequency, co-dominance, multi-allelic nature, reproducibility, extensive genome coverage, and easiness in detection (Sharma et al. 2020). These markers have already been successfully used in the genetic diversity characterization of A. coreanum, A. carmichaelii, A. gymnandrum, A. napellus L, A. austrokoreense, and many other species (Won et al. 2012; Yang and Zhou 2017; Lee et al. 2018). Various genetic diversity studies available in Aconitum species using different marker systems are presented in Table 2.
Table 2.
Genetic diversity studies in different species of Aconitum using diverse markers
| Species name | Sample size and location | Marker type used | Diversity and other description | References |
|---|---|---|---|---|
| A. noveboracense & A. columbianum | USA | Isozyme and ISSR | PP = 86% by Isozymes and 89.7% by RAPD, high level of FST = 0.24 and less gene flow (Nm = 0.68.) was recorded | Cole and Kuchenreuther (2001) |
| A. kusnezoffii, A. soongaricum A. carmichaelii & A. leucostomum | 3 population in Xinjiang Province | RAPD and ISSR | PP = 97.25% (RAPD) and 98.92% (ISSR). Hs was higher than Ht. GST was 0.4358 (RAPD) and 0.5005 (ISSR). Nm was 0.6473 for ISSR and 0.4991 for RAPD) | Zhao et al. (2015) |
| A. carmichaeli | Jiangyou, China | ISSR | PP was 69.39%, Na = 1.6939 Ne = 1.3715, H = 0.230 8, and I = 0.3530 | Luo et al. (1994) |
| A. kongboense L | Motuo, Tibet Plateau | ISSR | At species level, PP = 58.42%. Ne = 1.564; H = 0.320; and H = 0.469. All the cultivars cluster into 3 groups according to their geographical origin and genetic backgrounds | Meng et al. (2015) |
| A. plicatum & A. firmum | Carpathian Mountains Europe | AFLP | Gst was greater within the Sudetic A. plicatum (FST = 0.139, P < 0.001) than within the Carpathian A. firmum (FST = 0.062, P < 0.001) populations due to the long-lasting geographic isolation | Mitka et al. (2015) |
| A. brachypodum | Guizhou area | AFLP | Relatively high level of genetic diversity (He = 0.322 9 ± 0.179, I = 0.472 0 ± 0.251 7, PP = 80.57% at species level and Genetic differentiation index (Gst) was 0.864 2 | Du (2018) |
| A. kongboense | Eastern China | AFLP | NPL = 77, PPB = 68.75%, Na = 1.688, Ne = 1.412) and lowest in population 2 (NPL = 57, PPB = 50.89%, Na = 1.509, Ne = 1.273) | Meng et al. (2014) |
| A. coreanum | Korea | Microsatellite markers | Low gene flow (1.126), heterozygosity deficit, low level of among-population differentiation, small size of gene flow, and lack of sequence variation of the organelle suggest that species are reproductively isolated from other species | Won et al. (2012) |
| A. carmichaeli | Central china | Microsatellite markers | Ne = 3.25, Ho = 0.612, He = 0.493, I = 0.851 and h = 0.505. FST = 0.149) and Nm = 1.432 was limited | Yang and Zhou (2017) |
| A. austrokoreense | Korea | Microsatellite markers | High pairwise FST = 0.35 (mean) suggested significant differentiation among populations. Low within population genetic variation was recorded | Lee et al. (2018) |
NPL number of polymorphic loci, PP Percentage of Pymoorphic loci, Na observed number of alleles, Ne effective number of alleles, Ho observed heterozygosity, He expected heterozygosity, H Nei’s gene diversity, I Shannon’s index, Hs inter-population diversity Ht intra-population diversity, Nm gene flow, GST efficient of gene differentiation, h Nei’s genetic diversity index, FST genetic differentiation
Availability of genomic resources
Information on genetic resources provides insight into the expression, regulation, and heredity of traits, which can be harnessed for variety improvement and quality maintenance during molecular breeding efforts. Thus, the use of DNA markers has become useful assays for diversity analysis, phylogenetics, genetic mapping, marker-assisted breeding, genotyping, and genome analysis in Aconitum. The level of genetic diversity determined by various types of markers can be utilized for formulating conservation strategies for different species of Aconitum. Various efforts for genomic resource creation, such as microsatellite loci or SSR marker development, etc., are summarized in Table 3. To date, no efforts have been made the improvement of variety in Aconitum using the molecular marker; however, it might be beneficial in terms of time, labor, and cost-efficiency.
Table 3.
Microsatellite maker development in different species of Aconitum
| Name of species | Method for marker development | Marker detail | Application and validation | References |
|---|---|---|---|---|
| A. austrokoreense | Next-generation sequencing technology | 9 novel microsatellite markers were developed | These novel markers were found valuable for assessing the genetic diversity and germplasm conservation | Yun et al. (2015) |
| A. carmichaelii | Transcriptome sequencing | A total of 16,068 SSR across 14,168 unigenes | Potential genetic resource for genetic diversity assessment and comparative genetics across different Aconitum species | Rai et al. (2017) |
| A. brachypodum | Fast isolation by AFLP of sequences containing repeats | 12 microsatellite markers were developed | Among the 12 markers, three deviated from HW equilibrium significantly and have shown usefulness in population genetics | Li et al. (2015) |
| A. coreanum | Next-Generation Sequencing | 10 microsatellites were developed | Only two markers were polymorphic and showed FST of 0.205 and 0.275, respectively | Won et al. (2012) |
| A. gymnandrum | Direct genomic DNA sequencing | 19 polymorphic microsatellite loci were developed | Seven loci showed a significant deviation from Hardy–Weinberg equilibrium followinga Bonferroni correction, which might be due to non-random mating of individuals | Hou et al. (2020) |
| A. gymnandrum | Using the combined biotin capture method | 16 polymorphic microsatellite loci were identified | 50% loci were polymorphic and the number of alleles per locus ranged from 4 to 15 in 66 individuals from 6 population | Xu et al. (2011) |
| A. heterophyllum | Transcriptome sequencing | A total of 177,438 potential SSRs were identified | Functional markers could be identified for quality-related traits | Pal et al. (2015) |
| A. kusnezoffii | Genomic enrichment approach | A total of 19 microsatellite loci were developed | Among these, 13 loci polymorphic and 5 loci among the 6 amplified in A. barbatumvar. puberulumwere polymorphic, | Ge et al. (2016) |
| A. napellus | Sequencing of partial genomic library | 16 polymorphic microsatellite markers were characterized | These microsatellites offer an efficient tool for detailed investigations on population genetic structure of the species | Cadre et al. (2005) |
| A. pseudolaeve & A. longecassidatum | Complete sequencing of chloroplast (CP) genome | A total of 61 indels and 62 SNPs detected between both the species | A. pseudolaeve, A. barbatum and A. longecassidatum can be clearly distinguished using the novel indel markers [AcoTT (trnK-trnQ) and AcoYN (ycf1-ndhF)] | Park et al. (2017a) |
| A. reclinatum | Sequencing of a genomic library using Illumina HiSeq technology | 10 polymorphic primer pairs were developed | These microsatellites were found useful in genetic diversity and conservation genetics studies | Zhou et al. (2018) |
| A. reclinatum | Sequencing of a genomic library using Illumina HiSeq technology | 55 pairs of microsatellite primers were obtained | 14 pair (~ 25%) high polymorphism primers could be used for identification of genetic resources, genetic diversity analysis and DNA fingerprint | Yang & Zhou (2017) |
| A. vilmorinianum | Next-generation sequencing technology | 18 novel microsatellite markers were developed | These polymorphic microsatellite locican be useful for genetics studies | He et al. (2015) |
In A. kusnezoffii, 19 microsatellite loci were developed and these markers were found useful in quantifying male and female fitness in A. kusnezoffii and evaluating the effects of clonal growth on sexual reproduction (Ge et al. 2016). In A. brachypodum, 12 microsatellite markers were developed from two microsatellite-enriched libraries (AG, AC) constructed using an FIASCO method (Li et al. 2015). Hou et al. (2020) identified 19 polymorphic microsatellite loci for A. gymnandrum. Among these, seven loci showed significant deviation from Hardy–Weinberg equilibrium and showed potential for pollination ecology and population genetic studies in Qinghai–Tibet plateau. In A. gymnandrum, 16 out of the 32 loci were identified as polymorphism and were found useful in characterization in genetic diversity analysis (Xu et al. 2011). In A. coreanum (H. Lév.), ten microsatellites were developed, and only two markers were polymorphic and showed FST as 0.205 and 0.275, respectively (Won et al. 2012).
Currently, Next-Generation Sequencing (NGS) technology has been found useful in large number marker development in a shorter time. In A. reclinatum, sequencing of a genomic library using Illumina HiSeq technology, ten polymorphic primer pairs were developed. These microsatellites isolated were found useful in genetic diversity and conservation genetics studies (Zhou et al. 2018). Also, in A. austrokoreense, 9 novel microsatellite markers were developed from sequencing of genomic DNA and these novel markers were found valuable for assessing genetic diversity and for germplasm conservation (Yun et al. 2015). Similarly, in A. vilmorinianum, 18 novel microsatellite markers were developed using next-generation sequencing technology of genomic DNA (He et al. 2015). Cadre et al. (2005) identified and characterized 16 polymorphic microsatellite markers in A. napellus through sequencing of a partial genomic library and found them an efficient tool for detailed investigations of the population genetic structure of the species. These polymorphic microsatellite loci will be especially useful for genetics studies. In the future, these microsatellite loci can be valuable for further interpretation of the fine population structure and conservation strategies of this rare, threatened species.
Furthermore, NGS-based transcriptome analysis could provide a large dataset on the expression part of genomics and could be useful for the development of functionally relevant markers. This has explored the development and utilization of sequence-based markers for genome mapping and molecular breeding in a faster manner (Unamba et al. 2015). In comparative root and shoot transcriptome analysis of A. heterophyllum, a total of 177,438 potential SSRs were identified and these potential functional markers could be linked with quality-related traits (Pal et al. 2015). Similarly, in A. carmichaelii, the de novo transcriptome assembly yielded a total of 16,068 SSR from 14,168 unigenes motifs with at least ten repetitions of mono- to hexa-nucleotide (Rai et al. 2017) wherein mono-nucleotide repeats had the greater frequency (63.68%) followed by tri- and di-nucleotide repeats (22.3% and 12%, respectively). Based on the functional classification of SSR containing unigenes, the development of quality-related functional markers related to secondary metabolites, high-altitude adaptation, yield, and productivity could be developed.
Full genome sequencing has not been carried out in any species of Aconitum. However, the full sequence of the chloroplast and mitochondrial genome is available in many species of Aconitum. A comparative overview of full genome sequencing is summarized in Table 4. The chloroplast genome was found as a typical quadripartite structure with a Large Single-Copy region (LSC) and a Small Single-Copy (SSC) separated by a pair of inverted repeat regions. IR region was found more conserved as compared to the LSC and SSC regions (Chen et al. 2015; Sungyu, et al. 2016; Kong et al. 2017a, b; Park et al. 2017a; Park et al. 2017b; Meng et al. 2018; Kong et al. 2017a, b; Yang et al. 2018; Kim et al. 2019; Liu et al. 2020; Meng et al. 2019; Cheng et al.2020; Li et al. 2020; Lim et al. 2020; Wang and Li 2020; Zhang et al. 2021a, b; Ni et al. 2022;). Among all the species, the smallest size of cp genome (151,214 bp) was found in A. episcopale (Meng et al. 2018). These genomes were found good source of simple sequence repeats, and interestingly, among the different subgenus, a higher number of SSRs were obtained (64–62, average value 59) in subgus Lycoctonum as compared to subgenus Aconitum (40–60, average value 54). The maximum SSRs concentration was found in the LSC region (Kong et al. 2017a, b). However, the number of SSRs was found between 259 and 287 in a few other studies (Park et al. 2017b; Meng et al. 2018). Furthermore, cluster analysis based on the chloroplast genome revealed that two separate monophyletically groups formed according to their subgenus of Aconitum (Park et al. 2017a; Kong et al. 2017a, b; Meng et al. 2018). These full chloroplast genome sequences have shown potential applicability in phylogenic classification, evolutionary history, and divergence studies.
Table 4.
Available information in the full chloroplast genome of different Aconitum species
| Species name | Genome size (bp) | Number of genes | GC content | Other important feature | References |
|---|---|---|---|---|---|
| A. coreanum | 155,880 | 131 genes—86 protein-coding, 8 rRNA genes, and 37 tRNA genes | 38.13% | LSC (86,338 bp) and SSC (16,946 bp) regions separated by a pair of IR regions (26,294 bp) | Park et al. (2017b) |
| A. carmichaelii | 157,040p | 131 genes—86 protein-coding, 8 rRNA genes, and 37 tRNA genes | 37.99% | LSC (87,628 bp) and SSC (16,924 bp) regions separated by a pair of IR regions (26,244 bp) | Park et al. (2017b) |
| A. pseudolaeve | 155,628 | 112 genes—78 protein-coding, 4 rRNA genes, and 30 tRNA genes | 38.0% | A pair of inverted repeat regions separated by LSC (86,683 bp) and SSC (17,091) regions | Park et al. (2017a) |
| A. longecassidatum | 155,524 | 112 genes—78 protein-coding, 4 rRNA genes, and 30 tRNA genes | 38.10% | A pair of inverted repeat regions separated by LSC (86,466 bp) and SSC (16,950 bp) regions | Park et al. (2017a) |
| A. vilmorinianum | 155,761 | 132 genes—78 protein-coding, 4 rRNA genes, and 30 tRNA genes | 38.10% | A pair of IR regions of 26,209 bp), one LSC (86,394 bp) and one SSC region (16,949 bp) two pseudogenes (yrps19 and yycf1) found | Meng et al. (2018) |
| A. delavayi | 155,769 | 131 genes—78 protein-coding, 4 rRNA genes, and 30 tRNA genes | 38.1% | A pair of IR regions of 26,240 bp), one LSC (86,340 bp) and one SSC region (16,949 bp) | Meng et al. (2018) |
| A. episcopale | 151,214 | 131 genes—78 protein-coding, 4 rRNA genes, and 29 tRNA genes | 38.3% | A pair of IR regions of 26,156 bp and 26,217 bp), one LSC (83,182 bp) and one SSC region (15,598 bp), two pseudogenes (yrps19 and yycf1) found | Meng et al. (2018) |
| A. hemsleyanum | 155,684 | 132 genes—78 protein-coding, 4 rRNA genes, and 30 tRNA genes | 38.1% | A pair of IR regions of 26,235 bp), one LSC (86,929 bp) and one SSC region (16,922 bp), two pseudogenes (yrps19 and yycf1) found | Meng et al. (2018) |
| A. contortum | 155,653 | 132 genes—78 protein-coding, 4 rRNA genes, and 30 tRNA genes | 38.1% | A pair of IR regions of 26,221 bp), one LSC (86,267 bp) and one SSC region (16,944 bp), two pseudogenes (yrps19 and yycf1) found | Meng et al. (2018) |
| A. scaposum | 157,688 |
145 unique genes, 8 rRNA genes and 38 tRNA genes |
38.0% | A pair of IR regions of 26,156 bp and 26,232 bp), one LSC (69,309 bp) and one SSC region (16,917 bp) | Zhang et al. (2021a, b) |
| A. coreanum | 157,024 | 132 genes (86 protein-coding genes, 8 rRNAs, and 37 tRNAs) | 38.0% | 87,637 bp of LSC and 16,901 bp of SSC, are separated by two 26,243 bp IRs | Kim et al. (2019) |
| A. puchonroenicum | 155,631 | 111 genes (4 rRNA genes, 29 tRNA genes, and 78 protein-coding genes | 38.05% | 86,689 bp LSC and 17,088 bp SSC regions are separated by 25,927 bp of a pair of IR regions | Lim et al. (2020) |
| A. piepunense | 155,836 | 130 genes (8 rRNA genes, 37 tRNA genes, and 85 protein-coding genes | 37.8% | A large LSC, (86,433 bp), a small SSC, (16,945 bp), and a pair of IR regions (26,229 bp) | Ni et al. (2022) |
| A. reclinatum | 157,354 |
135 genes, including 87 protein-coding genes, 40 tRNA genes and 8 rRNA genes |
38.0% | A pair of 26,061 bp IRs regions separated by LSC of 88,269 bp and SSC of 16,963 bp | Kong et al. (2017a, b) |
| A. flavum | 155,654 | 129 genes, including 83 protein-coding genes, 8 rRNA, and 37 tRNA genes | 38.1% | A one LSC of 86,390 bp, one SSC region of 16,968 bp, and two IR regions of 26,148 bp | Liu et al. (2020) |
| A. austroyunnanense | 155,818 | 131 genes, including 85 protein-coding genes, 37 tRNA genes, 8 rRNA genes and a pseudogene | 38.1% | Two IRs, (26,128 bp) regions, one large LSC (86,555 bp) and one SSC (17,007 bp) | Cheng et al. (2020) |
| A. pendulum | 155,597 | 131 genes, consisting of 86 protein-encoding genes, 8 rRNA, and 37 tRNA | 38.1% | A copy of LSC and SSC regions of 86,336 and 16,961 bp, isolated by two IRs regions of 26,150 bp | Wang end Li (2020) |
| A. brachypodum | 155,651 | 132 genes including 85 protein-coding genes, 37 tRNA genes, 8 rRNA genes and 2 pseudogenes | 38.0% | A pair of 26,213 bp IRs) separated by LSC region of 86,292 bp and SSC region of 16,933 bp | Meng et al. (2019) |
| A. carmichaelii | 155,737 | 112 gene species (incl. 78 protein-coding, 30 tRNAs & 4 rRNAs | 38.1% | A pair of IR regions of 26,193 bp, separated by LSC region of 86,330 bp and SSC region of 17,021 bp | Yang et al. (2018) |
| A. tanguticum | 157,114 | 112 gene, including 78 protein-coding, 30 tRNA and rRNA gene | 38.0% |
A pair of IR regions (26,255 bp), separated by LSC region (87,559 bp) and SSC region (17,045 bp) |
Li et al. (2020) |
| A. volubile var. pubescens | 155,872 | 131 genes including 86 protein-encoding genes, 8 rRNA genes and 37 tRNA) genes | 38.12% | LSC (86,348 bp), SSC (16,944 bp) separated by pair of IRs (26,290 bp) | Sungyu, et al. (2016) |
| A. barbatumvarpuberulum | 1 56,749 | 130 genes, including 84 protein-coding genes, 34 tRNA genes and 8 rRNA genes | 38.7% | Include LSC region of 87,630 bp and SCR region of 16,941 bp separated by two IRs of 26,089 bp | Chen et al. (2015) |
| A. angustius | 156,109 | 126 genes with 84 protein-coding genes, 38 tRNA genes and 4 rRNA genes | 38.0% | A pair of IR regions of 26,225 bp, separated by LSC region of 86,719 bp and SSC region of 16,914 bp | Kong et al. (2017a, b) |
| A. finetianum | 155,625 | 126 genes with 84 protein-coding genes, 38 tRNA genes and 4 rRNA genes | 38.0% | A pair of IR regions of 25,927 bp, separated by LSC region of 86,664 bp and SSC region of 17,107 bp | Kong et al. (2017a, b) |
| A. sinomontanum | 157,215 | 126 genes with 84 protein-coding genes, 38 tRNA genes and 4 rRNA genes | 38.0% | A pair of IR regions of 26,090 bp separated by LSC region of 86,074 bp and SSC region of 16,926 bp | Kong et al. (2017a, b) |
Tissue culture studies
Mass propagation techniques
Standardization of culture medium is one of the important components of plant tissue culture. Giri et al. (1993) reported standardization of in vitro propagation of A. heterophyllum Wall. They induced callus on MS medium supplemented with either 1 mg/l (2,4-d), 0.5 mg/l Kinetin and 10% coconut water or with 5 mg/l NAA and 1 mg/l BAP. Somatic embryos were able to form when calli induced on 2,4-d and NAA were transferred onto MS medium supplemented with 1 mg/ l BAP and 0.1 mg/l NAA following two sub-culturing. Complete plantlets were regenerated from these somatic embryos following 4 weeks on medium with ¼ MS medium. Rooting of plantlets was achieved with 1.0 mg/l IBA. Jabeen et al. (2006) were able to induce callus on MS medium supplemented with a low concentration of NAA (0.5 mg/l) and BAP (0.25 mg/l). The shooting was achieved in MS medium supplemented with 0.25 mg/l NAA with 0.5 mg/l BAP. Likewise, the regeneration of plantlets from callus cultures of leaf and lateral buds was achieved (Singh et al. 1998).
Young leaf explants and lateral buds were cultured on MS medium supplemented with 0.8 mg/l of BAP and NAA each leading to callus induction and finally somatic embryos and shoots. Pandey et al. (2004) could induce callus from the leaf segment of in vitro axillary bud on MS medium supplemented with 4.5 µMBAP and 26.9 µMNAA. They were able to induce shoots with the same concentration of BAP and a lower concentration of NAA. In vitro shooting and rooting of plantlets was achieved with 1 µMBAP and 12.3 µM of IBA. Since in vitro propagated plants are more likely to have variation in morphology, genetic makeup, phytochemical content, or stages of development than seed-grown or vegetative propagated plants, Pandey et al. (2004) evaluated the difference in the chromosome, protein profile, and alkaloid content of in vitro tubers raised and seed raised plants of comparable age. Rawat et al. (2013a) studied secondary metabolite production in A. violaceum Jacq. Among all the tested hormone combinations, 2.5 µM 2,4-dichlorophenoxyacetic acid (2,4-d) and 0.25 µM kinetin (Kn) could promote callus formation. 6-Benzyl amino purine (6-BAP) was found to be effective for shoot regeneration and secondary metabolite production compared to thidiazuron (TDZ). The frequency of plantlet regeneration was highest when calli were transferred to an MS medium supplemented with 1 µM BAP and 0.5 µM α-naphthalene acetic acid (NAA). It was observed that when low concentrations of NAA were added to the culture medium, secondary metabolite production was reduced as compared to cytokinin only. Further, different cytokinin concentrations affected secondary metabolite production in some cases.
Callus-mediated organogenesis from both leaf and petiole segments was reported by Gondval et al. (2016). MS medium supplemented with 0.5 mg/l TDZ and 1.0 mg/l NAA yielded optimum callusing. The shoots and roots were developed on MS medium supplemented with a combination of 0.25 mg/l IAA and 0.25 mg/l NAA. The plantlets were acclimatized and successfully transferred to field conditions. In A. ferox, callus was initiated and maintained on MS medium supplemented with 2.26 µM 2,4-d. For the development of adventitious shoots, BAP was found to be responding among all the tested plant growth regulators. Synergistic interaction between IAA and BAP inducted plantlets with well-developed roots and shoots. However, the best rooting response was recorded when shoots were placed on a paper bridge in the liquid MS medium. Phytochemical analysis revealed similar antioxidant activity closer to wild plants. The study leads to the development of the method for sustainable utilization of A. ferox (Singh et al. 2020). Rafiq et al. (2021) developed an efficient micropropagation protocol for A. chasmanthum Stapf ex Holmes using nodal explants. Nodal explants showed direct multiple shoot regeneration with BAP (0.5 mg/l) and rhizome formation was achieved following 8 weeks in MS medium and finally development of plantlets. These protocols could be used for large-scale propagation and conservation of various Aconitum species. A summary of species, explant, and media used in tissue culture studies of Aconitum has been summarized in Table 5.
Table 5.
Response of different species of Aconitum in tissue culture conditions
| Species | Explant | Media & growth regulators | References |
|---|---|---|---|
| A. atrox (Bruhl). Muk | Leaf explants and lateral buds | MS + 0.8 mg/L BAP and NAA | Singh et al. (1998) |
| A. balfourii Stapf | Leaf segment of in vitro axillary bud | MS + 4.5 µM BAP + 26.9 µM NAA | Pandey et al (2004) |
| A. balfourii Stapf | Leaf and lateral buds, axillary buds, leaf | MS + 4.5 µM BAP + 26.9 µM NAA | Singh et al. (1998) |
| A. balfourii Stapf | Leaf and petiole segments | MS + 0.5 mg/L TDZ + 1.0 mg/L NAA | Gondval et al (2016) |
| A. bucovinense Zapał | Leaf | MS + 1.5 mg/L IBA + 1.0 mg/L BAP | Kocot et al. (2022) |
| A. carmichaeli | Shoot tips and axillary buds | MS + 1.0 mg/L BAP | Hatano et al (1988) |
| A. chasmanthum Stapf ex Holmes | Nodes, leaves and stems | MS + cytokinins (BAP, Kn), auxins (2,4-d, NAA) | Rafiq et al. (2021) |
| A. ferox | Seed raised plantlets | MS + 2.26 µM 2,4-d) | Singh et al. (2020) |
| A. heterophyllum Wall | Leaf and petiole | MS + 1 mg/L 2,4-d+ 0.5 mg/L kinetin + 10% coconut water (v/v) or with 5 mg/L NAA + 1 mg/L NAA | Giri et al (1993) |
| A. heterophyllum Wall | Nodal segments | 0.5 mg/L NAA and 0.25 mg/L BAP | Jabeen et al. (2006) |
| A. heterophyllum Wall | Leaf explants | MS + 1 mg/L BAP + 0.5 mg/L Kin + 0.1 mg/l GA3 | Mahajan et al. (2015) |
| A. napellus | Shoot tip, stem nodes | Shoots (4 mg/l BA); explants on rafts (0.25 mg/l BA) | Watad et al. (1995) |
| A. uncinatum | Shoot apex – multiple shoots, plantlets | 44.4 μM BA + 46.5PM kinetin | Lim and Kitto (1995) |
| A. violaceum Jacq | Nodal segment | MS + 0.5 µM 2,4-d + 0.25 µM kinetin | Rawat et al (2013a) |
| A. violaceum Jacq | Shoot tip, callus | MS + 0.1 µM BAP + 0.5 µM NAA | Chandra (2003) |
Secondary metabolites’ production from callus suspension culture
Micropropagation through plant tissue culture has also been used as a route for the production of secondary metabolites through induction of callus or in suspension cultures in presence of higher concentrations of auxin or in a combination of auxin and cytokinin. Production of important secondary metabolites has been successfully achieved in many Aconitum species. Tissue culture-raised plants showed a higher accumulation of aconitine content. In A. violaceum, higher aconitine content was recorded in tissue culture-raised hardened plants as compared to control plants (Rawat et al. 2013a). In A. carmichaelii Debx., MS medium supplemented with 0.5 mg/l IAA exhibited better rooting at 20 °C, and in these roots, aconitine-type alkaloids (mesaconitine and hypaconitine) were found more than 50% higher than plants cultured at 15 °C and 10 °C (Shiping et al. 1998). Also, in A. ferox, a higher level of antioxidant activity was found in roots regenerated with the supplementation of 5MS + 6 µM BAP + 3 µM IAA, but total phenolic and flavonoid contents were obtained as lower than in control plants (Singh et al. 2020).
Cell suspension cultures can be efficient for aconitine alkaloids or related secondary metabolite production. In A. napellus, various culture conditions exhibited a positive impact on cell biomass and aconitine accumulation. Callus induced in leaf explants was transferred into liquid MS medium with 5% sucrose and showed significant improvement in cell growth and aconitine accumulation after 8 weeks. Salicylic acid and yeast extract supplementation in the MS medium induced a higher accumulation of aconitine and reached up to 0.043% (dry weight basis) (Hwang et al. 2004). In A. heterophyllum, total alkaloid (aconites) content was recorded as 3.75 times higher in Agrobacterium rhizogenes-mediated transformed roots (2.96%) as compared to non-transformed (control) roots (Giri et al. 1997). In other species, such studies are largely lacking; however, such biotechnological interventions could be promising tools for important metabolite production.
Future prospects and conclusion
More than 350 Aconitum have been found worldwide, and the phytochemical and pharmacological studies are still restricted to only a few species. Such a vast diversity in genetic resources created an opportunity for the discovery of novel molecules with higher efficient diterpene alkaloids or with different biological activities. Limited molecules present in Aconitum have been explored for pharmacological activities. From pharmacological activities, studies have been carried out in limited areas, such as anti-inflammatory, analgesic, anti-rheumatic, anti-nociceptive, hypoglycemic, multidrug resistance inhibitor, cytotoxic, anti-tumorous, immune stimulant, and antioxidant activities (Ameri 1998a; Ameri 1998b; Xu et al. 2006; Dasyukevich and Solyanik 2007; Verma et al. 2010; Huang et al. 2011; Wang et al 2012; Xing et al. 2014; Nesterova et al 2014; Zhang et al. 2015; Guo et al 2017; Zhang et al 2020; Zhang et al 2021a, b; Liang et al 2016). However, lead from these preliminary finding needs more in-depth investigations for novel drug discoveries. For example, most of the diterpene alkaloids have been validated for anticancer or anti-inflammatory activities; however, after the screening of all the existing diterpene alkaloids, the identification of the most potent molecule and activity–structure relationship might provide a breakthrough in drug discovery. Also, most of the ethnopharmacological uses of the genus need to be validated using suitable model systems followed by clinical trials. The processed A. carmichaelii hydrogel patch prepared by matrix prescription optimized through the central composite design-response surface method was found better in quality in terms of appearance, adhesion and in vitro release (Wu et al 2018) and such drug delivery systems can be more useful for determining the new dosage of aconite-type diterpene alkaloids.
High genetic diversity within the genus, hybridization compatibility in nature (existence of natural hybrids), large geographical distribution, and presence of a large array of aconitine-type diterpene alkaloid and other important secondary metabolites indicated tremendous possibilities of variety development in Aconitum. Only, A. carmichaelii Debeaux has been cultivated on farms in western China without any information on quality attributes. In India, A. heterophyllum is cultivated in high-altitude Himalayan regions to some extent. Diversity in quantitative traits at the genetic level provides the basis for successful breeding efforts during variety improvement. Thus, genetic characterization is necessary; however, such studies are largely unavailable in most of the Aconitum species. Genetic diversity studies at the global level in the species are only available in a few species (Cadre et al 2005; Xu et al. 2011; Won et al 2012; Li et al 2015; Pal et al 2015; He et al 2015; Yun et al 2015; Ge et al 2016; Rai et al 2017; Park et al 2017a; Zhou et al 2018; Yang and Zhou 2017; Hou et al 2020). Also, due to the scarcity of genomic information in most of the Aconitum species, trait-specific functional markers are not available in Aconitum species, which can be important for elite parental group identification, gene targeting, mapping studies, and marker-assisted breeding. Modern sequencing platforms, simulation modeling, and computational advancement can reduce time and expense in future Aconitum research. Thus, opportunities for genetic improvement in species through molecular breeding or core genotype selection for cultivation are insignificant in Aconitum.
Most of the Aconitum species preferred patchy distribution, and thus, low population density exists in nature. Extensive extraction of most of the species in Asia causes pressure on the wild population leading to its extinction. Therefore, regular monitoring of the population status in the wild to ascertain the existence of the species on the earth is required. Many species of Aconitum, e.g., A. chasmanthum (critically endangered), A. heterophyllum (endangered), A. violaceum (vulnerable), A. corsicum (vulnerable), A. austrokoreense (near threatened), A. firmum subsp. moravicum (near threatened), and A. lasiocarpum (near threatened), are categorized under the threat list of IUCN (IUCN 2020). However, many species restricted to very small geographic regions have yet not been evaluated and have a larger threat of extinction. Propagation protocols (tissue culture or conventional) are only available for a few species (Hatano et al 1987, 1988; Giri et al 1993; Watad et al 1995; Lim and Kitto 1995; Singh et al. 1998; Chandra 2003; Pandey et al 2004; Jabeen et al. 2006; Rawat et al. 2013b; Gondval et al. 2016; Singh et al. 2020; Rafiq et al. 2021) and robust method of propagation for commercial cultivation need to be developed. Although, seed germination methods have been standardized for many important species (Beigh et al 2006; Vandelook et al 2009; Shang et al 2011; Priyanka and Priyanka 2012; Rana and Sreenivasulu 2013); however, agro-techniques are only available in few species such as in A. carmichaelii (Gao et al 2021).
The availability of huge diversity in diterpene alkaloids and variable bioactivity induces synergistic effect on many disease conditions and improve overall therapeutic effects.However, a quantitative assessment of diterpene alkaloids in many Aconitum species has not yet been carried out. Instead, chemical derivatization of existing molecules became the area of modern research; however, these more effective modern aconitine-derived semi-synthetic compounds required screening of full clinical parameters as it may induce other side effects. Low content of alkaloid causes problems of individual compound isolation and its pharmacological characterization. Thus, the increased production of important secondary metabolites through cell culture or Agrobacterium rhizogenes-mediated transformed roots can reduce production costs. Small-scale protocols for suspension culture or Agrobacterium rhizogenes-mediated transformed roots for metabolite production have been successfully developed in a few Aconitum species with promising results (Giri et al. 1997; Shiping et al. 1998; Hwang et al. 2004; Rawat et al. 2013b; Singh et al. 2020; Nguyen et al. 2021). However, commercial production of cell or hairy root cultures in bioreactors required precise process development methods. Cell cultures are susceptible to shear stress that causes disorganization of cells leading to callus formation and lower biomass productivity. Although, it has been found useful for alkaloid production from Catharanthus trichophyllus, C. roseus, and many other transformed plants (Davioud et al 1989; Toivonen et al. 1989; Eibl and Eibl 2002; Sevón and Oksman-Caldentey 2002). Currently, newly emerged mathematical modeling for large-scale bioreactors can be helpful for designing the scale-up conditions and projecting the impact of a large-scale environment on biomass and metabolites’ production (Straathof et al 2019). Furthermore, pathway engineering, culturing of genetically transformed cells, and use of elicitors are other research areas that need to be explored in Aconitum species.
More than 70 traditional or modern processing methods are available for the detoxification of aconitum poison, and it is claimed that this processing increased the efficacy of extract of preparations after the structural modification in molecules. The stability and bioactivity of diterpene alkaloids under different temperature conditions and processing methods have not been carried out. Thus, more emphasis is needed on the analysis of derivatives of diterpene alkaloids generated through processing. Without proper analytical mapping and quantification, the safety and efficacy of aconitine-containing drugs for human use cannot be determined (Borcsa et al. 2011). A quantitative assessment of potent molecules after different processing techniques and assurance of quality will reduce the risk associated with the toxicity of the plant. Furthermore, optimization of suitable potent molecule recovery methods from different Aconitum species in extraction using Response Surface Methodology (RSM), Taguchi method, or other techniques might be useful for improving yield and economic value (Wu et al. 2018).
Thus, the review analysis revealed that the anti-inflammatory, analgesic activity, anticancer, and cytotoxicity activities of extract and some diterpene alkaloids are well explored. Further, research needs to be carried out on the pharmacological activity of many isolated compounds. Identification of suitable drug delivery systems for reducing the toxic effects on the body and bioprocessing methods for reducing the toxicity needs to be explored and developed. Production of important metabolite through calls, callus, or Agrobacterium rhizogenes-mediated genetic transformation or other advanced techniques, and their optimum recovery process needs to be developed for production of higher content of active metabolites. Among the available options, exploration of existing genetic resources, and information on genomic resources might be very important. Identification of candidate genes and transcription factors determining and regulating biosynthetic pathways related to quality might be important. Lack of genomic information is a hurdle in the current scenario, and modern genomics, transcriptomics or metabolomics techniques can help understand the inheritance of quality-related traits, trait-specific functional marker development for genetic characterization, and molecular breeding. Development of varieties for cultivation, agro-technologies for maintaining the quality of products, robust propagation methods for propagation for farmers, and temporal monitoring of the population status in wild for ascertaining conservation have been identified for future research. Thus, this review would be helpful in new drug discoveries, disease management, conservation of genetic resources, and economic upliftment of farmers.
Acknowledgements
SR thanks Director GBPNIHE for providing facilities and encouragement. This work received partial financial support from the Institute as In-house Project No-04.
Author contributions
SR and ST conceived the idea and designed the study. ST, PK, BS, AKP, and SR compiled the database and wrote the manuscript. All authors contributed to the editing and critical revision of the manuscript.
Funding
MoEF&CC (Project number 02).
Data availability statement
Authors confirmed that no primary or secondary data was used in this review.
Declarations
Conflict of interest
The authors declared that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- Achmatowicz O, Jr, Marion L. The structures of two new alkaloids: chasmaconitine and chasmanthinine. Can J Chem. 1964;42:154–159. doi: 10.1139/v64-021. [DOI] [Google Scholar]
- Adams SJ, Kuruvilla GR, Krishnamurthy KV, Nagarajan M, Venkatasubramanian P. Pharmacognostic and phytochemical studies on Ayurvedic drugs Ativisha and Musta. Rev Bras. 2013;23:398–409. doi: 10.1590/S0102-695X2013005000040. [DOI] [Google Scholar]
- Ahmad M, Ahmad W, Ahmad M, Zeeshan M, Obaidullah Shaheen F. Norditerpenoid alkaloids from the roots of Aconitum heterophyllum Wall with antibacterial activity. J Enzyme Inhib Med Chem. 2008;23:1018–1022. doi: 10.1080/14756360701810140. [DOI] [PubMed] [Google Scholar]
- Ali S, Chouhan R, Sultan P, Hassan QP, Gandhi SG. A comprehensive review of phytochemistry, pharmacology and toxicology of the genus Aconitum L. Adv Tradit Med. 2021;2021:534. doi: 10.1007/s13596-021-00565-8. [DOI] [Google Scholar]
- Ameri A. Effects of the alkaloids 6-benzoylheteratisine and heteratisine on neuronal activity in rat hippocampal slices. Neuropharmacology. 1997;36:1039–1046. doi: 10.1016/s00283908(97)00095-6. [DOI] [PubMed] [Google Scholar]
- Ameri A. The effects of Aconitum alkaloids on the central nervous system. Prog Neurobiol. 1998;56:211–235. doi: 10.1016/s0301-0082(98)00037-9. [DOI] [PubMed] [Google Scholar]
- Ameri A. Structure-dependent inhibitory action of the Aconitum alkaloids 14-benzoyltalitasamine and talitasamine in rat hippocampal slices. Naunyn Schmiedebergs Arch Pharmacol. 1998;357:585–592. doi: 10.1007/pl00005212. [DOI] [PubMed] [Google Scholar]
- 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. doi: 10.1038/sj.bjp.0702100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atta-ur-Rahman FN, Akhtar F, Choudhary MI, Khalid A. New norditerpenoid alkaloids from Aconitum falconeri. J Nat Prod. 2000;63:1393–1395. doi: 10.1021/np9905315. [DOI] [PubMed] [Google Scholar]
- Bai S, Sartagnuud S, Wang TY, Bao GH, Bao SY, Ao W. De novo transcriptome sequencing identifies genes involved in aconitine-type alkaloids biosynthesis in Aconitum kusnezoffii Reichb. Pharmacol Res-Mod Chin Med. 2022;2:100063. doi: 10.1016/j.prmcm.2022.100063. [DOI] [Google Scholar]
- Been A. Aconitum: Genus of powerful and sensational plants. Pharm Hist. 1992;34(1):35–39. [PubMed] [Google Scholar]
- Beigh SY, Nawchoo IA, Iqbal M. Cultivation and conservation of Aconitum heterophyllum: A critically endangered medicinal herb of the northwest Himalayas. J Herbs Spices Med Plants. 2006;11:47–56. doi: 10.1300/J044v11n04_06. [DOI] [Google Scholar]
- Bello-Ramírez AM, Buendía-Orozco J, Nava-Ocampo AA. A QSAR analysis to explain the analgesic properties of Aconitum alkaloids. Fundam Clin Pharmacol. 2003;17:575–580. doi: 10.1046/j.1472-8206.2003.00189.x. [DOI] [PubMed] [Google Scholar]
- Bessonova IA, Saidkhodzhaeva SA. Hetisane-type diterpenoid alkaloids. Chem Nat Compd. 2000;36:419–477. doi: 10.1023/A:1002808721838. [DOI] [Google Scholar]
- Bisht VK, Negi JS, Bhandari AK, Sundriyal RC. Traditional use of medicinal plants in district Chamoli, Uttarakhand, India. J Med Plants Res. 2013;7:918–929. doi: 10.5897/JMPR13.2599. [DOI] [Google Scholar]
- Borcsa B, Csupor D, Forgo P, Widowitz U, Bauer R, Hohmann J. Aconitum lipo-alkaloids–Semisynthetic products of the traditional medicine. Nat Prod Commun. 2011;6:527–536. doi: 10.1177/1934578X1100600413. [DOI] [PubMed] [Google Scholar]
- Brinckmann JA. Sustainable Sourcing: Markets for certified Chinese medicinal and aromatic Plants. Geneva: International Trade Centre; 2016. p. 22. [Google Scholar]
- Brink DE, Woods RS, Stern KR. Bulbiferous Aconitum (Ranunculaceae) of the western United States. Sida. 1994;16:9–15. [Google Scholar]
- Cadre SL, Boisselier-Dubayle MC, Lambourdiere J, Machon N, Moret J, Samadi S. Polymorphic microsatellites for the study of Aconitum napellus L. (Ranunculaceae), a rare species in France. Mol Ecol Notes. 2005;5:358–360. doi: 10.1111/j.1471-8286.2005.00925.x. [DOI] [Google Scholar]
- Chan TY. Aconite poisoning. Clin Toxicol. 2009;47:279–285. doi: 10.1080/15563650902904407. [DOI] [PubMed] [Google Scholar]
- Chan TY. Aconitum alkaloid poisoning related to the culinary uses of aconite roots. Toxins. 2014;6:2605–2611. doi: 10.3390/toxins6092605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandra B (2003) Studies on propagation, agrotechnology and phytochemical evaluation of some alpine medicinal plants of Himalayan region. Ph.D. thesis, Kumaun University, Nainital
- Chen X, Li Q, Li Y, Qian J, Han J. Chloroplast genome of Aconitum barbatum var. puberulum (Ranunculaceae) derived from CCS reads using the PacBio RS platform. Front Plant Sci. 2015;6:42. doi: 10.3389/fpls.2015.00042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng ZD, He J, Zhang YM, Yang CW, Ma XX, Li GD. The complete chloroplast genome sequence of Aconitum austroyunnanense WT Wang (Ranunculaceae): a medicinal plant endemic to China. Mitochondrial DNA Part B. 2020;5:248–249. doi: 10.1080/23802359.2019.1700195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cherney EC, Lopchuk JM, Green JC, Baran PS. A unified approach to ent-atisane diterpenes and related alkaloids: synthesis of (−)-methyl atisenoate, (−)-isoatisine and the hetidine skeleton. J Am Chem Soc. 2014;136:12592–12595. doi: 10.1021/ja507321j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chhetree RR, Dash GK, Mondal S, Acharyya S. Studies on the hypoglycaemic activity of Aconitum napellus l. roots. Drug Invent Today. 2010;2:343–346. [Google Scholar]
- Chodoeva A, Bosc JJ, Guillon J, Decendit A, Petraud M, Absalon C, Vitry C, Jarry C, Robert J. 8-O-Azeloyl-14-benzoylaconine: a new alkaloid from the roots of Aconitum karacolicum Rapcs and its anti-proliferative activities. Bioorg Med Chem. 2005;13:6493–6501. doi: 10.1016/j.bmc.2005.07.015. [DOI] [PubMed] [Google Scholar]
- Cole CT, Kuchenreuther MA. Molecular markers reveal little genetic differentiation among Aconitum noveboracense and A. columbianum (Ranunculaceae) populations. Am J Bot. 2001;88:337–347. doi: 10.2307/2657023. [DOI] [PubMed] [Google Scholar]
- Colombo ML, Bravin M, Tome F. A study of the diterpene alkaloids of Aconitum napellus ssp. neomontanum during its onthogenetic cycle. Pharmacol Res Commun. 1988;20:123–128. doi: 10.1016/s0031-6989(88)80856-7. [DOI] [PubMed] [Google Scholar]
- Csupor D, Forgo P, Csedo K, Hohmann J. C19 and C20 Diterpene Alkaloids from Aconitum toxicum RCHB. Helv Chim Acta. 2006;89:2981–2986. doi: 10.1002/hlca.200690267. [DOI] [Google Scholar]
- Dar GH, Naqshi AR. Threatened flowering plants of the Kashmir Himalaya–a checklist. Orient Sci. 2001;6:23–53. [Google Scholar]
- Dar GH, Nordenstam B. Asteraceae in the Flora of Sind Valley, Kashmir Himalaya, India. Nelumbo. 2014;56:14–88. [Google Scholar]
- Dasyukevich OI, Solyanik GI. Comparative study of anticancer efficacy of aconitine containing agent BC1 against ascite and solid forms of Ehrlich’s carcinoma. Exp Oncol. 2007;29:317–319. [PubMed] [Google Scholar]
- Davioud E, Kan C, Hamon J, Tempe J, Husson HP. Production of indole alkaloids by in vitro root cultures from Catharanthus trichophyllus. Phytochemistry. 1989;28:2675–2680. doi: 10.1016/S0031-9422(00)98066-X. [DOI] [Google Scholar]
- Devkota KP, Sewald N. Terpenoid alkaloids derived by amination reaction. In: Ramawat K, Mérillon JM, editors. Natural products. Berlin, Heidelberg: Springer; 2013. pp. 923–951. [Google Scholar]
- Druka A, Potokina E, Luo Z, Jiang N, Chen X, Kearsey M, Waugh R. Expression quantitative trait loci analysis in plants. Plant Biotechnol J. 2010;8:10–27. doi: 10.1111/j.1467-7652.2009.00460.x. [DOI] [PubMed] [Google Scholar]
- Du CH. AFLP analysis of genetic diversity of Aconitum brachypodum. Chin Tradit Herbal Drugs. 2018 doi: 10.1111/j.1467-7652.2009.00460.x. [DOI] [Google Scholar]
- Eibl R, Eibl D (2002) Bioreactors for plant cell and tissue cultures. In: Plant biotechnology and transgenic plants. CRC Press. pp 152–183
- Faber GM, Rudy Y. Action potential and contractility changes in [Na+]i overloaded cardiac myocytes: a simulation study. Biophys J. 2000;78:2392–2404. doi: 10.1016/S0006-3495(00)76783-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fico G, Braca A, Morelli I, Tome F. Flavonol glycosides from Aconitum Vulparia. Fitoterapia. 2003;74:420–422. doi: 10.1016/s0367-326x(03)00045-5. [DOI] [PubMed] [Google Scholar]
- Frejat FOA, Xu W, Shan L, Zhou X, Zhou XL. Three new lactone-type diterpenoid alkaloids from Aconitum rotundifolium Kar, Kir. Heterocycles. 2017;94:1903–1908. doi: 10.3987/COM-17-13768. [DOI] [Google Scholar]
- Fu YP, Li CY, Peng X, Zou YF, Rise F, Paulsen BS, et al. Polysaccharides from Aconitum carmichaelii leaves: Structure, immunomodulatory and anti-inflammatory activities. Carbohyd Polym. 2022;291:119655. doi: 10.1016/j.carbpol.2022.119655. [DOI] [PubMed] [Google Scholar]
- Gao LM, Yan HY, He YQ, Wei XM. Norditerpenoid alkaloids from Aconitum spicatum Stapf. J Integr Plant Biol. 2006;48:364–369. doi: 10.1080/14786419.2015.1114941. [DOI] [Google Scholar]
- Gao T, Bi H, Ma S, Lu J. The antitumor and immunostimulating activities of water soluble polysaccharides from Radix Aconiti, Radix Aconiti Lateralis and Radix Aconiti Kusnezoffii. Nat Prod Commun. 2010 doi: 10.1177/1934578X1000500322. [DOI] [PubMed] [Google Scholar]
- Gao T, Ma S, Song J, Bi H, Tao Y. Antioxidant and immunological activities of water-soluble polysaccharides from Aconitum kusnezoffii Reichb. Int J Biol Macromol. 2011;49:580–586. doi: 10.1016/j.ijbiomac.2011.06.017. [DOI] [PubMed] [Google Scholar]
- Gao F, Li YY, Wang D, Huang X, Liu Q. Diterpenoid alkaloids from the Chinese traditional herbal “Fuzi” and their cytotoxic activity. Molecules. 2012;17:5187–5194. doi: 10.3390/molecules17055187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao H, Huang Z, Li M, Zhang X, Yan Y, Cui L. Quantitative assays of two soil-borne pathogens of Aconitum carmichaelii Debx, Sclerotium rolfsii and Mucor circinelloides, in the main cultivation areas of China. J Appl Res Med Aromat Plants. 2021;25:100343. doi: 10.1016/j.jarmap.2021.100343. [DOI] [Google Scholar]
- Ge XY, Tian H, Liao WJ. Characterization of 19 microsatellite loci in the clonal monkshood Aconitum kusnezoffii (Ranunculaceae) Appl Plant Sci. 2016;4:1500141. doi: 10.3732/apps.1500141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giri A, Ahuja PS, Kumar PVA. Somatic embryogenesis and plant regeneration from callus cultures of Aconitum heterophyllum Wall. Plant Cell Tissue Organ Cult. 1993;32:213–218. doi: 10.1007/BF00029845. [DOI] [Google Scholar]
- Giri A, Banerjee S, Ahuja PS, Giri C. Production of hairy roots in Aconitum heterophyllum Wall: using Agrobacterium rhizogenes. In Vitro Cell Dev Biol Plant. 1997;33:280–284. doi: 10.1007/s11627-997-0050-6. [DOI] [Google Scholar]
- Gondval M, Chaturvedi P, Gaur AK. Thidiazuron-induced high frequency establishment of callus cultures and plantlet regeneration in Aconitum balfourii Stapf.: an endangered medicinal herb of North-West Himalayas. Indian J Biotech. 2016;15:251–255. [Google Scholar]
- 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. doi: 10.1248/cpb.c21-00262. [DOI] [Google Scholar]
- Gupta AK, Souravi K. Access and benefit sharing and threatened medicinal plants. In: Rajasekharan PE, Wani SH, editors. Conservation and utilization of threatened medicinal plants. Cham: Springer; 2020. pp. 513–529. [Google Scholar]
- Gupta R, Saxena R, Malviya N. Investigation of anti-inflammatory activity of ethanolic extract of Aconitum napellus Linn against carrageenan induced paw edema in rats. J Drug Deliv Therap. 2019;9:470–472. doi: 10.22270/jddt.v9i3.2892. [DOI] [Google Scholar]
- Hanuman JB, Katz A. Isolation and identification of four norditerpenoid alkaloids from processed and unprocessed root tubers of Aconitum ferox. J Nat Prod. 1993;56:801–809. doi: 10.1021/NP50096A001. [DOI] [Google Scholar]
- Hao DC, Gu XJ, Xiao PG (2015) Chemical and biological studies of Aconitum pharmaceutical resources. In: Medicinal plants: chemistry. Biology and Omics Elsevier, Cambridge, pp 253–292. 10.1016/B978-0-08-100085-4.00007-4
- Hatano K, Shoyama Y, Nishioka I. Somatic embryogenesis and plant regeneration from the anther of Aconitum carmichaeli Debx. Plant Cell Rep. 1987;6:446–448. doi: 10.1007/BF00272779. [DOI] [PubMed] [Google Scholar]
- Hatano K, Kamura K, Shoyama Y, Nishioka I. Clonal multiplication of Aconitum carmichaeli by tip tissue culture and alkaloid contents of clonally propagated plant. Planta Med. 1988;54:152–155. doi: 10.1055/s-2006-962375. [DOI] [PubMed] [Google Scholar]
- He J, Zhang ZR, Yang JB, Wang H, Meng J. Isolation and characterization of 18 microsatellites for Aconitum vilmorinianum Kom. (Ranunculaceae) using next-generation sequencing technology. Conserv Genet Resour. 2015;7:579–581. doi: 10.1007/s12686-015-0432-8. [DOI] [Google Scholar]
- Hong Y, Luo Y, Gao Q, Ren C, Yuan Q, Yang QE. Phylogeny and reclassification of Aconitum subgenus Lycoctonum (Ranunculaceae) PLoS ONE. 2017;12:e0171038. doi: 10.1371/journal.pone.0171038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hou M, Du GZ, Zhao ZG. Development of genomic microsatellite markers for Aconitum gymnandrum(Ranunculaceae) by next generation sequencing (NGS) Mol Biol Rep. 2020;47:727–729. doi: 10.1007/s11033-019-05160-4. [DOI] [PubMed] [Google Scholar]
- Huang Q, Wang D, Dong L (2011) Analgesic and anti-inflammatory activities of extracts from Aconitum pendulum Busch in Mice. Ningxia Med J 11
- Huang XJ, Ren W, Li J, Chen LY, Mei ZN. Anti-inflammatory and anticancer activities of ethanol extract of pendulous monkshood root in vitro. Asian Pac J Cancer Prev. 2013;14(6):3569–3573. doi: 10.7314/apjcp.2013.14.6.3569. [DOI] [PubMed] [Google Scholar]
- Hu ZX, Tang HY, Yan XH, Zeng YR, Aisa HA, Zhang Y, Hao XJ. Five new alkaloids from Aconitum apetalum (Ranunculaceae) Phytochem Lett. 2019;29:6–11. doi: 10.1016/j.phytol.2018.10.017. [DOI] [Google Scholar]
- Hwang SJ, Kim YH, Pyo BS. Optimization of aconitine production in suspension cell cultures of Aconitum napellus L. Korean J Med Crop Sci. 2004;12:366–371. [Google Scholar]
- Isono T, Oyama T, Asami A, Suzuki Y, Hayakawa Y, Ikeda Y, et al. The analgesic mechanism of processed Aconiti tuber: the involvement of descending inhibitory system. Am J Chin Med. 1994;22:83–94. doi: 10.1142/S0192415X94000115. [DOI] [PubMed] [Google Scholar]
- Ito K, Ohyama Y, Hishinuma T, Mizugaki M. Determination of Aconitum alkaloids in the tubers of Aconitum japonicum using gas chromatography/selected ion monitoring. Planta Med. 1996;62:57–59. doi: 10.1055/s-2006-957798. [DOI] [PubMed] [Google Scholar]
- IUCN (2020) The IUCN red list of threatened species. Version 2020–1. https://www.iucnredlist.org. Accessed Apr 4 2022
- Jabeen N, Shawl AS, Dar GH, Jan A, Sultan P. Callus induction and organogenesis from explants of Aconitum heterophyllum medicinal plant. Biotechnology. 2006;5:287–291. doi: 10.3923/biotech.2006.287.291. [DOI] [Google Scholar]
- Jabeen N, Kozgar MI, Dar GH, Shawl AS, Khan S. Distribution and taxonomy of genus Aconitum in Kashmir: potent medicinal resource of Himalayan valley. Chiang Mai J Sci. 2013;40(2):173–186. [Google Scholar]
- Jiang Q, Pelletier SW. Two new diterpenoid alkaloids from Aconitum palmatum. J Nat Prod. 1991;54:525–531. doi: 10.1021/np50074a027. [DOI] [Google Scholar]
- Jiang SH, Wang HQ, Li YM, Lin SJ, Tan JJ. Two new C18-norditerpenoid alkaloids from Aconitum delavayi. Chin Chem Lett. 2007;18:409–411. doi: 10.1016/j.cclet.2007.01.031. [DOI] [Google Scholar]
- Jiang ZB, Guo HH, Hu YQ, Zhou LR, Deng CF, Nan ZD, Ma XL, Wu XL (2022) Classification of diterpenoid alkaloids from Aconitum kusnezoffii Reichb. by liquid chromatography- tandem mass spectrometry- based on molecular networking. J Sep Sci 45:739–751. 10.1002/jssc.202100651 [DOI] [PubMed]
- Ju HJ, Yoo TK, Jin S, Kim H, Hyun TK. In vitro evaluation of the pharmacological properties of crude methanol extract and its fractions of Aconitum austrokoreense aerial parts. Revista Mexicana De Ingeniería Química. 2020;19:1341–1350. doi: 10.24275/rmiq/Bio1067. [DOI] [Google Scholar]
- Jung HS, Song BY, Lee CH, Yook TH. Effects of Cinnamomum cassia and Aconitum carmichaeli's Pharmacopuncture and oral administration on blood sugar in type II diabetic mice. J Acupunct Res. 2010;27:1–12. [Google Scholar]
- Kadota Y (1987) A revision of Aconitum subgenus Aconitum (Ranunculaceae) in East Asia. Sanwa Shoyaku Company
- Kawasaki R, Motoya W, Atsumi T, Mouri C, Kakiuchi N, Mikage M. The relationship between growth of the aerial part and alkaloid content variation in cultivated Aconitum carmichaeli Debeaux. J Nat Med. 2011;65:111–115. doi: 10.1007/s11418-010-0466-x. [DOI] [PubMed] [Google Scholar]
- Khetwal KS. Constituents of high altitude Himalayan herbs. part XX. A C-19 diterpenoid alkaloid from Aconitum balfourii. Indian J Chem. 2007;46B:1364–1366. [Google Scholar]
- Khetwal KS, Pande S. Constitutes of high altitude Himalayan herbs part XV: a new norditerpenoid alkaloid from the roots of Aconitum balfourii. Nat Prod Res. 2004;18:129–133. doi: 10.1080/1487641031000149885. [DOI] [PubMed] [Google Scholar]
- Kim DK, Kwon HY, Lee KR, Rhee DK, Zee OP. Isolation of a multidrug resistance inhibitor from Aconitum pseudo-laeve var. erectum. Arch Pharmacal Res. 1998;21:344–347. doi: 10.1007/BF02975299. [DOI] [PubMed] [Google Scholar]
- Kim JH, Lee SY, Kwon OJ, Park JH, Lee JY. Anti-aging and anti-diabetes effects of Aconitum pesudo-laeve var. erectum extracts. J Life Sci. 2013;23:616–621. doi: 10.5352/JLS.2013.23.5.616. [DOI] [Google Scholar]
- Kim Y, Yi JS, Min J, Xi H, Kim DY, Son J, et al. The complete chloroplast genome of Aconitum coreanum (H. Lév.) Rapaics (Ranunculaceae) Mitochondrial DNA Part B. 2019;4:3404–3406. doi: 10.1080/23802359.2019.1674213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kita Y, Ito M. Nuclear ribosomal ITS sequences and phylogeny of East Asian Aconitum subgen. Aconitum (Ranunculaceae), with special reference to extensive polymorphism in individual plants. Plant Syst Evol. 2000;225:1–13. doi: 10.1007/BF00985455. [DOI] [Google Scholar]
- Kocot D, Nowak B, Sitek E, Starzyńska-Janiszewska A, Mitka J. In vitro shoot regeneration from organogenic callus culture and rooting of Carpathian endemic Aconitum bucovinense Zapał. Research Square. 2022 doi: 10.21203/rs.3.rs-1428440/v1. [DOI] [Google Scholar]
- Kong H, Liu W, Yao G, Gong W. A comparison of chloroplast genome sequences in Aconitum (Ranunculaceae): a traditional herbal medicinal genus. PeerJ. 2017;5:e4018. doi: 10.7717/peerj.4018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong H, Zhang Y, Hong Y, Barker MS. Multilocus phylogenetic reconstruction informing polyploid relationships of Aconitum subgenus Lycoctonum (Ranunculaceae) in China. Plant Syst Evol. 2017;303:727–744. doi: 10.1007/s00606-017-1406-y. [DOI] [Google Scholar]
- Kuchenreuther MA. The natural history of Aconitum noveboracense Gray (northern monkshood), a federally threatened species. J Iowa Acad Science. 1996;103:57–62. [Google Scholar]
- Kumar V, Malhotra N, Pal T, Chauhan RS. Molecular dissection of pathway components unravel atisine biosynthesis in a non-toxic Aconitum species, A. heterophyllum Wall. 3 Biotech. 2016;6:106. doi: 10.1007/s13205-016-0417-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumari A, Singh D, Kumar S. Biotechnological interventions for harnessing podophyllotoxin from plant and fungal species: current status, challenges, and opportunities for its commercialization. Crit Rev Biotechnol. 2017;37:739–753. doi: 10.1080/07388551.2016.1228597. [DOI] [PubMed] [Google Scholar]
- Lan YJ, Sam NB, Cheng MH, Pan HF, Gao J. Progranulin as a potential therapeutic target in immune-mediated diseases. J Inflamm Res. 2021;14:6543. doi: 10.2147/JIR.S339254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee SR, Choi JE, Lee BY, Yu JN, Lim CE. Genetic diversity and structure of an endangered medicinal herb: implications for conservation. AoB Plants. 2018;10:ply021. doi: 10.1093/aobpla/ply021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y. Dynamic changes of alkaloids in growth cycle in stems and leaves of Aconitum carmichaelii by HPLC-MS/MS. Chin Tradit Herbal Drugs. 2019;24:1985–1991. doi: 10.7501/j.issn.0253-2670.2019.08.032. [DOI] [Google Scholar]
- Li LQ, Kadota Y. Aconitum. St. Louis: Missouri Botanical Garden Press; 2001. pp. 149–222. [Google Scholar]
- Li M, He J, Jiang LL, Ng ESK, Wang H, Lam FFY, et al. The anti-arthritic effects of Aconitum vilmorinianum, a folk herbal medicine in Southwestern China. J Ethnopharmacol. 2013;147:122–127. doi: 10.1016/j.jep.2013.02.018. [DOI] [PubMed] [Google Scholar]
- Li YQ, Meng Y, Zhang J. Development of 12 microsatellite markers for Aconitum brachypodum (Ranunculaceae), a critically endangered and endemic medicinal plant. Biochem Syst Ecol. 2015;61:462–464. doi: 10.1016/j.bse.2015.06.036. [DOI] [Google Scholar]
- Li YF, Zheng YM, Yu Y, Gan Y, Gao ZB. Inhibitory effects of lappaconitine on the neuronal isoforms of voltage-gated sodium channels. Acta Pharmacol Sin. 2019;40:451–459. doi: 10.1038/s41401-018-0067-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Q, Li X, Qieyang R, Nima C, Dongzhi D, Duojie GX. Characterization of the complete chloroplast genome of the Tangut monkshood Aconitum tanguticum (Ranunculales: Ranunculaceae) Mitochondrial DNA Part B. 2020;5:2306–2307. doi: 10.1080/23802359.2020.1773338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S, Yu L, Shi Q, Liu Y, Zhang Y, Wang S, Lai X. An insight into current advances on pharmacology, pharmacokinetics, toxicity and detoxification of aconitine. Biomed Pharmacother. 2022;151:113115. doi: 10.1016/j.biopha.2022.113115. [DOI] [PubMed] [Google Scholar]
- Liang M, Li S, Shen B, Cai J, Li C, Wang Z, et al. Anti-hepatocarcinoma effects of Aconitum coreanum polysaccharides. Carbohyd Polym. 2012;88:973–976. doi: 10.1016/j.carbpol.2012.01.050. [DOI] [Google Scholar]
- Liang Y, Wu JL, Li X, Guo MQ, Leung ELH, Zhou H, et al. Anti-cancer and anti-inflammatory new vakognavine-type alkaloid from the roots of Aconitum carmichaelii. Tetrahedron Lett. 2016;57:5881–5884. doi: 10.1016/j.tetlet.2016.11.065. [DOI] [Google Scholar]
- Liang Y, Yan GY, Wu JL, Zong X, Liu Z, Zhou H, et al. Qualitative and quantitative analysis of lipo-alkaloids and fatty acids in Aconitum carmichaelii using LC–MS and GC–MS. Phytochem Anal. 2018;29:398–405. doi: 10.1002/pca.2760. [DOI] [PubMed] [Google Scholar]
- Liang-Qian L. On distributional features of the genus Aconitum in Sino-Himalayan Flora. J Syst Evol. 1988;26:189–204. [Google Scholar]
- Lim CC, Kitto SL. Micropropagation of Aconitum uncinatum: growth regulator and antioxidant screening using the surface response analysis method. HortScience. 1995;30:186. doi: 10.21273/HORTSCI.30.2.186a. [DOI] [Google Scholar]
- Lim CE, Ryul BK, Lee JD, Jung KD, Noh TK, Lee BY. The complete chloroplast genome of Aconitum puchonroenicum Uyeki & Sakata (Ranunculaceae), a rare endemic species in Korea. Mitochondrial DNA Part B. 2020;5:1284–1285. doi: 10.1080/23802359.2020.1734497. [DOI] [Google Scholar]
- Lin MW, Wang YJ, Liu SI, Lin AA, Lo YC, Wu S. Characterization of aconitine-induced block of delayed rectifier K+ current in differentiated NG108-15 neuronal cells. Neuropharmacology. 2008;54:912–923. doi: 10.1016/j.neuropharm.2008.01.009. [DOI] [PubMed] [Google Scholar]
- Liu WL, Liu ZQ, Song FR, Liu SY. Specific conversion of diester-diterpenoid Aconitum alkaloids components into hydrolysis monoester-diterpenoid alkaloids components and lipo-alkaloids components. Chem J Chin Univ. 2007;3:717–720. [Google Scholar]
- Liu YG, Liu Q, Zhang HG. Studies on hydrolysates of aconitine by HPLC-MS. Chin New Drugs J. 2007;16:303–305. [Google Scholar]
- Liu XX, Jian XX, Cai XF, Chao RB, Chen QH, Chen DL, et al. Cardioactive C19-diterpenoid alkaloids from the lateral roots of Aconitum carmichaeli “Fu Zi”. Chem Pharm Bull. 2012;60:144–149. doi: 10.1248/cpb.60.144. [DOI] [PubMed] [Google Scholar]
- Liu S, Li F, Li Y, Li W, Xu J, Du H. A review of traditional and current methods used to potentially reduce toxicity of Aconitum roots in Traditional Chinese Medicine. J Ethnopharmacol. 2017;207:237–250. doi: 10.1016/j.jep.2017.06.038. [DOI] [PubMed] [Google Scholar]
- Liu Y, Yu S, You F. Characterization of the complete chloroplast genome of Aconitum flavum (Ranunculaceae) Mitochondrial DNA Part B. 2020;5:2982–2983. doi: 10.1248/cpb.60.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lone PA, Bhardwaj AK, Shah KW, Tabasum S. Ethnobotanical survey of some threatened medicinal plants of Kashmir Himalaya, India. J Med Plants Res. 2014;8:1362–1373. doi: 10.5897/JMPR2014.5649. [DOI] [Google Scholar]
- Luo Q, Ma D, Wang Y. ISSR identification of genetic diversity in Aconitum carmichaeli. Chin Tradit Herbal Drugs. 1994;24:wpr-575843. [Google Scholar]
- Luo Y, Zhang FM, Yang QE. Phylogeny of Aconitum subgenus Aconitum (Ranunculaceae) inferred from ITS sequences. Plant Syst Evol. 2005;252:11–25. doi: 10.1007/s00606-004-0257-5. [DOI] [Google Scholar]
- Mahajan R, Kapoor N, Singh I. Effect of growth regulators on in vitro cultures of Aconitum heterophyllum: an endangered medicinal plant. Int J Pure Appl Biosci. 2015;3:50–55. doi: 10.18782/2320-7051.2138. [DOI] [Google Scholar]
- Malhotra N, Sood H, Chauhan RS. Transcriptome-wide mining suggests conglomerate of genes associated with tuberous root growth and development in Aconitum heterophyllum Wall. 3 Biotech. 2016;6:1–8. doi: 10.1007/s13205-016-0466-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazur NA, Ivanova LA, Pavlova TS. Results of the clinical study of a new anti-arrhythmia preparation allapinin. Biull Vsesoiuznogo Kardiol Nauchn Tsentra AMN SSSR. 1986;9:30–33. [PubMed] [Google Scholar]
- Meng F, Peng M, Wang R, Wang C, Guan F. Analysis of genetic diversity in Aconitum kongboense L. revealed by AFLP markers. Biochem Syst Ecol. 2014;57:388–394. doi: 10.1016/j.bse.2014.09.013. [DOI] [Google Scholar]
- Meng F, Wang R, Peng M, Wang C, Wang Z, Guan F, Li Y. Evaluation of genetic diversity among Kongpo Monkshood ( Aconitum kongboense L.) germplasm accessions revealed by inter simple sequence repeat markers. HortScience. 2015;50:940–943. doi: 10.21273/HORTSCI.50.7.940. [DOI] [Google Scholar]
- Meng J, Li X, Li H, Yang J, Wang H, He J. Comparative analysis of the complete chloroplast genomes of four Aconitum medicinal species. Molecules. 2018;23:1015. doi: 10.3390/molecules23051015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng J, Zhang L, Li X, He J. The complete plastid genome sequence of Aconitum brachypodum (Ranunculaceae): an endangered species endemic to China. Mitochondrial DNA Part B. 2019;4:130–131. doi: 10.1080/23802359.2018.1540264. [DOI] [Google Scholar]
- Mitka J. Systematyka Aconitum subgen. Aconitum w Karpatach Wschodnich [Systematics of Aconitum subgen. Aconitum in the Eastern Carpathians] Roczniki Bieszczadzkie. 2000;9:79–116. [Google Scholar]
- Mitka J, Boron P, Wróblewska A, Baba W. AFLP analysis reveals infraspecific phylogenetic relationships and population genetic structure of two species of Aconitum in Central Europe. Acta Soc Bot Pol. 2015;84:267–276. doi: 10.5586/asbp.2015.012. [DOI] [Google Scholar]
- Mitka J, Novikov A, Rottensteiner WK. The taxonomic circumscription of Aconitum subgenus Aconitum (Ranunculaceae) in Europe. Webbia J Plant Taxon Geogr. 2021;76:11–45. doi: 10.36253/jopt-10006. [DOI] [Google Scholar]
- Mizugaki M, Ito K. Aconite toxins. In: Suzuki O, Watanabe K, editors. Drugs and poisons in humans—a handbook of practical analysis. New York: Springer; 2005. pp. 456–467. [Google Scholar]
- Mu ZQ, Gao H, Huang ZY, Feng XL, Yao XS. Puberunine and puberudine, two new C18-diterpenoid alkaloids from Aconitum barbatum var. puberulum. Org Lett. 2012;14:2758–2761. doi: 10.1021/ol3008217. [DOI] [PubMed] [Google Scholar]
- Nesterova YV, Povetieva TN, Suslov NI, Zyuz'Kov GN, Aksinenko SG, Pushkarskii SV, Krapivin AV. Anti-inflammatory activity of diterpene alkaloids from Aconitum baikalense. Bull Exp Biol Med. 2014;156:665. doi: 10.1007/s10517-014-2421-4. [DOI] [PubMed] [Google Scholar]
- Nguyen TNL, Hoang TTH, Nguyen HQ, Tu QT, Tran TH, Lo TMT, et al. Agrobacterium tumefaciens–mediated genetic transformation and overexpression of the flavonoid 3′ 5′-hydroxylase gene increases the flavonoid content of the transgenic Aconitum carmichaelii Debx. plant. In Vitro Cell Dev Biol Plant. 2021;58:93–102. doi: 10.1007/s11627-021-10190-4. [DOI] [Google Scholar]
- Ni X, Li J, Li Y, Zhang H, Duan B, Chen X, Xia C. The complete chloroplast genome of Aconitum piepunense (Ranunculaceae) and its phylogenetic analysis. Mitochondrial DNA Part B. 2022;7:115–117. doi: 10.1080/23802359.2021.2011448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nigmatullaev AM, Salimov BT. Method of isolating and separating individual alkaloids from the above-ground parts of Aconitum zeravschanicum Steinb. Rastitel'nye Resursy. 2000;36:118–121. [Google Scholar]
- Nisar M, Obaidullah Ahmad M, Wadood N, Lodhi MA, Shaheen F, Choudhary MI. New diterpenoid alkaloids from Aconitum heterophyllum Wall: selective butyrylcholinestrase inhibitors. J Enzyme Inhib Med Chem. 2009;24:47–51. doi: 10.1080/14756360801906202. [DOI] [PubMed] [Google Scholar]
- 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. doi: 10.1111/jphp.12310. [DOI] [PubMed] [Google Scholar]
- Olsen CS, Larsen HO. Alpine medicinal plant trade and Himalayan mountain livelihood strategies. Geogr J. 2003;169:243–254. doi: 10.1111/1475-4959.00088. [DOI] [Google Scholar]
- Pal T, Malhotra N, Chanumolu SK, Chauhan RS. 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. doi: 10.1007/s00425-015-2304-6. [DOI] [PubMed] [Google Scholar]
- Pandey H, Nandi SK, Kumar A, Palni UT, Chandra B, Palni LMS. In vitro propagation of Aconitum balfourii Stapf.: an important aconite of the Himalayan alpines. J Hortic Sci Biotechnol. 2004;79:34–41. doi: 10.1080/14620316.2004.11511733. [DOI] [Google Scholar]
- Park KH, Park M, Choi SE, Jeong MS, Kwon JH, Oh MH, et al. The Anti-oxidative and Anti-inflammatory Effects of Caffeoyl Derivatives from the Roots of Aconitum koreanum R. R AYMOND. Biolog Pharmac Bull. 2009;32:2029–2033. doi: 10.1248/bpb.32.2029. [DOI] [PubMed] [Google Scholar]
- Park I, Kim WJ, Yang S, Yeo SM, Li H, Moon BC. The complete chloroplast genome sequence of Aconitum coreanum and Aconitum carmichaelii and comparative analysis with other Aconitum species. PLoS ONE. 2017;12:e0184257. doi: 10.1371/journal.pone.0184257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park I, Yang S, Choi G, Kim WJ, Moon BC. The complete chloroplast genome sequences of Aconitum pseudolaeve and Aconitum longecassidatum, and development of molecular markers for distinguishing species in the Aconitum Subgenus Lycoctonum. Molecules. 2017;22:2012. doi: 10.3390/molecules22112012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parvez M, Gul W, Anwar S. Chasmanthinine. Acta Crystallogr C. 1998;54:125–126. doi: 10.1107/S0108270197013449. [DOI] [Google Scholar]
- Prasad SK, Jain D, Patel DK, Sahu AN, Hemalatha S. Antisecretory and antimotility activity of Aconitum heterophyllum and its significance in treatment of diarrhea. Indian J Pharmacol. 2014;46:82. doi: 10.4103/0253-7613.125182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Priyanka S, Priyanka S. Optimization of conditions for in vitro seed germination and shoot multiplication of Aconitum heterophyllum Wall. Int J Med Aromat Plants. 2012;2:481–487. [Google Scholar]
- Pullaiah T. Encyclopaedia of world medicinal plants. New Delhi: Regency Publications; 2006. [Google Scholar]
- Rafiq S, Wagay NA, Bhat IA, Kaloo ZA, Rashid S, Lin F, El-Abedin TKZ, Wani SH, Mahmoud EA, Almutairi KF, Elansary HO (2021) In vitro propagation of Aconitum chasmanthum Stapf Ex Holmes: an endemic and critically endangered plant species of the western Himalaya. Horticulturae 7:586. 10.3390/horticulturae7120586
- Rai M, Ra A, Kawan N, Yoshimats K, Takahash H, Suzuki H, et al. De novo RNA sequencing and expression analysis of Aconitum carmichaelii to analyze key genes involved in the biosynthesis of diterpene alkaloids. Molecules. 2017;22:2155. doi: 10.3390/molecules22122155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rana B, Sreenivasulu Y. Protein changes during ethanol induced seed germination in Aconitum heterophyllum. Plant Sci. 2013;198:27–38. doi: 10.1016/j.plantsci.2012.09.013. [DOI] [PubMed] [Google Scholar]
- Rawat JM, Rawat B, Chandra A, Nautiyal S. Influence of plant growth regulators on indirect shoot organogenesis and secondary metabolite production in Aconitum violaceum Jacq. Afric J Biotech. 2013;12:6287–6293. doi: 10.5897/AJB2013.13390. [DOI] [Google Scholar]
- Rawat JM, Rawat B, Agnihotri RK, Chandra A, Nautiyal S. In vitro propagation, genetic and secondary metabolite analysis of Aconitum violaceum Jacq.: a threatened medicinal herb. Acta Physiol Plant. 2013;35:2589–2599. doi: 10.1007/s11738-013-1294-x. [DOI] [Google Scholar]
- Sevón N, Oksman-Caldentey KM. Agrobacterium rhizogenes-mediated transformation: root cultures as a source of alkaloids. Planta Med. 2002;68:859–868. doi: 10.1055/s-2002-34924. [DOI] [PubMed] [Google Scholar]
- Shah NC. Conservation aspects of Aconitum species in the Himalayas with special reference to Uttaranchal (India) Med Plant Conserv. 2005;11:9–15. [Google Scholar]
- Shaheen F, Ahmad M, Khan MTH, Jalil S, Ejaz A, Sultankhodjaev MN, et al. Alkaloids of Aconitum laeve and their anti-inflammatory, antioxidant and tyrosinase inhibition activities. Phytochemistry. 2005;66:935–940. doi: 10.1016/j.phytochem.2005.02.010. [DOI] [PubMed] [Google Scholar]
- Shang ZH, Tang Y, Long RJ. Allelopathic effect of Aconitum pendulum (Ranunculaceae) on seed germination and seedlings of five native grass species in the Tibetan plateau. Nord J Bot. 2011;29:488–494. doi: 10.1111/j.1756-1051.2011.01120.x. [DOI] [Google Scholar]
- Sharma H, Kumar P, Singh A, Aggarwal K, Roy J, Sharma V, Rawat S. Development of polymorphic EST-SSR markers and their applicability in genetic diversity evaluation in Rhododendron arboreum. Mol Biol Rep. 2020;47:2447–2457. doi: 10.1007/s11033-020-05300-1. [DOI] [PubMed] [Google Scholar]
- Shen Y, Liang WJ, Shi YN, Kennelly EJ, Zhao DK. Structural diversity, bioactivities, and biosynthesis of natural diterpenoid alkaloids. Nat Prod Rep. 2020;37:763–796. doi: 10.1039/d0np00002g. [DOI] [PubMed] [Google Scholar]
- Sheng LH, Xu M, Xu LQ, Xiong F. Cytotoxic effect of lappaconitine on non-small cell lung cancer in vitro and its molecular mechanism. J Chin Med Mater. 2014;37:840–843. [PubMed] [Google Scholar]
- Shiping C, Shan SJ, Tanaka H, Shoyama Y. Effects of culture temperature on micro-tuber formation of Aconitum carmichaelii Debx. and aconitine type Alkaloid contents. Biotronics. 1998;27:15–20. [Google Scholar]
- Shoaib A, Salem-Bekhit MM, Siddiqui HH, Dixit RK, Bayomi M, Khalid M, Shakeel F. Antidiabetic activity of standardized dried tubers extract of Aconitumnapellus in streptozotocin-induced diabetic rats. 3 Biotech. 2020;10:56. doi: 10.1007/s13205-019-2043-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shrestha BB, Jha PK. Life history and population status of the endemic Himalayan Aconitum naviculare. Mt Res Dev. 2010;30:353–364. doi: 10.1659/MRD-JOURNAL-D-10-00003.1. [DOI] [Google Scholar]
- Shyaula SL. Phytochemicals, traditional uses and processing of Aconitum species in Nepal. Nepal J Sci Technol. 2011;12:171–178. doi: 10.3126/njst.v12i0.6496. [DOI] [Google Scholar]
- Shyaula L, Tamang T, Ghouri N, Adhikari A, Marasini S, Bajracharya GB, et al. Antileishmanial diterpenoid alkaloids from Aconitum spicatum (Bruhl) Stapf. Nat Prod Res. 2016;30:2590–2593. doi: 10.1080/14786419.2015.1114941. [DOI] [PubMed] [Google Scholar]
- Sinam YM, Devi GS. Seasonal variation of bioactive alkaloid content in Aconitum spp. from Manipur. India Bioscan. 2011;6:439–442. [Google Scholar]
- Singh A, Kuniyal CP, Lata H, Rajasekaran C, Prasad P, Bhadula SK, Purohit AN. In vitro propagation of Aconitum atrox (Bruhl). Muk, a threatened medicinal herb from Garhwal Himalaya. Physiol Mol Biol Plants. 1998;4:171–174. [Google Scholar]
- Singh M, Chettri A, Pandey A, Sinha S, Singh KK, Badola HK. In vitro propagation and phytochemical assessment of Aconitum ferox wall: a threatened medicinal plant of Sikkim Himalaya. Proc Natl Acad Sci B. 2020;90(2):313–321. doi: 10.1007/s40011-019-01104-x. [DOI] [Google Scholar]
- Solyanik GI, Fedorchuk AG, Pyaskovskaya ON, Dasyukevitch OI, Khranovskaya NN, Aksenov GN, Sobetsky VV. Anticancer activity of aconitine-containing herbal extract BC1. Exp Oncol. 2004;26:307–311. [PubMed] [Google Scholar]
- Song JZ, Han QB, Qiao CF, But PPH, Xu HX. Development and validation of a rapid capillary zone electrophoresis method for the determination of aconite alkaloids in aconite roots. Phytochem Anal. 2010;21:137–143. doi: 10.1002/pca.1168. [DOI] [PubMed] [Google Scholar]
- Srivastava N, Sharma V, Kamal B, Jadon VS. Aconitum: need for sustainable exploitation (with special reference to Uttarakhand) Int J Green Pharmacy. 2010;4:220–228. doi: 10.22377/ijgp.v4i4.151. [DOI] [Google Scholar]
- Straathof AJ, Wahl SA, Benjamin KR, Takors R, Wierckx N, Noorman HJ. Grand research challenges for sustainable industrial biotechnology. Trends Biotechnol. 2019;37:1042–1050. doi: 10.1016/j.tibtech.2019.04.002. [DOI] [PubMed] [Google Scholar]
- Subash AK, Augustine A. Hypolipidemic effect of methanol fraction of Aconitum heterophyllum wall ex Royle and the mechanism of action in diet-induced obese rats. J Adv Pharmac Technol Res. 2012;3:224. doi: 10.4103/2231-4040.104713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sungyu Y, Wook-jin K, Inkyu P, Sang-min Y, Byeong-cheol M. The complete chloroplast genome sequence of a medicinal plant Aconitum volubile var. pubescens Regel (Ranunculaceae) Korean Herbal Med Inf. 2016;4:21–25. [Google Scholar]
- Suyal R, Rawat S, Rawal RS, Bhatt ID. Variability in morphology, phytochemicals, and antioxidants in Polygonatum verticillatum (L.) All. populations under different altitudes and habitat conditions in Western Himalaya, India. Environ Monit Assess. 2019;191:783. doi: 10.1007/s10661-019-7687-6. [DOI] [PubMed] [Google Scholar]
- Tang P, Chen QH, Wang FP. Atropurpuran, a novel diterpene with an unprecedented pentacyclic cage skeleton, from Aconitum hemsleyanum var. atropurpureum. Tetrahedron Lett. 2009;50:460–462. doi: 10.1016/j.tetlet.2008.11.028. [DOI] [Google Scholar]
- Tang M, Zhao W, Xing M, Zhao J, Jiang Z, You J, Ni B, Ni Y, Liu C, Li J, Chen X. Resource allocation strategies among vegetative growth, sexual reproduction, asexual reproduction and defense during growing season of Aconitum kusnezoffii Reichb. Plant J. 2021;105:957–977. doi: 10.1111/tpj.15080. [DOI] [PubMed] [Google Scholar]
- Teng G, Zhang F, Li Z, Zhang C, Zhang L, Chen L, et al. Quantitative electrophysiological evaluation of the analgesic efficacy of two lappaconitine derivatives: a window into anti-nociceptive drug mechanisms. Neurosci Bull. 2021;37:1555–1569. doi: 10.1007/s12264-021-00774-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toivonen L, Balsevich J, Kurz W. Indole alkaloid production by hairy root cultures of Catharanthus roseus. Plant Cell Tissue Organ Cult. 1989;18:79–93. doi: 10.1002/bit.260370709. [DOI] [Google Scholar]
- Turabekova MA, Rasulev BF, Dzhakhangirov FN, Leszczynska D, Leszczynski J. Aconitum and Delphinium alkaloids of curare-like activity. QSAR analysis and molecular docking of alkaloids into AChBP. Eur J Med Chem. 2010;45:3885–3894. doi: 10.1016/j.ejmech.2010.05.042. [DOI] [PubMed] [Google Scholar]
- Ulubelen A, Meriçli AH, Meriçli F, Yilmaz F. Diterpenoid alkaloids from Aconitum orientale. Phytochemistry. 1996;41:957–961. doi: 10.1016/0031-9422(95)00670-2. [DOI] [Google Scholar]
- Ulubelen A, Mericli AH, Mericli F, Kolak U, Arfan M, Ahmad M, Ahmad H. Norditerpenoid alkaloids from the roots of Aconitum leave Royle. Pharmazie. 2002;57(6):427–429. [PubMed] [Google Scholar]
- Unamba CI, Nag A, Sharma RK. Next generation sequencing technologies: the doorway to the unexplored genomics of non-model plants. Front Plant Sci. 2015;6:1074. doi: 10.3389/fpls.2015.01074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vandelook F, Lenaerts J, Jozef AVA. The role of temperature in post-dispersal embryo growth and dormancy break in seeds of Aconitum lycoctonum L. Flora. 2009;204:536–542. doi: 10.1016/j.flora.2008.11.003. [DOI] [Google Scholar]
- Venkatasubramanian P, Balasubramani SP, Nandi SK, Tariq M. Bioactive metabolite profiling for identification of elite germplasms: a conservation strategy for threatened medicinal plants. Curr Sci. 2018;114:554–561. doi: 10.18520/cs/v114/i03/554-561. [DOI] [Google Scholar]
- Verma S, Ojha S, Raish M. Anti-inflammatory activity of Aconitum heterophyllum on cotton pellet-induced granuloma in rats. J Med Plants Res. 2010;4:1566–1569. doi: 10.5897/JMPR09.502. [DOI] [Google Scholar]
- Wada K, Hazawa M, Takahashi K, Mori T, Kawahara N, Kashiwakura I. Inhibitory effects of diterpenoid alkaloids on the growth of A172 human malignant cells. J Nat Prod. 2007;70:1854–1858. doi: 10.1021/np070270w. [DOI] [PubMed] [Google Scholar]
- Wada K, Hazawa M, Takahashi K, Mori T, Kawahara N, Kashiwakura I. Structure–activity relationships and the cytotoxic effects of novel diterpenoid alkaloid derivatives against A549 human lung carcinoma cells. J Nat Med. 2011;65:43–49. doi: 10.1007/s11418-010-0452-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wai AH, Rahman MM, Waseem M, Cho LH, Naing AH, Jeon JS, Lee DJ, Kim CK, Chung MY. Comprehensive genome-wide analysis and expression pattern profiling of PLATZ gene family members in Solanum lycopersicum L. under multiple abiotic stresses. Plants. 2022;11:3112. doi: 10.1007/s11676-022-01519-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J, Chen B, Ali S, Zhang T, Wang Y, Zhang H, et al. Epigenetic modification associated with climate regulates betulin biosynthesis in birch. J for Res. 2023;34:21–35. doi: 10.1007/s11676-021-01424-7. [DOI] [Google Scholar]
- Wang ZH, Li YQ. Characterization of the complete chloroplast genome of Aconitum pendulum (Ranunculaceae), an endemic medicinal herb. Mitochondrial DNA Part B. 2020;5:382–383. doi: 10.1080/23802359.2019.1703592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Li Z, Yang B. Trans-2, 2′, 4, 4′-tetramethyl-6, 6′-dinitroazobenzene from Aconitum sungpanense. Fitoterapia. 2004;75:789–791. doi: 10.1016/j.fitote.2004.09.016. [DOI] [PubMed] [Google Scholar]
- Wang Z, Wen J, Xing J, He Y. Quantitative determination of diterpenoid alkaloids in four species of Aconitum by HPLC. J Pharm Biomed Anal. 2006;40:1031–1034. doi: 10.1016/j.jpba.2005.08.012. [DOI] [PubMed] [Google Scholar]
- Wang DP, Lou HY, Huang L, Hao XJ, Liang GY, Yang ZC, Pan WD. A novel franchetine type norditerpenoid isolated from the roots of Aconitum carmichaeli Debx. with potential analgesic activity and less toxicity. Bioorg Med Chem Lett. 2012;22:4444–4446. doi: 10.1016/j.bmcl.2012.04.132. [DOI] [PubMed] [Google Scholar]
- Wangchuk P, Bremner JB, Skelton BW, White AH, Rattanajak R, Kamchonwongpaisan S. Antiplasmodial activity of atisinium chloride from the Bhutanese medicinal plant, Aconitum orochryseum. J Ethnopharmacol. 2010;130:559–562. doi: 10.1016/j.jep.2010.05.057. [DOI] [PubMed] [Google Scholar]
- Wani TA, Kaloo ZA, Dangroo NA. Aconitum heterophyllum Wall. ex Royle: A critically endangered medicinal herb with rich potential for use in medicine. J Integr Med. 2022;20:104–113. doi: 10.1016/j.joim.2021.12.004. [DOI] [PubMed] [Google Scholar]
- Watad AA, Kochba M, Nissim A, Gaba V. Improvement of Aconitum napellus micropropagation by liquid culture on floating membrane rafts. Plant Cell Rep. 1995;14:345–348. doi: 10.1007/BF00238594. [DOI] [PubMed] [Google Scholar]
- Weber M, Owens K, Sarpong R. Atropurpuran-missing biosynthetic link leading to the hetidine and arcutine C20-diterpenoid alkaloids or an oxidative degradation product? Tetrahedron Lett. 2015;56:3600–3603. doi: 10.1016/j.tetlet.2015.01.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen RQ, Li DH, Zhao X, Wang JB, Zhao YL, Zhang P, Sun ZY, Yan D, Xiao XH, Ren YZ. Rationality of the processing methods of aconiti lateralis radix (fuzi) based on chemical analysis. Acta Pharmaceutica Sinica. 2013;48:286–290. [PubMed] [Google Scholar]
- Won H, Yun YE, Kwak M, Han JE. Genetic diversity assessment of Aconitum coreanum (H. Lév.) Rapaics (Ranunculaceae), an endangered plant species in Korea, using microsatellite markers. J Species Res. 2012;1:224–231. doi: 10.12651/JSR.2012.1.2.224. [DOI] [Google Scholar]
- Wu G, Jiang S, Zhu D. Norditerpenoid alkaloids from roots of Aconitum finetianum. Phytochemistry. 1996;42:1253–1255. doi: 10.1016/0031-9422(95)00918-3. [DOI] [Google Scholar]
- Wu G, Du L, Zhao L, Shang R, Liu D, Jing Q, et al. The total alkaloids of Aconitum tanguticum protect against lipopolysaccharide-induced acute lung injury in rats. J Ethnopharmacol. 2014;155:1483–1491. doi: 10.1016/j.jep.2014.07.041. [DOI] [PubMed] [Google Scholar]
- Wu L, Wu W, Jiang T, Wang X, Yang H. Study on optimization of formulation matrix of processed Aconitum carmichaelii hydrogel patch and its in vitro drug release. China Pharmacy. 2018;12:37–41. [Google Scholar]
- Xiao PG, Wang WP, Gao F, Yan LP, Chen DL, Liu Y. A pharmacophylogenetic study of Aconitum L. (Ranunculaceae) from China. J Syst Evol. 2006;44:1–46. doi: 10.1360/aps050046. [DOI] [Google Scholar]
- Xie Y, Jiang ZH, Zhou H, Xu HX, Liu L. Simultaneous determination of six Aconitum alkaloids in proprietary Chinese medicines by high-performance liquid chromatography. J Chromatogr A. 2005;1093:195–203. doi: 10.1016/j.chroma.2005.07.071. [DOI] [PubMed] [Google Scholar]
- Xing BN, Jin SS, Wang H, Tang QF, Liu JH, Li RY, et al. New diterpenoid alkaloids from Aconitum coreanum and their anti-arrhythmic effects on cardiac sodium current. Fitoterapia. 2014;94:120–126. doi: 10.1016/j.fitote.2014.01.022. [DOI] [PubMed] [Google Scholar]
- Xu H, Arita H, Hayashida M, Zhang L, Sekiyama H, Hanaoka K. Pain-relieving effects of processed Aconiti tuber in CCI-neuropathic rats. J Ethnopharmacol. 2006;103:392–397. doi: 10.1016/j.jep.2005.08.050. [DOI] [PubMed] [Google Scholar]
- Xu T, Chen J, Zhu L, Li Z. Development of microsatellite loci for Aconitum gymnandrum (Ranunculaceae), a species endemic to the Qinghai-Tibetan Plateau. Am J Bot. 2011;98(1):e7–e9. doi: 10.3732/ajb.1000418. [DOI] [PubMed] [Google Scholar]
- Yan L, Qin-Er Y. Taxonomic revision of Aconitum (Ranunculaceae) from Sichuan. China Acta Phytotaxonomica Sinica. 2005;43(4):289–386. doi: 10.1360/aps040102. [DOI] [Google Scholar]
- Yan G, Sun H, Sun W, Zhao L, Meng X, Wang X. Rapid and global detection and characterization of aconitum alkaloids in Yin Chen Si Ni Tang, a traditional Chinese medical formula, by ultra performance liquid chromatography–high resolution mass spectrometry and automated data analysis. J Pharm Biomed Anal. 2010;53:421–431. doi: 10.1016/j.jpba.2010.05.004. [DOI] [PubMed] [Google Scholar]
- Ya-Nan HE, Shui-Ping OU, Xiong X, Yuan PAN, Jin PEI, Run-Chun XU, et al. Stems and leaves of Aconitum carmichaelii Debx. as potential herbal resources for treating rheumatoid arthritis: chemical analysis, toxicity and activity evaluation. Chin J Nat Med. 2018;16:644–652. doi: 10.1016/S1875-5364(18)30104-3. [DOI] [PubMed] [Google Scholar]
- Yang H, Zhou T. The development of novel microsatellite markers and genetic diversity study for Aconitum carmichaeli. J Agric Biotechnol. 2017;25:58–66. [Google Scholar]
- Yang CL, Huang ZF, Zhang YH, Liu YH, Chen Y, Yi JH. Effects of steaming and baking on content of alkaloids in Aconite Lateralis Radix (Fuzi) China J Chin Materia Med. 2014;39:4798–4803. [PubMed] [Google Scholar]
- Yang J, Zeng X, Guo S. Characterization of the complete chloroplast genome of the perennial herb Aconitum carmichaelii (Ranunculales: Ranunculaceae) Conserv Genet Resour. 2018;10:605–608. doi: 10.1007/s12686-017-0875-1. [DOI] [Google Scholar]
- Yang Y, Hu P, Zhou X, Wu P, Si X, Lu B, Zhu Y, Xia Y. Transcriptome analysis of Aconitum carmichaelii and exploration of the salsolinol biosynthetic pathway. Fitoterapia. 2020;140:104412. doi: 10.1016/j.fitote.2019.104412. [DOI] [PubMed] [Google Scholar]
- Yin T, Zhou H, Cai L, Ding Z. Non-alkaloidal constituents from the genus Aconitum: a review. RSC Adv. 2019;9:10184–10194. doi: 10.1039/C9RA01219B. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang X, Xu Q, Le L, Zhou T, Yu W, Wang G, Fu FF, Cao F. Comparative histology, transcriptome, and metabolite profiling unravel the browning mechanisms of calli derived from ginkgo (Ginkgo biloba L.) J for Res. 2022;2022:384. doi: 10.3390/plants11223112. [DOI] [Google Scholar]
- 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. doi: 10.1016/j.phytol.2018.04.001. [DOI] [Google Scholar]
- Yu M, Cao LL, Yang YX, Guan LL, Gou LL, Shu XY, Huang J, Liu D, Zhang H, Hou DB. Genetic diversity and marker–trait association analysis for agronomic traits in Aconitum carmichaelii Debeaux. Biotechnol Biotechnol Equip. 2017;31:905–911. doi: 10.1080/13102818.2017.1355747. [DOI] [Google Scholar]
- Yue H, Pi Z, Song F, Liu Z, Cai Z, Liu S. Studies on the aconitine-type alkaloids in the roots of Aconitum carmichaeli Debx. by HPLC/ESIMS/MSn. Talanta. 2009;77:1800–1807. doi: 10.1016/j.talanta.2008.10.022. [DOI] [PubMed] [Google Scholar]
- Yue XF, Zhang YN, Zhang J, Zhang ZQ. Free fatty acids profile analysis of alcohol extract of Aconitum taipeicum Hand.-Mazz. with gas chromatography-mass spectrometry. Anal Methods. 2010;2:668–672. doi: 10.1039/B9AY00307J. [DOI] [Google Scholar]
- Yun YE, Yu JN, Nam GH, Ryu SA, Kim S, Oh K, Lim CE. Next-generation sequencing identification and characterization of microsatellite markers in Aconitum austrokoreense Koidz, an endemic and endangered medicinal plant of Korea. Genet Mol Res. 2015;14:4812–4817. doi: 10.4238/2015.May.11.13. [DOI] [PubMed] [Google Scholar]
- Zhang SY, Jiang Y, Bi YF, Yan WJ, Zhang YB. Diterpenoid alkaloids from Aconitum kirinense. J Asian Nat Prod Res. 2013;15:78–83. doi: 10.1080/10286020.2012.744212. [DOI] [PubMed] [Google Scholar]
- Zhang H, Guo Z, Han L, You X, Xu Y. The antitumor effect and mechanism of taipeinine A, a new C19-diterpenoid alkaloid from Aconitum taipeicum, on the HepG2 human hepatocellular carcinoma cell line. Jbuon. 2014;19:705–712. [PubMed] [Google Scholar]
- Zhang Y, Shu Z, Yin L, Ma L, Wang X, Fu X. Anti-inflammatory and antinociceptive activities of non-alkaloids fractions from Aconitum flavum in vivo. Rev Bras Farmacog. 2015;25:47–52. doi: 10.1016/j.bjp.2014.11.013. [DOI] [Google Scholar]
- Zhang JF, Li Y, Gao F, Shan LH, Zhou XL. Four new C20-diterpenoid alkaloids from Aconitum rotundifolium. J Asian Nat Prod Res. 2018;21:716–724. doi: 10.1080/10286020.2018.1473384. [DOI] [PubMed] [Google Scholar]
- Zhang L, Miao X, Li Y, Dai H, Shang X, Hu F, Fan Q. Toxic and active material basis of Aconitum sinomontanum Nakai based on biological activity guidance and UPLC-Q/TOF-MS technology. J Pharm Biomed Anal. 2020;188:113374. doi: 10.1016/j.jpba.2020.113374. [DOI] [PubMed] [Google Scholar]
- Zhang L, Siyiti M, Zhang J, Yao M, Zhao F. Antiinflammatory and anti-rheumatic activities in vitro of alkaloids separated from Aconitum soongoricum Stapf. Exp Ther Med. 2021;21:493. doi: 10.3892/etm.2021.9924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang M, Luo J, Su L, Ding Q, Yin X, Hou F, Gao J, Peng C. The complete chloroplast genome of Aconitum scaposum. Mitochondrial DNA Part B. 2021;6:2149–2150. doi: 10.1080/23802359.2021.1944380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao C, Li M, Luo Y, Wu W. Isolation and structural characterization of an immunostimulating polysaccharide from fuzi, Aconitum carmichaeli. Carbohyd Res. 2006;341:485–491. doi: 10.1016/j.carres.2005.11.032. [DOI] [PubMed] [Google Scholar]
- Zhao YY, Zhang Y, Lin RC, Sun WJ. An expeditious HPLC method to distinguish Aconitum kusnezoffii from related species. Fitoterapia. 2009;80:333–338. doi: 10.1016/j.fitote.2009.04.005. [DOI] [PubMed] [Google Scholar]
- Zhao F, Nie J, Chen M, Wu G. Assessment of genetic characteristics of Aconitum germplasms in Xinjiang Province (China) by RAPD and ISSR markers. Biotechnol Biotechnol Equip. 2015;29:309–314. doi: 10.1080/13102818.2015.1004899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao D, Shen Y, Shi Y, Shi X, Qiao Q, Zi S, Zhao E, Yu D, Kennelly EJ. Probing the transcriptome of Aconitumcarmichaelii reveals the candidate genes associated with the biosynthesis of the toxic aconitine-type C19-diterpenoid alkaloids. Phytochemistry. 2018;152:113–124. doi: 10.1016/j.phytochem.2018.04.022. [DOI] [PubMed] [Google Scholar]
- 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 Aconitumcarmichaelii Debeaux. J Ethnopharmacol. 2015;160:173–193. doi: 10.1016/j.jep.2014.11.043. [DOI] [PubMed] [Google Scholar]
- Zhou J, Liu W, Kong H, Gong W. Identification and characterization of microsatellites in Aconitumreclinatum (Ranunculaceae), a rare species endemic to North America. Appl Plant Sci. 2018;6:e01161. doi: 10.1002/aps3.1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zi J, Mafu S, Peters RJ. To gibberellins and beyond! Surveying the evolution of (di) terpenoid metabolism. Annu Rev Plant Biol. 2014;65:259–286. doi: 10.1146/annurev-arplant-050213-035705. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data availability statement
Authors confirmed that no primary or secondary data was used in this review.
Authors confirmed that no primary or secondary data was used in this review.






