Next Article in Journal
A Comparison between Bulgarian Tanacetum parthenium Essential Oil from Two Different Locations
Previous Article in Journal
Sc-Modified C3N4 Nanotubes for High-Capacity Hydrogen Storage: A Theoretical Prediction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Biomedical Potential of Terpenoid Alkaloids: Sources, Structures, and Activities

1
School of Pharmacy, Yantai University, Yantai 264005, China
2
School of Life Sciences, Yantai University, Yantai 264005, China
3
College of Pharmacy, University of Utah, Salt Lake City, UT 84108, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 3 April 2024 / Revised: 20 April 2024 / Accepted: 21 April 2024 / Published: 25 April 2024

Abstract

:
Terpenoid alkaloids are recognized as a class of compounds with limited numbers but potent biological activities, primarily derived from plants, with a minor proportion originating from animals and microorganisms. These alkaloids are synthesized from the same prenyl unit that forms the terpene skeleton, with the nitrogen atom introduced through β-aminoethanol, ethylamine, or methylamine, leading to a range of complex and diverse structures. Based on their skeleton type, they can be categorized into monoterpenes, sesquiterpenes, diterpenes, and triterpene alkaloids. To date, 289 natural terpenoid alkaloids, excluding triterpene alkaloids, have been identified in studies published between 2019 and 2024. These compounds demonstrate a spectrum of biological activities, including anti-inflammatory, antitumor, antibacterial, analgesic, and cardioprotective effects, making them promising candidates for further development. This review provides an overview of the sources, chemical structures, and biological activities of natural terpenoid alkaloids, serving as a reference for future research and applications in this area.

1. Introduction

Alkaloids, a diverse class of secondary metabolites, are widely distributed in nature, with more than 27,000 species identified to date, predominantly originating from the plant kingdom, though relatively few are found in the animal and microbial kingdoms [1]. They typically exhibit strong biological activities, including antitumor, antibacterial, insecticidal, and analgesic effects [2,3,4]. Among the numerous classes of alkaloids, terpenoid alkaloids (TeAs) occupy a pivotal position. These alkaloids are formed from terpenoids through amination reactions, making them aminated terpenes [5]. TeAs are classified as pseudo alkaloids primarily because their biosynthetic origins do not involve the amino acid pathway. Instead, terpenoid moieties in TeAs are biosynthesized from isoprene through the methylerythritol phosphate (MEP) pathway, while nitrogen atoms are typically introduced into the structures of terpenoids in the form of β-aminoethanol, ethylamine, or methylamine [1].
Despite the vast variety of alkaloids and terpenoids isolated from nature, only a tiny proportion of them conform to the structural features of TeAs. TeAs, as a natural product with diverse structures, are primarily divided into monoterpene, sesquiterpene, diterpene, and triterpene alkaloids according to the differences in their skeletons [6]. Among them, monoterpene alkaloids are derived from iridoid compounds, mainly concentrated in the plants of Bignoniaceae, Lamiaceae, Gentianaceae, and Scrophularia [7]. Sesquiterpene alkaloids are the least abundant class of TeAs, which are narrowly distributed in the plant kingdom and mainly concentrated in plants such as Dendrobium [5]. Diterpenoid alkaloids (DAs) are the most complex and numerous compounds in TeAs, mainly concentrated in the Aconitum and Delphinium plants of Ranunculaceae [8]. In addition, marine sponges are also an important source of diterpenoid alkaloids.
Although small in number, these alkaloids are widely bioactive. For example, incarvillateine, a monoterpene alkaloid with strong analgesic activity, isolated from the traditional Chinese medicine Incarvillae sinensis LAM., has become a significant lead compound in the development of new non-narcotic pain medications [9]. DAs have been used for many years as traditional medicines in China, Japan, Russia, Mongolia, and India [10]. Because of their severe toxicity, in ancient times, Aconitum roots were often used to hydrolyze highly toxic DAs (e.g., aconitine) into less toxic derivatives (e.g., benzylaconine) by soaking, boiling, or other processing methods [8,11]. Modern pharmacological studies have shown that diterpene alkaloids have significant anti-inflammation, analgesia, anticancer, and anti-arrhythmia effects [8]. Moreover, as a diterpenoid alkaloid, Crassicauline A has been clinically utilized as an anti-arrhythmic drug [12]. Similarly, cyclovirobuxine-D, a triterpene alkaloid, is also used clinically as an antiarrhythmic drug [13] and has been recognized as a lead compound for innovative analgesics [14].
The research significance and medical value of TeAs as a class of natural products with unique structures and a wide range of biological activities are clear. Given the complexity and variability of triterpenoid alkaloids’ structures and the constraints of space, this paper will focus on the sources, chemical structures, and biological activities of natural TeAs, excluding triterpene alkaloids, discovered in the past five years, hoping to provide a reference for the further research and application of TeAs.

2. Classes of Terpenoidal Alkaloids

2.1. Monoterpenoid Alkaloids

Monoterpenoid alkaloids represent a distinct class of alkaloids derived from iridoid glycosides, typically originating from loganin and secologanin after amination. According to Wang’s classification of monoterpene alkaloids, they can be divided into two categories: iridoids and secoiridoids [15]. This section discusses 26 monoterpenoid alkaloids isolated from plants, including 24 iridoid-type alkaloids (124) and two secoiridoid-type alkaloids (2526). Specific plant sources are listed in Table 1. The chemical structure details are shown in Figure 1.

2.1.1. Iridoid-Type Alkaloids (124)

The biosynthetic precursors of these alkaloids are iridoid glycosides. Based on the level of hydrogenation within their nitrogen-containing six-membered rings, they can be classified into four subtypes: pyridine ring type, piperidine ring type, dihydropyridine ring type, and tetrahydropyridine ring type [15].
Alstochonines A (1) and B (2) were isolated from the branches of Alstonia scholaris (Apocynaceae). Alstochonine A (1) was the first reported C-4 methylated nor-monoterpenoid alkaloid. Alstochonine B (2), processing a cyclopentyl[c]pyridine skeleton, is believed to be biosynthesized from iridotrial by ammonification, aromatization, and oxidation reactions [16].
(R)-10-hydroxyl-4-noractinidine (3) was extracted and isolated from Rauvolfia vomitoria’s trunk, the first reported monoterpene alkaloid in R. vomitoria (Apocynaceae) [17].
Delavatines C-E (911), three monoterpene alkaloids with a cyclopentane[c]piperidine skeleton, were isolated from whole plants of Incarvillea delavayi (Bignoniaceae) [18].
Incarvine G (12), a novel monoterpene alkaloid isolated from Incarvillea sinensis Lam., is an ester composed of a monoterpene alkaloid with a cyclopentane[c]piperidine skeleton and glucose [19].
Isoxerine (13), isolated from the roots of Scrophularia ningpoensis, was named due to its absolute configuration of C-7 being 7S, differing from oxerine [20].
Figure 1. Structures of monoterpene alkaloids (126).
Figure 1. Structures of monoterpene alkaloids (126).
Molecules 29 01968 g001
Forsyqinlingines C (14) and D (15) were isolated from the ripe fruits of Forsythia suspensa (Oleaceae), with the structures determined by analysis of spectra including HR-ESI and NMR. Both alkaloids belong to a rare class of planar C9-monoterpenoid alkaloids [21].
The dimeric monoterpene alkaloids (±)-Caryopterisines A (19) and B (20) were identified as racemates isolated from Caryopteris glutinosa Rehder (Lamiaceae), and their absolute configurations were determined using calculated ECD spectra and X-ray diffraction analysis. They are two novel dimers with a 6/5/5/5/6 pentacyclic system. In addition, they can be biosynthesized by oxerine dehydration or oxygenation and subsequent Diels–Alder reactions [22].
Caryopterisines F-I (47) and caryopterisines C-E (1618) were subsequently isolated from C. glutinosa Rehder. Caryopterisines F-I (47) are four monoterpene alkaloids containing the cyclopentyl[c]pyridine skeleton, while caryopterisines C-E (1618) represent three novel dimeric monoterpene alkaloids and are believed to be biosynthesized via the Diels–Alder reaction followed by aromatization rearrangement and a series of subsequent reactions [23]. Of these, caryopterisine C (16) has an unprecedented 6/5/6/6/5 pentacyclic ring framework, while caryopterisines D (17) and E (18) both have 6/6/6/5 fused ring frameworks.
Compounds 8 and 2124 were isolated from the aerial parts of Caryopteris mongolica Bunge (Lamiaceae), a medicinal plant in Mongolia. Among them, (5S*,7R*)-7-Ethoxy-6, 7-dihydro-7-methyl-5H-cyclopenta[c]pyridin-5-ol (8) is a monoterpene alkaloid with a cyclopenta[c]pyridine framework [24]. (5aR*,6S*,10S*,11R*,11aR*)-10,11a-Dimethyl-6,7,9,10,11,11a-hexahydro-5H-6,11-epoxycyclopenta [6,7]azuleno [1,2-c]pyridin-5,8(5aH)-dione(21) and (5aR*,6S*,7aR*,8S*,11aR*)-10-Hydroxy-7a,11a-dimethyl-5a,6,7,7a,8,11a-hexahydro-5H-6,8-epoxycyclopenta [6,7]azuleno [1,2-c] pyridin-5-one(22) could be formed by fusion of the cyclopenta[c]pyridine and 4-Demethyliridoid, and (5R*,5aR*,10bS*,11R*)-5-Hydroxy-10b,11-dimethyl-5,5a,10b,11-tetrahydro-6H-5,11-methanopyrido [3′,4′:3,4]cyclopenta [1,2-g]isoquinolin-6-one(23) and (6S*,6aR*,11R*,11aS*)-6a-Hydroxy-11,11a-dimethyl-6,6a,11,11a-tetrahydro-5H-6,11-methanopyrido [3′,4′:4,5]cyclopenta [1,2-h]isoquinolin-5-one(24) are dimerization products of two cyclopenta[c]pyridine [24].
Table 1. Names and plant sources of monoterpene alkaloids (126).
Table 1. Names and plant sources of monoterpene alkaloids (126).
No.Compound NamesSourcesPlant PartsRef.
1Alstochonine AAlstonia scholarisbranch[16]
2Alstochonine BAlstonia scholarisbranch[16]
3(R)-10-hydroxyl-4-noractinidineRauvolfia vomitoriatrunk[17]
4Caryopterisine FCaryopteris glutinosawhole plant[23]
5Caryopterisine GCaryopteris glutinosawhole plant[23]
6Caryopterisine HCaryopteris glutinosawhole plant[23]
7Caryopterisine ICaryopteris glutinosawhole plant[23]
8(5S*,7R*)-7-Ethoxy-6,7-dihydro-7-methyl-5H-cyclopenta[c]pyridin-5-ol.Caryopteris mongolica Bungeaerial part[24]
9Delavatine CIncarvillea delavayiwhole plant[18]
10Delavatine DIncarvillea delavayiwhole plant[18]
11Delavatine EIncarvillea delavayiwhole plant[18]
12Incarvine GIncarvillea sinensiswhole herb[19]
13IsoxerineScrophularia ningpoensisroot[20]
14Forsyqinlingine CForsythia suspensafruit[21]
15Forsyqinlingine DForsythia suspensafruit[21]
16Caryopterisine CCaryopteris glutinosawhole plant[23]
17Caryopterisine DCaryopteris glutinosawhole plant[23]
18Caryopterisine ECaryopteris glutinosawhole plant[23]
19(±)-Caryopterisine ACaryopteris glutinosawhole plant[22]
20(±)-Caryopterisine BCaryopteris glutinosawhole plant[22]
21(5aR*,6S*,10S*,11R*,11aR*)-10,11a-Dimethyl-6,7,9,10,11,11a-hexahydro-5H-6,11-epoxycyclopenta [6,7]azuleno [1,2-c]pyridin-5,8(5aH)-dione.Caryopteris mongolica Bungeaerial part[24]
22(5aR*,6S*,7aR*,8S*,11aR*)-10-Hydroxy-7a,11a-dimethyl-5a,6,7,7a,8,11a-hexahydro-5H-6,8-epoxycyclopenta [6,7]azuleno [1,2-c] pyridin-5-one.Caryopteris mongolica Bungeaerial part[24]
23(5R*,5aR*,10bS*,11R*)-5-Hydroxy-10b,11-dimethyl-5,5a,10b,11-tetrahydro-6H-5,11-methanopyrido [3′,4′:3,4]cyclopenta [1,2-g]isoquinolin-6-one.Caryopteris mongolica Bungeaerial part[24]
24(6S*,6aR*,11R*,11aS*)-6a-Hydroxy-11,11a-dimethyl-6,6a,11,11a-tetrahydro-5H-6,11-methanopyrido [3′,4′:4,5]cyclopenta [1,2-h]isoquinolin-5-oneCaryopteris mongolica Bungeaerial part[24]
25LongiflorineUncaria longiflora var. pteropodaleaf[25]
26Lomatogonin CLomatogonium carinthiacumwhole plant[26]

2.1.2. Secoiridoid-Type Alkaloids (2526)

This kind of alkaloid is mainly distributed in Gentianaceae plants and derived from secoiridoid glycosides [15]. Longiflorine (25), isolated from the leaves of Uncaria longiflora var. Pteropoda (Rubiaceae), is a monoterpenoid alkaloid with a lactam ring derived from secologanin [26]. Lomatogonin C (26), isolated from dried whole plants of Lomatogonium carinthiacum (Gentianaceae), is a natural monoterpene alkaloid derived from secoiridoid [25].

2.2. Sesquiterpene Alkaloids (2732)

Sesquiterpene alkaloids represent the least abundant class of TeAs derived from sesquiterpenes and incorporate nitrogen atoms in the basic skeleton of sesquiterpenes [27]. This subsection mainly describes six sesquiterpene alkaloids from nature, including a rare alkaloid from the ocean. The plant sources are listed in Table 2, and the chemical structure details are shown in Figure 2.
Table 2. Names and plant sources of sesquiterpene alkaloids (2732).
Table 2. Names and plant sources of sesquiterpene alkaloids (2732).
No.Compound NameSourcesPlant PartsRef.
27Commipholactam AResina Commiphoramyrrh[28]
28Dendrofindline ADendrobium findlayanumstem[29]
29Dendrofindline BDendrobium findlayanumstem[29]
30Findlayine DDendrobium findlayanumstem[30]
31Findlayine FDendrobium findlayanumstem[30]
32Echinoflorinegorgonian Echinogorgia flora/[31]
/: did not report.
Commipholactam A (27) was isolated from the dried myrrh of Resina Commiphora and represented a rare cadinane-type sesquiterpenoid. Unlike typical cadinane sesquiterpenoids, where ring C is usually present as a lactone, compound 27 appears as a lactam ring [28].
Dendrofindlines A-B (2829) and Findlayines D-F (3031) were all obtained from the dried stems of Dendrobium findlayanum and belong to the dendrobine-type alkaloid group [29,30]. Notably, Dendrofindline A (28) and Findlayine D (30) belong to the seco-lactone dendrobine-type alkaloid group. Findlayine D (30) is the first reported dendrobine-type alkaloid to feature a 2-ethoxy-2-oxoethyl group attached at C-2. Moreover, Dendrofindline B(29) is identified as a dendrobine-type alkaloid with a nitrogen-containing ring cleavage [29]. Findlayine F(31) is a nor-dendrobine-type alkaloid with a 5-decarboxylated structure [30].
Echinoflorine (32), isolated from the Gorgonian Echinogorgia flora, is a guaipyridine-type alkaloid with a γ-lactone-cyclohepta[c]pyridine skeleton, which is different from the cyclohepta[b]pyridine skeleton derived from terrestrial organisms [31].

2.3. Diterpenoid Alkaloids (DAs) (33289)

DAs are the most abundant and structurally complex TeAs, characterized by numerous stereocenters. They typically originate from the amination of tetracyclic or pentacyclic diterpenes, forming heterocyclic systems possessing β-aminoethanol, methylamine, or ethylamine nitrogen atoms [32]. Based on the number of carbon atoms in the skeleton of DAs, they can be classified into three major categories: C18, C19, and C20 [33]. Shen Yong comprehensively reviewed the classification of diterpenoid alkaloids in 2020 [8]. Consequently, this article will not delve into an extensive discussion of this classification but will focus only on the classification of new members discovered in the past five years. This section describes 257 newly discovered natural DAs, including 11 C18-DAs, 139 C19-DAs, 84 C20-DAs, 14 Bis-DAs, and 9 other types of DAs. These DAs were predominantly isolated from the plants in the Aconitum and Delphinium genera, with only two new DAs isolated from the ripe fruits of Forsythia suspensa. Additionally, five new DAs were obtained from microorganisms and marine animals.

2.3.1. C18-DAs (3343)

Without C18 in the structure, these alkaloids predominantly feature a 4-OH or ester substitution, with a few compounds having 3,4-epoxy substitution. According to the presence or absence of oxygen-containing groups at C7, they are classified into lappaconitines and ranaconitines [32]. Eleven C18-DAs (3343) are described in this subsection. Plant sources are shown in Table 3. Detailed chemical structures are shown in Figure 3.
Compounds 3337 are identified as lappaconine-type alkaloids, with four originating from Aconitum and only Naviconine (33) derived from Delphinium [34,35,36]. Amino groups are generally uncommon in DAs, whereas Leucostosine C(35) is the first naturally occurring DA to feature an amino group attached at C-7 [36].
Compounds 3840, obtained from Aconitum, are classified as ranaconitine-type DAs. Kirisine A (40) and B (38) are rare DAs with 9,14-methylenedioxy group. In addition, kirisine B (38), containing a chlorine substituent at C-4, represents a fourth example of DAs with a chlorine substituent [35].
Compounds 41 and 42 are rearranged C18-DAs, where the C7-C17 bond was rearranged to a C8-C17 bond [37]. Compounds 41 and 42 are derived from the rearrangement of ranaconitine-type DAs, and both contain an oxygen-containing hydroxyl group at C7. Additionally, Barpubenine A (41) is the first reported N-oxide in C18-DAs [37].
1-N-deethyl-1,16-demethoxy-1,16-dihydroxypyranaconidine(43) is a C18-nor-diterpene alkaloid with a skeleton similar to ranaconidine except that it lacks an N-ethyl group [38]. Furthermore, a hydroxyl group exists at C-16 instead of a methoxy group, which is common in C18-DAs [38].
Table 3. Names and plant sources of C18-DAs (3343).
Table 3. Names and plant sources of C18-DAs (3343).
No.Compound NameSourcesPlant PartsRef.
33NaviconineDelphinium naviculare var. lasiocarpumwhole plant[34]
34Kirisine EAconitum kirinense Nakairoot[35]
35Leucostosine CAconitum leucostomum Woroschroot[36]
36Leucostosine DAconitum leucostomum Woroschroot[36]
37Kirisine DAconitum kirinense Nakairoot[35]
38Kirisine BAconitum kirinense Nakairoot[35]
39Kirisine CAconitum kirinense Nakairoot[35]
40Kirisine AAconitum kirinense Nakairoot[35]
41Barpubenine AAconitum barbatum var. puberulum Ledeb.whole plant[37]
42Barpubenine BAconitum barbatum var. puberulum Ledeb.whole plant[37]
431-N-deethyl-1,16-demethoxy-1,16-dihydroxyranaconidineAconitum iochanicumoneroot[38]

2.3.2. C19-DAs (44182)

C19-DAs are the largest class of DAs. According to the differences in the skeleton, they can be divided into six types: aconitines, lycoctonines, lactones, 7,17-seco, franchetines, and rearranged class [39]. Aconitines and lycoctonines constitute the majority of C19-DAs. This subsection summarizes 139 new C19-DAs discovered in the past five years, including 70 aconitine-type C19-DAs, 40 lycotonine-type C19-DAs, 5 lactone-type C19-DAs, 10 franchetine-type C19-DAs, 7 Seco-type C19-DAs, and 7 rearranged C19-DAs.
  • Aconitine-type C19-DAs (44113)
Aconitine-type C19-DAs are alkaloids characterized by the absence of an oxygen group at C-7. When ester groups are present at C-8 and C-14, they exhibit acute toxicity [8]. In the past five years, 70 new compounds (44113) were discovered, with 65 obtained from Aconitum and 5 from Delphinium [32,34,38,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54]. The compounds were further discussed according to the presence of oxygen-containing groups connected at C-3, C-6, or C-15 positions. The plant sources are shown in Table 4. Detailed chemical structures are shown in Figure 4.
Compounds 4491 are alkaloids that lack oxygen-containing groups at C-3, C-6, and C-15. Notably, compounds 5057, featuring a double bond between the N and C-19, were isolated from Aconitum [38,40,50]. Compounds 5864 are linked to a 2-(2-methyl-4-oxoquinazolin-3-yl) benzoate moiety at C-18 [49,54]. Compounds 5863 exhibit rotational isomerism due to an unusual axial chiral phenyl-quinoline side chain at C-18, with the stereoisomerism studied by temperature-varying NMR techniques [49,54]. Compounds 68 and 69 have an unusual ketone group attached to C-19, with compound 68 uniquely featuring a ketone group at C-14 instead of a common methoxy or ester group. Compounds 7984 are new C19-DAs with a vaginatunine A fragment at C-18 [32,55]. Compounds 8991 lack a common methoxy group in DAs at C-16; however, there is a double bond between C-15 and C-16 [45,46,51].
Compounds 9294 possess oxygen-containing groups at C-3, C-6, and C-15 [48,56,57]. Interestingly, compound 94 is the third reported C19-DA with hydroxyl groups at C-3, C-13, and C-15 and with a fatty acid ester moiety at C-8 [57]. Compounds 9596 have hydroxyl groups at C-15 without oxygen-containing groups at C-3/C-6. Compounds 9799 are DAs with oxygen-containing groups at C-6, while there are no oxygen-containing groups at C-3 and C-15. Compounds 100101 have oxygen groups at C-3 but no oxygen groups at C-6 and C-15. Compounds 102106 have oxygen-containing groups at C-3 and C-6, whereas 105 and 106 contain a ketone group at C-3 [42,48,58]. Compounds 107113 have oxygen-containing groups at C-6 and C-15 but no oxygen-containing group at C-3 [59]. Interestingly, the acetyloxy group of compound 113 at the C-6 position is β-oriented, uncommon in aconitine-type DAs [48,59].
Table 4. Names and plant sources of aconitine-type C19-DAs (44113).
Table 4. Names and plant sources of aconitine-type C19-DAs (44113).
No.Compound NameSourcesPlant PartsRef.
44NaviconitineDelphinium naviculare var. lasiocarpumwhole plant[34]
45Acoapetaludine DAconitum apetalum (Huth) B.Fedtschwhole plant[40]
46Acoapetaludine EAconitum apetalum (Huth) B.Fedtschwhole plant[40]
47Acoapetaludine FAconitum apetalum (Huth) B.Fedtschwhole plant[40]
48Acoapetaludine GAconitum apetalum (Huth) B.Fedtschwhole plant[40]
49Forrestline DDelphinium forrestii var. viridewhole herb[53]
50Episcopaline CAconitum episcopaleroot[50]
51Acoapetaludine HAconitum apetalum (Huth) B.Fedtschwhole plant[40]
52Acoapetaludine IAconitum apetalum (Huth) B.Fedtschwhole plant[40]
53Acoapetaludine JAconitum apetalum (Huth) B.Fedtschwhole plant[40]
54Novolunine CAconitum novoluridumroot[49]
55Austroyunnanine CAconitum austroyunnanenseroot[45]
561-N-deethyl-1,16-demethoxy-1,16 dihydroxy-N(19)-en-austroconitine AAconitum iochanicumoneroot[38]
571-N-deethyl-1,16-demethoxy-1,16-dihydroxy-18-methoxy-N(19)-en-austroconitine AAconitum iochanicumoneroot[38]
58Brevicanine AAconitum brevicalcaratumroot[54]
59Novolunine AAconitum novoluridumroot[49]
60Novolunine BAconitum novoluridumroot[49]
61Brevicanine BAconitum brevicalcaratumroot[54]
62Brevicanine CAconitum brevicalcaratumroot[54]
63Brevicanine DAconitum brevicalcaratumroot[54]
64Forrestline BDelphinium forrestii var. viridewhole herb[53]
65Refractine AAconitum refractum var. circinatumwhole plant[41]
66Richardsonine BAconitum richardsonianum Lauenerroot[44]
67Richardsonine CAconitum richardsonianum Lauenerroot[44]
68Richardsonine AAconitum richardsonianum Lauenerroot[44]
69Acoapetaludine KAconitum apetalum (Huth) B.Fedtschwhole plant[40]
70Acoapetaludine BAconitum apetalum (Huth) B.Fedtschwhole plant[40]
71Acoapetaludine CAconitum apetalum (Huth) B.Fedtschwhole plant[40]
72Forrestline EDelphinium forrestii var. viridewhole herb[53]
73Brevicalcarine BAconitum brevicalcaratumroot[60]
74Brevicalcarine CAconitum brevicalcaratumroot[60]
75Rockidine AAconitum generaroot[43]
76Pseudostapine AAconitum pseudostapfianumroot[51]
77Refractine BAconitum refractum var. circinatumwhole plant[41]
78Austroyunnanine AAconitum austroyunnanenseroot[45]
79Apetalrine AAconitum apetalumaerial part[55]
80Apetalrine BAconitum apetalumaerial part[55]
81Apetalrine CAconitum apetalumaerial part[55]
82Apetalrine DAconitum apetalumaerial part[55]
83Apetalrine EAconitum apetalumaerial part[55]
84Brevicalcarine AAconitum brevicalcaratumroot[60]
85Nagarumine AAconitum nagarumroot[46]
86Nagarutine AAconitum nagarum Stapfroot[42]
87Episcopaline AAconitum episcopaleroot[50]
88Pseudostapine BAconitum pseudostapfianumroot[51]
89Pseudostapine CAconitum pseudostapfianumroot[51]
90Nagarumine BAconitum nagarumroot[46]
91Austroyunnanine BAconitum austroyunnanenseroot[45]
92Smirnotine AAconitum smirnovii Steinbaerial part[58]
93Pendulumine AAconitum pendulumrhizome[48]
94LipojesaconitineAconitum japonicum subsp. subcuneatumrhizoma[57]
956-demethoxyhypaconineAconitum carmichaelii Debx.lateral root[47]
96Carmichaeline KAconitum carmichaelii Debx.lateral root[47]
9710-hydroxychasmanineAconitum japonicum subsp. subcuneatumrhizoma[57]
98Rockidine BAconitum generaroot[43]
99GeordineAconitum georgei Comberroot[61]
1003-hydroxykaracolineAconitum japonicum subsp. subcuneatumrhizoma[57]
101Episcopine BAconitum episcopaleroot[52]
102Acotarine FAconitum taronenseroot[56]
103Acotarine GAconitum taronenseroot[56]
104Smirnotine BAconitum smirnovii Steinbaerial part[58]
105Pendulumine EAconitum pendulumrhizome[48]
106Nagarutine CAconitum nagarum Stapfroot[42]
1078-O-ethyl-benzoyldeoxyaconineAconitum carmichaelii Debx.lateral root[47]
108Pendulumine CAconitum pendulumrhizome[48]
109Pendulumine DAconitum pendulumrhizome[48]
110Nagarutine BAconitum nagarum Stapfroot[42]
111Nagarutine DAconitum nagarum Stapfroot[42]
112Pendulumine FAconitum pendulumrhizome[48]
113DelcarpumDelphinium peregrinum L. var. eriocarpum Boissaerial part[59]
  • Lycoctonine-type C19-DAs (114153)
Lycoctonine-type C19-DAs are oxidized at C-7 and C-8. Compounds 114153 are all novel members of the lycoctonine-type C19-DAs, with 38 compounds derived from Delphinium and only 2 from Aconitum [62,63,64,65,66,67,68]. The plant sources are shown in Table 5. Detailed chemical structures are shown in Figure 5.
According to the different oxygen-containing groups at C-7 and C-8, they can be divided into two subtypes. Eleven new compounds (115125) feature a C-7 and C-8 diol. Compounds 114 and 126 have rare methoxy and acetoxy groups at C-8, respectively [34,69]. Compounds 115 and 116, isolated from Aconitum sczukinii, have very similar chemical structures, with the only difference being the presence of double bonds between C-2 and C-3 in compound 115 [65]. Compound 117 contains a carboxyl group attached to the nitrogen. Compounds 118 and 119 are identified as a pair of regioisomers [62]. Compound 121 from Delphinium ajacis is notable for its rare hydroxyl group at C-12 [70]. Compounds 124126 all featured a characteristic N=CH fragment, with compound 126 also possessing an additional nitrone group [68].
Twenty-seven novel compounds (127153) obtained from Delphinium species all have a 7,8-methylenedioxy group. Compounds 130 and 131 each have a rare aldehyde group attached to the N atom [67]. Compound 145 is unprecedented, with an ether bond between C-1 and C-19 [71]. Compounds 134, 138, and 153 have an N=CH fragment, with compounds 138 and 153 further possessing a nitrone group [64,67,72]. Compounds 136137 and 149152 have a characteristic keto group attached to C-19 [67,72]. Interestingly, compound 153 from Delphinium displays an unusual β-oriented 1-OMe [72].
Table 5. Names and plant sources of lycoctonine-type C19-DAs (114153).
Table 5. Names and plant sources of lycoctonine-type C19-DAs (114153).
No.Compound NameSourcesPlant PartsRef.
114NaviculineDelphinium naviculare var. lasiocarpumwhole plant[34]
115Sczukiniline DAconitum sczukinii Turczroot[65]
116Sczukiniline EAconitum sczukinii Turczroot[65]
117Grandifline CDelphinium grandiflorumaerial parts[73]
118Shawurenine CDelphinium shawurense W. T. Wangaerial parts[62]
119Shawurenine DDelphinium shawurense W. T. Wangaerial parts[62]
120Uncinatine-ADelphinium uncinatumwhole plant[66]
121Ajacisine GDelphinium ajacisseed[70]
122Grandiflonine FDelphinium grandiflorum L.whole plant[68]
123Ajacisine FDelphinium ajacisseed[70]
124Grandiflonine EDelphinium grandiflorum L.whole plant[68]
125Grandiflonine GDelphinium grandiflorum L.whole plant[68]
126Chrysotrichumine ADelphinium chrysotrichumaerial parts[69]
127ElapacilineDelphinium elatum cv. Pacific Giantseed[67]
128MeladineDelphinium elatum cv. Pacific Giantseed[67]
129N-deethyldelphelineDelphinium elatum cv. Pacific Giantseed[67]
130N-deethyl-N-formyleladineDelphinium elatum cv. Pacific Giantseed[67]
131N-deethyl-N-formyldelphelineDelphinium elatum cv. Pacific Giantseed[67]
132MelapacitineDelphinium elatum cv. Pacific Giantseed[67]
133N-deethylpacinineDelphinium elatum cv. Pacific Giantseed[67]
134IminoeladineDelphinium elatum cv. Pacific Giantseed[67]
13519-oxopacilineDelphinium elatum cv. Pacific Giantseed[67]
13619-oxopacinineDelphinium elatum cv. Pacific Giantseed[67]
137N-deethyl-19-oxoeladineDelphinium elatum cv. Pacific Giantseed[67]
138Brunodelphinine CDelphinium brunonianum Royleaerial parts[64]
139Grandifloline ADelphinium grandiflorum L.whole herb[63]
140Grandifloline BDelphinium grandiflorum L.whole herb[63]
141Grandifloline CDelphinium grandiflorum L.whole herb[63]
142Grandifloline EDelphinium grandiflorum L.whole herb[63]
143Grandifloline DDelphinium grandiflorum L.whole herb[63]
144Grandifloline FDelphinium grandiflorum L.whole herb[63]
145Liangshanine ADelphinium liangshanense W. T. Wangwhole plant[71]
146Liangshanine BDelphinium liangshanense W. T. Wangwhole plant[71]
147Kamaonensine ADelphinium kamaonense Huthwhole plant[72]
148Kamaonensine BDelphinium kamaonense Huthwhole plant[72]
149Kamaonensine CDelphinium kamaonense Huthwhole plant[72]
150Kamaonensine DDelphinium kamaonense Huthwhole plant[72]
151Kamaonensine EDelphinium kamaonense Huthwhole plant[72]
152Kamaonensine GDelphinium kamaonense Huthwhole plant[72]
153Kamaonensine FDelphinium kamaonense Huthwhole plant[72]
  • Lactone-type C19-DAs (154158)
Lactone-type C19-DAs are generally formed by oxidation of the 14-ketone in the C ring of aconitine-type DAs to form a six-membered lactone C ring. Only five new members (154158) belong to this type. The plant sources are shown in Table 6. Detailed chemical structures are shown in Figure 6.
Interestingly, these five newly discovered lactone-type C19-DAs have an unprecedented five-membered lactone D ring [74,75]. Compounds 154158 are formed by cleavage of the bond between C-15 and C-16, followed by subsequent lactonization. In addition, compounds 157 and 158 are C-13 epimers of each other, highlighting a unique aspect of their structural configuration [74].
Table 6. Names and plant sources of lactone-type C19-DAs (154158).
Table 6. Names and plant sources of lactone-type C19-DAs (154158).
No.Compound NameSourcesPlant PartsRef.
154Kusnezosine AAconitum kusnezoffii Reichb. var. gibbiferumroot[75]
155Kusnezosine BAconitum kusnezoffii Reichb. var. gibbiferumroot[75]
156Kusnezosine CAconitum kusnezoffii Reichb. var. gibbiferumroot[75]
157Stylosine AAconitum stylosumroot[74]
158Stylosine BAconitum stylosumroot[74]
  • Franchetine-type C19-DAs (159168)
Franchetine-type C19-DAs are distinguished from aconitines by an additional ether between C-6 and C-17. Ten novel alkaloids (159168) are classified as franchetine-type C19-DAs [43,56,76]. The plant sources are shown in Table 7. Detailed chemical structures are shown in Figure 7. Compounds 159, 161166, and 168 have a double bond between C-7 and C-8, whereas compounds 160 and 167 have a 7,8-epoxy unit [43,56,76]. In addition, Compounds 161 and 163 have a characteristic hydroxyl group at C-16 instead of the methoxy group joint in DAs.
  • 7,17-seco-type C19-DAs (169175)
7,17-seco-type C19-DAs are characterized by the cleavage of the C7-C17 bond, typically along with double bonds between C-7 and C-8. There are seven members (169175) of this class that have been identified over the past five years. Interestingly, compounds 169173 have a hemiacetal fragment, which results from the C7-C17 bond breaking, followed by the formation of an ether bond [64,73,77]. Compounds 169173 are the 7,17-secolycoctonine C19-DAs with a C7-O-C17 unit [73]. Compounds 170173 have an unprecedented N, O-diacetyl residue [77]. Among them, compounds 170 and 172 are isomers; the methoxy group is located at C-11 in 172 and at C-6 in 170. Moreover, compound 173, lacking a methoxy group at C-6, is a demethylation product of 170. The plant sources are shown in Table 8, and detailed chemical structures are shown in Figure 8.
  • Rearranged C19-DAs (176182)
Over the past five years, only seven members (176182) have been classified as rearranged C19-DAs [46,52,64,73,78]. Compounds 176 and 182 are unique alkaloids with a rearranged six-membered B ring formed by the C-8 and C-10 linkage [46,50]. Compounds 177 and 178 are two new rare rearranged aconitine-type C19-diterpenoid alkaloids whose C7-C17 bond rearranges to form a C8-C17 bond [79]. The N-C19 and C7-C17 bonds in compound 179 are broken and rearranged into N-C7 bonds, and C-19 was oxidized to carbonyl [73]. In addition, compound 179 belongs to a new rearranged subtype named grandiflodines, which possesses a C7-N-C17 unit and a C17-O-C19 unit. Compound 181 displays an unusual rearranged C19-DA skeleton with the cleavage of N-C19 and C7-C17 bonds and the construction of the N-C7 bond [78]. The plant sources are shown in Table 9. Detailed chemical structures are shown in Figure 9.

2.3.3. C20-DAs (183266)

C20-DAs are compounds with structures more complex than those of C18- and C19-DAs, with most C20-DAs having a characteristic exocyclic double bond between C-16 and C-17. Based on the variations in the skeletons, the vast majority of C20-DAs can be classified into seven types: atisines, deudatines, hetisines, hetidines, anopterines, napellines, and vakognavines [39]. This section describes 84 C20-DAs from nature, including 9 atisine-type C20-DAs, 26 hetisine-type C20-DAs, 11 hetidine-type C20-DAs, 13 deudatine-type C20-DAs, 8 napelline-type C20-DAs, 6 vakognavine-type C20-DAs, and 11 rearranged C20-DAs. Among these alkaloids, 62 were obtained from Aconitum plants and 22 from Delphinium plants.
  • Atisine-type C20-DA (183191)
Atisine-type C20-DAs are structurally characterized by their N atoms being linked to C-20 and C-19 and share the same carbon skeleton as atisine diterpenes. Only nine newly discovered compounds (183191) have been classified as atisine-type C20-DAs [37,53,69,80,81]. Compounds 183186, derived from Delphinium, feature an additional ether bond between C-20 and C-7. Among these, compounds 183185 have cyano groups at C-19, making the first reported cyano-containing DAs [80]. Furthermore, compound 186 is noted for bearing an oxazolidine ring F [69]. Compounds 187188 and 190191 have a double bond between N and C-20, with compound 188 possessing a rare ketone group at C-15 [37]. The plant sources are shown in Table 10. Detailed chemical structures are shown in Figure 10.
  • Hetisine-type C20-DAs (192217)
Compared to atisine-type C20-DAs, hetisine-type C20-DAs feature a hexacyclic with an additional C14-C20 bond and N-C6 bond [82]. This category is the largest, and the newly discovered compounds 192217 belong to this class of C20-DAs [37,59,68,83,84]. Among these, only compound 203, obtained from Aconitum, has a hydroxyl group at C-6 [85]. Compounds 204206 and 211212 have a hydroxyl group at C-15; notably, only compound 212 shows an α-oriented OH group at C-15 [86]. Compound 210 is the first hetisine-type C20-DA with one hexose substitution, identified as β-glucoside [47]. In addition, Compound 213 is a rare DA linked by the ether bond between C-17′ and C-2 between hetisine-type C20-DAs and hetidane-type diterpenes [84]. The plant sources are shown in Table 11. Detailed chemical structures are shown in Figure 11.
  • Hetidine-type C20-DAs (218228)
Hetidine-type C20-DAs possess an additional C14-C20 bond, distinguishing them structurally from the atisine class. Eleven new members (218228) have been classified as such [65,81,84,85,90,91]. Compounds 218219, 221, 223, and 225226 all feature a typical exocyclic double bond characteristic of C20-DAs, while compounds 220, 222, and 224 exhibit a distinct intra-ring double bond between C15 and C16. Moreover, compound 226 is notable for having an ester group between C-12 and C-14, forming a lactone ring D, representing a novel skeleton of hetidine-type C20-DAs [65]. The plant sources are shown in Table 12. Detailed chemical structures are shown in Figure 12.
  • Denudatine-type C20-DAs (229241)
Denudatine-type C20-DAs possess an additional C7-C20 bond compared to the atisine class. Thirteen new compounds (229241) isolated from the Aconitum genus have been classified as this class [35,37,93]. Compounds 229237 possess typical exocyclic double bonds between C-16 and C-17, while compounds 238241 display a hydroxyl group at C-16 and C-17. Compound 233 includes a rare ether bond between C-1 and C-19 [35]. The plant sources are shown in Table 13. Detailed chemical structures are shown in Figure 13.
  • Napelline-type C20-DAs (242249)
Napelline-type C20-DAs are structurally similar to kaurane diterpenes with the distinctive additional C7-C20 bond. The new compounds 242249 obtained from the genus Aconitum have been classified as this class [35,94,95,96]. Compounds 242245 have a typical exocyclic double bond. Among these, compound 242 is a rare N-oxide of natural napelline-type C20-DAs, and compound 245 presents a C20-DA with an iminium methine moiety [35,96]. Compounds 247248 are napelline-type hydrochloride C20-DAs with a characteristic methyl and hydroxyl group at C-16 instead of a typical exocyclic double bond [94]. Compound 249 contains a sulfonic acid unit [95]. The plant sources are shown in Table 14. Detailed chemical structures are shown in Figure 14.
  • Vakognavine-type C20-DAs (250255)
The fundamental skeleton structure of vakognavine-type C20-DAs is defined by the bond cleavage between N and C-19 of hetisines. Only six new compounds (250255) belong to this class [43,68,89,97]. Compound 253 is the first vakognavine-type C20-DA with a characteristic C2-O-C19 unit. Compounds 254 and 255 represent the first natural diterpenoid alkaloid at C-18, with an α-oriented methyl group [68]. The plant sources are shown in Table 15. Detailed chemical structures are shown in Figure 15.
  • Rearranged C20-DAs (256266)
Rearranged C20-DAs retain the characteristics of C20-DAs but with altered skeletons. Eleven new compounds (256266) obtained from Aconitum belong to this class [37,40,84,95,98,99]. Compounds 256258 are rearranged C20-DAs with racemulosine skeletons derived from denudatine–type DAs via double Wanger–Meerwein rearrangements of rings A and C [84]. Compounds 259260 are zwitterionic sulfonated C20-DAs with a rearranged atisane skeleton. Compounds 261262 are two sulfonated seco C20-DAs originating from the Criegee rearrangements; notably, compound 261 is a 13,16-seco-napelline DA, and 262 is a 12,13-seco-napelline DA [99]. Compounds 263264 are novel DAs derived through semipinacol rearrangements of the napelline-type DAs, which migrate through C13−C16 and C15−C6 bonds, respectively [95]. Compound 266 has a tetra-hydropyran ring system unlike napellines [40]. The plant sources are shown in Table 16. Detailed chemical structures are shown in Figure 16.

2.3.4. Bis-DAs (267280)

Bis-DAs are formed by condensing two molecules of diterpenoid alkaloids, typically linked via an O-ether linkage. Fourteen new members (267280), all obtained from the Aconitum plant, have been identified [84,92,100]. The plant sources are shown in Table 17. Detailed chemical structures are shown in Figure 17.
Compounds 267268 are classified as denudatine–atistine-type Bis-DAs, with denudatine and atistine fragments linked by an ether bond between C-17 and C-22′ [100]. Compound 269, with heteratisine and hetidine fragments linked by an ether bond between C-17 and C-18′, falls into the heteratisine–hetidine class of alkaloids [92]. Compounds 270275 belong to hetidine–hetisine class, whose hetidine fragments and hetisine fragments are linked by ether bonds [84]. Among these, Compounds 270274 are isomers, differing only in the position of the hydroxyl substitution in the hetisine moiety [92]. Compounds 277280, as hetidine-rearranged hetisine-class alkaloids, are bridged by a rare single bond between C-17 and C-17′ in Bis-DAs [84,92].

2.3.5. Other DAs (281289)

In addition to the diterpenoid alkaloids mentioned above, some diterpenoid alkaloids have novel structures, such as compounds 281289 [100,101,102,103]. The sources are shown in Table 18. Detailed chemical structures are shown in Figure 18.
Compounds 281283, isolated from the marine sponge Spongia sp., a marine invertebrate, represent three novel DAs with γ-lactam rings [101]. Compounds 284285 are novel DAs with a guanacastane skeleton isolated from the endophytic fungus Trichoderma koningii A729 [102].
Compounds 286287 are two C17-labdane DAs isolated from ripe fruits of Forsythia suspensa. The structure of 286 was determined as 3-hydroxyl-4,4,10,13-tetramethyl-1(2),3(4),5(10),6(7)-octahydrobenzo[f]quinolin, and 287 was obtained by oxidation of the hydroxyl group at C-3 in 286 [103]. Compounds 288289, isolated from lateral roots of Aconitum carmichaelii, represent the first reported natural C21-DAs. The structure of 288 is similar to that of denudatine- and napelline-type C20-DAs. Compound 289 is an isomer of 288, a 12,16-seco derivative of 288 [104].

3. Biological Activity

Terpenoid alkaloids, a class of compounds with far-reaching pharmacological significance, exhibit unique pharmacological effects and extensive biological activities. This section provides an overview of the biological activities of TeAs that have been newly discovered in the past five years, including anti-inflammatory activity, analgesic effect, anticancer activity, and antibacterial and antiviral properties. A table of TeAs’ biological activities is provided (Table 19).

3.1. Anti-Inflammatory Activity

The anti-inflammatory activity of TeAs has been well documented, with compounds like gentianine and benzoylaconitine among those reported [105,106]. This paper highlights 16 new anti-inflammatory members in TeAs over the past five years, including 4 monoterpene alkaloids and 12 DAs.
Delavatines C (9) and E (11) showed more significant inhibition of NO production in lipopolysaccharide (LPS)-stimulated BV2 cells compared to aminoguanidine bicarbonate, with IC50 values of 25.62 and 17.29 μM, respectively, and no cytotoxicity [18]. Stylosine A (157) showed significant inhibitory activity against LPS-induced production of inflammatory cytokines (IL-1β, COX-2, and TNF-α) in RAW264.7 cells at a dose of 0.1 μg/mL without cytotoxicity [74]. Geordine (103) exhibited specific anti-inflammatory activity and inhibited LPS-induced NO production in RAW264.7 cells at 50 μM, with an inhibition rate of 29.75% [61]. Ajacisines F-H (121, 123, and 214) showed strong anti-inflammatory activity by inhibiting LPS-induced NO production in BV-2 cells, with inhibition rates of 80% at 50 μM and no cytotoxicity [70]. Anthoroidine B (277) inhibited the production of NO and TNF-α, with IC50 values of 357.68 and 67.56 μM, respectively [84].
Forqinlingines C-D (1415) and forsyqinlingines A-B (286287) showed anti-inflammatory activities by inhibiting the release of β-glucuronidase in polymorphonuclear leukocytes (PMNs) induced by platelet-activating factor (PAF), with inhibition rates of 45.2%, 40.1%, 56.7%, and 58.6%, respectively [21].
Kamaonensines B (148) and F (153) showed more robust anti-inflammatory activities than the positive drug indomethacin (9.0 ± 1.3 μM), with IC50 values of 2.7 ± 0.5 and 0.9 ± 0.2 μM, respectively. Network pharmacological studies indicated that the anti-inflammatory mechanism may be related to the MAPK signaling pathway. In addition, molecular docking results showed that the infrequent amides and methylenedioxy groups could be the two critical pharmacophores in 148 and 153 [72].
Aconicumine A (170) exhibited anti-inflammatory activity by inhibiting LPS-activated NO production in RAW264.7 cells (IC50 = 19.7 ± 1.1 μM). Structure–activity relationship studies identified the methoxy group at its C-6 position is an effective group for anti-inflammatory activity [77].
Forrestline F (190) significantly inhibited NO activity in RAW264.7 cells (IC50 = 9.57 ± 1.34 μM). Further studies showed modulating anti-inflammatory effects through inhibiting ROS production and NF-κB, MAPK, and Nrf2 signaling pathways [53].

3.2. Analgesic Activity

Opioids and non-steroidal anti-inflammatory drugs (NSAIDs) are the primary drugs for pain treatment [107]. However, both drug classes can cause severe adverse reactions in clinical use. As an essential class of TeAs, the analgesic activity of DAs has been widely studied. Several new compounds with analgesic activity have been reported in the past five years of research. Compounds 89, 91, 176, 180, 182, 241, 259264, and 288 showed analgesic activity by inhibiting acetic acid-induced abdominal contractions in mice.
Compounds 89, 91, 176, 180, and 182 exhibited more excellent analgesic activity than the positive controls aspirin and acetaminophen. Episcopaline B (182), Pseudostapine C (89), Austroyunnanine B (91), and Episcopine A (180) significantly reduced acetic acid-induced abdominal contractions in mice in a dose-related manner, with the ID50 values of 55.0, 60.3, 48.0, and 66.1 μmol/kg, respectively [45,50,51,52]. Nagarumine C (176) demonstrated significant analgesic activity and inhibited acetic acid-induced writhing in mice at 76.0 μmol/kg [46].
Aconicatisulfonines A (259) and B (260) showed significant analgesic activity against acetic acid-induced writhing in mice, with inhibition rates of 43.2% and 64.7% (morphine, 66.8%) at 0.3 mg/kg, respectively [98]. Aconicarnine E (241) inhibited acetic acid-induced writhing in mice by 43.8% at 1.0 mg/kg [93]. Aconapelsulfonines A (261) and B (262) showed specific analgesic effects at the dose of 0.3 mg/kg, with inhibition rates of 63.6% and 19.3% (morphine, 84.6%), respectively [99]. Aconicarmisulfonines B (263) and C (264) displayed analgesic effects in mice, with inhibition rates of 31.26% and 26.84%, respectively [95]. Aconidenusulfonine A (288) showed analgesic activity, reducing acetic acid-induced writhing in mice by 26.35% at 2.0 mg/kg (i.p.), and its structure-activity relationship indicated that the analgesic activity might be related to a single bond between C-12 and C-16 [104].
The transient receptor vanilloid 1 (TRPV1) channel is a crucial target in developing new analgesics for pain management [108]. Acosinomonine B (178) showed a strong inhibitory effect on the activation of the TRPV1 channel in HEK-293 cells mediated by capsaicin (0.5 μM), with an inhibition rate of 31.78% at the concentration of 10 μM, making compound 178 a promising analgesic lead structure [79].

3.3. Antitumor Activity

TeAs have proven to be effective chemotherapeutic drugs for various cancers. For example, paclitaxel and its derivatives docetaxel and cabazitaxel have been clinically used for cancer treatment [109]. Over the past five years, studies have identified eight new members of TeAs with potential anticancer activity.
Incarvine G (12) showed cytotoxicity with the IC50 value of 60.29 μM against MDA-MB-231 cells and inhibited the migration and invasion of breast cancer cells. Further mechanistic studies showed that Incarvine G inhibited the migration and invasion of MDA-MB-231 cells by inhibiting actin cytoskeleton formation [19]. (±)-Caryopterisines A (19) and B (20) reduced kynurenine (Kyn) biosynthesis in HeLa cells by inhibiting indoleamine 2,3-dioxygenase (IDO) at doses of 10 μM with inhibition ratios of 25.7% and 29.8%, respectively [22]. Given the role of IDO cancer immunotherapy [110], compounds 19 and 20 are highlighted for their potent anticancer activities via IDO inhibition.
Commipholactam A (27) showed cytotoxicity against HepG2 and A549 cells, with IC50 values of 21.73 ± 2.86 μM and 128.50 ± 17.06 μM, respectively [28]. 8-O-ethyl-benzoyldeoxyaconine (107) demonstrated strong anticancer activity with an IC50 of 12.58 ± 1.82 and 12.76 ± 2.10 μM against human non-small-cell lung cancer A549 and H460 cells, respectively [47]. Lipojesaconitine (98) displayed significant cytotoxicity against four cell lines (A549, MDA-MB-231, MCF-7, and KB) with IC50 values ranging from 6.0 to 7.3 μM. However, it showed weak cytotoxicity against KB-VIN (IC50 = 18.6 μM), suggesting potential efflux by P-gp [57].
Brunonianines B (182) and C (183) showed significant cytotoxicity on Caco-2 (colon cancer) and Skov-3 (human ovarian cancer) cell lines. Compounds 182 and 183 showed comparable cytotoxicity to hydroxycamptothecin (HCPT) against the Caco-2 cell line, with IC50 values of 3.14 ± 0.37 and 2.41 ± 0.35 μM, respectively. Moreover, compound 182 showed stronger cytotoxicity than the HCPT (2.29 μM) Skov-3 cell line, with an IC50 value of 2.20 μM, likely due to its 19-S conformation. Further mechanistic studies showed that compound 182 could activate the Bax/Bcl-2/caspase-3 signaling pathway to induce apoptosis and effectively inhibit Skov-3 cell proliferation, migration, and invasion [80]. Ceylonamide G (281) was cytotoxic to DU145 cells, a human prostate cancer cell line, with an IC50 value of 6.9 μM and a minimum effective concentration (MEC) of 10 μM [101].

3.4. Cardioprotective Activity

Cardioprotective activity is a unique biological activity of DAs, such as Guan fu base A, which has been clinically developed to treat arrhythmias. Two new DAs with cardioprotective activity have been discovered in the past five years.
Smirnotine A (94) has some preventive effects on aconitine-induced arrhythmia in mice. The occurrence of ventricular tachycardia and ventricular flutter was significantly prolonged at 8 mg/kg, and ventricular flutter, ventricular fibrillation, and survival time of mice were prolonged considerably at 16 mg/kg [58]. Gyalanunine A (253) showed significant cardiotonic activity after perfusion in frog hearts and significantly inhibited myocardial contraction when combined with β-blockers in isolated frog hearts, suggesting that its mechanism of action may be related to epinephrine β receptors [89]. The existence of a hemiacetal moiety might be the critical structural feature necessary for the cardiac effect of 253 [89].

3.5. Antimicrobial Activity

Natural alkaloids have been proven to possess excellent antimicrobial activity. In the past five years, it has been found that several TeAs exhibit antimicrobial activity, such as antibacterial, antiviral, and antiplasmodial activities.

3.5.1. Antiviral Activity

Forsyqinlingines C-D (1415) and forsyqinlingine A-B (286287) showed antiviral activities against influenza A virus (H1N1) and respiratory syncytial virus (RSV). They exhibited IC50 values of 11.9, 15.1, 6.9, and 7.7 μM, alongside EC50 values of 13.5, 14.0, and 5.0 μM, respectively [21,103]. Tanguticulines A (199) and E (203) were effective against H1N1, inhibiting the cytopathic effect with IC50 values of 2.9 and 2.4 μg/mL, respectively [85].

3.5.2. Antibacterial Activity

Acoapetaludines D (45) and E (46) showed weak anti-Helicobacter pylori activity with minimum inhibitory concentrations (MICs) of 100 and 50 μg/mL, respectively [40]. Stylosines A (157) and B (158) exhibited antibacterial activity against Staphylococcus aureus with MIC of 2.00 and 32.00 μg/mL, respectively [74]. Koninginols A (284) and B (285) exhibited significant antibacterial activities (CMCC 63501) with MIC values of 10 and 2 μg/mL, respectively [102].

3.5.3. Antiplasmodial Activity

2-O-cinnamoyl hetisine (209) showed antimalarial activity against the Plasmodium falciparum strains Pf INDO and Pf 3D7, with the IC50 values of 1.92 μM and 10.8 μM, respectively [88].

3.6. Other Activity

In addition to the widely recognized anti-inflammatory, analgesic, antitumor, and antimicrobial activities mentioned above, recent research reported over the past five years has uncovered several TeAs with other significant biological activities, including vascular relaxation activity, antifibrosis activity, and neuroprotective activities.
Alstochonines A (1) and B (2) showed moderate vasorelaxant activity with rates of 73.6 ± 2.8% and 95.4 ± 3.7%, making the first report of the vasorelaxant activity of monoterpene alkaloids [16]. Lomatogonin C(26) displayed immunosuppressive activity, further evidenced by the inhibition of T cell proliferation and secretion of its cytokine IFN–γ in T cells stimulated with the anti-CD3/CD28 antibody [26]. (±)-Caryopterisines A (19) and B (20) inhibited estrogen E2 biosynthesis in human ovarian granulosa-like KGN cells by 57.2% and 39.9% at a dose of 10 μM, respectively.
Caryopterisine C (16) showed potential antifibrotic activity without cytotoxicity by inhibiting collagen accumulation (IC50 = 14.26 ± 1.46 μM) in NIH3T3 cells (murine embryo fibroblasts). Further studies into proteins involved in transforming growth factor-β-activated signaling pathways revealed that caryopterisine C (16) reduced collagen accumulation by inhibiting ERK1/2, P38, and SMAD2/3 phosphorylation. Apetalrine B (82) showed neuroprotective activity, with a neuroprotective rate on H2O2-induced SH-SY5Y cell injury of 77.4%. Its neuroprotective effect is believed to be achieved by inhibiting apoptosis [55]. Uncinatine-A (120) showed significant acetylcholinesterase (AChE) inhibitory activity, with IC50 values of 207.73 ± 0.3 μM [66]. Anthoroidines G (204) and I (206) showed certain AChEI activity, with IC50 values of 6.3 ± 1.6 and 9.3 ± 3 μM, respectively [84].

4. Conclusions

This review summarizes the sources, chemical structures, and biological activities of 289 TeAs discovered between 2019 and 2024, including 26 monoterpenoid alkaloids, 6 sesquiterpenoid alkaloids, and 257 DAs. DAs are the most abundant class of terpenoid alkaloids widely distributed in Aconitum and Delphinium. Seven novel DAs (283289) were obtained from Spongia sp., Trichoderma koningii A729, and Forsythia suspensa in the last five years, respectively. Monoterpene alkaloids are mainly distributed in Apocynaceae, Scrophulariaceae, and Gentianaceae, while sesquiterpene alkaloids, the least common class, are found in Dendrobium.
The majority of TeAs exhibit anti-inflammatory, antitumor, and antimicrobial properties. Among the terpenoid alkaloids discovered in the past five years, the analgesic activity is unique to diterpenoid alkaloids. Distinctively, the analgesic activity has been identified exclusively in diterpenoid alkaloids, with several DAs demonstrating analgesic effects in mice superior to standard drugs like aspirin and acetaminophen, including Episcopaline B, Pseudostapine C, and Austroyunnanine B. These findings support the potential for developing novel analgesics. Moreover, TeAs show promise as therapeutic agents for various cancers, exhibiting inhibitory effects on breast, intestinal, liver, lung, and cervical cancer cells in vitro. The efficacy of these compounds in vivo remains an area for future research. TeAs also hold potential as cardiovascular medications, exemplified by compounds like Smirnotine A, which has shown heart-protective activity.
While current research has revealed a broad spectrum of TeAs’ biological activities, most of these studies have been limited to in vitro cell viability assessments. There is a significant need for further in vivo pharmacological studies to understand the therapeutic potential of TeAs against various diseases comprehensively. This review has compiled the sources, structural characteristics, and biological activities of newly discovered compounds within the last five years, aiming to serve as a valuable resource for the continued exploration and application of terpenoid alkaloids in therapeutic contexts.

Author Contributions

Conceptualization, S.L.; methodology, X.W. and J.X.; software, X.W. and J.X.; validation, X.W. and J.X.; formal analysis, L.S. and Y.S.; investigation, X.W. and J.X.; resources, S.L.; data curation, Y.X. and J.X.; writing—original draft preparation, X.W. and J.X.; writing—review and editing, F.Z., C.N. and S.L.; visualization, C.N. and S.L.; supervision, C.N. and S.L.; project administration, S.L.; funding acquisition, S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation Project of Shandong Province, grant number ZR2023QH088, and the Research Start-up Fund for Doctor in Yantai University, grant number YX20B03.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

TeAsTerpenoid alkaloids
MEPMethylerythritol phosphate
NMRNuclear magnetic resonance
HR-ESIHigh-resolution mass spectrometry
ECDElectron capture detector
DAsDiterpenoid alkaloids
TNF-αTumor necrosis factor-α
IL-1βInterleukin-1β
NF-κBNuclear factor-κB
TRPV1Transient receptor vanilloid 1
MECMinimum effective concentration
HEK-293Human Embryonic Kidney 293 Cells
LPSLipopolysaccharide
COX-2Cyclooxygenase-2
PMNsPolymorphonuclear leukocytes
PAFPlatelet-activating factor
MAPKMitogen-activated protein kinase
AChEAcetylcholinesterase
MICsMinimum inhibitory concentrations
IC50Half maximal inhibitory concentration
ID50Half infectious dose
ERK1/2Extracellular signal-regulated kinase 1/2
ROSReactive oxygen species
Nrf2NF-E2-related factor 2
KynKynurenine
IDOIndoleamine 2,3-dioxygenase
P-gpP-glycoprotein
HCPTHydroxy camptothecin
RSVRespiratory syncytial virus
H1N1Influenza A virus
IFN–γInterferon-γ
BaxBcl-2-associated X protein
Bcl-2B-cell lymphoma-2
P38p38 mitogen-activated protein kinase (MAPK)
SMAD 2/3Mothers against decapentaplegic homolog 2/3
CMCC 63501Bacillus subtilis
MeMethyl
EtEthylic
IbutIsobutyryl
Mb2-metylbutyryl
BzBenzoyl
AsAnisoyl
VrVeratroyl
CinnCinnamoyl
AntAnthranoyl
GlcGlucose
AcAcetyl
pHbp-hydroxybenzoyl
lipoStearoyl,oleoyl,linolenoyl,linoleoyl,palmitoly

References

  1. Pereira, A.G.; Cassani, L.; Garcia-Oliveira, P.; Otero, P.; Mansoor, S.; Echave, J.; Xiao, J.; Simal-Gándara, J.; Prieto, M.A. Plant Alkaloids: Production, Extraction, and Potential Therapeutic Properties. In Natural Secondary Metabolites; Springer International Publishing: Cham, Switzerland, 2023; pp. 157–200. [Google Scholar]
  2. Wei, X.; Khan, A.; Song, D.; Dai, Z.; Liu, Y.P.; Yu, H.F.; Wang, B.; Zhu, P.F.; Ding, C.F.; Zhao, X.D.; et al. Three New Pyridine Alkaloids from Vinca major Cultivated in Pakistan. Nat. Prod. Bioprospecting 2017, 7, 323–327. [Google Scholar] [CrossRef] [PubMed]
  3. Debnath, B.; Singh, W.S.; Das, M.; Goswami, S.; Singh, M.K.; Maiti, D.; Manna, K. Role of plant alkaloids on human health: A review of biological activities. Mater. Today Chem. 2018, 9, 56–72. [Google Scholar] [CrossRef]
  4. Yan, Y.; Li, X.; Zhang, C.; Lv, L.; Gao, B.; Li, M. Research Progress on Antibacterial Activities and Mechanisms of Natural Alkaloids: A Review. Antibiotics 2021, 10, 318. [Google Scholar] [CrossRef] [PubMed]
  5. Cherney, E.C.; Baran, P.S. Terpenoid-Alkaloids: Their Biosynthetic Twist of Fate and Total Synthesis. Isr. J. Chem. 2011, 51, 391–405. [Google Scholar] [CrossRef]
  6. Kukula-Koch, W.A.; Widelski, J. Chapter 9-Alkaloids. In Pharmacognosy; Badal, S., Delgoda, R., Eds.; Academic Press: Boston, MA, USA, 2017; pp. 163–198. [Google Scholar]
  7. Zhang, X.; Tao, F.; Cui, T.; Luo, C.; Zhou, Z.; Huang, Y.; Tan, L.; Peng, W.; Wu, C. Sources, Transformations, Syntheses, and Bioactivities of Monoterpene Pyridine Alkaloids and Cyclopenta[c]pyridine Derivatives. Molecules 2022, 27, 7187. [Google Scholar] [CrossRef]
  8. Shen, Y.; Liang, W.-J.; Shi, Y.-N.; Kennelly, E.J.; Zhao, D.-K. Structural diversity, bioactivities, and biosynthesis of natural diterpenoid alkaloids. Nat. Prod. Rep. 2020, 37, 763–796. [Google Scholar] [CrossRef]
  9. Wang, M.L.; Yu, G.; Yi, S.P.; Zhang, F.Y.; Wang, Z.T.; Huang, B.; Su, R.B.; Jia, Y.X.; Gong, Z.H. Antinociceptive effects of incarvillateine, a monoterpene alkaloid from Incarvillea sinensis and possible involvement of the adenosine system. Sci. Rep. 2015, 5, 16107. [Google Scholar] [CrossRef] [PubMed]
  10. Wang, F.-P.; Liang, X.-T. C20-diterpenoid alkaloids. In The Alkaloids: Chemistry and Biology; Academic Press: Boston, MA, USA, 2002; Volume 59, pp. 1–280. [Google Scholar]
  11. Chodoeva, A.; Bosc, J.-J.; Robert, J. Aconitum Alkaloids and Biological Activities. In Natural Products: Phytochemistry, Botany and Metabolism of Alkaloids, Phenolics and Terpenes; Ramawat, K.G., Mérillon, J.-M., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 1503–1523. [Google Scholar]
  12. Tao, P.; Wang, Y.; Wang, Y. Attenuation and Structural Transformation of Crassicauline A During Sand Frying Process and Antiarrhythmic Effects of its Transformed Products. Front. Pharmacol. 2021, 12, 734671. [Google Scholar] [CrossRef]
  13. He, X.L.; Wang, S.B.; Du, G.H. Cyclovirobuxine. In Natural Small Molecule Drugs from Plants; Du, G.H., Ed.; Springer: Singapore, 2018; pp. 25–29. [Google Scholar]
  14. Munikishore, R.; Liu, R.; Zhang, S.; Zhao, Q.S.; Nian, Y.; Zuo, Z. Structurally modified Cyclovirobuxine-D Buxus alkaloids as effective analgesic agents through Cav3.2 T-Type calcium channel inhibition. Bioorganic Chem. 2023, 135, 106493. [Google Scholar] [CrossRef]
  15. Wang, F.P. Alkaloid Chemistry; Natural Products Chemistry Series; Chemical Industry Press: Beijing, China, 2008; pp. 378–431. [Google Scholar]
  16. Zhang, F.X.; Liu, H.H.; Yang, K.L.; Yang, T.; Zhou, R.X.; Miao, R.K.; Zhan, G.Q.; Guo, Z.J. New phenylpropanoids and monoterpene alkaloids with vasorelaxant activities from the branches of Alstonia scholaris. Fitoterapia 2022, 158, 105143. [Google Scholar] [CrossRef]
  17. Zhan, G.; Yang, K.; Yang, T.; Li, X.; Zhou, R.; Miao, R.; Guan, Y.; Teng, Y.; Guo, Z. A New Monoterpene Alkaloid From the Stems of Rauvolfia vomitoria. Rec. Nat. Prod. 2023, 17, 232–240. [Google Scholar] [CrossRef]
  18. Shen, X.P.; Chen, H.; Li, S.S.; Li, J.Y.; Li, X.; Zu, X.P.; Xu, X.K.; Li, X.; Shen, Y.H. Monoterpene Alkaloids from Incarvillea delavayi Bureau et Franchet and Their Inhibition against LPS Induced NO Production in BV2 Cells. Chem. Biodivers. 2022, 19, e202101013. [Google Scholar] [CrossRef] [PubMed]
  19. Song, W.; Zhang, J.; Zhao, P.; Huang, Y.; Zhang, S. A New Monoterpene Alkaloid from Incarvillea sinensis with Migration Inhibitory Activity on Cancer Cell. Chem. Biodivers. 2023, 20, e202201203. [Google Scholar] [CrossRef]
  20. Huo, Y.-F.; Wang, H.-L.; Wei, E.-H.; Jia, P.-S.; Liu, H.-Q.; Fan, X.-L.; Yana, K.-L. Two new compounds from the roots of Scrophularia ningpoensis and their anti-inflammatory activities. J. Asian Nat. Prod. Res. 2019, 21, 1083–1089. [Google Scholar] [CrossRef] [PubMed]
  21. Li, W.; Sun, L.T.; Zhao, L.; Yue, X.D.; Dai, S.J. New C9-Monoterpenoid Alkaloids Featuring a Rare Skeleton with Anti-Inflammatory and Anti-viral Activities from Forsythia suspensa. Chem. Biodivers. 2021, 19, e202100668. [Google Scholar]
  22. Zhang, X.; Luo, G.; Cao, Y.; Mishig, D.; Lodonjav, M.; Pan, D.; Yao, X.; Wang, F.; Zhang, G.; Luo, Y. (±)-Caryopterisines A and B, dimeric monoterpene alkaloids with unprecedented 6/5/5/5/6 pentacyclic rings scaffold from Caryopteris Glutinosa. Bioorganic Chem. 2021, 116, 105364. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, X.; Cao, Y.; Pan, D.; Yao, X.; Wang, F.; Zhang, G.; Luo, Y. Anti-fibrotic pyridine-containing monoterpene alkaloids from Caryopteris glutinosa. Phytochemistry 2022, 203, 113378. [Google Scholar] [CrossRef] [PubMed]
  24. Mishig, D.; Gruner, M.; Lübken, T.; Ganbaatar, C.; Regdel, D.; Knölker, H.J. Isolation and structure elucidation of pyridine alkaloids from the aerial parts of the Mongolian medicinal plant Caryopteris mongolica Bunge. Sci. Rep. 2021, 11, 13740. [Google Scholar] [CrossRef] [PubMed]
  25. Guo, K.; Ren, X.; Zhou, T.T.; Li, T.; Liu, Y.C.; Tao, Y.; Hu, H.; Li, D.; Liu, Y.; Li, S.H. Secondary metabolites from the Mongolian medicine Lomatogonium carinthiacum. Fitoterapia 2023, 165, 105402. [Google Scholar] [CrossRef]
  26. Salim, F.; Yunus, Y.M.; Anouar, E.H.; Awang, K.; Langat, M.; Cordell, G.A.; Ahmad, R. Absolute Configuration of Alkaloids from Uncaria longiflora through Experimental and Computational Approaches. J. Nat. Prod. 2019, 82, 2933–2940. [Google Scholar] [CrossRef]
  27. Brocksom, T.; de Oliveira, K.; Desiderá, A. The Chemistry of the Sesquiterpene Alkaloids. J. Braz. Chem. Soc. 2017, 28, 933–942. [Google Scholar] [CrossRef]
  28. Zhu, S.S.; Qin, D.P.; Wang, S.X.; Yang, C.; Li, G.P.; Cheng, Y.X. Commipholactam A, a cytotoxic sesquiterpenoidal lactam from Resina Commiphora. Fitoter. 2019, 134, 382–388. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, G.Y.; Tan, L.; Cheng, L.; Ding, L.S.; Zhou, Y.; Deng, Y.; He, Y.Q.; Guo, D.L.; Xiao, S.J. Dendrobine-type alkaloids and bibenzyl derivatives from Dendrobium findlayanum. Fitoterapia 2020, 142, 104497. [Google Scholar] [CrossRef] [PubMed]
  30. Yang, D.; Cheng, Z.Q.; Hou, B.; Yang, L.; Zi, C.T.; Dong, F.W.; Hu, J.M.; Zhou, J. Two unusual dendrobine-type alkaloids from Dendrobium findlayanum. Fitoterapia 2020, 144, 104607. [Google Scholar] [CrossRef] [PubMed]
  31. Luo, X.; Wu, R.; Han, X.; Tang, X.; Wang, Q.; Li, P.; Li, G. Guaiane sesquiterpenes from the gorgonian Echinogorgia flora collected in the South China Sea. RSC Adv. 2022, 12, 2662–2667. [Google Scholar] [CrossRef]
  32. Thawabteh, A.M.; Thawabteh, A.; Lelario, F.; Bufo, S.A.; Scrano, L. Classification, Toxicity and Bioactivity of Natural Diterpenoid Alkaloids. Molecules 2021, 26, 4103. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, F.P.; Chen, Q.H.; Liu, X.Y. Diterpenoid alkaloids. Nat. Prod. Rep. 2010, 27, 529–570. [Google Scholar] [CrossRef] [PubMed]
  34. Xue, W.J.; Zhao, B.; Zhao, J.Y.; Sagdullaev, S.S.; Akber Aisa, H. Three new diterpenoid alkaloids from Delphinium naviculare var. lasiocarpum W. T. Wang. Phytochem. Lett. 2019, 33, 12–16. [Google Scholar] [CrossRef]
  35. Jiang, G.Y.; Qin, L.L.; Gao, F.; Huang, S.; Zhou, X.L. Fifteen new diterpenoid alkaloids from the roots of Aconitum kirinense Nakai. Fitoterapia 2020, 141, 104477. [Google Scholar] [CrossRef]
  36. Yang, H.B.; Luo, Y.Y.; Xu, J.B.; Liu, Y.; Gao, F.; Huang, S.; Chen, L.; Zhou, X.L. Two New C18-Diterpenoid Alkaloids from Aconitum leucostomum Worosch. Chem. Biodivers. 2022, 19, e202200483. [Google Scholar] [CrossRef]
  37. Ablajan, N.; Zhao, B.; Zhao, J.Y.; Wang, B.L.; Sagdullaev, S.S.; Aisa, H.A. Diterpenoid alkaloids from Aconitum barbatum var. puberulum Ledeb. Phytochemistry 2021, 181, 112567. [Google Scholar] [CrossRef] [PubMed]
  38. Hu, J.; Lv, Y.; Li, Q.; Mao, X.; Peng, T.; Gao, X.; Jin, N.; Yin, S.; Shi, X.; Wang, W. Diterpenoid Alkaloids from the Roots of Aconitum iochanicum. Chem. Nat. Compd. 2020, 56, 492–495. [Google Scholar] [CrossRef]
  39. Liu, X.Y.; Ke, B.W.; Qin, Y.; Wang, F.P. Chapter One-The diterpenoid alkaloids. In The Alkaloids: Chemistry and Biology; Knölker, H.J., Ed.; Academic Press: Boston, MA, USA, 2022; Volume 87, pp. 1–360. [Google Scholar]
  40. Hu, Z.X.; An, Q.; Tang, H.Y.; Chen, Z.H.; Aisa, H.A.; Zhang, Y.; Hao, X.J. Acoapetaludines A-K, C20 and C19-diterpenoid alkaloids from the whole plants of Aconitum apetalum (Huth) B. Fedtsch. Phytochemistry 2019, 167, 112111. [Google Scholar] [CrossRef] [PubMed]
  41. Wan, Z.L.; Zhang, M.; Xiao, Y.; Chen, L.; Xie, J.; Huang, S.; Zhou, X.L. C19-diterpenoid alkaloids from Aconitum refractum var. circinatum. Nat. Prod. Res. 2023, 1–6. [Google Scholar] [CrossRef] [PubMed]
  42. Ding, N.; Wang, S.; Xiao, Y.; Huang, S.; Chen, L.; Xie, J.; Zhou, X.L. Four New C19-Diterpenoid Alkaloids from Aconitum nagarum Stapf. Chem. Biodivers. 2023, 20, e202300058. [Google Scholar] [CrossRef] [PubMed]
  43. Zhou, X.; Chen, L.; Yang, C.; Ren, J.; Feng, Y.; Huang, S. Diterpenoid Alkaloids from the Roots of Aconitum rockii and Their Antifeedant Activity. Chin. J. Org. Chem. 2022, 42, 1856. [Google Scholar]
  44. Xing, F.; Xie, J.; Luo, Y.Y.; Huang, S.; Gao, F.; Chen, L.; Zhou, X.L. C19-Diterpenoid alkaloids from Aconitum richardsonianum. Phytochem. Lett. 2022, 51, 149–154. [Google Scholar] [CrossRef]
  45. Hu, J.; Wu, Q.; Li, Q.; Lv, T.; Peng, T.-F.; Yin, S.; Jin, H.-Z. Antinociceptive diterpenoid alkaloids from the roots of Aconitum Austroyunnanense. J. Asian Nat. Prod. Res. 2022, 25, 132–138. [Google Scholar] [CrossRef]
  46. Hu, J.; Li, S.F.; Li, Q.; Lv, T.; Peng, T.F.; Yin, S.; Min, Y. Antinociceptive C19–diterpenoid alkaloids from the roots of Aconitum nagarum. J. Asian Nat. Prod. Res. 2022, 25, 540–546. [Google Scholar] [CrossRef]
  47. Yu, Y.; Wu, S.; Zhang, J.; Li, J.; Yao, C.; Wu, W.; Wang, Y.; Ji, H.; Wei, W.; Gao, M.; et al. Structurally diverse diterpenoid alkaloids from the lateral roots of Aconitum carmichaelii Debx. and their anti-tumor activities based on in vitro systematic evaluation and network pharmacology analysis. RSC Adv. 2021, 11, 26594–26606. [Google Scholar] [CrossRef]
  48. Wang, J.; Lou, H.; Li, J.; Liu, Y.; Han, H.; Yang, Z.; Pan, W.; Chen, Z. C19-diterpenoid alkaloids from the rhizomes of Aconitum pendulum. Fitoterapia 2021, 151, 104887. [Google Scholar] [CrossRef] [PubMed]
  49. Lu, J.; Xu, J.B.; Li, X.; Zhou, X.L.; Zhang, C.; Gao, F. Three New C19-Diterpenoid Alkaloids from Aconitum novoluridum. Chem. Pharm. Bull. 2021, 69, 811–816. [Google Scholar] [CrossRef] [PubMed]
  50. Hu, J.; Li, J.X.; Li, Q.; Mao, X.; Peng, T.F.; Liu, H.Q.; Yin, S.; Yuan, H.J. Antinociceptive C19–diterpenoid alkaloids from the root of Aconitum episcopale. J. Asian Nat. Prod. Res. 2021, 24, 617–623. [Google Scholar] [CrossRef] [PubMed]
  51. Hu, J.; Li, J.X.; Li, Q.; Mao, X.; Peng, T.F.; Jin, N.H.; Yin, S.; Tang, Y. Antinociceptive C19–diterpenoid alkaloids isolated from Aconitum pseudostapfianum. J. Asian Nat. Prod. Res. 2021, 23, 637–643. [Google Scholar] [CrossRef] [PubMed]
  52. Hu, J.; Li, J.; Li, Q.; Mao, X.; Peng, T.; Jin, N.; Yin, S.; Shi, X.; Li, Y. Antinociceptive C19-Diterpenoid Alkaloids from Aconitum episcopale. Chem. Nat. Compd. 2021, 57, 503–506. [Google Scholar] [CrossRef]
  53. Song, Z.; Gao, C.; Jiang, Q.; Xu, J.; Xiong, L.; Liu, K.; Sun, D.; Li, H.; Chen, L. Diterpenoid alkaloids from Delphinium forrestii var. viride and their anti-inflammation activity. Phytochemistry 2021, 192, 112971. [Google Scholar]
  54. Wang, Z.S.; Chen, W.; Jiang, H.Y.; Gao, F.; Zhou, X.L. Semi-synthesis and structural elucidation of brevicanines A–D, four new C19-diterpenoid alkaloids with rotameric phenomenon from Aconitum brevicalcaratum. Fitoterapia 2019, 134, 404–410. [Google Scholar] [CrossRef]
  55. Wan, L.X.; Zhang, J.F.; Zhen, Y.Q.; Zhang, L.; Li, X.; Gao, F.; Zhou, X.L. Isolation, Structure Elucidation, Semi-Synthesis, and Structural Modification of C19-Diterpenoid Alkaloids from Aconitum apetalum and Their Neuroprotective Activities. J. Nat. Prod. 2021, 84, 1067–1077. [Google Scholar] [CrossRef]
  56. Si, Y.; Ding, X.; Adelakuna, T.A.; Zhang, Y.; Hao, X.J. Acotarines A-G, new diterpenoid alkaloids from Aconitum taronense induce lysosomal biogenesis. Fitoterapia 2020, 147, 104738. [Google Scholar] [CrossRef]
  57. Yamashita, H.; Miyao, M.; Hiramori, K.; Kobayashi, D.; Suzuki, Y.; Mizukami, M.; Goto, M.; Lee, K.H.; Wada, K. Cytotoxic diterpenoid alkaloid from Aconitum japonicum subsp. subcuneatum. J. Nat. Med. 2019, 74, 83–89. [Google Scholar] [CrossRef]
  58. Zhao, B.; Ablajan, N.; Zhao, J.Y.; Kodirova, D.R.; Sagdullaev, S.S.; Aisa, H.A. Two new C19-diterpenoid alkaloids from Aconitum smirnovii. Phytochem. Lett. 2020, 38, 96–100. [Google Scholar] [CrossRef]
  59. Alhilal, M.; Sulaiman, Y.A.M.; Alhilal, S.; Gomha, S.M.; Ouf, S.A. Antifungal Activity of New Diterpenoid Alkaloids Isolated by Different Chromatographic Methods from Delphinium peregrinum L. var. eriocarpum Boiss. Molecules 2021, 26, 1375. [Google Scholar] [CrossRef] [PubMed]
  60. Zhou, X.; Chen, L.; Xie, J.; Ding, H.; Wan, D.; Li, Q.; Zeng, H.; Xing, F. Three New C19-Diterpenoid Alkaloids from Aconitum brevicalcaratum. Chin. J. Org. Chem. 2023, 43, 2245. [Google Scholar]
  61. Yao, L.L.; Zhang, S.Q.; Guo, C.; Li, B.X.; Yang, H.J.; Yin, T.P.; Cai, L. A new C19-diterpenoid alkaloid in Aconitum georgei Comber. Nat. Prod. Res. 2022, 38, 85–90. [Google Scholar] [CrossRef]
  62. Ablajan, N.; Xue, W.J.; Zhao, J.Y.; Sardorbek, A.; Boymirzayevich Begmatov, N.; Sagdullaev, S.; Zhao, B.; Akber Aisa, H. Two New C19-Diterpenoid Alkaloids from Delphinium shawurense. Chem. Biodivers. 2023, 20, e202200936. [Google Scholar] [CrossRef] [PubMed]
  63. Yan, Y.; Jiang, H.; Yang, X.; Ding, Z.; Yin, T. Grandiflolines A–F, new anti-inflammatory diterpenoid alkaloids isolated from Delphinium grandiflorum. Front. Chem. 2022, 10, 1012874. [Google Scholar] [CrossRef]
  64. Ma, H.; Ma, Y.; Dawa, Z.; Yao, Y.; Wang, M.; Zhang, K.; Zhu, C.; Liu, F.; Lin, C. Diterpenoid Alkaloids Isolated from Delphinium brunonianum and Their Inhibitory Effects on Hepatocytes Lipid Accumulation. Molecules 2022, 27, 2257. [Google Scholar] [CrossRef]
  65. Li, J.M.; Li, X.; Gao, F.; Cai, L.; Liang, X.X.; Chen, L.; Zhou, X.L. Five new diterpenoid alkaloids from Aconitum sczukinii Turcz. Phytochem. Lett. 2022, 47, 120–124. [Google Scholar] [CrossRef]
  66. Ahmad, S.; Gul, N.; Ahmad, M.; Almehmadi, M.; Shafie, A.; Shah, S.A.A.; Rahman, N.U.; Ahmad, H. Isolation, crystal structure, DFT calculation and molecular docking of uncinatine-A isolated from Delphinium uncinatum. Fitoterapia 2022, 162, 105268. [Google Scholar] [CrossRef]
  67. Yamashita, H.; Doi, N.; Hanawa, N.; Itoh, M.; Nakano, W.; Takahashi, E.; Mizukami, M.; Kaneda, K.; Suzuki, Y.; Goto, M.; et al. Eleven new C19-diterpenoid alkaloids from Delphinium elatum cv. Pacific Giant. J. Nat. Med. 2021, 76, 161–170. [Google Scholar]
  68. Xu, J.B.; Li, Y.Z.; Huang, S.; Chen, L.; Luo, Y.Y.; Gao, F.; Zhou, X.L. Diterpenoid alkaloids from the whole herb of Delphinium grandiflorum L. Phytochemistry 2021, 190, 112866. [Google Scholar] [CrossRef]
  69. He, Y.; Zhang, D.; West, L.M. Delphatisine D and Chrysotrichumine A, two new diterpenoid alkaloids from Delphinium chrysotrichum. Fitoterapia 2019, 139, 104407. [Google Scholar] [CrossRef]
  70. Tang, Q.; Shen, X.; Hao, Y.K.; Yang, S.Y.; Fu, J.T.; Wu, T.Y.; Zhao, H.Y.; Qin, B.; Li, Y.L.; Zhang, Y.B.; et al. Diterpenoid Alkaloids from Delphinium ajacis and Their Anti-inflammatory Activity. Chem. Biodivers. 2024, 21, e202301958. [Google Scholar] [CrossRef] [PubMed]
  71. Zhou, X.; Chen, L.; Liu, Y.; Zhao, Y.D.; Zhong, H.Y.; Chen, S.Y.; Huang, S. Diterpenoid Alkaloids from Delphinium liangshanense. Chem. Biodivers. 2024, e202301923. [Google Scholar] [CrossRef]
  72. Jing, D.; Zhang, Y.; Gong, C.; Du, K.; Wang, Y.; Lai, L.; Meng, D. Kamaonensine A-G: Lycaconitine-type C19-diterpenoid alkaloids with anti-inflammatory activities from Delphinium kamaonense Huth. Phytochemistry 2023, 215, 113822. [Google Scholar] [CrossRef]
  73. Wang, Y.; Sun, D.; Chen, Y.; Xu, J.; Xu, Y.; Yue, X.; Jia, J.; Li, H.; Chen, L. Alkaloids of Delphinium grandiflorum and their implication to H2O2-induced cardiomyocytes injury. Bioorganic Med. Chem. 2021, 37, 116113. [Google Scholar] [CrossRef]
  74. Shu, Y.; Wang, J.P.; Cai, X.Y.; Li, X.L.; Hu, J.T.; Sun, C.T.; Cai, L.; Ding, Z.T. Stylosines A and B, anti-inflammatory diterpenoid alkaloids from Aconitum stylosum. Tetrahedron 2020, 76, 131520. [Google Scholar] [CrossRef]
  75. Li, Y.Z.; Qin, L.L.; Gao, F.; Shan, L.H.; Zhou, X.L. Kusnezosines A-C, three C19-diterpenoid alkaloids with a new skeleton from Aconitum kusnezoffii Reichb. var. gibbiferum. Fitoterapia 2020, 144, 104609. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, N.; Xia, F.; Li, S.Y.; Nian, Y.; Wei, L.X.; Xu, G. Diterpenoid Alkaloids from the Aerial Parts of Aconitum flavum Hand.-Mazz. Nat. Prod. Bioprospecting 2021, 11, 421–429. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, D.B.; Liang, Y.N.; Wang, Z.; Shi, L.K.; Zhang, Z.; Tang, Z.S.; Huang, L.Q. Aconicumines A–D, an advanced class of norditerpenoid alkaloids with an unprecedented N,O-diacetal motif from Aconitum taipeicum Hand.-Mazz., exhibit anti-inflammatory properties in vitro. Phytochemistry 2023, 210, 113675. [Google Scholar] [CrossRef]
  78. Li, X.; Ye, M.; Gao, F.; Zhou, X.; Chen, L.; Huang, S. A new diterpenoid alkaloid from Delphinium gyalanum C. Marquand & Airy Shaw. Nat. Prod. Res. 2021, 37, 130–135. [Google Scholar] [PubMed]
  79. Li, Y.; Zeng, J.; Tian, Y.H.; Hou, Y.; Da, H.; Fang, J.; Gao, K. Isolation, identification, and activity evaluation of diterpenoid alkaloids from Aconitum sinomontanum. Phytochemistry 2021, 190, 112880. [Google Scholar] [CrossRef] [PubMed]
  80. Li, Q.; Wang, Z.W.; Wang, M.X.; Yu, H.L.; Chen, L.; Cai, Z.; Zhang, Y.; Gu, M.M.; Shao, Y.L.; Han, H.P.; et al. Brunonianines A-C, C20-diterpenoid alkaloids with cyano group from Delphinium brunonianum Royle. Phytochemistry 2024, 219, 113987. [Google Scholar] [CrossRef] [PubMed]
  81. Ahunova, H.; Bobakulov, K.M.; Turgunov, K.K.; Tashkhodzhaev, B.; Mukarramov, N.I.; Abdullaev, S.V. Alkaloids from Delphinium oreophilum. The New Diterpene Alkaloid 15-Epinaviculine B. Chem. Nat. Compd. 2023, 59, 1137–1141. [Google Scholar] [CrossRef]
  82. Yin, T.; Zhang, H.; Zhang, W.; Jiang, Z. Chemistry and biological activities of hetisine-type diterpenoid alkaloids. RSC Adv. 2021, 11, 36023–36033. [Google Scholar] [CrossRef]
  83. Pu, Y.L.; Tian, L.F.; Chen, L.; Deng, M.Y.; Xie, J.; Huang, S.; Zhou, X.-L. Diterpenoid alkaloids from Delphinium trichophorum. J. Asian Nat. Prod. Res. 2023, 25, 1175–1183. [Google Scholar] [CrossRef] [PubMed]
  84. Huang, S.; Zhang, J.F.; Chen, L.; Gao, F.; Zhou, X.L. Diterpenoid alkaloids from Aconitum anthoroideum that offer protection against MPP±Induced apoptosis of SH-SY5Y cells and acetylcholinesterase inhibitory activity. Phytochemistry 2020, 178, 112459. [Google Scholar] [CrossRef] [PubMed]
  85. Fan, X.; Yang, L.; Liu, Z.; Lin, L.; Li, C.; Guo, S.; Wang, Z.; Wang, Z.; Sui, F. Diterpenoid alkaloids from the whole plant of Aconitum tanguticum (Maxim.) Stapf. Phytochemistry 2019, 160, 71–77. [Google Scholar] [CrossRef] [PubMed]
  86. Yan, Y.F.; Wang, Y.R.; Jiang, H.J.; Ding, Z.B.; Yin, T.P. New diterpenoid alkaloids from Delphinium pachycentrum Hemsl. Nat. Prod. Res. 2022, 38, 1487–1493. [Google Scholar] [CrossRef]
  87. Xu, J.B.; Luo, Y.Y.; Huang, S.; Gao, F.; Zhou, X.L. Four New Diterpenoid Alkaloids from The Roots of Aconitum coreanum. Chem. Biodivers. 2020, 17, e1900600. [Google Scholar] [CrossRef]
  88. Anmol; Kumari, S.; Kumar, R.; Singh, R.; Aggarwal, G.; Agrawal, P.; Sahal, D.; Sharma, U. Antiplasmodial diterpenoid alkaloid from Aconitum heterophyllum Wall. ex Royle: Isolation, characterization, and UHPLC-DAD based quantification. J. Ethnopharmacol. 2022, 287, 114931. [Google Scholar] [CrossRef] [PubMed]
  89. Ye, M.Z.; Li, X.Y.; Xie, J.; Chen, L.; Gao, F.; Zhou, X.L.; Huang, S. Gyalanunines A and B, two new C20-diterpenoid alkaloids from Delphinium gyalanum. Tetrahedron Lett. 2022, 108, 154153. [Google Scholar] [CrossRef]
  90. Li, H.Y.; Yan, B.C.; Wei, L.X.; Sun, H.D.; Puno, P.T. Tangutidines A-C, Three Amphoteric Diterpene Alkaloids from Aconitum tanguticum. Nat. Prod. Bioprospecting 2021, 11, 459–464. [Google Scholar] [CrossRef] [PubMed]
  91. Kurbanov, U.K.; Levkovich, M.G.; Mukarramov, N.I. New Diterpene Alkaloid from Delphinium paradoxum. Chem. Nat. Compd. 2024, 60, 115–118. [Google Scholar] [CrossRef]
  92. Ye, X.; Wang, Y.; Chen, Q.; Feng, Q.; Liu, T.; Feng, W.; Liang, Y.; Liu, X.; Li, C.; Wang, Z. Anti-inflammatory diterpenoid alkaloids from Aconitum tanguticum (Maxim.) Stapf. Phytochemistry 2023, 206, 113524. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, H.; Shao, S.; Xia, H.; Wu, Y.Z.; Zhu, C.G.; Xu, C.B.; Zhang, T.T.; Guo, Q.L.; Shi, J.G. Denudatine-type diterpenoid alkaloids from an aqueous extract of the lateral root of Aconitum carmichaelii. J. Asian Nat. Prod. Res. 2021, 23, 615–626. [Google Scholar] [CrossRef] [PubMed]
  94. Li, H.; Xu, J.; Zhang, X.; Liu, C.; Chen, N.; Naruo, A.; Qin, Z.; Ao, M. New napelline-type diterpenoid alkaloids from Aconiti kusnezoffii roots: Structure elucidation, plausible biogenetic pathway and biological activities. Phytochem. Lett. 2021, 43, 53–59. [Google Scholar] [CrossRef]
  95. Guo, Q.; Xia, H.; Wu, Y.; Shao, S.; Xu, C.; Zhang, T.; Shi, J. Structure, property, biogenesis, and activity of diterpenoid alkaloids containing a sulfonic acid group from Aconitum carmichaelii. Acta Pharm. Sin. B 2020, 10, 1954–1965. [Google Scholar] [CrossRef]
  96. Wang, J.; Meng, X.H.; Chai, T.; Yang, J.L.; Shi, Y.P. Diterpenoid Alkaloids and One Lignan from the Roots of Aconitum pendulum Busch. Nat. Prod. Bioprospecting 2019, 9, 419–423. [Google Scholar] [CrossRef]
  97. Huang, S.; Wang, J.Z.; Guo, Q.J.; Xiao, Y.; Feng, Y.M.; Chen, L.; Xie, J.; Zhou, X.L. Diterpenoid alkaloids from two species of Delphinium. J. Asian Nat. Prod. Res. 2022, 25, 718–730. [Google Scholar] [CrossRef]
  98. Wu, Y.; Shao, S.; Guo, Q.; Xu, C.; Xia, H.; Zhang, T.; Shi, J. Aconicatisulfonines A and B, Analgesic Zwitterionic C20-Diterpenoid Alkaloids with a Rearranged Atisane Skeleton from Aconitum Carmichaelii. Org. Lett. 2019, 21, 6850–6854. [Google Scholar] [CrossRef]
  99. Liu, H.; Wu, Y.; Guo, Q.; Shao, S.; Xu, C.; Zhang, T.; Shi, J. Aconapelsulfonines A and B, seco C20-diterpenoid alkaloids deriving via Criegee rearrangements of napelline skeleton from Aconitum carmichaelii. Chin. Chem. Lett. 2021, 32, 33–36. [Google Scholar] [CrossRef]
  100. He, J.R.; Zhang, L.M.; Lou, H.B.; Lv, D.; Zuo, A.X.; Shen, Y. Two new bis-diterpenoid alkaloids from Aconitum weixiense. J. Asian Nat. Prod. Res. 2022, 25, 842–848. [Google Scholar] [CrossRef]
  101. Jomori, T.; Setiawan, A.; Sasaoka, M.; Arai, M. Cytotoxicity of New Diterpene Alkaloids, Ceylonamides G-I, Isolated From Indonesian Marine Sponge of Spongia sp. Nat. Prod. Commun. 2019, 14, 1934578X19857294. [Google Scholar] [CrossRef]
  102. Chen, S.; Li, H.; Chen, Y.; Li, S.; Xu, J.; Guo, H.; Liu, Z.; Zhu, S.; Liu, H.; Zhang, W. Three new diterpenes and two new sesquiterpenoids from the endophytic fungus Trichoderma koningiopsis A729. Bioorganic Chem. 2019, 86, 368–374. [Google Scholar] [CrossRef] [PubMed]
  103. Li, W.; Zhao, L.; Sun, L.T.; Xie, Z.P.; Zhang, S.M.; Yue, X.D.; Dai, S.J. Trinorlabdane diterpenoid alkaloids featuring an unprecedented skeleton with anti-inflammatory and antiviral activities from Forsythia suspensa. RSC Adv. 2021, 11, 29684–29689. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, J.; Lei, X.; Wei, Y.; Liu, H.; Guo, Q.; Zhang, T.; Shi, J. Two unique C21-diterpenoid alkaloids from Aconitum carmichaelii. Chin. Chem. Lett. 2022, 33, 5047–5050. [Google Scholar] [CrossRef]
  105. Wang, N.; Liu, Y.; Jia, C.; Gao, C.; Zheng, T.; Wu, M.; Zhang, Q.; Zhao, X.; Li, Z.; Chen, J.; et al. Machine learning enables discovery of Gentianine targeting TLR4/NF-κB pathway to repair ischemic stroke injury. Pharmacol. Res. 2021, 173, 105913. [Google Scholar] [CrossRef]
  106. Yu, H.H.; Li, M.; Li, Y.B.; Lei, B.B.; Yuan, X.; Xing, X.K.; Xie, Y.F.; Wang, M.; Wang, L.; Yang, H.J.; et al. Benzoylaconitine Inhibits Production of IL-6 and IL-8 via MAPK, Akt, NF-κB Signaling in IL-1β-Induced Human Synovial Cells. Biol. Pharm. Bull. 2020, 43, 334–339. [Google Scholar] [CrossRef]
  107. Sisignano, M.; Geisslinger, G. Rethinking the use of NSAIDs in early acute pain. Trends Pharmacol. Sci. 2023, 44, 193–195. [Google Scholar] [CrossRef]
  108. Basso, L.; Aboushousha, R.; Fan, C.Y.; Iftinca, M.; Melo, H.; Flynn, R.; Agosti, F.; Hollenberg, M.D.; Thompson, R.; Bourinet, E.; et al. TRPV1 promotes opioid analgesia during inflammation. Sci. Signal. 2019, 12, eaav0711. [Google Scholar] [CrossRef] [PubMed]
  109. Sousa-Pimenta, M.; Estevinho, L.M.; Szopa, A.; Basit, M.; Khan, K.; Armaghan, M.; Ibrayeva, M.; Sönmez Gürer, E.; Calina, D.; Hano, C.; et al. Chemotherapeutic properties and side-effects associated with the clinical practice of terpene alkaloids: Paclitaxel, docetaxel, and cabazitaxel. Front. Pharmacol. 2023, 14, 1157306. [Google Scholar] [CrossRef] [PubMed]
  110. Le Naour, J.; Galluzzi, L.; Zitvogel, L.; Kroemer, G.; Vacchelli, E. Trial watch: IDO inhibitors in cancer therapy. OncoImmunology 2020, 9, 1777625. [Google Scholar] [CrossRef] [PubMed]
Figure 2. Structures of sesquiterpene alkaloids (2732).
Figure 2. Structures of sesquiterpene alkaloids (2732).
Molecules 29 01968 g002
Figure 3. Structures of C18-DAs (3343).
Figure 3. Structures of C18-DAs (3343).
Molecules 29 01968 g003
Figure 4. Structures of aconitine-type C19-DAs (44113).
Figure 4. Structures of aconitine-type C19-DAs (44113).
Molecules 29 01968 g004
Figure 5. Structures of lycoctonine-type C19-DAs (114153).
Figure 5. Structures of lycoctonine-type C19-DAs (114153).
Molecules 29 01968 g005
Figure 6. Structures of lactone-type C19-DAs (154158).
Figure 6. Structures of lactone-type C19-DAs (154158).
Molecules 29 01968 g006
Figure 7. Structures of franchetine-type C19-DAs (159168).
Figure 7. Structures of franchetine-type C19-DAs (159168).
Molecules 29 01968 g007
Figure 8. Structures of 7,17-seco-type C19-DAs (169175).
Figure 8. Structures of 7,17-seco-type C19-DAs (169175).
Molecules 29 01968 g008
Figure 9. Structures of rearranged C19-DAs (176182).
Figure 9. Structures of rearranged C19-DAs (176182).
Molecules 29 01968 g009
Figure 10. Structures of atisine-type C20-DAs (183191).
Figure 10. Structures of atisine-type C20-DAs (183191).
Molecules 29 01968 g010
Figure 11. Structures of hetisine-type C20-DAs (192217).
Figure 11. Structures of hetisine-type C20-DAs (192217).
Molecules 29 01968 g011
Figure 12. Structures of hetidine-type C20-DAs (218228).
Figure 12. Structures of hetidine-type C20-DAs (218228).
Molecules 29 01968 g012
Figure 13. Structures of denudatine-type C20-DAs (229241).
Figure 13. Structures of denudatine-type C20-DAs (229241).
Molecules 29 01968 g013
Figure 14. Structures of napelline-type C20-DAs (242249).
Figure 14. Structures of napelline-type C20-DAs (242249).
Molecules 29 01968 g014
Figure 15. Structures of vakognavine-type C20-DAs (250255).
Figure 15. Structures of vakognavine-type C20-DAs (250255).
Molecules 29 01968 g015
Figure 16. Structures of rearranged C20-DAs (256266).
Figure 16. Structures of rearranged C20-DAs (256266).
Molecules 29 01968 g016
Figure 17. Structures of Bis-DAs (267280).
Figure 17. Structures of Bis-DAs (267280).
Molecules 29 01968 g017
Figure 18. Structures of other DAs (281289).
Figure 18. Structures of other DAs (281289).
Molecules 29 01968 g018
Table 7. Names and plant sources of franchetine-type C19-DAs (159168).
Table 7. Names and plant sources of franchetine-type C19-DAs (159168).
No.Compound NameSourcesPlant PartsRef.
159Acotarine AAconitum taronenseroot[56]
160Acotarine BAconitum taronenseroot[56]
161Acotarine CAconitum taronenseroot[56]
162Acotarine DAconitum taronenseroot[56]
163Acotarine EAconitum taronenseroot[56]
164Flavumoline AAconitum flavum Hand.-Mazzaerial parts[76]
165Flavumoline BAconitum flavum Hand.-Mazzaerial parts[76]
166Flavumoline CAconitum flavum Hand.-Mazzaerial parts[76]
167Flavumoline DAconitum flavum Hand.-Mazzaerial parts[76]
168Rockidine CAconitum generaroot[43]
Table 8. Names and plant sources of 7,17-seco-type C19-DAs (169175).
Table 8. Names and plant sources of 7,17-seco-type C19-DAs (169175).
No.Compound NameSourcesPlant PartsRef.
169Grandifline BDelphinium grandiflorumaerial part[73]
170Aconicumine AAconitum taipeicum Hand.-Mazz.root[77]
171Aconicumine BAconitum taipeicum Hand.-Mazz.root[77]
172Aconicumine CAconitum taipeicum Hand.-Mazz.root[77]
173Aconicumine DAconitum taipeicum Hand.-Mazz.root[77]
174Brunodelphinine BDelphinium brunonianum Royleaerial part[64]
175Brunodelphinine DDelphinium brunonianum Royleaerial part[64]
Table 9. Names and plant sources of rearranged C19-DAs (176182).
Table 9. Names and plant sources of rearranged C19-DAs (176182).
No.Compound NameSourcesPlant PartsRef.
176Nagarumine CAconitum nagarumroot[46]
177Acosinomonine AAconitum sinomontanumroot[79]
178Acosinomonine BAconitum sinomontanumroot[79]
179Grandifline ADelphinium grandiflorum L.whole herb[73]
180Episcopine AAconitum episcopaleroot[52]
181Gyalanutine ADelphinium gyalanum C. Marquand & Airy Shawwhole plant[78]
182Episcopaline BAconitum episcopaleroot[50]
Table 10. Name and plant sources of atisine-type C20-DAs (183191).
Table 10. Name and plant sources of atisine-type C20-DAs (183191).
No.Compound NameSourcesPlant PartsRef.
183Brunonianine ADelphinium brunonianum.whole plant[80]
184Brunonianine BDelphinium brunonianum.whole plant[80]
185Brunonianine CDelphinium brunonianum.whole plant[80]
186Delphatisine DDelphinium chrysotrichumaerial part[69]
187Barpubesine AAconitum barbatum var. puberulum Ledebwhole plant[37]
188Barpubesine BAconitum barbatum var. puberulum Ledebwhole plant[37]
189Barpubesine CAconitum barbatum var. puberulum Ledebwhole plant[37]
190Forrestline FDelphinium forrestii var. viridewhole plant[53]
191Brunodelphinine EDelphinium brunonianumaerial parts[64]
Table 11. Names and plant sources of hetisine-type C20-DAs (192217).
Table 11. Names and plant sources of hetisine-type C20-DAs (192217).
No.Compound NameSourcesPlant PartsRef.
192Trichophorine ADelphinium trichophorum Franch.whole herb[83]
193Trichophorine BDelphinium trichophorum Franch.whole herb[83]
194Trichophorine CDelphinium trichophorum Franch.whole herb[83]
195Coreanine AAconitum coreanumroot[87]
196Coreanine BAconitum coreanumroot[87]
197Coreanine CAconitum coreanumroot[87]
198Coreanine DAconitum coreanumroot[87]
199Tanguticuline AAconitum tanguticum (Maxim.) Stapfwhole plant[85]
200Tanguticuline BAconitum tanguticum (Maxim.) Stapfwhole plant[85]
201Tanguticuline CAconitum tanguticum (Maxim.) Stapfwhole plant[85]
202Tanguticuline DAconitum tanguticum (Maxim.) Stapfwhole plant[85]
203Tanguticuline EAconitum tanguticum (Maxim.) Stapfwhole plant[85]
204Anthoroidine GAconitum anthoroideum DC.whole plant[84]
205Anthoroidine HAconitum anthoroideum DC.whole plant[84]
206Anthoroidine IAconitum anthoroideum DC.whole plant[84]
207Grandiflonine CDelphinium grandiflorum L.whole plant[68]
208Grandiflonine DDelphinium grandiflorum L.whole plant[68]
2092-O-cinnamoyl hetisineAconitum heterophyllumroot[88]
210hetisane-15β-O-β-d-glucosideAconitum carmichaelii Debx.root[47]
211HydrodavisineDelphinium peregrinum L. var. eriocarpum Boissaerial part[59]
212PachycentineDelphinium pachycentrum Hemslwhole herb[86]
213Anthoroidine AAconitum anthoroideum DC.whole plant[84]
214Ajacisine HDelphinium ajacisseed[70]
215Tanguticuline FAconitum tanguticum (Maxim.) Stapfwhole plant[85]
216Tanguticuline GAconitum tanguticum (Maxim.) Stapfwhole plant[85]
217Gyalanunine BDelphinium gyalanum C. Marquand & Airy Shawwhole plant[89]
Table 12. Names and plant sources of hetidine-type C20-DAs (218228).
Table 12. Names and plant sources of hetidine-type C20-DAs (218228).
No.Compound NameSourcesPlant PartsRef.
21815-epinaviculine BDelphinium oreophilumaerial part[81]
219Tangutidine DAconitum tanguticum (Maxim.) Stapfwhole plant[92]
220Tangutidine EAconitum tanguticum (Maxim.) Stapfwhole plant[92]
221ParadoxineDelphinium paradoxum Bungeaerial part[91]
222Tangutidine AAconitum tanguticumwhole plant[90]
223Tangutidine BAconitum tanguticumwhole plant[90]
224Tangutidine CAconitum tanguticumwhole plant[90]
225Anthoroidine FAconitum anthoroideum DC.whole plant[84]
226Sczukiniline AAconitum sczukinii Turczroot[65]
227Sczukiniline BAconitum sczukinii Turczroot[65]
228Sczukiniline CAconitum sczukinii Turczroot[65]
Table 13. Names and plant sources of denudatine-type C20-DAs (229241).
Table 13. Names and plant sources of denudatine-type C20-DAs (229241).
No.Compound NameSourcesPlant PartsRef.
229Kirisine FAconitum kirinense Nakairoot[35]
230Kirisine GAconitum kirinense Nakairoot[35]
231Kirisine HAconitum kirinense Nakairoot[35]
232Kirisine IAconitum kirinense Nakairoot[35]
233Kirisine JAconitum kirinense Nakairoot[35]
234Kirisine KAconitum kirinense Nakairoot[35]
235Kirisine LAconitum kirinense Nakairoot[35]
236Barpubesine DAconitum barbatum var. puberulumwhole plant[37]
237Aconicarnine CAconitum carmichaeliilateral root[93]
238Aconicarnine AAconitum carmichaeliilateral root[93]
239Aconicarnine BAconitum carmichaeliilateral root[93]
240Aconicarnine DAconitum carmichaeliilateral root[93]
241Aconicarnine EAconitum carmichaeliilateral root[93]
Table 14. Names and plant sources of napelline-type C20-DAs (242249).
Table 14. Names and plant sources of napelline-type C20-DAs (242249).
No.Compound NameSourcesPlant PartsRef.
242Kirisine MAconitum kirinense Nakairoot[35]
243Kirisine NAconitum kirinense Nakairoot[35]
244Kirisine OAconitum kirinense Nakairoot[35]
24512-epi-aconicarmichinium AAconitum pendulum Buschroot[96]
246Napelline CAconiti kusnezoffii Radixroot[94]
247Napelline DAconiti kusnezoffii Radixroot[94]
248Napelline EAconiti kusnezoffii Radixroot[94]
249Chuanfusulfonine AAconitum carmichaeliilateral root[95]
Table 15. Names and plant sources of vakognavine-type C20-DAs (250255).
Table 15. Names and plant sources of vakognavine-type C20-DAs (250255).
No.Compound nameSourcesPlant partsRef.
250Rockisine AAconitum generaroot[43]
251Umbrodine ADelphinium umbrosum Hand.-Mazz.whole plant[97]
252KingiadineDelphinium kingianum Bruhl. ex Huth.whole plant[97]
253Gyalanunine ADelphinium gyalanum C. Marquand & Airy Shawwhole plant[89]
254Grandiflonine ADelphinium grandiflorum L.whole plant[68]
255Grandiflonine BDelphinium grandiflorum L.whole plant[68]
Table 16. Names and plant sources of rearranged C20-DAs (256266).
Table 16. Names and plant sources of rearranged C20-DAs (256266).
No.Compound NameSourcesPlant PartsRef.
256Anthoroidine CAconitum anthoroideum DC.whole plant[84]
257Anthoroidine DAconitum anthoroideum DC.whole plant[84]
258Anthoroidine EAconitum anthoroideum DC.whole plant[84]
259Aconicatisulfonine AAconitum carmichaeliilateral root[98]
260Aconicatisulfonine BAconitum carmichaeliilateral root[98]
261Aconapelsulfonine AAconitum carmichaeliilateral root[99]
262Aconapelsulfonine BAconitum carmichaeliilateral root[99]
263Aconicarmisulfonine BAconitum carmichaeliilateral root[95]
264Aconicarmisulfonine CAconitum carmichaeliilateral root[95]
265BarpuberudineAconitum barbatum var. puberulum Ledebwhole plant[37]
266Acoapetaludine AAconitum apetalum (Huth) B.Fedtschwhole plant[40]
Table 17. Names and plant sources of Bis-DAs (267280).
Table 17. Names and plant sources of Bis-DAs (267280).
No.Compound NameTypesSourcesPlant PartsRef.
267Weisaconitine Edenudatine-atisineAconitum weixienseroot[100]
268Weisaconitine Fdenudatine-atisineAconitum weixienseroot[100]
269Tangirine Aheteratisine-hetidineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
270Tanguticinine Ahetidine-hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
271Tanguticinine Bhetidine-hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
272Tanguticinine Chetidine-hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
273Tanguticinine Dhetidine-hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
274Tanguticinine Ehetidine-hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
275Tanguticinine Fhetidine-hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
276Tanguticinine Ghetidine-atisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
277Anthoroidine Bhetidine-rearranged hetisineAconitum anthoroideum DC.whole plant[84]
278N-oxide anthoroidine Bhetidine-rearranged hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
2795-deoxyanthoridine Bhetidine-rearranged hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
280N-oxide 5-deoxyanthoroidine Bhetidine-rearranged hetisineAconitum tanguticum (Maxim.) Stapf.whole plant[92]
Table 18. Names and plant sources of other DAs (281289).
Table 18. Names and plant sources of other DAs (281289).
No.Compound NameSourcesRef.
281Ceylonamide GSpongia sp.[101]
282Ceylonamide HSpongia sp.[101]
283Ceylonamide ISpongia sp.[101]
284Koninginol ATrichoderma koningiopsis A729[102]
285Koninginol BTrichoderma koningiopsis A729[102]
286Forsyqinlingine AForsythia suspensa[103]
287Forsyqinlingine BForsythia suspensa[103]
288Aconidenusulfonine AAconitum carmichaelii[104]
28912,16-secoaconidenusulfonine AAconitum carmichaelii[104]
Table 19. Biological activity of terpenoid alkaloids.
Table 19. Biological activity of terpenoid alkaloids.
TeAs TypesActivitiesResearch MethodPossible MechanismRef.
Monoterpenoid Alkaloids
Alstochonine A (1)VasorelaxantIn vitroVasorelaxant activity against phenylephrine-induced contraction of rat mesenteric arteries with rates of 73.6 ± 2.8% and 95.4 ± 3.7% (IC50 = 93.30 ± 10.81, 60.56 ± 3.66 μM)[16]
Alstochonine B (2)In vitro[16]
Delavatine C (9)Anti-inflammatoryIn vitroInhibition of NO production in LPS-stimulated BV2 cells (IC50 = 25.62, 17.29 μM)[18]
Delavatine E (11)In vitro[18]
Forsyqinlingine C (14)Anti-inflammatory and antiviralIn vitroAnti-inflammatory activities by inhibiting the release of β-glucuronidase in PMNs with inhibition rates of 45.2% and 40.1%, and antiviral activities against H1N1 virus (IC50 = 11.9, 15.1 μM) and RSV (EC50 = 13.5, 14.0 μM)[21]
Forsyqinlingine D (15)In vitro[21]
Incarvine G (12)AntitumorIn vitroCytotoxicity (IC50 = 60.29 μM) against MDA-MB-231 cells and inhibiting actin cytoskeleton formation[19]
(±)-Caryopterisine A (19)In vitroReduction of Kyn biosynthesis in HeLa cells by inhibiting IDO at 10 μM with inhibition ratios of 25.7% and 29.8%, respectively[22]
(±)-Caryopterisine B (20)In vitro[22]
Caryopterisine C (16)AntifibroticIn vitroInhibition of collagen accumulation (IC50 = 14.26 ± 1.46 μM) in NIH3T3 cells and phosphorylation of ERK1/2, P38, and SMAD2/3[23]
Lomatogonin C (26)ImmunosuppressiveIn vitroInhibition of T cell proliferation (21.62 ± 3.06%) and IFN–γ secretion (37.59 ± 5.41%) at 20 μM[26]
Sesquiterpene alkaloids
Commipholactam A (27)AntitumorIn vitroCytotoxicity against HepG2 (IC50 = 21.73 ± 2.86 μM) and A549 (IC50 = 128.50 ± 17.06 μM) cells[28]
Diterpenoid alkaloids
Geordine (103)Anti-inflammatoryIn vitroInhibition of NO production (29.75%) in LPS-induced RAW264.7 cells at 50 μM[61]
Stylosine A (157)In vitroInhibition of production of IL-1β, COX-2, and TNF-α in LPS-induced RAW264.7 cells in a dose-dependent manner[74]
Ajacisine F (121)In vitroInhibition of NO production in LPS-induced BV-2 cells with inhibition rates of 80% at 50 μM[70]
Ajacisine G (123)In vitro[70]
Ajacisine H (214)In vitro[70]
Kamaonensine B (148)In vitroInhibition of NO production in LPS-stimulated RAW264.7 cells (IC50 = 2.7 ± 0.5 and 0.9 ± 0.2 μM) and might be mediated by the regulation of some related proteins in the MAPK signaling pathways[72]
Kamaonensine F (153)In vitro[72]
Aconicumine A (170)In vitroInhibition of LPS-activated NO production in RAW264.7 cells (IC50 = 19.7 ± 1.1 μM)[77]
Forrestline F (190)In vitroInhibition of NO production in RAW264.7 cells (IC50 = 9.57 ± 1.34 μM) through inhibiting NF-κB, MAPK, and Nrf2 signaling pathways[53]
Anthoroidine B (277)In vitroInhibition of the production of NO and TNF-α in LPS-stimulated RAW264.7 cells, with IC50 values of 357.68 and 67.56 μM[84]
Forsyqinlingine A (286)Anti-inflammatory and antiviralIn vitroAnti-inflammatory activities by inhibiting the release of β-glucuronidase in PMNs with inhibition rates of 56.7 and 58.6%, and antiviral activities against H1N1 virus (IC50 = 6.9 and 7.7 μM) and RSV (EC50 = 5.0 and 4.8 μM)[103]
Forsyqinlingine B (287)In vitro[103]
Pseudostapine C (89)AnalgesicIn vivoReduction of acetic acid-induced abdominal contractions in mice in a dose-related manner, with the ID50 values of 66.1, 60.3, 48.0, and 55.0 μmol/kg, respectively[51]
Austroyunnanine B (91)In vivo[45]
Episcopine A (180)In vivo[52]
Episcopaline B (182)In vivo[50]
Nagarumine C (176)In vivoInhibition of acetic acid-induced writhing in mice (ED50 = 76.0 μmol/kg)[46]
Acosinomonine B (178)In vitroStrong inhibitory effect on the activation of the TRPV1 channel in HEK-293 cells mediated by capsaicin, with an inhibition rate of 31.78% at 10 μM[79]
Aconicatisulfonine A (259)In vivoReduction in acetic acid-induced writhing in mice by 43.2%, 64.7%, 63.6%, and 19.3% at 0.3 mg/kg, respectively[98]
Aconicatisulfonine B (260)In vivo[98]
Aconapelsulfonine A (261)In vivo[99]
Aconapelsulfonine B (262)In vivo[99]
Aconicarmisulfonine B (263)In vivoReduction in acetic acid-induced writhing in mice by 31.26%, 26.84%, and 43.8% at 1.0 mg/kg (i.p.)[95]
Aconicarmisulfonine C (264)In vivo[95]
Aconicarnine E (241)In vivo[93]
Aconidenusulfonine A (288)In vivoReduction in acetic acid-induced writhing in mice by 26.35% at 2.0 mg/kg (i.p.)[104]
Lipojesaconitine (98)AntitumorIn vitroCytotoxicity against A549, MDA-MB-231, MCF-7, and KB with IC50 values ranging from 6.0 to 7.3 μM[57]
8-O-ethyl-benzoyldeoxyaconine (107)In vitroAnticancer activity against A549 (IC50 = 12.58 ± 1.82 μM) and H460 (IC50 = 12.76 ± 2.10 μM) cells[47]
Brunonianine B (182)In vitroCytotoxicity on Caco-2 (IC50 = 3.14 ± 0.37 μM) and Skov-3 (IC50 = 2.20 ± 0.21 μM) cells and activation of the Bax/Bcl-2/caspase-3 signaling pathway[80]
Brunonianine C (183)In vitroCytotoxicity on Caco-2 (IC50 = 2.41 ± 0.35 μM) and Skov-3 (IC50 = 6.88 ± 0.81 μM) cells[80]
Ceylonamide G (281)In vitroCytotoxic to DU145 cells (IC50 = 6.9 μM, MEC = 10 μM)[101]
Smirnotine A (94) CardioprotectiveIn vivoSome preventive effects on aconitine-induced arrhythmia in mice[58]
Gyalanunine A (253)In vitroSignificant cardiotonic activity after perfusion in frog hearts and could be related to the β receptor[89]
Tanguticuline A (199)AntiviralIn vitroInhibition of the cytopathic effect against H1N1 with IC50 values of 2.9 and 2.4 μg/mL, respectively[85]
Tanguticuline E (203)In vitro[85]
Acoapetaludine D (45)AntibacterialIn vitroAnti-Helicobacter pylori (MIC = 100 μg/mL)[40]
Acoapetaludine E (46)In vitroAnti-Helicobacter pylori (MIC = 50 μg/mL)[40]
Stylosine A (157)In vitroAnti-Staphylococcus aureus (MIC = 2.00 μg/mL)[74]
Stylosine B (158)In vitroAnti-Staphylococcus aureus (MIC = 32.00 μg/mL)[74]
Koninginol A (284)In vitroAnti-Bacillus subtilis (MIC = 10.00 μg/mL)[102]
Koninginol B (285)In vitroAnti-Bacillus subtilis (MIC = 2.00 μg/mL)[102]
2-O-cinnamoyl hetisine (209)AntiplasmodialIn vitroAnti-Plasmodium falciparum strains Pf INDO (IC50 = 1.92 μM) and the Pf 3D7 (IC50 = 10.8 μM)[88]
Apetalrine B (82)Neuroprotective In vitroNeuroprotective activity (77.4%) on H2O2-induced SH-SY5Y cell[55]
Uncinatine-A (120)AChEI activityIn vitroAcetyl-cholinesterase inhibitory activity (IC50 = 207.73 ± 0.3 μM[66]
Anthoroidine G (204)In vitroAcetyl-cholinesterase inhibitory activity (IC50 = 6.3 ± 1.6 μM)[84]
Anthoroidine I (206)In vitroAcetyl-cholinesterase inhibitory activity (IC50 = 9.3 ± 3 μM)[84]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, X.; Xin, J.; Sun, L.; Sun, Y.; Xu, Y.; Zhao, F.; Niu, C.; Liu, S. Exploring the Biomedical Potential of Terpenoid Alkaloids: Sources, Structures, and Activities. Molecules 2024, 29, 1968. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules29091968

AMA Style

Wang X, Xin J, Sun L, Sun Y, Xu Y, Zhao F, Niu C, Liu S. Exploring the Biomedical Potential of Terpenoid Alkaloids: Sources, Structures, and Activities. Molecules. 2024; 29(9):1968. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules29091968

Chicago/Turabian Style

Wang, Xuyan, Jianzeng Xin, Lili Sun, Yupei Sun, Yaxi Xu, Feng Zhao, Changshan Niu, and Sheng Liu. 2024. "Exploring the Biomedical Potential of Terpenoid Alkaloids: Sources, Structures, and Activities" Molecules 29, no. 9: 1968. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules29091968

Article Metrics

Back to TopTop