Next Article in Journal / Special Issue
Recent Advances in Grayanane Diterpenes: Isolation, Structural Diversity, and Bioactivities from Ericaceae Family (2018–2024)
Previous Article in Journal
Synthesis of 4′-Thionucleoside Analogues Bearing a C2′ Stereogenic All-Carbon Quaternary Center
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Progress on Sesquiterpene Compounds from Artabotrys Plants of Annonaceae

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.
Molecules 2024, 29(7), 1648; https://doi.org/10.3390/molecules29071648
Submission received: 29 February 2024 / Revised: 27 March 2024 / Accepted: 4 April 2024 / Published: 6 April 2024
(This article belongs to the Special Issue Plant Sourced Compounds: Extraction, Identification and Bioactivity)

Abstract

:
Artabotrys, a pivotal genus within the Annonaceae family, is renowned for its extensive biological significance and medicinal potential. The genus’s sesquiterpene compounds have attracted considerable interest from the scientific community due to their structural complexity and diverse biological activities. These compounds exhibit a range of biological activities, including antimalarial, antibacterial, anti-inflammatory analgesic, and anti-tumor properties, positioning them as promising candidates for medical applications. This review aims to summarize the current knowledge on the variety, species, and structural characteristics of sesquiterpene compounds isolated from Artabotrys plants. Furthermore, it delves into their pharmacological activities and underlying mechanisms, offering a comprehensive foundation for future research.

1. Introduction

Annonaceae, a prominent family within the tropical flora, is classified under the Ranunculaceae. It contains approximately 130 genera and over 2100 species, featuring a rich diversity of tropical trees, shrubs, and climbing plants [1]. Many species within Annonaceae family have been widely used in ethnobotany to treat a myriad of health conditions [2]. For instance, Polyalthia, one of the largest and most famous genera within Annonaceae, has been widely used in the treatment of rheumatic fever, peptic ulcer, and systemic pain [3]. The chemical diversity present in Annonaceae species is vast, yielding a plethora of natural compounds such as alkenes [4], terpenoids [5], alkaloids [6,7,8,9,10], and phenols [11]. These compounds demonstrate a broad spectrum of pharmacological activities, including anti-mosquito [12], anti-cancer [13,14,15], antibacterial [16], anti-protozoal [17,18], and antifungal [19]. Among them, Annonaceous acetogenins stand out for their potent anti-tumor potential, making them one of the most promising natural product discoveries [20].
Artabotrys, belonging to the Annonaceae family, comprises about 110 species of plants around the world, predominantly distributed in tropical and subtropical regions such as Southeast Asia, Indonesia, and Malaysia. The plants of this genus are climbing shrubs. The leaves of these plants are usually compound, the flowers are small and clustered on the raceme, and the fruits are drupe-shaped. There are many traditional uses of this genus, such as the treatment of cholera, malaria, and other diseases [21]. The plants of this genus have a wide range of biological significance and medicinal value. An examination of data from the Plants of the World Online database facilitated a detailed summary of Artabotrys species and their distribution (Table 1).
The genus Artabotrys, within the Annonaceae family, is distinguished by its wealth of chemical components [22]. To date, research has identified a diverse array of compounds from these plants, including alkaloids [23,24], volatile oils [25,26], cyclohexenes [27,28], phenylpropanoids [29], flavonoids [30], quinones [31], and sesquiterpenes [32]. Among these, sesquiterpenes stand out as one of the principal active components, heralded for their significant medical value and importance in research. So far, there have been many research articles on the plants of Artabotrys; however, the majority have focused on individual compounds or relatively extensive research overviews. Comprehensive reviews specifically addressing the sesquiterpene compounds derived from the plants of the genus are notably scarce. Therefore, this paper aims to fill this gap by reviewing the current research progress of sesquiterpene compounds derived from the plants of Artabotrys in Annonaceae. It meticulously summarizes the variety, species, and structural characteristics of sesquiterpene compounds identified within these plants and explores their pharmacological activities and underlying mechanisms, offering a comprehensive foundation for future research.

2. Chemical Constitution

Sesquiterpenes, a diverse class of natural organic compounds, are characterized by a basic carbon skeleton comprising 15 carbon atoms arranged in three isoprene units. Based on the number of carbon rings in the structure, sesquiterpenes can be divided into five structural types: acyclic sesquiterpenes [33], monocyclic sesquiterpenes [34], bicyclic sesquiterpenes [35,36], tricyclic sesquiterpenes [37], and tetracyclic sesquiterpenes [38]. Acyclic sesquiterpenes encompass linear sesquiterpenes [39] and unsaturated acyclic sesquiterpenes [40] whereas monocyclic sesquiterpenes include germacrane [41], cyclofarnesane [42], bisabolane [43], and elemane [44]. Bicyclic sesquiterpenes feature structures like eudesmane [45], isodaucane [46], guaiane [47], acorane [48], and eremophilane [49,50]. Tricyclic sesquiterpenes include aristolane [51], and aromadendrane [52]. Tetracyclic sesquiterpenes include camphane, labdane, and ginkgolide [53,54].
Sesquiterpenes represent a distinguished class of natural organic compounds, notable for their widespread natural sources. These compounds are predominantly derived from a range of plants [55,56], especially those known for their aromatic properties, as well as from fungi [57,58,59], and marine organisms [60,61]. Sesquiterpenes have a variety of biological activities, encompassing antimalarial [62], antioxidant [63], anti-inflammatory [64,65], antibacterial [66,67], and anti-tumor effects [68,69]. Therefore, sesquiterpenes have displayed significant therapeutic potential in the pharmaceutical sector, while their unique properties also make them invaluable to the perfume industry.
Extensive research into the sesquiterpenes extracted from Artabotrys plants reveals a remarkable diversity within this genus. To date, investigations have identified over 80 distinct sesquiterpene types isolated from Artabotrys, underscoring the genus’s rich contribution to the pool of naturally occurring sesquiterpenes. A detailed breakdown of these sesquiterpenes reveals a wide array of structural types, including 19 bisabolane-type, 15 eudesmane-type, 8 norbisabolane-type, 6 guaiane-type, 4 aromadendrane-type, aristolane-type, and cadinane-type, 3 eremophilane-type, 2 isodaucane-type and acorane-type, 1 germacrane-type, alongside a multitude of other sesquiterpene variants.
These findings further attest to the extraordinary potential of the plant as a ‘natural drug bank’, such as for the development of innovative anti-tumor and anti-inflammatory drugs. Each sesquiterpene identified offers unique insights into potential pharmacological applications and holds the promise of playing a pivotal role in devising novel therapeutic strategies.

2.1. Bisabolane-Type Sesquiterpenes

Bisabolane-type sesquiterpenes, a subclass of monocyclic sesquiterpenes, are characterized by their six-membered carbon rings and side chains. These compounds boast a plethora of natural sources, including marine invertebrates, terrestrial plants, and microorganisms. Notably, bisabolane-type sesquiterpenes exhibit a wide range of biological activities, such as anti-inflammatory and antibacterial [70,71]. To date, more than 350 kinds of bisabolane-type sesquiterpenes have been isolated from various plant families, including Compositae and Zingiberaceae [72]. In the context of the Artabotrys genus, several bisabolane-type sesquiterpenes have also been successfully extracted (Table 2).
Among the isolated compounds, compounds 1 and 2 were extracted from the roots of Artabotrys uncinatus in 1979; their structures were elucidated by spectroscopic methods [73,74]. Compounds 3 and 4 were also derived from A. uncinatus [75]. Subsequently, researchers isolated 13 bisabolane-type sesquiterpenes (517 in Table 2) from the roots of A. hexapetalus in 2017 [76]. Moreover, 25 monomeric compounds, including two bisabolane-type sesquiterpenes chlospicate E (18) and arbisabol-9-en-7,11-diol (19) [77], were isolated from Artabotrys pilosus by a combination of chromatographic separation methods and spectral identification techniques. The structural details of the related compounds are shown in Figure 1.

2.2. Norbisabolane-Type Sesquiterpenes

Norbisabolane-type sesquiterpenes, another subset of monocyclic sesquiterpenes, known for their spiroketal structures, have primarily been isolated from Phyllanthus spp. within the Euphorbiaceae [78]. From the extracts of Artabotrys plants, several norbisabolane-type sesquiterpenes (Table 3) were successfully purified by a series of chromatographic techniques, and the structures were elucidated via comprehensive analysis of nuclear magnetic resonance (NMR), mass spectrometry (MS) and other technical means. Among them, compound 20 was isolated from A. hexapetalus [76], while compounds 2127 were isolated from the branches and leaves of A. hongkongensis in 2017 [79]. The detailed structures of the compounds are shown in Figure 2.

2.3. Eudesmane-Type Sesquiterpenes

Eudesmane-type sesquiterpenes, classified as bicyclic sesquiterpenes, are notable for their widespread distribution in nature. Eudesmane-type sesquiterpenes are characterized by a core structure comprising two six-membered rings and four substituents with a total of 15 carbon atoms, leading to a considerable structural diversity primarily attributed to variations in the substituents’ positioning and the double bonds within the rings. Studies have shown that these compounds displayed anti-inflammatory [80], anti-fungal [81], anti-cancer [82], anti-diabetic nephropathy [83], and the ability to inhibit the proliferation of leukemia cell lines [84].
A significant number of eudesmane-type sesquiterpenes have been isolated from Artabotrys (Table 4), with compounds 2842 representing this variety. Among them, compounds 2834, a series of seven eudesmane-type sesquiterpenes, were isolated from Artabotrys hongkongensis Hance in 2020 [85]. The compound 7-trinoreudesma-4(15),8-dien-1β-ol-7-one (45) was isolated from the ethyl acetate extract of the 90% ethanol extract of the branches and leaves of A. pilosus by various modern chromatographic separation techniques. Its identification as colorless oil soluble in chloroform was identified by structural identification, affirming its classification as an eudesmane-type sesquiterpene [77]. Additionally, the other eight eudesmane-type sesquiterpenes (3542) were isolated from Artabotrys hainanensis [86], A. hongkongensis [79], and A. pilosus [77] by various separation techniques. The distinctive structures of eudesmane-type sesquiterpenes from the Artabotrys genus plants are depicted in Figure 3.

2.4. Guaiane-Type Sesquiterpenes

Guaiane-type sesquiterpenes, a subclass of bicyclic sesquiterpenes, are distinguished by their unique structural framework, which features a seven-membered ring fused with a five-membered lactone ring, augmented by two methyl groups and one isopropyl group. These compounds are prevalent across more than 30 families of plants, demonstrating a broad spectrum of biological activities, including anti-tumor, anti-inflammatory, antibacterial, and antioxidant [87,88]. The genus Artabotrys plants, known for its rich chemical diversity, also harbors guaiane-type sesquiterpenes. Compounds 4348 represent guaiane-type sesquiterpenes isolated from various Artabotrys species (Table 5). Guaiane pogostol O-methyl ether (46) from Artabotrys stenopetalus in 1997 marked the beginning of the identification of such compounds within the genus [89]. Compounds 43 and 44 are two sesquiterpenes isolated from the 90% ethanol extract of the branches and leaves of A. hainanensis, both identified as guaiane-type sesquiterpenes [86]. Compound 45, a colorless oily substance isolated from A. pilosus [77], was confirmed as guaianediol through NMR data analysis and comparison with existing literature [90]. Additionally, alismol (47) and alismoxide (48) were derived from the stem [91] and flower of A. hainanensis [86], respectively, with the latter previously identified in Alisma orientalis [92]. The structures of these guaiane-type sesquiterpenes are depicted in Figure 4.

2.5. Eremophilane-Type Sesquiterpenes

Eremophilane-type sesquiterpenes, derived from the biosynthetic precursor farnesyl diphosphate (FPP), represent a distinct group within the bicyclic sesquiterpene compound family. These compounds are characterized by their unique irregular bicyclic structures, with structural variations primarily arising from various oxidations on the bicyclic skeleton and the isopropyl side chain [93]. Related studies have shown that these compounds displayed anti-inflammatory effects and can inhibit the NO produced by lipopolysaccharide (LPS)-induced RAW 264.7 macrophages [94]. In studying the chemical constituents of the Artabotrys genus, researchers have successfully isolated several eremophilane-type sesquiterpenes (Table 6). Among them, compounds 49 and 50 are two eremophilane-type sesquiterpenes obtained from the branches and leaves of A. hongkongensis in the same research process [79], while compound 51 was obtained from the branches and leaves of A. hainanensis in another study one year later [86]. The chemical structures of these three eremophilane-type sesquiterpenes with serial numbers 4951 are shown in Figure 5.

2.6. Isodaucane-Type Sesquiterpenes

Isodaucane-type sesquiterpenes, which belong to bicyclic sesquiterpenes, are distinguished by their distinctive structural configuration, featuring a five-membered ring coupled with a seven-membered ring. Despite their relatively scarce occurrence in nature compared with other common types of sesquiterpenes, dedicated research efforts have led to the successful isolation of two isodaucane-type sesquiterpenes from the Artabotrys genus (Table 7). Compounds 52 and 53 were isolated from the branches and leaves of A. hongkongensis and the stem bark of A. stenopetalus, respectively [79,89]. The structures of the two compounds are shown in Figure 6.

2.7. Acorane-Type Sesquiterpenes

Acorane-type sesquiterpenes are distinguished by their spiro [4.5] decane skeleton, featuring an isopropyl unit at C-1 and a dimethyl substitution at C-4 and C-8 [95]. This unique natural product category falls within the bicyclic sesquiterpene compound, known for its wide range of pharmacological activities, such as antiviral activity [96] and anti-inflammatory activity [97,98]. Despite their notable bioactivity, acorane-type sesquiterpenes are exceedingly rare in both plants and microorganisms. In a significant discovery, two acorane-type sesquiterpenes were successfully isolated from the genus of Artabotrys (Table 8). Compounds 54 and 55 were isolated from the roots of A. hexapetalus in 2017 alongside 13 bisabolane-type sesquiterpenes (517 in Table 2) [76]. The structures of the two compounds are shown in Figure 7.

2.8. Cadinane-Type Sesquiterpenes

Cadinane-type sesquiterpenes, a class of bicyclic sesquiterpenes, are synthesized through the catalytic action of sesquiterpene synthase (STS) on FPP [99]. These compounds have complex stereochemistry and a wide range of pharmacological activities, such as hypoglycemic [100], antifungal [101], and anti-inflammatory [102]. To date, a considerable diversity of cadinane-type sesquiterpenes with diverse structures and biological activities have been isolated and identified from a variety of plants and microorganisms. Furthermore, with the continuous advancement of modern biotechnology, the biosynthetic pathways of representative cadinene-type sesquiterpenes have been substantially elucidated [103]. The following compounds are cadinene-type sesquiterpenes obtained from the genus of Artabotrys (Table 9). Notably, 10β, 15-hydroxy-α-cadinol (56) was isolated from both A. pilosus [77] and A. hainanensis [86]. Additionally, amorph-4-en-10α-ol (57) was isolated from the branches and leaves of A. hainanensis [86]. Compounds 58 and 59, further enriching the variety of cadinene-type sesquiterpenes, were derived from the branches and leaves of A. pilosus [77]. The detailed structures of these compounds are depicted in Figure 8.

2.9. Aristolane-Type Sesquiterpenes

Aristolane-type sesquiterpenes are naturally occurring sesquiterpenes, primarily obtained from Nardostachys, Axinyssa, and Russula [104]. Aristolane-type sesquiterpenes usually contain a gem-dimethyl cyclopropane structure [105], which belongs to the tricyclic sesquiterpenes. These compounds play a pivotal role in regulating serotonin transporter (SERT) to enhance or inhibit SERT [106], which offers therapeutic potential for the treatment of neuropsychiatric and digestive diseases. Advances in research and technology have enabled the isolation of several aristolane-type sesquiterpenes from Artabotrys plants (Table 10). 10-hydroxyaristolan-9-one (60), initially isolated from the stems of A. uncinatus in 2007, has also been found in the branches and leaves of A. hongkongensis in another study a few years later [79,107], alongside compounds 6163 [79]. The structures of aristolane-type sesquiterpenes involved are shown in Figure 9.

2.10. Aromadendrane-Type Sesquiterpenes

Aromadendrane-type sesquiterpenes, akin to the aristolane-type sesquiterpenes mentioned earlier, belong to the tricyclic sesquiterpenes family, noted for their anti-inflammatory [108]. Studies have found that certain aromadendrane-type sesquiterpenes compounds can interact with benzoquinone to form heterodimers, offering cytoprotective effects on glutamate-induced neurological deficits [109]. The following three compounds (6465) are classified as aromadendrane-type sesquiterpenes obtained from Artabotrys (Table 11). Compound 64 was obtained from branches and leaves of A. hainanensis [86]. The remaining compound (-)-ent-4β-hydroxy-10α-methoxyaromadendrane (65) was obtained from the stem of A. uncinatus by numerous efforts of researchers in 2007 [107]. Compounds 66 and 67 are two sesquiterpenes obtained from the flowers of A. hexapetalus [110]. Figure 10 shows the detailed structures of the five aromadendrane-type sesquiterpenes.

2.11. Other Types of Sesquiterpenes

Beyond the previously mentioned sesquiterpenes, many other types of sesquiterpenes have also been obtained from the plants of Artabotrys, as detailed in Table 12.
Notably, β-caryophyllene oxide (68), caryophyllene-type sesquiterpenes with a unique polycyclic structure, were isolated from the stem bark of A. stenopetalus [89]. Compounds 74 and 76 in Table 12 also belong to this class of sesquiterpenes. Compounds 69 and 70, derived by reducing some carbon atoms in cadinane-type sesquiterpenes, represent a class of bicyclic sesquiterpene. 4-hydroxy-4,7-dimethyl-1-tetralone (69), reduced by 3 carbons, and oxyphyllone D (70), reduced by 1 carbon, have been isolated from the branches and leaves of A. pilosus [77] and A. hainanensis [86], respectively. Additionally, 1β-hydroxy-4(15),5E,10(14)-germacratriene (71), a germacrane-type sesquiterpene, was isolated from the branches and leaves of A. hainanensis [86] and belongs to monocyclic sesquiterpenes. artahongkongol A (72), a unique trinoreudesmane sesquiterpene derived from the corresponding eudesmane-type sesquiterpenes by removing a propyl group, was obtained from the stems and leaves of A. hongkongensis [85]. (4R,5R,7R)-1(10)-spirovetiven-11-ol-2-one (81), a rare natural spirovetivane-type sesquiterpene, was first isolated from the flower of A. hainanensis [86]. Compounds 82, 83, and 84 are three bisabolene-type sesquiterpenes isolated from the roots of A. hexapetalus, with compounds 83 and 84 identified as a pair of enantiomers [32]. The remaining compounds listed in Table 12, not described in detail here, represent unique sesquiterpenes with special structural types rare in nature isolated from Artabotrys plants. The specific structures of the related compounds are illustrated as follows (Figure 11).

3. Pharmacological Activities

Sesquiterpenes, with their distinct carbon skeletons and roles in diverse biochemical processes, play a pivotal role in drug discovery and development. Their unique structures enable a wide array of biological and pharmacological actions, making them invaluable in modern medicinal research. In particular, sesquiterpenes from the plants of the genus Artabotrys have shown significant activity across numerous pharmacological studies. The following are some key pharmacological activities attributed to sesquiterpenes isolated from Artabotrys plants.

3.1. Antimalarial Activity

Malaria is one of the oldest diseases in humans. It is a disease caused by parasites [111] mainly transmitted to humans through mosquito bites [112]. Malaria is an infectious disease caused by malaria parasites [113,114,115]. Predominantly prevalent in tropical and subtropical regions, especially in Africa, South Asia, Southeast Asia, and Central America [116], malaria accounts for more than 200 million cases worldwide each year [117]. The development of effective antimalarial drugs can reduce the spread and infection of malaria and accelerate the early recovery of patients [118]. Therefore, it is of great significance to find more effective antimalarial drugs. In the process of studying antimalarial drugs, researchers have found that some sesquiterpenes and some other natural components derived from Artabotrys plants have shown antimalarial activity. Notably, yingzhaosu A (1) is the first antimalarial drug with a clear structure containing an endoperoxide structure in history [119]. This discovery has spurred further research and the synthesis of new antimalarial drugs, although the exact mechanism of yingzhaosu A’s antimalarial action remains partially understood. Current research suggests that yingzhaosu A’s mechanism of action may involve two primary processes. Firstly, in the presence of oxygen and iron (II), yingzhaosu A will undergo a degradation reaction due to the induction of iron (II), forming unsaturated ketones and cyclohexyl radicals, respectively. The active substances produced in this process may be the reason for its antimalarial effect [120].
Secondly, a recent study found that when yingzhaosu A plays a role in the body, it is attacked by heme, which destroys its peroxide structure, produces tertiary oxygen-centered radicals, and rearranges to remove the side chain. Therefore, the yingzhaosu A is split into two parts. Heme is an important marker of malaria parasites. Based on the above findings, a heme-activatable probe has been successfully developed, which will play an important role in the field of antimalarial [121].
Beyond yingzhaosu A (1), related compounds such as yingzhaosu B (2), yingzhaosu C (3), and yingzhaosu C (4) have also demonstrated antimalarial effect, expanding the library of potential antimalarial agents derived from natural sources [122,123,124].

3.2. Antibacterial and Antifungal Activity

Bacterial infections significantly impact global health, causing widespread morbidity and mortality, and placing a significant burden on health care systems [125,126]. At present, many bacteria are resistant to antibiotics, which has become an extremely important public health problem [127,128,129]. However, due to the increase in global antimicrobial resistance, the efficacy of some treatments for bacterial infections is reduced or even ineffective. Therefore, it is particularly important to find new therapeutic drugs and design new treatment strategies in the field of antibacterial [130,131].
Among the promising candidates, sesquiterpenes derived from Artabotrys plants have demonstrated antibacterial effects through different mechanisms, showing potential against a variety of bacterial and fungal pathogens. Notably, isodaucane-type sesquiterpene artabotrol (53), isolated from the stem bark of A. stenopetalus, a plant belonging to the genus Artabotrys, exhibits a specific inhibitory effect on Cryptococcus neoformans [132].
Furthermore, globulol (66), isolated from the flowers of A. hexapetalus and the fruit of Eucalyptus globulus Labill, has been shown to inhibit several fungi, including Alternaria solani, Fusarium oxysporum, Fusarium graminearum, Rhizoctonia solani, and Venturia pirina, with a half maximal inhibitory concentration (IC50) values of 47.1 μM, 114.3 μM, 53.4 μM, 56.9 μM, 32.1 μM, and 21.8 μM, respectively. In addition, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay results showed that globulol (66) also had inhibitory effects on Xanthomonas vesicatoria and Bacillus subtilis, with IC50 values of 158.0 μM and 737.2 μM, respectively [133]. Another compound, dihydroactinidiolide (78) showed antibacterial activity against Bacillus cereus and Vibrio parahaemolyticus in related studies [134].

3.3. Antitumor Activity

Some sesquiterpenoids have been shown to have antitumor activity [135]. A notable example is the sesquiterpene (−)-8R-Artaboterpenoids B (83) isolated from the root of A. hexapetalus, which exhibited cytotoxicity against five tumor cells including HCT-116, Hep G2, A2780, NCI-H1650, and BGC-823 with IC50 values of 1.38, 3.30, 6.51, 8.19 and 2.14 μM, indicating its potential as an anticancer agent [32]. Similarly, another study identified seven sesquiterpenoids, chlospicate E (18), 1β, 6α-dihydroxy-4α (15)-epoxyeudesmane (41), guaianediol (45), 10β,15-hydroxy-α-cadinol (56), 15-hydroxy-t-muurolol (58), 10α-hydroxycadin-4-en-15-al (59), and 10β-hydroxyisodauc-6-en-14-al (79) from A. pilosus showed significant inhibitory activity against HL-60, SMMC-7721, A-549, MCF-7, and SW480 human tumor cells. These compounds have the potential to develop new anti-tumor drugs as lead compounds. According to the relevant experimental results, the IC50 values of chlospicate E (18) were 14.25, 21.32, 25.34, 16.23, 10.21 μM, the IC50 values of 1β, 6α-dihydroxy-4α (15)-epoxyeudesmane (41) were 18.25, 9.65, 8.27, 4.63, 8.64 μM, the IC50 values of guaianediol (45) were 10.23, 8.64, 9.23, 10.42, 15.22 μM, the IC50 values of 10β,15-hydroxy-α-cadinol (56) were 10.11, 5.14, 4.38, 6.32, 3.28 μM, the IC50 values of 15-hydroxy-t-muurolol (58) were 2.36, 4.02, 7.32, 6.41, 5.23 μM, the IC50 values of 10α-hydroxycadin-4-en-15-al (59) were 5.23, 6.87, 4.96, 5.86, 4.20 μM and the IC50 values of 10β-hydroxyisodauc-6-en-14-al (79) were 15.23, 6.26, 10.23, 9.32, 5.49 μM, respectively. Among these compounds, 10β, 15-hydroxy-α-cadinol (56) had the strongest inhibitory effect on SW480 cells with an IC50 value of 3.28 μM, and 1β, 6α-dihydroxy-4α (15)-epoxyeudesmane (41) had the strongest inhibitory effect on MCF-7 cells with an IC50 value of 4.63 μM [77].
Further research in 2018 unveiled seven eudesmane-type sesquiterpenes (2834) and one trinoreudesmane-type sesquiterpene (72) from the genus Artabotrys, showing cytotoxicity and inhibitory effects on five human tumor cell lines (IC50 values of 0.57 to 15.68 μM), with some compounds outperforming the antitumor drug doxorubicin [85]. In addition, yingzhaosu C (3) also demonstrated tumor inhibitory effects on HCT-116, HepG 2, and A 2780 cell lines, with IC50 values of 3.24, 3.23, and 3.14 μM, respectively [76]. In related studies, compound 24 was found to have a general inhibitory effect on A-549, MCF-7, HT-29, A-498, Pc-3, and PACA-2 human tumor cells, but its effect was not significant, and its IC50 values were 11.3, 12.3, 14.5, 16.6, 24.3, and 19.6 μM, respectively [136].
Dihydroactinidiolide (78) also has significant anti-tumor activity against four human tumor cell lines, epithelial cell carcinoma (Hela), human prostate cancer (PC-3), breast cancer (MCF-7), and hepatocellular carcinoma (HePG-2) [137]. Additionally, β-caryophyllene oxide (68) has been studied for its antitumor mechanism. It is well known that phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR)/ribosomal protein S6 kinase 1 (S6K1) and mitogen-activated protein kinase (MAPK) signaling cascades play an important role in many physiological processes of tumor cells, including cell proliferation, survival, angiogenesis, and metastasis of tumor cells. Through Western blot analysis, MTT assay, and other research methods, it was found that β-caryophyllene oxide (68) not only inhibited the constitutive activation of PI3K/AKT/mTOR/S6K1 signaling cascade in human prostate cancer PC-3 and breast cancer MCF-7 cells; it also causes the activation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 MAPK in tumor cells, and down-regulates various gene products related to cell proliferation, anti-apoptosis, and metastasis. In addition, in different tumor cells, β-caryophyllene oxide (68) can simultaneously target PI3K/AKT/mTOR/S6K1 and MAPK signaling pathways, inhibit the proliferation of related tumor cells and induce the apoptosis of tumor cells by activating caspase-3 and releasing cytochrome c. These results suggest that β-caryophyllene oxide (68) is a potential candidate drug for the prevention and treatment of cancer [138,139,140].

3.4. Anti-Inflammatory and Analgesic Activity

Inflammation is a complex immune response, which is the body’s defense mechanism against injury and infection [141,142]. The five main symptoms of inflammation are pain, fever, redness, swelling, and loss of function. Inflammation can be divided into acute and chronic inflammation [143]. If inflammation is left unchecked, it may lead to autoimmune diseases, neurodegenerative diseases, etc. [144]. At present, there are many effective anti-inflammatory drugs, which are also the most common clinical treatment drugs. However, the commonly used anti-inflammatory drugs will have some side effects during the treatment [145,146]. Therefore, in addition to using traditional non-steroidal anti-inflammatory drugs to treat inflammation, some compounds isolated from natural sources are also considered new options for treating inflammatory diseases [147,148,149,150].
Among these, some natural sesquiterpenes obtained from the Artabotrys genus have demonstrated promising anti-inflammatory and analgesic activities. For instance, caryolane-1,9β-diol (76), which was found in A.uncinatus in 2007, exhibits significant anti-inflammatory activity in a dose-dependent manner [107,151]. Similarly, spathulenol (64), isolated from the twigs and leaves of A. hainanensis and previously found in other species such as Psidium guineense Sw. has shown notable inhibitory effect on the related pathological symptoms of the Cg-induced paw edema and pleurisy model in mice established in the experiment [152].
Additionally, alismol (47) also has anti-inflammatory effects, reducing the levels of NO and prostaglandin E2 in cells and inhibiting the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) stimulated by lipopolysaccharide in the body. It also inhibits the messenger RNA (mRNA) and protein expression of pro-inflammatory cytokines including interleukin and tumor necrosis factor α (TNF-α) [153].

3.5. Antiviral Activity

The ongoing threat of viral infections, such as influenza virus [154], coronavirus disease 2019 (COVID-19) virus [155], and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus [156], underscores the importance of effective antiviral therapies in preventing disease spread, mitigating viral damage, and facilitating patient recovery. Antiviral drugs not only help control outbreaks and improve treatment outcomes but also minimize the risk of viral mutations and drug resistance. In this context, sesquiterpenes, a class of compounds derived from natural products, hold significant promise for antiviral drug development. Their potential for inhibiting viral activity, supporting drug development, and boosting immunity offers valuable insights for future antiviral strategies.
Research indicates that sesquiterpene compounds 3, 11, 12, 17, 54, and 55 have inhibitory effects on Coxsackievirus B3 and influenza A virus. Specifically, compounds 3, 54, and 55 have moderate antiviral activity against Coxsackievirus B3, with IC50 values ranging from 6.41 to 33.33 μM. Meanwhile, compounds 12 and 17 showed weak inhibitory activity against the influenza A virus with IC50 values ranging from 19.24 to 33.33 μM [76]. Furthermore, guaianediol (45) obtained from A. pilosus in 2016 displayed anti-human immunodeficiency virus type 1 (anti-HIV-1) virus activity. In previous related studies, a variety of research methods have been used to explore its anti-HIV-1 activity, such as human immunodeficiency virus type 1 reverse transcriptase (HIV-1 RT) assay, syncytium assay, and other research methods. In addition to its significant anti-HIV-1 activity in syncytium assay, the results suggest that Guaianediol may inhibit HIV-1 RT, though its exact IC50 value requires further investigation [157].

3.6. Antioxidant Activity

Studies have shown that some sesquiterpene compounds have significant antioxidant activity [158,159]. These compounds can exhibit antioxidant properties in vivo through a variety of mechanisms, including scavenging free radicals, increasing antioxidant enzyme activity, and regulating oxidation-reduction balance. Among these, spathulenol (64) not only exhibits an anti-inflammatory effect but also demonstrates a significant antioxidant effect, with its IC50 value ranging from 26.13 to 85.60 μM [152]. Furthermore, studies on the dichloromethane extract of dihydroactinidiolide (78) have revealed its free radical scavenging activity, underscoring the antioxidant potential of sesquiterpenes [137].

3.7. Discussion on Structure-Activity Relationships

In general, compounds with the same skeleton structure are often possessed of similar biological activities and pharmacological effects. Through comparison and analysis of some compounds with the same skeleton structure, as well as known activities, the possible structure-activity relationship of some sesquiterpene compounds with the same skeleton structure is discussed.
Compounds 56, 58, and 59 are cadinane-type sesquiterpenes, with the same skeleton structure. They are all possessed of anti-tumor activities, but the inhibitory effect on the same tumor cells is different. The difference in the structure of compounds 56 and 58 structure is only the difference in the hydrogen atom configuration at the C-1 position. The hydrogen atom at the C-1 position of compound 56 is the R configuration, and the hydrogen atom at the C-1 position of 58 is the S configuration. It is speculated that it may be the main factor affecting the pharmacological activity of the two. When the hydrogen atom at the C-1 position of the two is the R configuration, this may have a better inhibitory effect on the tumor cells of A-549, MCF-7, and SW480.

4. Conclusions

Artabotrys, a prominent genus within the Annonaceae family, is renowned for its vast global presence and rich chemical diversity, including flavonoids, alkaloids, and terpenoids. Many of the chemical components have shown good pharmacological activity and have high research value.
This paper presents a comprehensive review of the sesquiterpene compounds identified in plants and their pharmacological activities, aiming to provide a solid scientific foundation for further exploring and utilizing this genus. It also seeks to deepen the understanding of sesquiterpene compounds’ pharmacological actions and mechanisms. An extensive review of research literature has cataloged approximately 85 sesquiterpene compounds and their sources from Artabotrys plants, categorizing them according to their structural characteristics. In addition to the common types of sesquiterpenes, such as bisabolane-type sesquiterpenes and eudesmane-type sesquiterpenes, which are rich in plant and microbial sources, this genus also harbors sesquiterpenes with special structures that are relatively rare. Pharmacological research reveals that these compounds exhibit a broad spectrum of activities, including antimalarial, anti-inflammatory, antiviral, and antitumor effects, underscoring their significant medicinal potential and positioning them as potential leads for drug development.
Despite the promising pharmacological activity, the mechanism behind the activities of sesquiterpenes from Artabotrys plants remains insufficiently explored, posing challenges to their clinical application of the related sesquiterpenes. Furthermore, many sesquiterpene components in the genus of Artabotrys remain undiscovered, suggesting vast opportunities for future research. It is anticipated that ongoing studies will uncover new sesquiterpene compounds and elucidate their mechanisms of action, enhancing the therapeutic value of Artabotrys sesquiterpenes.

Author Contributions

Conceptualization, S.L.; methodology, Y.S. and J.X.; software, Y.S. and J.X.; validation, Y.S. and J.X.; formal analysis, Y.S. and J.X.; investigation, Y.S. and J.X.; resources, S.L.; data curation, Y.X. and X.W.; writing—original draft preparation, Y.S. and J.X.; writing—review and editing, F.Z., C.N. and S.L.; visualization, C.N. and S.L.; supervision, F.Z., 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 Doctors in Yantai University, grant number YX20B03.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

NMRNuclear magnetic resonance
FPPFarnesyl diphosphate
LPSLipopolysaccharide
STSSesquiterpene synthase
SERTRegulating serotonin transporter
IC50Half maximal inhibitory concentration
MTT3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
PI3KPhosphoinositide 3-kinase
AKTProtein kinase B
mTORMammalian target of rapamycin
S6K1Ribosomal protein S6 kinase 1
MAPKMitogen-activated protein kinase
ERKExtracellular signal-regulated kinase
JNKc-Jun N-terminal kinase
iNOSInducible nitric oxide synthase
COX-2Cyclooxygenase-2
mRNAMessenger RNA
TNF-αTumor necrosis factor α
COVID-19Coronavirus disease 2019
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
anti-HIV-1Anti-human immunodeficiency virus type 1
HIV-1 RTHuman immunodeficiency virus type 1 reverse transcriptase

References

  1. Cascaes, M.M.; Carneiro, O.D.; Nascimento, L.D.; de Moraes, Â.A.; de Oliveira, M.S.; Cruz, J.N.; Guilhon, G.M.; Andrade, E.H. Essential Oils from Annonaceae Species from Brazil: A Systematic Review of Their Phytochemistry, and Biological Activities. Int. J. Mol. Sci. 2021, 22, 12140. [Google Scholar] [CrossRef]
  2. Jourjine, I.A.P.; Bauernschmidt, C.; Müller, C.; Bracher, F. A GC-MS Protocol for the Identification of Polycyclic Aromatic Alkaloids from Annonaceae. Molecules 2022, 27, 8217. [Google Scholar] [CrossRef]
  3. Yao, L.J.; Jalil, J.; Attiq, A.; Hui, C.C.; Zakaria, N.A. The medicinal uses, toxicities and anti-inflammatory activity of Polyalthia species (Annonaceae). J. Ethnopharmacol. 2019, 229, 303–325. [Google Scholar] [CrossRef]
  4. Santos, A.C.d.; Nogueira, M.L.; Oliveira, F.P.d.; Costa, E.V.; Bezerra, D.P. Essential Oils of Duguetia Species A. St. Hill (Annonaceae): Chemical Diversity and Pharmacological Potential. Biomolecules 2022, 12, 615. [Google Scholar] [CrossRef] [PubMed]
  5. Araújo, C.d.S.; Nery, D.A.; Oliveira, A.P.d.; Oliveira-Júnior, R.G.d.; Rolim, L.A.; Lopes, N.P.; Silva, M.F.d.S.; Pessoa, C.d.Ó.; Braz-Filho, R.; Dutra, L.M.; et al. New ent-kaurene-type nor-diterpene and other compounds isolated from Annona vepretorum Mart. (Annonaceae). Nat. Prod. Res. 2023, 37, 1565–1572. [Google Scholar] [CrossRef]
  6. Lorenzo, V.P.; Scotti, L.; da Silva Almeida, J.R.G.; Scotti, M.T. Annonaceae Family Alkaloids as Agents against Leishmaniasis: A Review and Molecular Docking Evaluation. Curr. Drug Metab. 2020, 21, 482–492. [Google Scholar] [CrossRef] [PubMed]
  7. Lúcio, A.S.; Almeida, J.R.; Da-Cunha, E.V.; Tavares, J.F.; Barbosa Filho, J.M. Alkaloids of the Annonaceae: Occurrence and a compilation of their biological activities. Alkaloids Chem. Biol. 2015, 74, 233–409. [Google Scholar] [PubMed]
  8. Lorenzo, V.P.; Lúcio, A.S.; Scotti, L.; Tavares, J.F.; Filho, J.M.; Lima, T.K.; Rocha, J.D.; Scotti, M.T. Structure-and Ligand-Based Approaches to Evaluate Aporphynic Alkaloids from Annonaceae as Multi-Target Agent against Leishmania donovani. Curr. Pharm. Des. 2016, 22, 5196–5203. [Google Scholar] [CrossRef]
  9. Nguemdjo Chimeze, V.W.; Bankoglu, E.E.; Zühlke, S.; Fannang, V.S.; Eckelmann, D.; Chi Shirri, J.; Djuidje, E.N.; Djama, C.M.; Stopper, H.; Wandji, J. Cytotoxic and genotoxic properties of artathomsonine, a new oxoberberine alkaloid from Artabotrys thomsonii (annonaceae). Nat. Prod. Res. 2022, 36, 2791–2799. [Google Scholar] [CrossRef]
  10. Huynh, N.V.; Nguyen Huu, D.M.; Huynh, N.T.; Chau, D.H.; Nguyen, C.D.; Nguyen Truong, Q.D.; Mai, D.T.; Dang, P.H. Anonazepine, a new alkaloid from the leaves of Annona muricata (Annonaceae). Z. Naturforschung C J. Biosci. 2022, 78, 247–251. [Google Scholar] [CrossRef]
  11. de Moraes, M.R.; Ryan, S.M.; Godoy, H.T.; Thomas, A.L.; Maia, J.G.S.; Richards, K.M.; Tran, K.; Smith, R.E. Phenolic Compounds and Metals in Some Edible Annonaceae Fruits. Biol. Trace Elem. Res. 2020, 197, 676–682. [Google Scholar] [CrossRef] [PubMed]
  12. Maia, D.S.; Lopes, C.F.; Saldanha, A.A.; Silva, N.L.; Sartori, Â.L.B.; Carollo, C.A.; Sobral, M.G.; Alves, S.N.; Silva, D.B.; de Siqueira, J.M. Larvicidal effect from different Annonaceae species on Culex quinquefasciatus. Environ. Sci. Pollut. Res. 2020, 27, 36983–36993. [Google Scholar] [CrossRef] [PubMed]
  13. Rady, I.; Bloch, M.B.; Chamcheu, R.N.; Banang Mbeumi, S.; Anwar, M.R.; Mohamed, H.; Babatunde, A.S.; Kuiate, J.R.; Noubissi, F.K.; El Sayed, K.A.; et al. Anticancer Properties of Graviola (Annona muricata): A Comprehensive Mechanistic Review. Oxid. Med. Cell. Longev. 2018, 2018, 1826170. [Google Scholar] [CrossRef] [PubMed]
  14. Costa, E.V.; de Souza, C.A.S.; Galvão, A.F.C.; Silva, V.R.; Santos, L.d.S.; Dias, R.B.; Rocha, C.A.G.; Soares, M.B.P.; da Silva, F.M.A.; Koolen, H.H.F.; et al. Duguetia pycnastera Sandwith (Annonaceae) Leaf Essential Oil Inhibits HepG2 Cell Growth In Vitro and In Vivo. Molecules 2022, 27, 5664. [Google Scholar] [CrossRef] [PubMed]
  15. Galvão, A.F.C.; Araújo, M.d.S.; Silva, V.R.; Santos, L.d.S.; Dias, R.B.; Rocha, C.A.G.; Soares, M.B.P.; Silva, F.M.A.d.; Koolen, H.H.F.; Zengin, G.; et al. Antitumor Effect of Guatteria olivacea R. E. Fr. (Annonaceae) Leaf Essential Oil in Liver Cancer. Molecules 2022, 27, 4407. [Google Scholar] [CrossRef] [PubMed]
  16. Terezan, A.P.; Junqueira, J.G.M.; Wakui, V.G.; Kato, L.; Oliveira, C.M.A.; Martins, C.H.G.; Santiago, M.B.; Severino, V.G.P. Qualitative analysis of the acetogenins from Annona coriacea (Annonaceae) leaves by HPLC-Q-Orbitrap and their antibacterial potential against oral pathogens. Nat. Prod. Res. 2022, 36, 765–771. [Google Scholar] [CrossRef] [PubMed]
  17. Aminimoghadamfarouj, N.; Nematollahi, A.; Wiart, C. Annonaceae: Bio-resource for tomorrow’s drug discovery. J. Asian Nat. Prod. Res. 2011, 13, 465–476. [Google Scholar] [CrossRef]
  18. Okpekon, T.A.; Kabran, F.A.; Say, V.M.; Evanno, L.; Maciuk, A.; Loiseau, P.; Champy, P.; Figadère, B. Apoprunellelactone (APL), an antiprotozoal lactone from the stem barks of Isolona cooperi Hutch. & Dalziel (Annonaceae). Nat. Prod. Res. 2021, 35, 5112–5119. [Google Scholar]
  19. Kayo, M.T.; Simo, M.K.; Tagatsing Fotsing, M.; Talla, E.; Laurent, S.; Elst, L.V.; Henoumont, C.; Yankep, E.; Alfred Ngenge, T.; Keumoe, R.; et al. Antifungal potential of extracts, fractions and compounds from Uvaria comperei (Annonaceae) and Oxyanthus unilocularis (Rubiaceae). Nat. Prod. Res. 2021, 35, 5732–5736. [Google Scholar] [CrossRef]
  20. Tundis, R.; Xiao, J.; Loizzo, M.R. Annona species (Annonaceae): A rich source of potential antitumor agents? Ann. N. Y. Acad. Sci. 2017, 1398, 30–36. [Google Scholar] [CrossRef]
  21. Andriamadio, J.H.; Rasoanaivo, L.H.; Benedec, D.; Vlase, L.; Gheldiu, A.M.; Duma, M.; Toiu, A.; Raharisololalao, A.; Oniga, I. HPLC/MS analysis of polyphenols, antioxidant and antimicrobial activities of Artabotrys hildebrandtii O. Hffm. extracts. Nat. Prod. Res. 2015, 29, 2188–2196. [Google Scholar] [CrossRef] [PubMed]
  22. Quang Hop, N.; The Son, N. Botanical Description, Traditional Uses, Phytochemistry, and Pharmacology of the Genus Artabotrys: A Review. Chem. Biodivers. 2022, 19, e202200725. [Google Scholar] [CrossRef] [PubMed]
  23. Hsieh, T.J.; Chang, F.R.; Chia, Y.C.; Chen, C.Y.; Lin, H.C.; Chiu, H.F.; Wu, Y.C. The Alkaloids of Artabotrys uncinatus. J. Nat. Prod. 2001, 64, 1157–1161. [Google Scholar] [CrossRef] [PubMed]
  24. Tan, K.K.; Khoo, T.J.; Rajagopal, M.; Wiart, C. Antibacterial alkaloids from Artabotrys crassifolius Hook.f. & Thomson. Nat. Prod. Res. 2015, 29, 2346–2349. [Google Scholar] [PubMed]
  25. Wong, H.F.; Brown, G.D. β-Methoxy-γ-methylene-α,β-unsaturated-γ-butyrolactones from Artabotrys hexapetalus. Phytochemistry 2002, 59, 99–104. [Google Scholar] [CrossRef] [PubMed]
  26. Murphy, B.T.; Cao, S.; Brodie, P.J.; Miller, J.S.; Ratovoson, F.; Birkinshaw, C.; Rakotobe, E.; Rasamison, V.E.; Tendyke, K.; Suh, E.M.; et al. Antiproliferative compounds of Artabotrys madagascariensis from the Madagascar rainforest. Nat. Prod. Res. 2008, 22, 1169–1175. [Google Scholar] [CrossRef] [PubMed]
  27. Tang, J.Y.; Liu, Y.P.; Ju, P.K.; Luo, X.L.; Zhang, Z.J.; Ren, P.; Lai, L.; Chen, G.Y.; Fu, Y.H. A new polyoxygenated cyclohexene derivative from Artabotrys hainanensis. Nat. Prod. Res. 2018, 32, 1727–1732. [Google Scholar] [CrossRef]
  28. Liu, Y.P.; Tang, J.Y.; Hua, Y.; Lai, L.; Luo, X.L.; Zhang, Z.J.; Yin, W.Q.; Chen, G.Y.; Fu, Y.H. Bioactive polyoxygenated seco-cyclohexenes from Artabotrys hongkongensis. Bioorg. Chem. 2018, 76, 386–391. [Google Scholar] [CrossRef] [PubMed]
  29. Jingguang, Y.; Tongmei, L.; Lan, S.; Xiuzhen, L. Neo-Lignans and Hemiterpenoid from the Seeds of Artabostrys hexapetalus (Annonaceae). J. Chin. Pharm. Sci. 2002, 11, 4–10. [Google Scholar]
  30. Somanawat, J.; Talangsri, N.; Deepolngam, S.; Kaewamatawong, R. Flavonoid and megastigmane glycosides from Artabotrys hexapetalus leaves. Biochem. Syst. Ecol. 2012, 44, 124–127. [Google Scholar] [CrossRef]
  31. Sharma, M.; Singh, N.; Jafri, M.; Mehta, B. Anthraquinones from Artabotrys odoratissimus (Leaves). Indian J. Chem. 2005, 44, 1740–1741. [Google Scholar]
  32. Xi, F.M.; Ma, S.G.; Liu, Y.B.; Li, L.; Yu, S.S. Artaboterpenoids A and B, Bisabolene-Derived Sesquiterpenoids from Artabotrys hexapetalus. Org. Lett. 2016, 18, 3374–3377. [Google Scholar] [CrossRef]
  33. Achoub, H.; Mencherini, T.; Esposito, T.; Luca, R.; Aquino, R.; Gazzerro, P.; Zaiter, L.; Benayache, F.; Benayache, S. New sesquiterpenes from Asteriscus graveolens. Nat. Prod. Res. 2021, 35, 2190–2198. [Google Scholar] [CrossRef] [PubMed]
  34. Eddin, L.B.; Jha, N.K.; Goyal, S.N.; Agrawal, Y.O.; Subramanya, S.B.; Bastaki, S.M.A.; Ojha, S. Health Benefits, Pharmacological Effects, Molecular Mechanisms, and Therapeutic Potential of α-Bisabolol. Nutrients 2022, 14, 1370. [Google Scholar] [CrossRef] [PubMed]
  35. Scandiffio, R.; Geddo, F.; Cottone, E.; Querio, G.; Antoniotti, S.; Gallo, M.P.; Maffei, M.E.; Bovolin, P. Protective Effects of (E)-β-Caryophyllene (BCP) in Chronic Inflammation. Nutrients 2020, 12, 3273. [Google Scholar] [CrossRef] [PubMed]
  36. Su, W.; Zhao, J.P.; Hu, J.; Yang, M.; Jacob, M.; Cai, X.; Zeng, R.; Chen, S.H.; Huang, H.Y.; Khan, I.; et al. Two new bicyclic sesquiterpenes from the stems of Kadsura heteroclita. Nat. Prod. Res. 2014, 28, 1197–1201. [Google Scholar] [CrossRef] [PubMed]
  37. Le Bideau, F.; Kousara, M.; Chen, L.; Wei, L.; Dumas, F. Tricyclic Sesquiterpenes from Marine Origin. Chem. Rev. 2017, 117, 6110–6159. [Google Scholar] [CrossRef]
  38. Hemtasin, C.; Kanokmedhakul, S.; Kanokmedhakul, K.; Hahnvajanawong, C.; Soytong, K.; Prabpai, S.; Kongsaeree, P. Cytotoxic Pentacyclic and Tetracyclic Aromatic Sesquiterpenes from Phomopsis archeri. J. Nat. Prod. 2011, 74, 609–613. [Google Scholar] [CrossRef] [PubMed]
  39. Dai, Q.; Zhang, F.L.; Feng, T. Sesquiterpenoids Specially Produced by Fungi: Structures, Biological Activities, Chemical and Biosynthesis (2015–2020). J. Fungi 2021, 7, 1026. [Google Scholar] [CrossRef]
  40. Durairaj, J.; Di Girolamo, A.; Bouwmeester, H.J.; de Ridder, D.; Beekwilder, J.; van Dijk, A.D. An analysis of characterized plant sesquiterpene synthases. Phytochemistry 2019, 158, 157–165. [Google Scholar] [CrossRef]
  41. Yan, H.; Xu, L.L.; Zheng, X.F.; Zou, X.F.; Xiao, L.G.; Zhou, Y.S.; He, L.; Liu, H.Y. Sesquiterpenes from Chloranthus holostegius with anti-inflammatory activities. Fitoterapia 2024, 172, 105766. [Google Scholar] [CrossRef] [PubMed]
  42. Rajachan, O.a.; Sangdee, A.; Kanokmedhakul, K.; Tontapha, S.; Amornkitbamrung, V.; Kanokmedhakul, S. Cyclofarnesane sesquiterpenoids from the fungus Sanghuangporus sp. Phytochem. Lett. 2020, 37, 17–20. [Google Scholar] [CrossRef]
  43. Zou, J.X.; Song, Y.P.; Liu, X.H.; Li, X.N.; Ji, N.Y. Bisabolane, cadinane, and cyclonerane sesquiterpenes from an algicolous strain of Trichoderma asperelloides. Bioorg. Chem. 2021, 115, 105223. [Google Scholar] [CrossRef] [PubMed]
  44. Ding, L.F.; Liu, J.X.; Xie, Z.Q.; Wang, D.S.; Nie, W.; Song, L.D.; Wu, X.D.; Zhao, Q.S. Magnograndins J-M, elemane sesquiterpenoids from the leaves of Magnolia grandiflora and their inhibitory effects on nitric oxide production. Phytochem. Lett. 2019, 31, 121–124. [Google Scholar] [CrossRef]
  45. Wu, Q.X.; Shi, Y.P.; Jia, Z.J. Eudesmane sesquiterpenoids from the Asteraceae family. Nat. Prod. Rep. 2006, 23, 699–734. [Google Scholar] [CrossRef] [PubMed]
  46. Tan, Y.P.; Savchenko, A.I.; Agnew-Francis, K.A.; Boyle, G.M.; Bernhardt, P.V.; Fraser, J.A.; Williams, C.M. Kalparinol, a Salvialane (Isodaucane) Sesquiterpenoid Derived from Native Australian Dysphania Species That Suggests a Putative Biogenetic Link to Zerumbone. J. Nat. Prod. 2020, 83, 1473–1479. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, J.M.; Zhang, D.W.; Du, W.Y.; Zhang, M.; Zhao, J.L.; Chen, R.D.; Xie, K.B.; Dai, J.G. Sesquiterpenes from the endophytic fungus Periconia sp. F-31. J. Asian Nat. Prod. Res. 2022, 24, 397–402. [Google Scholar] [CrossRef] [PubMed]
  48. Li, J.; Zhao, J.; Wang, W.; Li, L.; Zhang, L.; Zhao, X.F.; Liu, Q.R.; Liu, F.; Yang, M.; Khan, I.A.; et al. New Acorane-Type Sesquiterpene from Acorus calamus L. Molecules 2017, 22, 529. [Google Scholar] [CrossRef] [PubMed]
  49. Yuyama, K.T.; Fortkamp, D.; Abraham, W.R. Eremophilane-type sesquiterpenes from fungi and their medicinal potential. Biol. Chem. 2017, 399, 13–28. [Google Scholar] [CrossRef]
  50. Arizmendi, N.; Hou, C.; Guo, F.; Li, Y.; Kulka, M. Bicyclic eremophilane-type petasite sesquiterpenes potentiate peroxisome proliferator-activated receptor γ activator-mediated inhibition of dendritic cells. Int. J. Immunopathol. Pharmacol. 2018, 32, 2058738418787739. [Google Scholar] [CrossRef]
  51. Wang, Q.; Tang, X.; Liu, H.; Luo, X.; Sung, P.J.; Li, P.; Li, G. Clavukoellians G–K, New Nardosinane and Aristolane Sesquiterpenoids with Angiogenesis Promoting Activity from the Marine Soft Coral Lemnalia sp. Mar. Drugs 2020, 18, 171. [Google Scholar] [CrossRef]
  52. Buechi, G.; Hofheinz, W.; Paukstelis, J.V. Synthesis of (-)-aromadendrene and related sesquiterpenes. J. Am. Chem. Soc. 1969, 91, 6473–6478. [Google Scholar] [CrossRef]
  53. Li, C.; Liu, K.; Liu, S.; Aerqin, Q.; Wu, X. Role of Ginkgolides in the Inflammatory Immune Response of Neurological Diseases: A Review of Current Literatures. Front. Syst. Neurosci. 2020, 14, 45. [Google Scholar] [CrossRef] [PubMed]
  54. Hien, N.T.; Cuc, D.T.; Thuy, N.T.T.; Hiep, H.; Huyen, V.T.; Ai, D.T.T.; Nhiem, N.X. Labdane- type diterpenoids and sesquiterpenes from Curcuma aromatica and their nitric oxide inhibitory activity in lipopolysaccharide-stimulated RAW264.7 macrophages. J. Asian Nat. Prod. Res. 2023, 26, 387–393. [Google Scholar] [CrossRef]
  55. Park, H.J.; Kwon, S.H.; Yoo, K.O.; Sohn, I.C.; Lee, K.T.; Lee, H.K. Sesquiterpenes from the leaves of Ligularia fischeri var. spiciformis. Planta Med. 2000, 66, 783–784. [Google Scholar] [CrossRef] [PubMed]
  56. Descoins, C., Jr.; Bazzocchi, I.L.; Ravelo, A.G. New Sesquiterpenes from Euonymus europaeus (Celastraceae). Chem. Pharm. Bull. 2002, 50, 199–202. [Google Scholar] [CrossRef]
  57. Yang, X.Y.; Niu, W.R.; Li, R.T.; Cui, X.M.; Liu, J.K. Two new sesquiterpenes from cultures of the higher fungus Pholiota nameko. Nat. Prod. Res. 2019, 33, 1992–1996. [Google Scholar] [CrossRef]
  58. Shi, Z.Z.; Fang, S.T.; Miao, F.P.; Yin, X.L.; Ji, N.Y. Trichocarotins A–H and trichocadinin A, nine sesquiterpenes from the marine-alga-epiphytic fungus Trichoderma virens. Bioorg. Chem. 2018, 81, 319–325. [Google Scholar] [CrossRef]
  59. Rodriguez, S.; Kirby, J.; Denby, C.M.; Keasling, J.D. Production and quantification of sesquiterpenes in Saccharomyces cerevisiae, including extraction, detection and quantification of terpene products and key related metabolites. Nat. Protoc. 2014, 9, 1980–1996. [Google Scholar] [CrossRef]
  60. Song, Y.P.; Fang, S.T.; Miao, F.P.; Yin, X.L.; Ji, N.Y. Diterpenes and Sesquiterpenes from the Marine Algicolous Fungus Trichoderma harzianum X-5. J. Nat. Prod. 2018, 81, 2553–2559. [Google Scholar] [CrossRef]
  61. Torii, M.; Kato, H.; Hitora, Y.; Angkouw, E.D.; Mangindaan, R.E.P.; de Voogd, N.J.; Tsukamoto, S. Lamellodysidines A and B, Sesquiterpenes Isolated from the Marine Sponge Lamellodysidea herbacea. J. Nat. Prod. 2017, 80, 2536–2541. [Google Scholar] [CrossRef] [PubMed]
  62. Thebtaranonth, C.; Thebtaranonth, Y.; Wanauppathamkul, S.; Yuthavong, Y. Antimalarial sesquiterpenes from tubers of Cyperus rotundus: Structure of 10,12-Peroxycalamenene, a sesquiterpene endoperoxide. Phytochemistry 1995, 40, 125–128. [Google Scholar] [CrossRef] [PubMed]
  63. Bartikova, H.; Hanusova, V.; Skalova, L.; Ambroz, M.; Bousova, I. Antioxidant, pro-oxidant and other biological activities of sesquiterpenes. Curr. Top. Med. Chem. 2014, 14, 2478–2494. [Google Scholar] [CrossRef] [PubMed]
  64. Nuermaimaiti, M.; Turak, A.; Yang, Q.; Tang, B.; Zang, Y.; Li, J.; Aisa, H.A. Sesquiterpenes from Artemisia Sieversiana and their anti-inflammatory activities. Fitoterapia 2021, 154, 104996. [Google Scholar] [CrossRef] [PubMed]
  65. Chi, J.; Li, B.C.; Dai, W.F.; Liu, L.; Zhang, M. Highly oxidized sesquiterpenes from Artemisia austro-yunnanensis. Fitoterapia 2016, 115, 182–188. [Google Scholar] [CrossRef] [PubMed]
  66. Liang, Y.; Xu, W.; Liu, C.; Zhou, D.; Liu, X.; Qin, Y.; Cao, F.; Li, J.; Yang, R.; Qin, J. Eremophilane sesquiterpenes from the endophytic fungus Xylaria sp. GDG-102. Nat. Prod. Res. 2019, 33, 1304–1309. [Google Scholar] [CrossRef] [PubMed]
  67. Hansson, D.; Menkis, A.; Himmelstrand, K.; Thelander, M.; Olson, Å.; Stenlid, J.; Karlsson, M.; Broberg, A. Sesquiterpenes from the conifer root rot pathogen Heterobasidion occidentale. Phytochemistry 2012, 82, 158–165. [Google Scholar] [CrossRef]
  68. Lim, P.C.; Ali, Z.; Khan, I.A.; Khan, S.I.; Kassim, N.K.; Awang, K.; Shaari, K.; Ismail, A. Cytotoxic constituent of Melicope latifolia (DC.) T. G. Hartley. Nat. Prod. Res. 2022, 36, 1416–1424. [Google Scholar] [CrossRef] [PubMed]
  69. Wu, Z.l.; Li, J.Y.; Sun, Z.S.; Yang, Y.X.; Xu, X.K.; Li, H.L.; Zhang, W.D. Vlasouliodes A-D, four new C30 dimeric sesquiterpenes exhibiting potential inhibition of MCF-7 cells from Vladimiria souliei. Fitoterapia 2022, 161, 105234. [Google Scholar] [CrossRef]
  70. Li, C.S.; Liu, L.T.; Yang, L.; Li, J.; Dong, X. Chemistry and Bioactivity of Marine-Derived Bisabolane Sesquiterpenoids: A Review. Front. Chem. 2022, 10, 881767. [Google Scholar] [CrossRef]
  71. Hu, S.; Ma, Y.L.; Guo, J.M.; Wen, Q.; Yan, G.; Yang, S.; Fu, Y.H.; Liu, Y.P. Bisabolane sesquiterpenes from Clausena sanki with their potential anti-inflammatory activities. Nat. Prod. Res. 2020, 34, 3499–3505. [Google Scholar] [CrossRef]
  72. Shu, H.Z.; Peng, C.; Bu, L.; Guo, L.; Liu, F.; Xiong, L. Bisabolane-type sesquiterpenoids: Structural diversity and biological activity. Phytochemistry 2021, 192, 112927. [Google Scholar] [CrossRef]
  73. Liang, X.T.; Yu, D.Q.; Wu, W.L.; Deng, H.C. The structure of yingzhaosu A. Acta Chim. Sin. 1979, 37, 215–230. [Google Scholar]
  74. Liang, X.T.; Yu, D.Q.; Pan, W.D. The structure of yingzhaosu B. Acta Chim. Sin. 1979, 37, 231–240. [Google Scholar]
  75. Zhang, L.; Zhou, W.S.; Xu, X. A new sesquiterpene peroxide (yingzhaosu C) and sesquiterpenol (yingzhaosu D) from Artabotrys unciatus (L.) Meer. J. Chem. Soc. Chem. Commun. 1988, 523–524. [Google Scholar]
  76. Xi, F.M.; Liu, Y.B.; Qu, J.; Li, Y.; Tang, Z.H.; Li, L.; Li, Y.H.; Chen, X.G.; Ma, S.G.; Yu, S.S. Bioactive sesquiterpenoids from the roots of Artabotrys hexapetalus. Tetrahedron 2017, 73, 571–582. [Google Scholar] [CrossRef]
  77. Wang, T.W. Study on Structures and Antitumor Activities of Chemical Constituents from Artabotrys pilosus. Master’s Thesis, Jilin Agricultural University, Changchun, China, 2016. [Google Scholar]
  78. Gu, C.; Yin, A.P.; Yuan, H.Y.; Yang, K.; Luo, J.; Zhan, Y.J.; Yang, C.R.; Zuo, D.M.; Li, H.Z.; Xu, M. New anti-HBV norbisabolane sesquiterpenes from Phyllantus acidus. Fitoterapia 2019, 137, 104151. [Google Scholar] [CrossRef] [PubMed]
  79. Wu, S.L.; Liu, Y.P.; Chen, G.Y.; Han, C.R.; Song, X.P.; Zhong, X.; Fu, Y.H. Sesquiterpenes from Artabotrys hongkongensis. China J. Chin. Mater. Med. 2017, 42, 3146–3151. [Google Scholar]
  80. Jang, H.J.; Lee, S.; Lee, S.J.; Lim, H.J.; Jung, K.; Kim, Y.H.; Lee, S.W.; Rho, M.C. Anti-inflammatory Activity of Eudesmane-Type Sesquiterpenoids from Salvia plebeia. J. Nat. Prod. 2017, 80, 2666–2676. [Google Scholar] [CrossRef] [PubMed]
  81. Li, W.; Cai, C.H.; Guo, Z.K.; Wang, H.; Zuo, W.J.; Dong, W.H.; Mei, W.L.; Dai, H.F. Five new eudesmane-type sesquiterpenoids from Chinese agarwood induced by artificial holing. Fitoterapia 2015, 100, 44–49. [Google Scholar] [CrossRef]
  82. Alarif, W.M.; Al-Footy, K.O.; Zubair, M.S.; Halid Ph, M.; Ghandourah, M.A.; Basaif, S.A.; Al-Lihaibi, S.S.; Ayyad, S.E.N.; Badria, F.A. The role of new eudesmane-type sesquiterpenoid and known eudesmane derivatives from the red alga Laurencia obtusa as potential antifungal–antitumour agents. Nat. Prod. Res. 2016, 30, 1150–1155. [Google Scholar] [CrossRef]
  83. Zhang, J.; Wang, Y.; Zhu, R.; Li, Y.; Li, Y.; Qiao, Y.; Zhou, J.; Lou, H. Cyperane and eudesmane-type sesquiterpenoids from Chinese liverwort and their anti-diabetic nephropathy potential. RSC Adv. 2018, 8, 39091–39097. [Google Scholar] [CrossRef] [PubMed]
  84. Ma, L.F.; Xu, H.; Wang, J.D.; Tong, X.M.; Zhan, Z.J.; Ying, Y.M.; Wang, J.W.; Zhang, H.; Shan, W.G. Three new eudesmane sesquiterpenoids and a new dimer from the aerial part of Salvia plebeia R. Br. Phytochem. Lett. 2018, 25, 122–125. [Google Scholar] [CrossRef]
  85. Wen, Q.; Liu, Y.P.; Yan, G.; Yang, S.; Hu, S.; Hua, J.; Yin, W.Q.; Chen, G.Y.; Fu, Y.H. Bioactive Eudesmane sesquiterpenes from Artabotrys hongkongensis Hance. Nat. Prod. Res. 2020, 34, 1687–1693. [Google Scholar] [CrossRef] [PubMed]
  86. Tang, J.Y. Study on the Chemical Constituents and Antitumor Activities of Artabotrys hainanensis R.E.Fries. Master’s Thesis, Fujian University of Tradition Chinese Medicine, Fuzhou, China, 2018. [Google Scholar]
  87. Li, Y.; Liu, J.; Wu, Y.; Li, Y.; Guo, F. Guaiane-type sesquiterpenes from Curcuma wenyujin. Phytochemistry 2022, 198, 113164. [Google Scholar] [CrossRef]
  88. Ma, G.H.; Chen, K.X.; Zhang, L.Q.; Li, Y.M. Advance in biological activities of natural guaiane-type sesquiterpenes. Med. Chem. Res. 2019, 28, 1339–1358. [Google Scholar] [CrossRef]
  89. Fleischer, T.C.; Waigh, R.D.; Waterman, P.G. Pogostol O-methyl ether and artabotrol: Two novel sesquiterpenes from the stem bark of Artabotrys stenopetalus. J. Nat. Prod. 1997, 60, 1054–1056. [Google Scholar] [CrossRef]
  90. Achenbach, H.; Schwinn, A. Aporphinoid alkaloids and terpenoids from Piptostigma fugax. Phytochemistry 1995, 38, 1037–1048. [Google Scholar] [CrossRef]
  91. Chen, G.Y.; Zhu, G.Y.; Han, C.R.; Li, Q.Y.; Fang, H.X.; Bi, H.P. Studies on sesquiterpenoids from the flowers of Artabotrys hainanensis. Chin. Tradit. Herb. Drugs 2005, 36, 1619–1620. [Google Scholar]
  92. Peng, G.P.; Tian, G.; Huang, X.F.; Lou, F.C. Guaiane-type sesquiterpenoids from Alisma orientalis. Phytochemistry 2003, 63, 877–881. [Google Scholar] [CrossRef]
  93. Ai, H.L.; Lv, X.; Ye, K.; Wang, M.X.; Huang, R.; Shi, B.B.; Li, Z.H. Four New Highly Oxygenated Eremophilane Sesquiterpenes from an Endophytic Fungus Boeremia exigua Isolated from Fritillaria hupehensis. J. Fungi 2022, 8, 492. [Google Scholar] [CrossRef]
  94. Niu, S.; Liu, D.; Shao, Z.; Proksch, P.; Lin, W. Eremophilane-type sesquiterpenoids in a deep-sea fungus Eutypella sp. activated by chemical epigenetic manipulation. Tetrahedron 2018, 74, 7310–7325. [Google Scholar] [CrossRef]
  95. Zhang, W.; Meng, Q.; Wu, J.; Cheng, W.; Liu, D.; Huang, J.; Fan, A.; Xu, J.; Lin, W. Acorane sesquiterpenes from the deep-sea derived Penicillium bilaiae fungus with anti-neuroinflammatory effects. Front. Chem. 2022, 10, 1036212. [Google Scholar] [CrossRef] [PubMed]
  96. Yong, J.Y.; Li, M.; Li, W.R.; Gao, R.M.; Su, G.Z.; Wang, H.Q.; Yang, J.; Li, L.; Li, Y.H.; Scott, P.; et al. Seco-Sesquiterpenes and acorane-type sesquiterpenes with antiviral activity from the twigs and leaves of Illicium henryi Diels. Bioorg. Chem. 2023, 131, 106324. [Google Scholar] [CrossRef] [PubMed]
  97. Guo, R.; Ren, Q.; Tang, Y.X.; Zhao, F.; Lin, B.; Huang, X.X.; Song, S.J. Sesquiterpenoids from the roots of Daphne genkwa Siebold et Zucc. with potential anti-inflammatory activity. Phytochemistry 2020, 174, 112348. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, M.; Zhao, J.L.; Liu, J.M.; Chen, R.D.; Xie, K.B.; Chen, D.W.; Feng, K.P.; Zhang, D.; Dai, J.G. Neural anti-inflammatory sesquiterpenoids from the endophytic fungus Trichoderma sp. Xy24. J. Asian Nat. Prod. Res. 2017, 19, 651–658. [Google Scholar] [CrossRef] [PubMed]
  99. Jiang, L.; Wen, Y.; Peng, Y.; Chen, T.; Chen, J.; Yang, J.; Gong, T.; Zhu, P. Advances in biosynthesis of cadinane sesquiterpenes. Chin. J. Biotechnol. 2021, 37, 1952–1967. [Google Scholar]
  100. Zhou, C.X.; Zhang, L.S.; Chen, F.F.; Wu, H.S.; Mo, J.X.; Gan, L.S. Terpenoids from Curcuma wenyujin increased glucose consumption on HepG2 cells. Fitoterapia 2017, 121, 141–145. [Google Scholar] [CrossRef] [PubMed]
  101. Shang, Z.C.; Han, C.; Xu, J.L.; Liu, R.H.; Yin, Y.; Wang, X.B.; Yang, M.H.; Kong, L.Y. Twelve formyl phloroglucinol meroterpenoids from the leaves of Eucalyptus robusta. Phytochemistry 2019, 163, 111–117. [Google Scholar] [CrossRef]
  102. Qin, D.P.; Pan, D.B.; Xiao, W.; Li, H.B.; Yang, B.; Yao, X.J.; Dai, Y.; Yu, Y.; Yao, X.S. Dimeric Cadinane Sesquiterpenoid Derivatives from Artemisia annua. Org. Lett. 2018, 20, 453–456. [Google Scholar] [CrossRef]
  103. Paddon, C.J.; Westfall, P.J.; Pitera, D.J.; Benjamin, K.; Fisher, K.; McPhee, D.; Leavell, M.D.; Tai, A.; Main, A.; Eng, D.; et al. High-level semi-synthetic production of the potent antimalarial artemisinin. Nature 2013, 496, 528–532. [Google Scholar] [CrossRef]
  104. Wang, L.X.; Jiang, X.J.; Li, X.M.; Mao, M.F.; Wei, G.Z.; Wang, F. Aristolane-type Sesquiterpenoids from Nardostachys chinensis and Revised Structure of Aristolanhydride. Nat. Prod. Bioprospect. 2019, 9, 149–155. [Google Scholar] [CrossRef] [PubMed]
  105. Durán-Peña, M.J.; Botubol Ares, J.M.; Hanson, J.R.; Collado, I.G.; Hernández-Galán, R. Biological activity of natural sesquiterpenoids containing a gem-dimethylcyclopropane unit. Nat. Prod. Rep. 2015, 32, 1236–1248. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, Y.P.; Ying, S.S.; Zheng, H.H.; Liu, Y.T.; Wang, Z.P.; Zhang, H.; Deng, X.; Wu, Y.J.; Gao, X.M.; Li, T.X.; et al. Novel serotonin transporter regulators: Natural aristolane- and nardosinane- types of sesquiterpenoids from Nardostachys chinensis Batal. Sci. Rep. 2017, 7, 15114. [Google Scholar] [CrossRef] [PubMed]
  107. Lan, Y.H.; Wang, H.Y.; Wu, C.C.; Chen, S.L.; Chang, C.L.; Chang, F.R.; Wu, Y.C. New Constituents from Stems of Artabotrys uncinatus. Chem. Pharm. Bull. 2007, 55, 1597–1599. [Google Scholar] [CrossRef] [PubMed]
  108. Shen, S.M.; Yang, Q.; Zang, Y.; Li, J.; Liu, X.; Guo, Y.W. Anti-inflammatory aromadendrane-and cadinane-type sesquiterpenoids from the South China Sea sponge Acanthella cavernosa. Beilstein J. Org. Chem. 2022, 18, 916–925. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, C.Y.; Zhang, J.Z.; Li, Y.L.; Xu, Z.J.; Qiao, Y.N.; Yuan, S.Z.; Tang, Y.J.; Lou, H.X. Heterodimers of Aromadendrane Sesquiterpenoid with Benzoquinone from the Chinese Liverwort Mylia nuda. J. Nat. Prod. 2024, 87, 132–140. [Google Scholar] [CrossRef]
  110. Mahidol, C.; Chimnoi, N.; Chokchaichamnankit, D.; Techasakul, S. Identification of volatile constituents in Artabotrys hexapetalus flowers using simple headspace solvent-trapping technique in combination with gas chromatography-mass spectrometry and retention indices. Acta Hortic. 2005, 667, 43–50. [Google Scholar] [CrossRef]
  111. Brabin, B.J. Malaria’s contribution to World War One–the unexpected adversary. Malar. J. 2014, 13, 497. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, Y.; He, Z.Q.; Wang, D.; Hu, Y.B.; Qian, D.; Yang, C.Y.; Zhou, R.M.; Li, S.H.; Lu, D.L.; Zhang, H.W. One Health approach to improve the malaria elimination programme in Henan Province. Adv. Parasitol. 2022, 116, 153–186. [Google Scholar]
  113. Garrido-Cardenas, J.A.; González-Cerón, L.; Manzano-Agugliaro, F.; Mesa-Valle, C. Plasmodium Genomics: An approach for learning about and ending human malaria. Parasitol. Res. 2019, 118, 1–27. [Google Scholar] [CrossRef]
  114. Barber, B.E.; Grigg, M.J.; Cooper, D.J.; van Schalkwyk, D.A.; William, T.; Rajahram, G.S.; Anstey, N.M. Clinical management of Plasmodium knowlesi malaria. Adv. Parasitol. 2021, 113, 45–76. [Google Scholar] [PubMed]
  115. Fuehrer, H.P.; Campino, S.; Sutherland, C.J. The primate malaria parasites Plasmodium malariae, Plasmodium brasilianum and Plasmodium ovale spp.: Genomic insights into distribution, dispersal and host transitions. Malar. J. 2022, 21, 138. [Google Scholar] [CrossRef] [PubMed]
  116. Cohen, S. Immunity to malaria. Proc. R. Soc. Lond. B Biol. Sci. 1979, 203, 323–345. [Google Scholar]
  117. Walker, I.S.; Rogerson, S.J. Pathogenicity and virulence of malaria: Sticky problems and tricky solutions. Virulence 2023, 14, 2150456. [Google Scholar] [CrossRef] [PubMed]
  118. White, N.J. The role of anti-malarial drugs in eliminating malaria. Malar. J. 2008, 7, S8. [Google Scholar] [CrossRef] [PubMed]
  119. Kong, L.Y.; Tan, R.X. Artemisinin, a miracle of traditional Chinese medicine. Nat. Prod. Rep. 2015, 32, 1617–1621. [Google Scholar] [CrossRef] [PubMed]
  120. Bailly, C.; Hénichart, J.P. Advocacy for the Medicinal Plant Artabotrys hexapetalus (Yingzhao) and Antimalarial Yingzhaosu Endoperoxides. Molecules 2022, 27, 6192. [Google Scholar] [CrossRef]
  121. Liu, S.; Wei, C.; Liu, T.; Ma, S.G.; Chen, C.; Lin, H.; Zhang, L.; Wang, H.; Zhang, C.J.; Yu, S.S. A heme-activatable probe and its application in the high-throughput screening of Plasmodium falciparum ring-stage inhibitors. Signal Transduct. Tar. 2022, 7, 160. [Google Scholar] [CrossRef]
  122. Boukouvalas, J.; Pouliot, R.; Fréchette, Y. Concise synthesis of yingzhaosu C and epi-yingzhaosu C by peroxyl radical cyclization. Assignment of relative configuration. Tetrahedron Lett. 1995, 36, 4167–4170. [Google Scholar] [CrossRef]
  123. Xu, X.; Xie, X. Total synthesis of Yingzhaosu B and its three diastereoisomers. Chin. J. Chem. 2010, 12, 381–384. [Google Scholar] [CrossRef]
  124. Xu, X.X.; Hu, Q.S. Synthesis of the diastereoisomeric Yingzhaosu D. Chin. J. Chem. 1992, 10, 285–288. [Google Scholar]
  125. Zhang, G.F.; Liu, X.; Zhang, S.; Pan, B.; Liu, M.L. Ciprofloxacin derivatives and their antibacterial activities. Eur. J. Med. Chem. 2018, 146, 599–612. [Google Scholar] [CrossRef] [PubMed]
  126. Li, Z.; Zhao, L.; Bian, Y.; Li, Y.; Qu, J.; Song, F. The Antibacterial Activity of Quinazoline and Quinazolinone Hybrids. Curr. Top. Med. Chem. 2022, 22, 1035–1044. [Google Scholar] [CrossRef] [PubMed]
  127. Kaur, R.; Rani, P.; Atanasov, A.G.; Alzahrani, Q.; Gupta, R.; Kapoor, B.; Gulati, M.; Chawla, P. Discovery and Development of Antibacterial Agents: Fortuitous and Designed. Mini Rev. Med. Chem. 2022, 22, 984–1029. [Google Scholar] [PubMed]
  128. D’Agostino, I.; Ardino, C.; Poli, G.; Sannio, F.; Lucidi, M.; Poggialini, F.; Visaggio, D.; Rango, E.; Filippi, S.; Petricci, E.; et al. Antibacterial alkylguanidino ureas: Molecular simplification approach, searching for membrane-based MoA. Eur. J. Med. Chem. 2022, 231, 114158. [Google Scholar] [CrossRef] [PubMed]
  129. Paulin, S.; Alm, R.A.; Beyer, P. A novel pre-clinical antibacterial pipeline database. PLoS ONE 2020, 15, e0236604. [Google Scholar] [CrossRef] [PubMed]
  130. Vila, J.; Moreno-Morales, J.; Ballesté-Delpierre, C. Current landscape in the discovery of novel antibacterial agents. Clin. Microbiol. Infect. 2020, 26, 596–603. [Google Scholar] [CrossRef] [PubMed]
  131. Cattoir, V.; Felden, B. Future Antibacterial Strategies: From Basic Concepts to Clinical Challenges. J. Infect. Dis. 2019, 220, 350–360. [Google Scholar] [CrossRef] [PubMed]
  132. Nyandoro, S.S.; Joseph, C.C.; Nkunya, M.H.H.; Hosea, K.M.M. New antimicrobial, mosquito larvicidal and other metabolites from two Artabotrys species. Nat. Prod. Res. 2013, 27, 1450–1458. [Google Scholar] [CrossRef]
  133. Tan, M.; Zhou, L.; Huang, Y.; Wang, Y.; Hao, X.; Wang, J. Antimicrobial activity of globulol isolated from the fruits of Eucalyptus globulus Labill. Nat. Prod. Res. 2008, 22, 569–575. [Google Scholar] [CrossRef]
  134. Chulalaksananukul, W. Chemical composition and antibacterial activity of extracts from freshwater green algae, Cladophora glomerata Kützing and Microspora floccosa (Vaucher) Thuret. J. BioScience Biotechnol. 2014, 3, 211. [Google Scholar]
  135. Debela, D.T.; Muzazu, S.G.Y.; Heraro, K.D.; Ndalama, M.T.; Mesele, B.W.; Haile, D.C.; Kitui, S.K.; Manyazewal, T. New approaches and procedures for cancer treatment: Current perspectives. SAGE Open Med. 2021, 9, 20503121211034366. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, X.; Tian, F.; Zhang, H.B.; Pilarinou, E.; McLaughlin, J.L. Biologically Active Blumenol A from the Leaves of Annona Glabra. Nat. Prod. Rep. 1999, 14, 77–81. [Google Scholar]
  137. Das, M.; Prakash, S.; Nayak, C.; Thangavel, N.; Singh, S.K.; Manisankar, P.; Devi, K.P. Dihydroactinidiolide, a natural product against Aβ25–35 induced toxicity in Neuro2a cells: Synthesis, in silico and in vitro studies. Bioorg. Chem. 2018, 81, 340–349. [Google Scholar] [CrossRef] [PubMed]
  138. Park, K.R.; Nam, D.; Yun, H.M.; Lee, S.G.; Jang, H.J.; Sethi, G.; Cho, S.K.; Ahn, K.S. β-Caryophyllene oxide inhibits growth and induces apoptosis through the suppression of PI3K/AKT/mTOR/S6K1 pathways and ROS-mediated MAPKs activation. Cancer Lett. 2011, 312, 178–188. [Google Scholar] [CrossRef]
  139. Martin, G.S. Cell signaling and cancer. Cancer Cell 2003, 4, 167–174. [Google Scholar] [CrossRef]
  140. Castaneda, C.A.; Cortes-Funes, H.; Gomez, H.L.; Ciruelos, E.M. The phosphatidyl inositol 3-kinase/AKT signaling pathway in breast cancer. Cancer Metastasis Rev. 2010, 29, 751–759. [Google Scholar] [CrossRef] [PubMed]
  141. Sanjeewa, K.K.A.; Herath, K.H.I.N.M.; Yang, H.W.; Choi, C.S.; Jeon, Y.J. Anti-Inflammatory Mechanisms of Fucoidans to Treat Inflammatory Diseases: A Review. Mar. Drugs 2021, 19, 678. [Google Scholar] [CrossRef] [PubMed]
  142. Hou, C.; Chen, L.; Yang, L.; Ji, X. An insight into anti-inflammatory effects of natural polysaccharides. Int. J. Biol. Macromol. 2020, 153, 248–255. [Google Scholar] [CrossRef]
  143. Rocha, D.H.A.; Pinto, D.C.G.A.; Silva, A.M.S. Macroalgae Specialized Metabolites: Evidence for Their Anti-Inflammatory Health Benefits. Mar. Drugs 2022, 20, 789. [Google Scholar] [CrossRef]
  144. Dinarello, C.A. Anti-inflammatory Agents: Present and Future. Cell 2010, 140, 935–950. [Google Scholar] [CrossRef] [PubMed]
  145. Kazemi, S.; Shirzad, H.; Rafieian-Kopaei, M. Recent Findings in Molecular Basis of Inflammation and Anti-inflammatory Plants. Curr. Pharm. Des. 2018, 24, 1551–1562. [Google Scholar] [CrossRef] [PubMed]
  146. Gautam, R.; Jachak, S.M. Recent developments in anti-inflammatory natural products. Med. Res. Rev. 2009, 29, 767–820. [Google Scholar] [CrossRef]
  147. Cruz-Martins, N. Molecular Mechanisms of Anti-Inflammatory Phytochemicals. Int. J. Mol. Sci. 2022, 23, 11016. [Google Scholar] [CrossRef]
  148. Pekacar, S.; Bulut, S.; Özüpek, B.; Orhan, D.D. Anti-Inflammatory and Analgesic Effects of Rosehip in Inflammatory Musculoskeletal Disorders and Its Active Molecules. Curr. Mol. Pharmacol. 2021, 14, 731–745. [Google Scholar] [CrossRef]
  149. Elbandy, M. Anti-Inflammatory Effects of Marine Bioactive Compounds and Their Potential as Functional Food Ingredients in the Prevention and Treatment of Neuroinflammatory Disorders. Molecules 2023, 28, 2. [Google Scholar] [CrossRef]
  150. Jang, J.H.; Lee, T.J. Mechanisms of Phytochemicals in Anti-Inflammatory and Anti-Cancer. Int. J. Mol. Sci. 2023, 24, 7863. [Google Scholar] [CrossRef] [PubMed]
  151. Delgado, G.; del Socorro Olivares, M.; Chávez, M.I.; Ramírez-Apan, T.; Linares, E.; Bye, R.; Espinosa-García, F.J. Antiinflammatory Constituents from Heterotheca inuloides. J. Nat. Prod. 2001, 64, 861–864. [Google Scholar] [CrossRef]
  152. do Nascimento, K.F.; Moreira, F.M.F.; Alencar Santos, J.; Kassuya, C.A.L.; Croda, J.H.R.; Cardoso, C.A.L.; Vieira, M.D.C.; Góis Ruiz, A.L.T.; Ann Foglio, M.; de Carvalho, J.E.; et al. Antioxidant, anti-inflammatory, antiproliferative and antimycobacterial activities of the essential oil of Psidium guineense Sw. and spathulenol. J. Ethnopharmacol. 2018, 210, 351–358. [Google Scholar] [CrossRef]
  153. Shi, D.; Song, X.; Guo, Y.; Xu, J.; Liu, Y.; Zhang, J.; Cui, C.A.; Jin, D.Q. Alismol, a Sesquiterpenoid Isolated from Vladimiria souliei, Suppresses Proinflammatory Mediators in Lipopolysaccharide-Stimulated Microglia. J. Mol. Neurosci. 2017, 62, 106–113. [Google Scholar] [CrossRef]
  154. Kumari, R.; Sharma, S.D.; Kumar, A.; Ende, Z.; Mishina, M.; Wang, Y.; Falls, Z.; Samudrala, R.; Pohl, J.; Knight, P.R.; et al. Antiviral Approaches against Influenza Virus. Clin. Microbiol. Rev. 2023, 36, e0004022. [Google Scholar] [CrossRef] [PubMed]
  155. Gudima, G.; Kofiadi, I.; Shilovskiy, I.; Kudlay, D.; Khaitov, M. Antiviral Therapy of COVID-19. Int. J. Mol. Sci. 2023, 24, 8867. [Google Scholar] [CrossRef] [PubMed]
  156. Beheshtirouy, S.; Khani, E.; Khiali, S.; Entezari-Maleki, T. Investigational antiviral drugs for the treatment of COVID-19 patients. Arch. Virol. 2022, 167, 751–805. [Google Scholar] [CrossRef] [PubMed]
  157. Chareonkla, A.; Pohmakotr, M.; Reutrakul, V.; Yoosook, C.; Kasisit, J.; Napaswad, C.; Tuchinda, P. A new diarylheptanoid from the rhizomes of Zingiber mekongense. Fitoterapia 2011, 82, 534–538. [Google Scholar] [CrossRef] [PubMed]
  158. Yanda, L.; Tatsimo, S.J.N.; Tamokou, J.D.D.; Matsuete-Takongmo, G.; Meffo-Dongmo, S.C.; Meli Lannang, A.; Sewald, N. Antibacterial and Antioxidant Activities of Isolated Compounds from Prosopis africana Leaves. Int. J. Anal. Chem. 2022, 2022, 4205823. [Google Scholar] [CrossRef]
  159. Chaudhary, A.K.; Ahmad, S.; Mazumder, A. Isolation, structural elucidation and in vitro antioxidant activity of compounds from chloroform extract of Cedrus deodara (Roxb.) Loud. Nat. Prod. Res. 2015, 29, 268–273. [Google Scholar] [CrossRef]
Figure 1. The structures of bisabolane-type sesquiterpenes from Artabotrys.
Figure 1. The structures of bisabolane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g001
Figure 2. The structures of norbisabolane-type sesquiterpenes from Artabotrys.
Figure 2. The structures of norbisabolane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g002
Figure 3. The structures of eudesmane-type sesquiterpenes from Artabotrys.
Figure 3. The structures of eudesmane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g003
Figure 4. The structures of guaiane-type sesquiterpenes from Artabotrys.
Figure 4. The structures of guaiane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g004
Figure 5. The structures of eremophilane-type sesquiterpenes from Artabotrys.
Figure 5. The structures of eremophilane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g005
Figure 6. The structures of isodaucane-type sesquiterpenes from Artabotrys.
Figure 6. The structures of isodaucane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g006
Figure 7. The structures of acorane-type sesquiterpenes from Artabotrys.
Figure 7. The structures of acorane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g007
Figure 8. The structures of cadinane-type sesquiterpenes from Artabotrys.
Figure 8. The structures of cadinane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g008
Figure 9. The structures of aristolane-type sesquiterpenes from Artabotrys.
Figure 9. The structures of aristolane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g009
Figure 10. The structures of aromadendrane-type sesquiterpenes from Artabotrys.
Figure 10. The structures of aromadendrane-type sesquiterpenes from Artabotrys.
Molecules 29 01648 g010
Figure 11. The structures of other types of sesquiterpenes from Artabotrys.
Figure 11. The structures of other types of sesquiterpenes from Artabotrys.
Molecules 29 01648 g011
Table 1. Distribution of Artabotrys plant resources.
Table 1. Distribution of Artabotrys plant resources.
No.SpeciesDistribution
1Artabotrys aereus AstVietnam
2Artabotrys antunesii Engl. & DielsAngola
3Artabotrys arachnoides J.SinclairNew Guinea
4Artabotrys atractocarpus I.M.TurnerBorneo
5Artabotrys aurantiacus Engl.Cameroon, Central African Repu, Congo, Gabon, Zaïre
6Artabotrys blumei Hook.f. & ThomsonChina South-Central, China Southeast, Hainan, Vietnam
7Artabotrys brachypetalus Benth.Botswana, Caprivi Strip, Malawi, Mozambique, Northern Provinces, Tanzania, Zambia, Zaïre, Zimbabwe
8Artabotrys brevipes CraibLaos, Thailand
9Artabotrys burmanicus A.DC.Assam, Myanmar
10Artabotrys byrsophyllus I.M.Turner & UtteridgeMalaya, Thailand
11Artabotrys cagayanensis Merr.Philippines
12Artabotrys camptopetalus DielsNew Guinea
13Artabotrys carnosipetalus JessupQueensland
14Artabotrys caudatus Wall. ex Hook.f. & ThomsonAssam, Bangladesh, East Himalaya
15Artabotrys chitkokoi K.Z.Hein, Naive & J.ChenMyanmar
16Artabotrys coccineus KeayNigeria
17Artabotrys collinus Hutch.Tanzania, Zambia
18Artabotrys congolensis De Wild. & T.DurandCameroon, Central African Repu, Congo, Gabon, Zaïre
19Artabotrys costatus KingBorneo, Malaya
20Artabotrys crassifolius Hook.f. & ThomsonMalaya, Myanmar, Thailand
21Artabotrys crassipetalus Pellegr.Gabon
22Artabotrys cumingianus S.VidalPhilippines
23Artabotrys darainensis Deroin & L.Gaut.Madagascar
24Artabotrys dielsianus Le ThomasCameroon
25Artabotrys fragrans Jovet-AstChina South-Central, China Southeast, Vietnam
26Artabotrys gossweileri Baker f.Cabinda
27Artabotrys gracilis KingBorneo, Malaya, Sumatera
28Artabotrys grandifolius KingMalaya, Sumatera
29Artabotrys hainanensis R.E.Fr.China Southeast, Hainan
30Artabotrys harmandii Finet & Gagnep.Cambodia, Laos, Thailand, Vietnam
31Artabotrys hexapetalus (L.f.) BhandariComoros, India, Laos, Sri Lanka
32Artabotrys hienianus BânVietnam
33Artabotrys hildebrandtii O.Hoffm.Madagascar
34Artabotrys hirtipes Ridl.Borneo
35Artabotrys hispidus Sprague & Hutch.Guinea, Ivory Coast, Liberia, Sierra Leone
36Artabotrys inodorus Zipp.New Guinea
37Artabotrys insignis Engl. & DielsBenin, Cameroon, Congo, Gabon, Ghana, Guinea, Ivory Coast, Liberia, Sierra Leone, Zaïre
38Artabotrys insurae Junhao Chen & EiadthongThailand
39Artabotrys jacques-felicis Pellegr.Cameroon, Central African Repu, Zaïre
40Artabotrys javanicus I.M.TurnerJawa
41Artabotrys jollyanus PierreCameroon, Guinea, Ivory Coast, Liberia
42Artabotrys kinabaluensis I.M.TurnerBorneo
43Artabotrys kurzii Hook.f. & ThomsonMyanmar
44Artabotrys lanuginosus Boerl.Borneo, Sulawesi, Sumatera
45Artabotrys lastoursvillensis Pellegr.Gabon, Uganda
46Artabotrys letestui Pellegr.Congo, Gabon
47Artabotrys libericus DielsLiberia
48Artabotrys likimensis De Wild.Burundi, Central African Repu, Kenya, Rwanda, Uganda, Zaïre
49Artabotrys longipetalus Junhao Chen & EiadthongThailand
50Artabotrys longistigmatus NurainasSumatera
51Artabotrys lowianus KingMalaya
52Artabotrys luteus ElmerPhilippines
53Artabotrys luxurians Ghesq. ex Cavaco & Keraudr.Madagascar
54Artabotrys macrophyllus Hook.f.Gulf of Guinea Is.
55Artabotrys macropodus I.M.TurnerBorneo
56Artabotrys madagascariensis Miq.Madagascar
57Artabotrys maingayi Hook.f. & ThomsonBorneo, Malaya
58Artabotrys manoranjanii M.V.Ramana, J.Swamy & K.C.MohanAndaman Is.
59Artabotrys modestus DielsTanzania
60Artabotrys monteiroae Oliv.Angola, Burundi, Ethiopia, Kenya, KwaZulu-Natal, Madagascar, Malawi, Mozambique, Northern Provinces, Rwanda, Sudan, Swaziland, Tanzania, Uganda, Zambia, Zaïre, Zimbabwe
61Artabotrys multiflorus C.E.C.Fisch.China South-Central, China Southeast, Myanmar, Thailand
62Artabotrys nicobarianus D.DasAndaman Is., Nicobar Is.
63Artabotrys oblanceolatus CraibThailand
64Artabotrys oblongus KingCambodia, Malaya
65Artabotrys ochropetalus I.M.TurnerBorneo
66Artabotrys oliganthus Engl. & DielsCameroon, Central African Repu, Gabon, Guinea, Ivory Coast, Liberia
67Artabotrys oxycarpus KingMalaya, Thailand
68Artabotrys pachypetalus B.Xue & Junhao ChenChina Southeast
69Artabotrys pallens AstVietnam
70Artabotrys palustris Louis ex BoutiqueZaïre
71Artabotrys pandanicarpus I.M.TurnerBorneo
72Artabotrys parkinsonii ChatterjeeMyanmar
73Artabotrys petelotii Merr.Laos, Vietnam
74Artabotrys phuongianus BânVietnam
75Artabotrys pierreanus Engl. & DielsCameroon, Congo, Gabon, Zaïre
76Artabotrys pilosus Merr. & ChunChina Southeast, Hainan
77Artabotrys pleurocarpus Maingay ex Hook.f. & ThomsonMalaya, Thailand
78Artabotrys polygynus Miq.Borneo
79Artabotrys porphyrifolius NurainasSumatera
80Artabotrys punctulatus C.Y.WuChina South-Central, Thailand
81Artabotrys rhynchocarpus C.Y.WuChina South-Central, China Southeast
82Artabotrys roseus Boerl.Borneo
83Artabotrys rufus De Wild.Benin, Cameroon, Central African Repu, Congo, Gabon, Nigeria, Togo, Zaïre
84Artabotrys rupestris DielsTanzania
85Artabotrys sahyadricus Robi, K.M.P.Kumar & HareeshIndia
86Artabotrys sarawakensis I.M.TurnerBorneo
87Artabotrys scortechinii KingMalaya
88Artabotrys scytophyllus (Diels) Cavaco & KeraudrenMadagascar
89Artabotrys sericeus Sujana & VadhyarIndia
90Artabotrys siamensis Miq.Myanmar, Thailand
91Artabotrys spathulatus Jun H.Chen, Chalermglin & R.M.K.SaundersThailand
92Artabotrys speciosus Kurz ex Hook.f. & ThomsonAndaman Is.
93Artabotrys spinosus CraibCambodia, Laos, Thailand, Vietnam
94Artabotrys suaveolens (Blume) BlumeBorneo, Jawa, Lesser Sunda Is., Malaya, Maluku, Myanmar, New Guinea, Nicobar Is., Philippines, Sulawesi, Sumatera, Thailand, Bangladesh
95Artabotrys sumatranus Miq.Borneo, Jawa, Sumatera
96Artabotrys tanaosriensis Jun H.Chen, Chalermglin & R.M.K.SaundersThailand
97Artabotrys taynguyenensis BânVietnam
98Artabotrys tetramerus BânVietnam
99Artabotrys thomsonii Oliv.Cabinda, Cameroon, Central African Repu, Congo, Gabon, Liberia, Nigeria, Zaïre
100Artabotrys tipulifer I.M.Turner & UtteridgeMalaya, Thailand
101Artabotrys tomentosus NurainasSumatera
102Artabotrys uniflorus (Griff.) CraibMyanmar, Thailand
103Artabotrys veldkampii I.M.TurnerBorneo
104Artabotrys velutinus Scott ElliotBenin, Cabinda, Cameroon, Central African Repu, Congo, Gabon, Ghana, Guinea, Guinea-Bissau, Ivory Coast, Liberia, Nigeria, Senegal, Sierra Leone, Uganda, Zaïre
105Artabotrys venustus KingBorneo, Malaya, Sumatera, Thailand
106Artabotrys vidalianus ElmerPhilippines
107Artabotrys vietnamensis BânVietnam
108Artabotrys vinhensis AstVietnam
109Artabotrys wrayi KingMalaya
110Artabotrys zeylanicus Hook.f. & ThomsonIndia, Sri Lanka
Table 2. Bisabolane-type sesquiterpenes from Artabotrys.
Table 2. Bisabolane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
1Yingzhaosu AA. uncinatus[73]
2Yingzhaosu BA. uncinatus[74]
3Yingzhaosu CA. uncinatus[75]
4Yingzhaosu DA. uncinatus[75]
5(4R,10S,11E)-Yingzhaosu FA. hexapetalus[76]
6(4S,10S,11E)-Yingzhaosu FA. hexapetalus[76]
7(1R,2S,3S,4E)-Yingzhaosu GA. hexapetalus[76]
8(1S,2R,3R,4E)-Yingzhaosu GA. hexapetalus[76]
9(4R,8E,11S,12S)-Yingzhaosu HA. hexapetalus[76]
10(4R,8E,11R,12R)-Yingzhaosu HA. hexapetalus[76]
11(4S,8S,10S,11S)-Yingzhaosu IA. hexapetalus[76]
12(4R,8S,10S,11S)-Yingzhaosu IA. hexapetalus[76]
13(4R,8E,11R,12S)-Yingzhaosu JA. hexapetalus[76]
14(4S,8E,11S,12R)-Yingzhaosu JA. hexapetalus[76]
15(4R,8Z,11S,12S)-Yingzhaosu KA. hexapetalus[76]
16(4S,8Z,11R,12R)-Yingzhaosu KA. hexapetalus[76]
17(1S,2R,4R,8S,10E)-Yingzhaosu LA. hexapetalus[76]
18Chlospicate EA. pilosus[77]
19Arbisabol-9-en-7,11-diolA. pilosus[77]
Table 3. Norbisabolane-type sesquiterpenes from Artabotrys.
Table 3. Norbisabolane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
20(1R,2S,4R,8R,10E)-Yingzhaosu MA. hexapetalus[77]
21Blumenol AA. hongkongensis[79]
224,5-Dihydroblumenol AA. hongkongensis[79]
23(6R,9S)-3-Oxo-α-ionolA. hongkongensis[79]
243-Hydroxy-β-iononeA. hongkongensis[79]
25DehydrovomifoliolA. hongkongensis[79]
26(3R,6R,7E)-3-Hydroxy-4,7-Megastigmadien-9-oneA. hongkongensis[79]
27Sarmentol FA. hongkongensis[79]
Table 4. Eudesmane-type sesquiterpenes from Artabotrys.
Table 4. Eudesmane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
281α-Hydroxy-5,11-eudesmadieneA. hongkongensis[85]
295-Eudesmene-1β,4α-diolA. hongkongensis[85]
301β,11-Dihydroxy-5-eudesmeneA. hongkongensis[85]
311β-Hydroxy-11-methoxy-5-eudesmeneA. hongkongensis[85]
322α-Hydroxy pterodontic acidA. hongkongensis[85]
331β,9β-Dihydroxy-4aH-eudesma-5,11(13)-Dien-12-oic acidA. hongkongensis[85]
341β,3α-Dihydroxyeudesma-5,11(13)-Dien-12-oic acidA. hongkongensis[85]
35CryptomeridiolA. hainanensis[86]
364,10-Epi-5β-hydroxydihydroeiidesmolA. hainanensis[86]
37Eudesm-4(14)-ene-3α,11-diolA. hainanensis[86]
38OplodiolA. hainanensis[86]
39β-EudesmolA. hainanensis
A. hongkongensis
[86]
[79]
40Trans-3β-(1-hydroxy-1-methylethyl)-8αβ-methyl-5-methylenedecalin-2-oneA. hongkongensis[79]
411β,6α-Dihydroxy-4α (15)-EpoxyeudesmaneA. pilosus[77]
427-Trinoreudesma-4(15),8-dien-1β-ol-7-oneA. pilosus[77]
Table 5. Guaiane-type sesquiterpenes from Artabotrys.
Table 5. Guaiane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
43Liguducin AA. hainanensis[86]
44AlpinenoneA. hainanensis[86]
45GuaianediolA. pilosus[77]
46Guaiane pogostol O-methyl etherA. stenopetalus[89]
47AlismolA. hainanensis[91]
48AlismoxideA. hainanensis[86]
Table 6. Eremophilane-type sesquiterpenes from Artabotrys.
Table 6. Eremophilane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
49FukinoneA. hongkongensis[79]
50PetasitoloneA. hongkongensis[79]
5111-Hydroxy-valenc-1(10)-en-2-oneA. hainanensis[86]
Table 7. Isodaucane-type sesquiterpenes from Artabotrys.
Table 7. Isodaucane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
5210-Oxo-isodauc-3-en-15-oic acidA. hongkongensis[79]
53ArtabotrolA. stenopetalus[89]
Table 8. Acorane-type sesquiterpenes from Artabotrys.
Table 8. Acorane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
54(3R,4S,8R,12R)-Yingzhaosu EA. hexapetalus[76]
55(3S,4R,8S,12S)-Yingzhaosu EA. hexapetalus[76]
Table 9. Cadinane-type sesquiterpenes from Artabotrys.
Table 9. Cadinane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
5610β,15-Hydroxy-α-cadinolA. pilosus
A. hainanensis
[77]
[86]
57Amorph-4-en-10α-olA. hainanensis[86]
5815-Hydroxy-t-muurololA. pilosus[77]
5910α-Hydroxycadin-4-en-15-alA. pilosus[77]
Table 10. Aristolane-type sesquiterpenes from Artabotrys.
Table 10. Aristolane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
6010-Hydroxyaristolan-9-oneA. uncinatus
A. hongkongensis
[107]
[79]
61Aristol-8-en-1-oneA. hongkongensis[79]
62Aristolan-9-en-1-oneA. hongkongensis[79]
63Aristolan-1,9-dieneA. hongkongensis[79]
Table 11. Aromadendrane-type sesquiterpenes from Artabotrys.
Table 11. Aromadendrane-type sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
64SpathulenolA. hainanensis[86]
65(-)-Ent-4β-hydroxy-10α-MethoxyaromadendraneA. uncinatus[107]
66GlobulolA. hexapetalus[110]
67β-GurjuneneA. hexapetalus[110]
Table 12. Other types of sesquiterpenes from Artabotrys.
Table 12. Other types of sesquiterpenes from Artabotrys.
No.Name of CompoundSourceReference
68β-Caryophyllene oxideA. stenopetalus[89]
694-Hydroxy-4,7-dimethyl-1-tetraloneA. pilosus
A. hainanensis
[77]
[86]
70Oxyphyllone DA. pilosus[77]
711β-Hydroxy-4(15),5E,10(14)-germacratrieneA.hainanensis[86]
72Artahongkongol AA. hongkongensis[85]
73Clovane-2β,9α-diolA. hainanensis[91]
74TricyclohumuladiolA. hainanensis[91]
751-Methoxy-9-caryolanolA. uncinatus[107]
76Caryolane-1,9β-diolA. uncinatus[107]
77Litseachromolaevane AA. hainanensis[86]
78DihydroactinidiolideA. hainanensis[86]
7910β-Hydroxyisodauc-6-en-14-alA. pilosus
A. hainanensis
[77]
[86]
80Homalomenol CA. hainanensis[86]
81(4R,5R,7R)-1(10)-spirovetiven-11-ol-2-oneA. hainanensis[86]
82(2R,4S,8S,10R)-Artaboterpenoid AA. hexapetalus[32]
83(−)-8R-Artaboterpenoid BA. hexapetalus[32]
84(+)-8S-Artaboterpenoid BA. hexapetalus[32]
85JunipediolA. hainanensis[91]
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

Sun, Y.; Xin, J.; Xu, Y.; Wang, X.; Zhao, F.; Niu, C.; Liu, S. Research Progress on Sesquiterpene Compounds from Artabotrys Plants of Annonaceae. Molecules 2024, 29, 1648. https://doi.org/10.3390/molecules29071648

AMA Style

Sun Y, Xin J, Xu Y, Wang X, Zhao F, Niu C, Liu S. Research Progress on Sesquiterpene Compounds from Artabotrys Plants of Annonaceae. Molecules. 2024; 29(7):1648. https://doi.org/10.3390/molecules29071648

Chicago/Turabian Style

Sun, Yupei, Jianzeng Xin, Yaxi Xu, Xuyan Wang, Feng Zhao, Changshan Niu, and Sheng Liu. 2024. "Research Progress on Sesquiterpene Compounds from Artabotrys Plants of Annonaceae" Molecules 29, no. 7: 1648. https://doi.org/10.3390/molecules29071648

Article Metrics

Back to TopTop