Open access peer-reviewed chapter

Diterpenes from Different Fungal Sources and Their 13C-NMR Data

Written By

Fozia Farhat, Arneeb Tariq, Annam Zikrea and Riffat Nasim Fatima

Submitted: 12 March 2018 Reviewed: 29 May 2018 Published: 07 November 2018

DOI: 10.5772/intechopen.79186

From the Edited Volume

Terpenes and Terpenoids

Edited by Shagufta Perveen and Areej Al-Taweel

Chapter metrics overview

1,831 Chapter Downloads

View Full Metrics

Abstract

Diterpenes are one of the classes of natural products with about 7000 structures. The basic skeleton of diterpene contains 20 carbon atoms. Microbes contain a large number of diterpenoid with many oxidized carbons and nitrogen atoms. To date, a number of secondary metabolites have been isolated from fungal sources, and some of these examples showed diverse structural features and interesting biological activities. These classes of compounds have attracted the interest of natural product scientist due to their potential biological activities. This chapter includes recently (2013–2018) isolated compounds from various fungal sources especially cythane, clerodanes, halimanes, abietane, and indole-type diterpenes. Biosynthetic pathway of plants and fungi diterpenes showed homology at initial steps but showed differences at latter steps. The biological activity and 13C-NMR data of these recently isolated compounds have been discussed. These diterpenes exhibited potential nitric oxide, anticancer, antioxidant, and antitumor properties. The diterpenes are clerodane, labdane, and kaurane derivatives. A brief discussion on the 13C-NMR chemical shifts of these diterpenes has been discussed at the end of each type.

Keywords

  • fungal
  • biosynthesis
  • diterpenes
  • biological activities
  • 13C-NMR

1. Introduction

Terpenoids comprise the largest, structurally most diverse family of natural products and play important roles in all living organisms [1, 2]. Fungi (Ascomycota and Basidiomycota) are prolific producers of structurally diverse terpenoid compounds. Classes of terpenoids identified in fungi include the sesqui-, di-, and triterpenoids.

As the largest group of documented natural products, terpenoids have attracted attention from a broad scientific community and have been heavily investigated due to their interesting structural characteristics and profound biological effects [3, 4, 5, 6].

Fungi are important source of potential bioactive compounds which play an important role in pharmacology industry [7, 8, 9, 10, 11]. Among fungi, mushrooms are the most attractive sources of bioactive compounds both of chemical and biomedical interests. Approximately 2000 mushrooms are safe for human consumption, and about 650 of them have medicinal properties out of 15,000 documented species of mushrooms [12]. These are also important in industrial processes to enhance composition of bioactive compounds in fermented grain assays [13, 14, 15].

Advertisement

2. Diterpene biosynthesis

Diterpenoid biosynthesis has been studied in plants, bacteria, and fungi [16]; still lots of work are required to clone many important respective genes to characterize and engineer diterpenoid pathways in these representative organisms which remain a big challenge [17]. Fungal di-TPS enzymes show homology to plant enzymes in terms of its size and the combination of biochemical studies with molecular genetics. This also facilitated the comparison of plant and fungal biochemical pathways leading to the formation of gibberellins in plants and fungi [18].

The first committed step in diterpenoid biosynthesis is the cyclization of GGPP to produce the diterpene scaffold, which occurs via a carbocation cascade. Classically, activation of the carbocation cascade by terpene synthases corresponds to the removal of the pyrophosphate group from the linear substrate. This ionization-dependent reaction is catalyzed by class I terpene synthases [19]. Fusicoccanes are potent phytotoxins known to be synthesized by a few fungal species. P. amygdali was the first monofunctional diterpene synthase cloned and characterized in E. coli [20, 21]. Diterpenoids cyclized by the first, one-step route involves a monofunctional class I diterpene synthase that catalyzes ionization-dependent diphosphate cleavage and subsequent carbocation migration and quenching using a mechanism similar to sesquiterpene synthases, except the prenyl chain is now longer by one isoprene unit [22].

Biosynthesis of labdane-type diterpenoids requires a two-step cyclization pathway involving first a protonation dependent cyclization of GGPP to form the characteristic labdane bicycle and, in the second step, ionization-dependent cyclization at a separate active site to generate the final cyclic product (Figure 1). Cyclization of GGPP to ent-CDP and then to the tetracyclic ent-kaurene generates the precursor for gibberellin (gibberellic acids, GA) phytohormones that are major regulators of plant growth and development. It is believed that because of its essential role in plants, ent-kaurene represents the ancestral diterpenoid cyclization pathway from which alternative cyclization routes evolved to generate the large diversity of labdane-type compounds known today [22]. In fact, it has been shown that single amino acid changes are sufficient to alter the product profile of the class I ent-kaurene synthase to form new cyclic scaffolds [23, 24].

Figure 1.

Overview of diterpenoid biosynthesis: (a) monofunctional-2,10(14)-diene is modified into different fusicoccane compounds and (b) bifunctional diterpene synthases make different labdane-related scaffolds that are modified into bioactive compounds.

Advertisement

3. Diterpenoid classification

3.1. Bicyclic diterpenoids

3.1.1. Clerodane diterpenes

Clerodane diterpenes are the natural group of secondary metabolites holding an utmost pharmacological significance. These are bicyclic structures consisting of a fused ring (decalin moiety from C-1 to C-10) with a six-carbon side chain (C-11 to C-16) attached at C-9. The rest of the carbons (C-17 to C-20) are bonded at C-8, C-4, C-5, and C-9, correspondingly [25]. Only 25% of the clerodane diterpenes showed 5:10 cis ring junction, while the rest possess 5:10 trans ring fusion as presented here in the form of columbin and clerodin, respectively. Columbin exhibited dose-dependent anti-inflammatory activity as well as chemopreventive activity against colorectal cancer [26, 27, 28]. During the last 25 years, over 1300 diterpenoids and nor-diterpenoids with the clerodane carbon skeleton have been isolated [29, 30]. The detailed classification of clerodane diterpenes is given in Figure 2.

Figure 2.

Clerodane skeleton, cis and trans structures of clerodane.

3.1.1.1. Clerodane diterpenes by biotransformation from endophytic fungi

Three strains of endophytic fungi L. gonubiensis, N. ribis, and P. stromaticum produced one known and five unknown compounds (B1–B4) through a process of biotransformation, while compounds Q1–Q2 are derived as chemical derivatization of compound 2 [30]. These compounds were actually isolated for the first time from Croton argyrophylloides (Euphorbiaceae) and further biotransformed by Cunninghamella echinulata and Rhizopus stolonifer fungi and produced a new diterpene, as previously described by Monte et al. [31] and Mafezoli et al. [32] (Figure 3).

Figure 3.

Chemical structures of the metabolites by and chemical derivatives of the (3R,4S,5S,8S,9R,10S)-3,12-dioxo-15,16-epoxy-4-hydroxycleroda-13(16),14-diene (compound 1) [35].

3.1.1.2. 13C-NMR date

13C-NMR spectra of substrate 1 and B2 suggested the C-7 hydroxylation making the signal at δ 70.1 (CH) in B2. Compound B2 is identified as new metabolite (4S,5S,7R,8R,9S,10S)-4,7-dihydroxy-15,16-epoxy-3,12-dioxocleroda-13(16),14-diene, and its molecular formula C20H28O5 is sorted by HRMS. Compound B3 was unique for cultures of N. ribis. The 13C-NMR spectrum of B3 showed the presence of one at δ 72.2 in the spectrum confirmed that compound 1 was regioselectively bioreduced at C-3. The new compound B3 was named (3R,4S,5S,7R,8S,9R,10S)-3,4,7-trihydroxy-15,16-epoxy-12-oxocleroda-13(16),14-diene, which is in agreement with the molecular formula C20H30O5. The biotransformation product B4 was obtained only in the P. stromaticum culture. The 13C-NMR spectrum of B4 showed no reduction of carbonyl group at δ 213.5 (C-3) and the appearance of carbinol methane group at δ 71.8 (C-6). And it is named as (4S,5R,6R,8S,9R,10S)-4,6-dihydroxy-15,16-epoxy-3,12-dioxocleroda-13(16),14-diene, which is in agreement with the molecular formula C20H28O5. The new compound Q1 was named (4S,5S,7R,8R,9S,10S)-7-propionyloxy-4-hydroxy-15,16-epoxy-3,12-dioxocleroda-13(16),14-diene. The new derivative Q2 was named (4S,5S,7R,8R,9S,10S)-7-benzyloxy-4-hydroxy-15,16-epoxy-3,12-dioxocleroda-13(16),14-diene [30].

3.1.1.3. Biological activity

Clerodane diterpenes possessed effective insect antifeeding and related insecticidal properties. There are approximately more than 400 natural and semisynthetic products that have been assayed in the laboratories showing potential antifeedant properties [33].

3.1.2. Labdanes

The labdane-related diterpenoids are a special group, consisting of over 7000 members, which are distinguished by their unique biosynthesis. Gibberellin phytohormones as well as antibiotics such as some of phytoalexins and phytoanticipins fall into this family [34]. Labdanolic acids have been identified as biomarkers for the botanical origin of French ambers [35], while copalic acid and its relatives have been associated with the biological activity of the resins from Copaifera species. The lanceolatins are a group of labdanes and abietanes which were obtained [40] from Cephalotaxus lanceolata (Cephalotaxaceae). Some of the abietanes were described in this chapter as well (Figure 4).

Figure 4.

Skeleton of labdanes.

3.2. Tricyclic diterpenoids

See Figure 5

Figure 5.

Classification of tricyclic abietane diterpenes.

3.2.1. Abietanes

Abietane is a class of diterpenoids with excellent metabolic profile. Compounds of this class showed broad spectrum antiviral, antibacterial, and antifungal activity [36, 37]. Abietane diterpenoids are extracted from few fungal species [38]. The intervention of quinone methides in the antioxidant activity of the phenolic diterpenoids ferruginol and carnosic acid has been discussed. The antifungal activity of some abietic acid esters in the context of their use as wood preservatives and the antiviral activity of podocarpic acid derivatives have been examined. In case of human cells, antiproliferative effect on tumor cells has been reported [39].

About 200 compounds of this family have been identified commonly known as dehydroabietic derivatives (dehydroabietanes) [40] assuming 20-carbon saturated aromatic ring I, abietane as standard (Figure 6).

Figure 6.

Abietane skeleton with some standard compounds [41].

3.2.1.1. Tricyclic abietatrienes

This group of abietane terpenes includes a tricyclic ring, three double bonds on B or C rings. Carboxylic acids are representatives of this group, of which the earliest example is the biologically active dehydroabietic acid (Figure 6), which possess an acid group at C-18 [42].

3.2.1.1.1. Abietic acid

Abietic acid was extensively studied in various organisms for its biological activities. Current data also suggest that abietic acid is an important compound for synthesis of novel metabolites. They contribute to the body of knowledge related to compound 1 and deepen the understanding of the potential and properties of 1 and its derivatives (Figure 7).

Figure 7.

Synthesis of compounds 2–10 [42].

3.2.1.1.2. Dehydroabietic acid

Dehydroabietic acid displays not only antiulcer and antimicrobial properties but also antitumor and anti-inflammatory effects (compound 2). Antimicrobial effects of DHA have been studied, specifically against methicillin-resistant strains of Staphylococcus aureus [43].

Biological activity: It also showed activity against other Gram-positive organisms such as Salmonella sp., Bacillus subtilis, and E. coli [44]. This latter study also described the inhibition of nitric oxide (NO) production by DHA, which was reported by other researchers as well [49]. Kawada et al. [45] have reported in relation with the inhibition of pro-inflammatory cytokines that DHA is useful for treating obesity-related diseases.

3.2.1.1.3. Ferruginol

Ferruginol (abieta-8, 11, 13-triene-12-ol) is the simplest phenolic abietane diterpenoid (3). This abietane occurs in plants belonging to the Podocarpaceae and Lamiaceae families [22].

Biological activity: This diterpene has attracted much attention since it has exhibited important bioactivities, such as antimicrobial [46], miticidal [47], cardioactive [48], and antioxidative [49]. Moreover, it accelerates the gastric ulcer healing process, and such effects have been related with the ability of ferruginol to increase the gastric prostaglandin content in vitro [50, 51, 52].

3.2.1.1.4. Callitrisic acid

Callitrisic acid is a diterpenoid acid contained in the resins of several Callitris species (Cupressaceae). It was simultaneously reported as a new natural product [53, 54]. This acid also occurs in plants of the genus Juniperus and Calceolaria, and it has also been found in the genus Illicium. Recently, a series of related acids to callitrisic acid having a C-19 carboxylic group have been isolated [55].

Biological activity: All these acids demonstrated important antiviral activity and significant anti-inflammatory activity [56].

3.2.2. Abietatriene 20-7 lactones

The abietatriene lactones are a group of compounds which possess an oxygen-containing ring which predominantly is in the form of lactones (i.e., abietatrien-20,7-olides). This group of abietanes are exemplified by carnosol (11,12-dihydroxy-8,11,13-20,7-olide). Carnosol possesses an aromatic C ring, carbon C-20 is a keto group, and carbons C-11 and C-12 are hydroxy groups. This abietane has displayed several biological activities. It displayed antioxidant, antimicrobial, anti-inflammatory, antitumor, and anti-HIV (IC50 = 8.0 μM) properties [57, 58].

3.2.3. Abietatetraenes

The abietatetraenes are a group of compounds which possess a fourth double bond which can be located at different positions. Among the 5,6-dehydro derivatives are coleon C and coleon U and related compounds (Figure 8). These metabolites are common in plants of the genus Coleus (synonym Plectranthus) and have described to possess antitumor, antimicrobial, and antiproliferative activity [59, 60] (Table 1).

Figure 8.

Representative member of tricyclic abietatrienes Callitris species.

Carbon number Compound B2a Compound B3b B4b Q1b Q2b 2b 3b 4b 5b 6a 7b 8b 9b 10b
1 24.6 21.4 23.5 23.5 23.5 14.3 14.2 9.8 14.3 14.3 14.3 14.3 16.3 16.3
2 37.6 30.2 35.8 36.1 36.1 17.1 17.1 14.3 17.1 17 17.1 17.1 18.6 18.6
3 215.7 72.2 213.5 214.4 214.3 18.4 18.1 17.1 18.4 18.4 18.4 18.4 21.1 21.0
4 83.0 79.0 83.0 81.4 81.3 21.0 18.4 18.4 21.0 21.0 21.0 21.0 23.9 23.9
5 46.7 43.1 48.9 45.6 45.6 21.6 19.1 21.0 21.6 21.6 21.6 21.6 25.2 23.9
6 41.7 41.4 71.9 37.3 37.1 22.7 20.9 21.6 22.2 22.6 22.6 22.6 30.0 25.2
7 70.1 70.0 34.4 72.2 77.4 25.4 21.5 22.6 22.6 25.3 25.4 25.5 33.4 30.0
8 45.9 44.9 35.3 41.8 41.8 27.6 22.6 25.4 23.0 27.6 26.3 27.6 37.1 33.4
9 44.0 42.8 42.0 42.7 42.7 34.8 25.5 25.6 25.2 34.8 26.4 34.8 37.2 37.1
10 42.7 42.0 41.2 41.0 40.9 35.0 27.5 27.6 25.4 35.0 26.5 35.1 37.9 37.2
11 48.3 47.4 46.8 47.1 47.0 37.7 31.3 34.8 27.6 37.7 27.6 37.5 45.6 37.9
12 197.4 194.8 196.8 194.5 194.3 38.5 34.7 35.0 34.8 38.1 32.7 38.4 47.4 45.6
13 131.0 129.6 129.6 129.5 129.5 41.7 35.0 37.7 35.0 38.4 33.7 45.9 52.6 47.4
14 109.4 108.8 108.8 108.8 108.8 45.9 37.8 38.4 37.8 45.7 34.6 46.6 54.8 52.6
15 146.1 144.5 144.7 144..6 144.7 46.5 38.4 45.8 38.5 46.5 34.8 51.1 63.6 54.8
16 149.9 147.0 147.1 147.0 147.1 51.1 45.6 46.5 41.8 51.1 35.0 53.0 123.9 63.5
17 12.2 11.8 16.3 11.6 11.6 52.4 46.6 51.1 45.8 52.4 37.7 55.3 124.0 123.9
18 22.2 16.0 22.0 22.0 21.9 120.7 51.0 52.14 46.5 53.4 38.5 63.9 126.8 124.0
19 16.7 16.1 9.4 15.9 19.0 122.6 52.1 53.6 51.1 120.7 40.1 120..5 134.5 126.8
20 19.2 18.8 17.8 19.0 15.8 135.7 57.3 120.6 52.3 122.6 45.8 122.5 145.7 134.5
21 145.3 120.5 122.6 120.7 127.3 46.5 135.7 146.7 145.7
22 170.3 122.6 135.7 122.6 128.8 50.4 145.5 171.1 146.8
23 178.8 135.6 145.3 135.7 129.4 51.1 171.3 171.2
24 145.2 173.4 145.3 135.6 52.4 179.4 179.1
25 172.9 178.2 174.0 136.3 120.6
26 178.2 178.3 145.2 122.6
27 172.6 135.7
28 178.1 145.3
29 174.1
30 178.2
Ref [30] [30] [30] [30] [30] [42] [42] [42] [42] [42] [42] [42] [42] [42]

Table 1.

13C-NMR data of New Clerodane Diterpenes from Fungal Biotransformation of the3,12-Dioxo-15,16-Epoxy-4-Hydroxycleroda-13(16),14-Diene and abietane diterpenoids.

13C-NMR at 125 MHz.


13C-NMR at 75 MHz in CDCl3.


3.3. Tetracyclic diterpenoids

3.3.1. Cythane diterpenes

Cyathus is a genus of fungi in the Nidulariaceae, a family collectively known as the bird’s nest fungi. Such compounds are named so, as they resemble tiny bird’s nests filled with “eggs,” structures large enough to have been mistaken in the past for seeds. The first cyathin A3 and allocyathin B3 were reported from fungus C. helenae in 1972, and since then a number of other diterpenes being isolated and documented from different species belong to genus Cyathus [61]. In particular, the species belonging to the genus Cyathus is recognized as prolific producer of bioactive cyathane diterpenoids with inimitable tricyclic ring skeleton [62]. Cyathane diterpenoids also represent a group of natural products with versatility both in structure and bioactivity [63, 64]. Cyathane diterpenes are important bioactive metabolites extracted from the genus Cyathus, Hericium, and Sarcodon. Genus Cyathus is beneficial in producing healthy food and possesses the potential of nitric oxide (NO) inhibition and antibacterial activities [64].

3.3.1.1. Diversity of cythane diterpenes

A number of other biologically significant cyathane diterpenoids have been isolated from the fruiting bodies of mushroom Sarcodon scabrosus [65, 66, 67], Sarcodon glaucopus [68, 69], and Sarcodon cyrneus [70, 71] and the culture of fungi C. helenae [72], C. africanus [73], C. earlei[74], C. striatius [75], Strobilurus tenacellus [76], and Hericium erinaceus [77, 78, 79, 80, 81, 82, 83, 84]. Some cyathane diterpenoids represented interesting and significant biological activities.

3.3.1.1.1. Cythane diterpenes from Cyathus gansuensis

Cyathus gansuensis was reported in 2002 and produced valuable bioactive metabolites from fermented grains of barley and rice [13] by transformation. Recently, seven new [85] metabolites (8–14) named have been isolated from fruiting body of C. gansuensis as presented in Figure 9. The L69 fungal strain was used to isolate these compounds (8–14).

Figure 9.

Representative members of abietatetraenes.

Biological activity: NO inhibition activity was tested on mouse monocyte, macrophages. Seven newly discovered cyathane diterpene derivatives showed inhibitory activity against the NO production in LPS-activated macrophages. The fungus can be a good choice for a transformation on a large scale to acquire enough pure metabolites for the future [85].

13C-NMR structural elucidation: The detail of 13C-NMR is presented in Table 2. 13C-NMR data for compounds 8–14 revealed 20 carbons ascribable for 4 methyls, 4 methylenes (one oxygenated), 4 methines (two oxygenated), two quaternary carbons, and six sp2 carbons. According to NMR and HRTOFMS at m/z 341.2079, [M + Na] + presented molecular formula of 8 and 9 (cyathin J and K) as C20H30O3 (six degrees of unsaturation), 10 (cyathin L) C22H32O5 (seven degrees of unsaturation degrees), 11 (cyathin M) C20H30O5 (six degrees of unsaturation), 12 (cyathin N) C20H28O5 (seven degrees of unsaturation), 13 (cyathin O) C20H30O5 (six degrees of unsaturation), and 14 (cyathin P) C20H28O5 (seven degrees of unsaturation) [85].

Carbon number Compound 8a 9a 10a 11a 12a 13a 14a 15c 16c 17c 18c 19c 20c 21c 22c 23c
1 145.9 145.9 39.8 83.7 84.3 53.4 84.5 38.6 39.9 38.8 34.9 33.8 33.7 83.8 82.1 53.0
2 129.0 129.2 29.4 209.7 209.0 211.1 209.6 29.3 29.7 29.7 23.7 23.2 22.8 209.8 75.3 210.6
3 142.3 142.9 140.4 141.8 140.7 145.0 142.6 140.8 144.9 141.8 79.2 77.4 77.5 141.9 140.7 144.4
4 146.2 147.3 139.2 176.2 177.0 177.1 172.5 140.6 144.0 139.9 78.5 77.0 76.6 176.4 146.7 179.6
5 44.1 41.6 41.7 46.8 142.6 41.0 41.4 38.5 152.7 36.5 36.0 37.6 31.8 47.0 45.8 45.6
6 45.7 44.8 45.1 45.0 49.4 46.5 45.3 47.2 48.6 45.1 46.2 43.6 42.8 45.2 43.4 58.2
7 29.2 34.1 31.0 28.9 29.7 29.5 29.8 35.8 33.8 35.1 33.7 28.0 32.3 29.1 28.9 35.5
8 33.6 34.1 38.4 37.2 35.3 37.3 35.2 36.2 38.0 17.5 31.2 31.7 30.8 37.4 38.1 39.0
9 55.8 55.8 50.6 47.7 47.6 43.0 47.1 51.2 50.3 50.9 43.6 42.6 42.2 47.9 50.9 43.6
10 29.1 38.5 30.5 28.2 124.4 27.6 32.2 30.3 121.3 32.1 24.8 26.2 25.7 28.4 29.2 27.0
11 130.0 72.6 72.0 128.6 118.2 74.6 76.6 160.2 135.8 72.2 159.9 160.5 69.9 128.8 129.8 39.2
12 143.1 147.8 129.7 143.7 149.6 55.9 49.3 144.7 130.1 146.8 145.2 142.4 146.0 143.8 143.3 156.5
13 69.2 125.9 150.5 69.1 71.4 71.8 70.2 78.5 33.5 159.1 78.2 79.5 157.6 69.3 69.1 127.3
14 81.5 76.7 76.2 80.6 81.9 106.4 107.4 75.5 76.5 77.4 74.8 80.2 76.0 80.8 91.3 209.2
15 66.6 64.8 169.4 66.3 63.1 60.6 62.2 196.2 172.1 194.7 196.1 194.2 192.9 66.5 66.6 26.9
16 17.8 17.3 17.2 17.8 24.9 13.6 13.4 17.7 27.0 16.7 18.9 19.7 17.7 18.0 18.0 16.4
17 19.2 19.3 25.1 20.3 21.0 26.3 22.3 24.8 24.2 24.7 19.9 20.0 19.5 20.4 20.6 23.9
18 27.4 27.4 28.4 27.0 26.8 26.3 26.3 28.5 28.2 28.5 29.1 28.1 28.3 27.2 28.4 27.2
19 22.6 23.2 21.9 19.8 20.5 20.7 20.2 21.8 21.9 22.2 19.7 19.4 19.3 20.0 22.5 19.8
20 23.4 23.4 22.4 20.3 21.3 20.8 21.4 22.6 22.0 22.6 19.9 19.6 19.8 21.6 23.2 21.2
21 172.3 28.0 57.3 57.9 59.3 57.1
22 21.1
Ref. [85] [85] [85] [85] [85] [85] [85] [87] [88] [88] [88] [88] [88] [89] [89] [89]
Carbon number Compound 24e 25d 26d 27d 28d 29d 30d 31d 32d 33d 34f 33a
1 213.7 53.1 88.5 47.0 82.8 84.5 90.5 215.4 216.0 53.3 38.5 38.3
2 127.0 211.1 83.5 72.8 37.2 209.5 83.3 125.8 126.4 210.9 28.4 29.1
3 188.5 144.2 140.2 77.8 137.2 141.6 139.6 192.3 193.2 144.9 139.9 140.7
4 84.0 181.7 141.4 78.2 137.8 174.0 140.1 53.3 53.5 177.0 136.6 139.8
5 43.7 43.2 39.0 37.1 41.6 43.3 41.3 38.9 37.5 40.5 40.4 36.1
6 52.9 45.8 56.6 54.6 42.6 44.9 42.8 42.7 41.9 46.0 40.6 44.5
7 33.1 32.0 34.8 34.7 31.0 30.5 30.9 29.7 29.9 29.7 30.4 34.3
8 34.5 39.3 30.2 33.7 36.2 35.3 36.1 30.3 30.8 37.1 37.0 37.1
9 54.0 43.5 48.7 42.6 49.9 47.3 47.2 50.2 50.4 42.9 49.2 50.4
10 34.8 36.6 37.0 32.3 28.3 27.6 28.2 29.9 33.3 31.5 30.1 35.0
11 73.0 71.6 72.4 72.3 80.0 79.9 79.9 74.6 76.9 76.3 72.4 62.1
12 157.5 145.9 157.0 157.3 149.0 149.2 149.2 64.0 49.7 49.8 138.6 148.2
13 122.6 127.3 123.2 123.2 126.6 126.9 126.6 57.0 70.6 70.2 158.2 154.6
14 209.7 76.2 210.6 210.1 111.2 110.8 111.1 104.8 108.2 107.4 85.4 85.3
15 64.4 65.0 64.5 64.4 58.9 58.9 58.9 59.1 62.1 62.0 192.9 194.2
16 17.4 17.1 15.6 17.9 12.0 12.2 12.2 14.6 16.1 13.2 16.4 16.4
17 14.5 24.3 23.6 20.7 17.3 22.4 19.6 20.5 22.4 26.0 24.5 24.7
18 31.2 26.7 28.2 29.8 27.5 26.3 27.2 33.0 33.0 26.2 27.0 27.7
19 22.2 21.0 24.3 20.5 21.2 20.2 19.3 23.4 21.0 20.5 21.5 22.0
20 25.3 20.1 19.6 20.4 22.6 21.2 24.6 21.0 23.3 20.6 21.8 22.3
21 56.6 ***
1’ 105.3 106.3
2’ 73.5 74.6
3’ 69.6 70.3
4’ 75.5 73.3
5’ 65.1 65.0
Ref. [90] [90] [90] [90] [90] [90] [90] [90] [90] [90] [96] [96]

Table 2.

13C-NMR data of cyathane diterpenoids.

13C NMR at 125 MHz.


13C NMR at 75 MHz in CDCl3.


13C NMR at 150 MHz CD3OD.


13C-NMR spectroscopic data for compounds 24–33 in MeOH at 200 MHz.


13C-NMR at 125 MHz.


13C-NMR 175 MHz in acetone-d6.


3.3.1.1.2. Cythane diterpenes from Cyathus africanus

Cyathus africanus is a medicinal basidiomycete fungus. Diterpenes have been reported to possess multiple bioactivities consisting of antimicrobial and anti-inflammatory properties [86]. The presently reported metabolites in this text are collected by the study of various scientists. Moreover, they have been characterized on the basis of their structural elucidation by spectroscopic methods and discussed in detail in this chapter (Table 2). Some of the new metabolites documented by various scientists are isolated from C. africanus. Cyathin Q (15) an important metabolite (Figure 10) showed autophagy-dependent apoptosis [87]. The gene sequence of this strain has also been reported and submitted to GenBank with an accession numbers JX103204. Sequences of analysis exhibited 100% homology with that of fungus C. africanus. Compounds 16–20 (D–H) structurally represented new group of metabolites, while neosarcodonin O (21), cyathatriol (22), and 11-O-acetylcyathatriol (23) are also known cyathane diterpenes. Five novel compounds are isolated from C. africanus and show potential NO inhibition and cytotoxicity against HeLa cell line in vitro [88]. The structural elucidation is also described in Figure 11.

Figure 10.

Newly (8–14) isolated metabolites from C. gansuensis [85].

Figure 11.

Structures of cyathane Q (15) isolated from C. africanus [87].

Ten new polyoxygenated cyathane diterpenoids, named as neocyathins (24–33), together with four known diterpenes are isolated from fungus Cyathus africanus (Figure 12). These compounds were isolated and identified by 13C-NMR technique [90] (Figure 13).

Figure 12.

Structures of metabolites isolated from C. africanus [88, 89].

Figure 13.

Structures of metabolites isolated from C. africanus [90].

Biological activity: Diterpenes with diverse bioactivities have been identified from plants and fungi [91]. Cyathin Q has the capacity to induce the apoptosis in HCT116 cells in a time- and dose-dependent manner. It was observed, when HCT116 cells exposed to 10 mM cyathin Q for 24 h exhibited apoptotic cells 82.07% [87]. This compound induced hallmarks of apoptotic events in HCT116 cells, including caspase activation, cytochrome c release, poly (ADP-ribose) polymerase (PARP) cleavage, and depolarization of the mitochondrial inner transmembrane potential. Nitric oxide has the capacity to react with aqueous oxygen, superoxide, and transition metals like iron or zinc-sulfur clusters, and overproduction of NO is involved in many pathogenic diseases, including inflammation and cancer. The inhibition of NO overproduction in cells may prevent the occurrence of inflammatory diseases and cancer. The inhibition capacity (IC50) was more pronounced for 16, 17, and 19 by exhibiting NO inhibition 79.44, 89.2 and 84.33% reduction, respectively [91]. Moreover, inhibition of NO is concentration dependent as compounds 16–23 showed no NO inhibition at concentration 100 μM [88, 89]. COX-2 and iNOS are two major inflammatory mediators in brain neurodegeneration [92, 93]. Compounds isolated from C. africanus [90] showed strong COX-2 and iNOS capacities. Western blot analysis demonstrated that compounds 24 and 28 significantly suppressed LPS-induced COX-2 expression, whereas compounds 27, 28, 30, 31, and 33 markedly inhibited LPS-induced iNOS expression. Among these compounds, 28 showed strong inhibitory effects on both COX-2 and iNOS. Interestingly, 30 abolished LPS-induced iNOS expression but did not affect LPS-induced COX-2 expression. In addition, we also assayed the activities of iNOS enzyme [90].

13C-NMR structural elucidation: The 13C-NMR spectrum of some of the compounds isolated from C. africanus presented in Table 2.

3.3.1.1.3. Cythane diterpene from Hericium erinaceus and H. flagellum

Hericium genus is among the most blessed medicinal and eatable mushrooms and known to produce secondary metabolites with the potential to treat neurodegenerative diseases. It enables improvement of many brain-related disorders [94]. In this regard, neurotrophins are nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) involved in survival, maintenance, and regeneration of specific neuronal populations in the adult brain [95]. Therefore, the metabolites extracted from Hericium are important source of metabolites and source as remedy in the fight against neurodegenerative diseases such as Parkinson’s, Alzheimer’s, and Huntington’s diseases, which are accompanied by decreased neurotrophic factor expression [102]. Two new potential metabolites have been isolated from H. erinaceus (strain STMA 06157B) and H. flagellum (strain CBS 103681) [96] (Figure 14).

Figure 14.

Structural elucidation of metabolites isolated from H. erinaceus and H. flagellum [96].

Biological activity and 13C-NMR analysis: All of the metabolites isolated from H. erinaceus and H. flagellum exhibited strong neutrotrophin capacity [95, 96]. Metabolites were also studied through 13C-NMR; compound 34 exhibited the presence of five non-proton-bearing carbons, including three olefinic (δC 139.9, 136.6, 138.6) and two aliphatic carbons (δC 40.6, 49.2). Furthermore, five methylene groups with corresponding carbons between δC 28.4 and 38.5 ppm, a further oxygenated methylene group at δC 65.1, vicinal to two aliphatic methines at δC 40.4, and six methines at δC 69.6–105.3 ppm were observed. 13C shifts and correlations of the HSQC-DEPT spectrum showed high similarity to 35 which was a derivative of the cyathane diterpenoid 34. The major difference between the two compound spectra was the missing methoxy group at C-11 in 35 (Figure 13). The detail of 13C-NMR data is described in Table 2.

3.3.2. Indole diterpenes

Indole diterpenes are the broad class of secondary metabolites with enormous structural and functional diversity. They mostly occur in filamentous fungal members having most abundance in Penicillium, Aspergillus, Neotyphodium, and Claviceps [97, 98]. This class of diterpenes is generally divided into two main groups, paxilline type and non-paxilline type [98], though it mainly consists of cyclic diterpenoid backbone in addition to an indole moiety.

3.3.2.1. Diversity of indole diterpenes

3.3.2.1.1. Indole diterpenes from Aspergillus nidulans

The marine fungi A. nidulans was reported to be the source of 19-hydroxypenitrem A (1) and 19-hydroxypenitrem E (2). The 13C-NMR spectrum of 19-hydroxypenitrem A (C37H44ClNO7) provided 37 resonance states from 5 methyl, 8 methylene (with 2 sp2 terminal), 1 sp2 and 7 sp3 methines (with 5 oxygenated), and 16 quaternary (with 5 oxygenated sp3 and 9 sp2) carbon atoms. In comparison, 19-hydroxypenitrem E (C37H45NO7) lack chlorine atom but have one additional hydrogen atom [98] (Figure 15).

Figure 15.

Indole diterpenes from A. nidulans [98].

3.3.2.1.2. Drechmeria sp.: a rich source of indole diterpenes

An endophytic fungi Drechmeria sp. was found to be the reservoir of diverse indole diterpenes including drechmerin A (38), drechmerin B (39), drechmerin C (40), drechmerin D (41), drechmerin E (42), drechmerin F (43), drechmerin G (44), terpendole A (45), terpendole C (46), terpendole I (47), and dehydroxypaxilline (48) [99](Figure 16).

Figure 16.

Indole diterpenes from (1–11) Drechmeria sp. [99].

The 13C-NMR spectrum of drechmerin exhibited 28 carbon resonances, comprising 8 aromatic carbons, 4 oxygenated carbons, 6 methylene carbons, 2 methine carbons, 3 quaternary carbons, and 5 methyl carbons [100]. The detail of 13C-NMR data is given in Table 3.

Carbon Number Compound 36g 37g 38c 39c 40c 41c 42c 43c 44c
2 152.2 151.3 152.6 152.2 152.3 153.8 153.8 153.5 150.7
3 116.7 116.4 54.7 53.8 53.9 52.1 52.1 51.8 51.9
4 132.7 131.0 41.1 40.9 40.9 43.9 43.9 43.7 46.6
5 123.4 125.9 34.0 34.1 34.1 27.5 27.5 27.4 33.2
6 122.6 118.9 26.5 26.4 26.5 29.8 29.8 29.5 32.3
7 110.4 110.3 79.3 77.7 77.9 73.1 73.1 73.0 84.5
8 120.3 121.1 *** *** *** *** *** *** ***
9 138.5 138.9 80.6 79.9 78.4 72.7 42.7 77.7 88.6
10 33.7 37.1 31.0 32.5 32.7 72.9 72.7 68.7 74.1
11 148.7 150.0 71.1 68.3 68.5 61.4 61.4 65.0 175.8
12 45.4 45.7 42.0 53.6 53.6 68.9 69.0 70.7 212.8
13 23.9 24.0 39.2 41.6 41.6 78.8 78.9 78.6 52.2
14 52.9 53.0 22.9 24.9 24.8 30.6 30.6 30.8 23.8
15 80.3 80.4 25.7 26.3 26.3 22.0 22.0 22.0 24.7
16 74.6 74.6 50.4 50.5 50.5 51.6 51.6 51.7 50.5
17 28.6 28.4 28.4 28.2 28.3 30.7 28.2
18 79.5 79.8 118.0 118.1 118.1 117.2 117.3 116.7 118.3
19 86.9 87.0 126.4 126.4 126.4 126.5 126.5 126.4 126.3
20 27.7 27.7 118.8 118.9 118.9 118.9 118.9 131.7 119.0
21 23.4 23.4 119.8 119.8 119.8 119.8 119.8 121.0 120.
22 75.9 76.0 120.8 120.9 120.9 120.8 120.8 121.0 121.2
23 64.8 64.8 112.8 112.8 112.8 112.7 112.7 110.8 112.8
24 60.0 60.0 142.2 142.2 142.2 141.9 141.9 142.0 142.2
25 64.7 64.7 15.0 15.0 15.0 16.6 16.6 16.5 15.0
26 73.0 73.0 20.4 17.0 17.1 19.1 19.1 19.0 16.4
27 *** *** 14.1 178.2 178.2 *** *** *** ***
28 69.7 69.8 73.0 72.8 77.7 76.2 76.1 73.7 16.4
29 29.0 29.0 25.5 26.4 23.7 28.7 28.8 27.5 84.0
30 27.9 28.0 25.9 25.5 22.1 17.2 17.1 25.2 27.8
1’ 42.8 42.8 60.1 95.5 96.0 36.4 22.9
2’ 53.8 53.9 123.7 77.5 77.6 78.4
3’ 106.8 105.3 136.3 77.9 77.8 79.0
4’ 19.3 19.3 18.2 22.6 22.4 22.0
5’ 29.0 29.0 26.1 21.3 21.7 20.6
36 19.8 19.8
37 142.5 142.5
38 110.7 110.7
39 19.9 19.8
40 16.9 17.1
Ref. [98] [98] [99] [99] [99] [99] [99] [99] [99]

Table 3.

13C-NMR data of indole diterpenoids.

13C-NMR at a 125 MHz.


13C-NMR at 75 MHz in CDCl3.


13C-NMR at 150 MHz CD3OD.


13C-NMR at 125 MHz for 13C, measured in DMSO-d6.


3.3.2.1.3. Indole diterpenes from marine A. flavus

The marine Aspergillus flavus had provided 4b-deoxy-β-aflatrem (1),9-isopentenyl paxilline (2), 6,8-di-O-methylcitreoisocoumarin (3), β-aflatrem (4), and paspaline (5). 4b-Deoxy-β-aflatrem (C32H39NO3) exhibits 14 degrees of unsaturation and consists of an indole chromophore and a carbonyl group. As per 13C-NMR spectrum, the respective structure owns resemblance to β-aflatrem, except that a methine replaced an oxygenated quaternary carbon, thereby resulting in an isopentenylated indole diterpenoid. Moreover, 9-isopentenyl paxilline (C32H39NO4) comprised hexacyclic indole diterpenoid skeleton [100] (Figure 17).

Figure 17.

Skeleton by NMR indole diterpenes from A. flavus [100].

3.3.2.1.4. Penitrem D from Penicillium crustosum

Penitrem D (C37H45NO4) was first isolated from P. crustosum in 1983. It is a complex structure with 9 rings, an indole core, and 11 stereocenters [101] (Figure 18).

Figure 18.

Structure and compound isolated from P. crustosum [102].

3.3.2.1.5. Emindole SB from Emericella striata

The mycelium of E. striata was reported to naturally produce emindole SB (C28H39NO). In its structure an indole unit fused to a tricyclic carbon scaffold, and it presented six stereocenters, including vicinal quaternary centers on the western cyclohexyl ring [103] (Figure 19).

Figure 19.

Structure and compound isolated from E. striata [103].

3.3.2.1.6. Paspaline obtained from Claviceps paspali

The ergot fungus Claviceps paspali was found to be the source of paspaline (C28H39NO2) in 1966. The structure owes similarity with emindole SB but contains one more ring comparatively [104] (Figure 20).

Figure 20.

Structure and compound isolate from Claviceps paspali [104].

3.3.2.2. Biological activity

The indole diterpenes, famously called tremorgenic mycotoxins, put forward promising insecticidal potential via regulation of their glutamategated chloride ion channels [105], antibiotic activity [107, 108], antiproliferative against human breast cancer cells [109], and antifungal efficacy [110].

References

  1. 1. Dairi T. Studies on biosynthetic genes and enzymes of isoprenoids produced by actinomycetes. The Journal of Antibiotics. 2005;58:227-243
  2. 2. Daum M, Herrmann S, Wilkinson B, Bechthold A. Genes and enzymes involved in bacterial isoprenoid biosynthesis. Current Opinion in Chemical Biology. 2009;13:180-188
  3. 3. Fraga BM. Natural Sesquiterpenes. Natural Product Reports. 2011;28:1580-1610
  4. 4. Xiao W, Li R, Huang S, Pu J, Sun H. Terpenoids from Schisandraceae family. Natural Product Reports. 2008;25:871-891
  5. 5. Geris R, Simpson TJ. Meroterpenoids produced by fungi. Natural Product Reports. 2009;26:1063-1094
  6. 6. Sun H, Li X, Meng L, Cui C, Gao S, Li C, et al. Asperolides A-C tetranorlabdane diterpenoids from marine algae- derived endophytic fungus Aspergillus wentii EN-48. Journal of Natural Products. 2012;75:148-152
  7. 7. Liang CH, Syu JL, Mau JL. Antioxidant properties of solid-state fermented adlay and rice by Phellinus linteus. Food Chemistry. 2009;116:841-845
  8. 8. Martins S, Mussatto SI, Martinez-Avila G, Montañez-Saenz J, Aguilar CN, Teixeira JA. Bioactive phenolic compounds: Production and extraction by solid-state fermentation. Biotechnology Advances. 2011;29:365-373
  9. 9. Okamura-Matsui T, Izuta H, Tomoda T, Noda H, Fukuda S, Ohsugi M. Fermented soybean with thrombosis preventing activity using mushroom mycelia as microbial source. Food Science and Technology Research. 2003;9:227-230
  10. 10. Zhang GP, Zhang F, Ru WM, Han JR. Solid-state fermentation of cornmeal with the ascomycete Morchella esculenta for degrading starch and upgrading nutritional value. World Journal of Microbiology and Biotechnology. 2010;26:15-20
  11. 11. Zhang ZY, Lei ZF, Lu Y, Lu ZZ, Chen Y, Lei ZF. Chemical composition and bioactivity changes in stale rice after fermentation with Cordyceps sinensis. Journal of Bioscience and Bioengineering. 2008;106:188-193
  12. 12. Rai M, Tidke G, Wasser SP. Therapeutic potential of mushrooms. Natural Product Radiance. 2005;4:246-257
  13. 13. Bao L, Li YX, Wang QX, Han JJ, Yang XL, Li HR, et al. Nutritive and bioactive components in rice fermented with the edible mushroom Pleurotus eryngii. Mycology. 2013;4: 96-102
  14. 14. Li YX, Bao L, Song B, Han JJ, Li HR, Zhao F, et al. A new benzoquinone and a new benzofuran from the edible mushroom Neolentinus lepideus and their inhibitory activity in NO production inhibition assay. Food Chemistry. 2013;141:1614-1618
  15. 15. Wang YQ, Bao L, Yang XL, Li L, Li SF, Gao H, et al. Bioactive sesquiterpenoids from the solid culture of the edible mushroom Flammulina velutipes growing on cooked rice. Food Chemistry. 2012;132(3):1346-1353
  16. 16. Joseph-Nathan Mulia P, Cuevas G, Quijano L. Journal of Natural Products. 2015;78:2580-2587
  17. 17. Tudznski B. Gibberellin biosynthesis in fungi: Genes, enzymes, evolution, and impact on biotechnology. Applied Microbiology and Biotechnology. 2005;66:597-611
  18. 18. Kirby J, Keasling JD. Biosynthesis of plant isoprenoids: Perspectives for microbial engineering. Annual Review of Plant Biology. 2009;60:335-355
  19. 19. Christianson DW. Chemical Reviews. 2006;106:3412-3442
  20. 20. De Boer AH, de Vries-van Leeuwen IJ. Fusicoccanes: Diterpenes with surprising biological functions. Trends in Plant Science. 2012;17(6):360-368
  21. 21. Toyomasu T et al. Fusicoccins are biosynthesized by an unusual chimera diterpene synthase in fungi. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(9):3084-3088
  22. 22. Peters RJ. Two rings in them all: The labdane-related diterpenoids. Natural Product Reports. 2010;27:1521-1530
  23. 23. Wilderman PR, Peters RJ. A single residue switch converts abietadiene synthase into a pimaradiene specific cyclase. Journal of the American Chemical Society. 2007;129(51):15736-15737
  24. 24. Xu M, Wilderman PR, Peters RJ. Following evolution’s lead to a single residue switch for diterpene synthase product outcome. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(18):7397-7401
  25. 25. Li R, Morris-Natschke SL, Lee KH. Clerodane diterpenes: Sources, structures and biological activities. Natural Product Reports. 2016;33:1166-1226
  26. 26. Monteiro AF, Batista JM, Machado MA, Severino RP, Bianah EW, Bolzani VS, et al. Diterpenoids of terrestrial origin. Journal of Natural Products. 2015;78:1451-1455
  27. 27. Wu H, Wang S, Xu Z, Sun S, Liu H, Wang J, et al. Two new clerodane diterpene from Tinospora sagitatta. Molecules. 2015;20:839-845
  28. 28. Annan K, Ekuadzi E, Asare C, Sarpong K, Pistorius D, Oberer L, et al. Phytochemistry Letters. Claerodane diterpene from Polyalthia logifolia (Sonn). 2015;11:28-31
  29. 29. Nohra YA, Perrichot V, Jeanneau L, LePolles L, Azar D. Chemical characterization and botanical origin of French ambers. Journal of Natural Products. 2015;78:1284-1293
  30. 30. Vasconcelos DHP, Barbosa FG, Oliveira NDCF, Lima MAS, Freire FDCO, et al. New clerodane diterpenes from fungal biotransformation of the 3,12-dioxo-15,16-epoxy-4-hydroxycleroda-13(16),14-diene. MOJ Bioorganic and Organic Chemistry. 2017;1(6):00036. DOI: 10.15406/mojboc.2017.01.00036
  31. 31. Monte FJQ, Dantas EMG, Braz-Filho R. New diterpenoids from croton argyrophylloides. Phytochemistry. 1988;27(10):3209-3212
  32. 32. Mafezoli J, Oliveira MC, Paiva JR, Sousa AH, Lima MA, et al. Stereo and regioselective microbial reduction of the clerodane diterpene 3,12-dioxo-15,16-epoxy-4-hydroxycleroda-13(16),14-diene. Natural Product Communications. 2014;9(6):759-762
  33. 33. Gebbinck K, Klein EA, Jansen BJM, de Groot A. Phytochemistry. 2002;61:737-770
  34. 34. Demetzos C, Konstantinos SD. Labdane type diterpenes: Chemistry and biological activity. Studies in Natural Products Chemistry. 2001;25:235-292
  35. 35. de Vargas FS, de Almeida PDO, Aranha ESP, de Boleti AP, Newton P, de Vasconcelios MC, et al. Biological activities and cytotoxicity of diterpenes from Copaifera spp. Oleorasins. Molecules. 2015;20:6194-6210
  36. 36. Leandro LF, Cardoso MJO, Silva SDC, Souza MGM, Veneziani RCS, Ambrosio SR, et al. Antibacterial activity of pinus elloitti and its major compounds, dehydroabietic acid against multidrug resistant strain. Medical Microbiology. 2014;63:1649
  37. 37. da Silva SD, Mendes de Souza MG, Oliveira Cardoso MJ, da Silva Moraes T, Ambrósio SR, Sola Veneziani RC, et al. Antibacterial activity of pinus elliotti against anaerobic bacteria. Anaerobe. 2014;30:146
  38. 38. Manner S, Vahermo M, Skogman ME, Krogerus S, Vuorela PM, Yli-Kauhaluoma J, et al. New derivatives of dehydroabietic acid target planktonic and biofilm bacteria in staphylococcus aureus and effectively disrupt bacterial membrane integrity. The European Journal of Medicinal Chemistry. 2015;102:68
  39. 39. Burgstahler AW, Marx JN. Digenesis of Tricyclic diterpenes. The Journal of Organic Chemistry. 1969;34:1562-1566
  40. 40. Gonzalez JN, Perez-Guaita D, Correa-Royero J, Zapata B, Agudelo L, Mesa-Arango M, et al. Synthesis and biological evolution of of novel Dehydoabietic acid dericative conjugated with acyl thiourea peptide moiety as antitumor agent. European Journal of Medicinal Chemistry. 2010;45:811-816
  41. 41. Gonzalez MA. Aromatic abietane diterpenoids: Their biological activity and synthesis. Natural Product Reports. 2012:1-17
  42. 42. Helfenstein A, Vahermo M, Nawrot DA, Demirci F, Gökalp I, Krogerus S, et al. Antibacterial profiling of abietane-type diterpenoids. Bioorganic and Medicinal Chemistry. 2017;25:132-137
  43. 43. Fallarero A, Skogman M, Kujala J, Rajaratnam M, Moreira VM, Yli-Kauhaluoma J, et al. The International Journal of Molecular Sciences. 2013;14:12054-12072
  44. 44. Jang HJ, Yang KS. Archives of Pharmacal Research. 2011;34:913-917
  45. 45. Kawada T, Hirai S, Goto T, Kuroyanagi K, Kim Y, Ohyama K, et al. BioFactors. 2009;35:442-448
  46. 46. Muhammad I, Mossa JS, El-Feraly FS. Phytotherapy Research. 1992;6:261-264
  47. 47. Chang S, Chen P, Wang S, Wu H. The Journal of Medical Entomology. 2001;38:455-457
  48. 48. Ulubelen A, Birman H, Oksuz S, Topcu G, Kolak U, Barla A, et al. Planta Medica. 2002;68:818-821
  49. 49. Ono M, Yamamoto M, Masuoka C, Ito Y, Yamashita M, Nohara T. Journal of Natural Products. 1999;62:1532-1537
  50. 50. Areche C, Theoduloz C, Yanez T, Souza-Brito ARM, Barbastefano V, de Paula D, et al. Journal of Pharmacy and Pharmacology. 2008;60:245-251
  51. 51. Iwamoto M, Minami T, Tokuda H, Ohtsu H, Tanaka R. Planta Medica, 2003;69:69-72
  52. 52. Bispo de Jesus M, Zambuzzi WF, Ruela de Sousa RR, Areche C, Santos de Souza AC, Aoyama H, et al. Biochimie. 2008;90:843-854
  53. 53. Gough LJ. Tetrahedron Letters. 1968;3:295-298
  54. 54. Carman RM, Deeth HC. The Australian Journal of Chemistry. 1967;20:2789-2793
  55. 55. Zhang GJ, Li YH, Jiang JD, Yu SS, Qu J, Ma SG, et al. Tetrahedron. 2013;69:1017-1023
  56. 56. Wang P, Deng G, Yuan W, Su Z. Bioorganic and Medicinal Chemistry Letters. 2013;23:6682-6687
  57. 57. Aruoma OI, Spencer JPE, Rossi R, Aeschbach R, Khan A, Mahmood N, et al. Food and Chemical Toxicology. 1996;34:449-456
  58. 58. Horiuchi K, Shiota S, Kuroda T, Hatano T, Yoshida T, Tsuchiya T. Biological and Pharmaceutical Bulletin. 2007;30:287-290
  59. 59. Xing V, Wu H, Wang X, Huang Y, Li Q, Li C, et al. Journal of Chemotherapy. 2008;20:238-245
  60. 60. Kobayashi K, Nishino C, Tomita H, Fukushima M. Phytochemistry. 1987;26:3175-3179
  61. 61. Ayer WA, Taube H. Metabolites of Cyathus helenae. Cyathin A3 and allocyathin B3, members of a new group of diterpenoids. Tetrahedron Letters. 1972;19:1917-1920
  62. 62. Wright DL, Whitehead CR. Recent progress on the synthesis of cyathane type diterpenes. Organic Preparations and Procedures International. 2000;32:307-331
  63. 63. Krzyczkowshi W, Malinowska E, Herold F. The structure, medicinal properties and biosynthesis of cyathane diterpenoids. Biotechnologia. 2008;1:146-147
  64. 64. Kamo T, Imura Y, Hagio T, Makabe H, Shibata H, Hirota M. Antiinflammatory cyathane diterpenoids from Sarcodon scabrosus. Bioscience, Biotechnology, and Biochemistry. 2004;68:1362-1365
  65. 65. Hirota M, Morimura K, Shibata H. Anti-inflammatory compounds from the bitter mushroom, Sarcodon scabrosus. Bioscience, Biotechnology, and Biochemistry. 2002;66:179-184
  66. 66. Shi XW, Liu L, Gao JM, Zhang AL. Cyathane diterpenes from Chinese mushroom Sarcodon scabrosus and their neurite outgrowth-promoting activity. European Journal of Medicinal Chemistry. 2011;46:3112-3117
  67. 67. Ma BJ, Liu JK. A new bitter diterpenoid from Sarcodon scabrosus. Journal of Basic Microbiology. 2005;45:328-330
  68. 68. Curini M, Maltese F, Marcotullio MC, Menghini L, Pagiotti R, Rosati O, et al. Glaucopines A and B, new cyathane diterpenes from the fruiting bodies of Sarcodon glaucopus. Planta Medica. 2005;71:194-196
  69. 69. Marcotullio MC, Pagiotti R, Campagna V, Maltese F, Fardella G, Altinier G, et al. Glaucopine C, a new diterpene from the fruiting bodies of Sarcodon glaucopus. Natural Product Research. 2006;20:917-921
  70. 70. Marcotullio MC, Pagiotti R, Maltese F, Oball-Mond Mwankie GN, Hoshino T, Obara Y, et al. Cyathane diterpenes from Sarcodon cyrneus and evaluation of their activities of neuritegenesis and nerve growth factor production. Bioorganic and Medicinal Chemistry. 2007;15:2878-2882
  71. 71. Marcotullio MC, Pagiott R, Maltese F, Obara Y, Hoshino T, Nakahata N, et al. Neurite outgrowth activity of cyathane diterpenes from Sarcodon cyrneus, cyrneines A and B. Planta Medica. 2006;72:819-823
  72. 72. Ayer WA, Browne LM, Mercer JR, Taylor DR, Ward DE. Metabolites of bird's nest fungi. Part 8. Some minor metabolites of Cyathus helenae and some correlations among the cyathins. Canadian Journal of Chemistry. 1978;56:717-721
  73. 73. Ayer WA, Yoshida T, van Schie DMJ. Metabolites of bird's nest fungi. Part 9. Diterpenoid metabolites of Cyathus africanus Brodie. The Canadian Journal of Chemistry. 1978;56:2113-2120
  74. 74. Ayer WA, Lee SP. Metabolites of bird's nest fungi. Part 11. Diterpenoid metabolites of Cyathus earlei. Canadian Journal of Chemistry. 1979;57:3332-3337
  75. 75. Hecht HJ. Striatin A, B, and C, novel diterpenoid antibiotics from Cyathus striatus, X-ray crystal structure of striatin A. Journal of the Chemical Society, Chemical Communications. 1978;15:665-666
  76. 76. Shiono Y, Hiramatsu F, Murayama T, Koseki T, Funakoshi T. Two cyathane-type diterpenoids from the liquid culture of Strobilurus tenacellus. Chemistry and Biodiversity. 2008;5:1811-1816
  77. 77. Kawagishi H, Simada A, Hosokawa S, Mori H, Sakamoto H, Ishiguro Y, et al. Erinacines E, F, and G, stimulators of nerve growth factor (NGF)-synthesis, from the mycelia of Hericium erinaceum. Tetrahedron Letters. 1996;37:7399-7402
  78. 78. Kawagishi H, Simada A, Shizuki K, Mori H, Sakamoto H, Furukawa S. Erinacine D, a stimulator of NGF-synthesis, from the mycelia of Hericium erinaceum. Heterocyclic Communications. 1996;2:51-54
  79. 79. Kawagishi H, Shimada A, Shirai R, Okamoto K, Ojima F, Sakamoto H, et al. Erinacines A, B and C, strong stimulators of nerve growth factor (NGF)-synthesis, from the mycelia of Hericium erinaceum. Tetrahedron Letters. 1994;35:1569-1572
  80. 80. Kawagishi H, Masui A, Tokuyamab S, Nakamura T. Erinacines J and K from the mycelia of Hericium erinaceum. Tetrahedron. 2006;62:8463-8466
  81. 81. Lee EW, Shizuki K, Hosokawa S, Suzuki M, Suganuma H, Inakuma T, et al. Two novel diterpenoids, erinacines H and I from the mycelia of Hericium erinaceum. Bioscience, Biotechnology, and Biochemistry. 2000;64:2402-2405
  82. 82. Kenmoku H, Sassa T, Kato N. Isolation of erinacine P, a new parental metabolite of cyathane-xylosides, from Hericium erinaceum and its biomimetic conversion into erinacines A and B. Tetrahedron Letters. 2000;41:4389-4393
  83. 83. Kenmoku H, Shimai T, Toyomasu T, Kato N, Sassa T. Erinacine Q, a new erinacine from Hericium erinaceum, and its biosynthetic route to erinacine C in the basidiomycete. Bioscience, Biotechnology, and Biochemistry. 2002;66:571-575
  84. 84. Kenmoku H, Kato N, Shimada M, Omoto M, Mori A, Mituhashi W, et al. Isolation of (−)-cyatha-3,12-diene, a common biosynthetic intermediate of cyathane diterpenoids, from an erinacine-producing basidiomycete, Hericium erinaceum. Tetrahedron Letters. 2001;42:7439-7422
  85. 85. Wang B, Han J, Xu W, Chen Y, Liu H. Production of bioactive cyathane diterpenes by a bird’s nest fungus Cyathus gansuensis growing on cooked rice. Food Chemistry. 2014;152(2014):169-176
  86. 86. Xu J, Jin DQ, Song H, Guo YQ, He YS. Lathyrane diterpenes from Euphorbia prolifera and their inhibitory activities on LPS-induced NO production. Fitoterapia. 2012;83:1205-1209
  87. 87. He L, Han J, Li B, Huang L, Ma K, Chen Q, et al. Identification of a new cyathane diterpene that induces mitochondrial and autophagy-dependent apoptosis and shows a potent in vivo anticolorectal cancer activity. European Journal of Medicinal Chemistry. 2016;111:183-192
  88. 88. Han JJ, Zhang L, Xu JK, Bao L, Zhao F, Chen YH, et al. Three new cyathane diterpenoids from the medicinal fungus Cyathus africanus. Journal of Asian Natural Products Research. 2013;17:541-549
  89. 89. Han J, Chen YH, Bao L, Yang XL, Liu D, et al. Anti-inflammatory and cytotoxic cyathane diterpenoids from the medicinal fungus Cyathus africanus. Fitoterapia. 2015;84:22-31
  90. 90. Wei J, Cheng Y, Guo WH, Wang DC, Zhang Q, Li D, et al. Molecular diversity and potential anti-neuroinflammatory activities of cyathane diterpenoids from the basidiomycete Cyathus africanus. Scientific Reports. 2017;21:8883. DOI: 10.1038/s41598-017-09118-z
  91. 91. Han J, Chen Y, Bao L, Yang X, Liu D, Li S, et al. Anti-inflammatory and cytotoxic cyathane diterpenoids from the medicinal fungus Cyathus africanus. Fitoterapia. 2013;84:22-31
  92. 92. Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism. Progress in Neurobiology. 2005;76:77-98. DOI: 10.1016/j.pneurobio.2005.06.004
  93. 93. Giovannini MG et al. Experimental brain inflammation and neurodegeneration as model of Alzheimer’s disease: Protective effects of selective COX-2 inhibitors. International Journal of Immunopathology and Pharmacology. 2003;16:31-40
  94. 94. Young BM. Antiplasmodial activity: The first proof of inhibition of hemecrystallization by marine isonitriles. European Journal of Medicinal Chemistry. 2015;93(2015):373-380
  95. 95. Allen SJ, Watson JJ, Shoemark DK, Barua NU, Patel NK. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacology and Therapeutics. 2013;138:155-175
  96. 96. Rupcic Z, Rascher M, Kanaki S, Köster RW, Stadler M, Wittstein K. Two new cyathane diterpenoids from mycelial cultures of the medicinal mushroom Hericium erinaceus and the rare species, Hericium flagellum. The International Journal of Molecular Sciences. 2018;19:740. DOI: 10.3390/ijms19030740
  97. 97. Saikia S, Nicholson MJ, Young C, Parker EJ, Scott B. The genetic basis for indole-diterpene chemical diversity in filamentous fungi. Mycological Research. 2008;112:184-199
  98. 98. Zhang P, Li XM, Li X, Wanga BG. New indole-diterpenoids from the algal-associated fungus Aspergillus nidulans. Phytochemistry Letters. 2015;12:182-185
  99. 99. Zhao JC, Wang YL, Zhang TY, Chen ZJ, Yang TM, Wu YY, et al. Indole diterpenoids from the endophytic fungus Drechmeria sp. as natural antimicrobial agents. Phytochemistry. 2018;148:21-28
  100. 100. Sun K, Li Y, Guo L, Wang Y, Liu P, Zhu W. Indole diterpenoids and isocoumarin from the fungus, Aspergillus flavus, isolated from the prawn, Penaeus vannamei. Marine Drugs. 2014;12:3970-3981
  101. 101. Sallam AA, Houssen WE, Gissendanner CR, Orabi KY, Foudah AI, El-Sayed KA. Bioguided discovery and pharmacophore modeling of the mycotoxic indole diterpene alkaloids penitrems as breast cancer proliferation, migration, and invasion inhibitors. Medicinal Chemistry Communications. 2013;4(10):1360-1369
  102. 102. Calvo AM, Cary JW. Association of fungal secondary metabolism and sclerotial biology. Frontiers in Microbiology. 2015;6:62
  103. 103. Tomoda H, Tabata N, Yang DJ, Takayanagi H, Omura S. Terpendoles, novel ACAT inhibitors produced by Albophoma yamanashiensis. III. Production, isolation and structure elucidation of new components. The Journal of Antibiotics. 1995;48(8):793-804
  104. 104. Jesus AE, Gorst-Allman CP, Steyn PS, Heerden FR, Vleggaar R, Wessels PL, et al. Tremorgenic mycotoxins from Penicillium crustosum: Biosynthesis of penitrem A. Journal of the Chemical Society, Perkin Transactions. 1983;1:1863-1868
  105. 105. George DT, Kuenstner EJ, Pronin SV. A concise approach to Paxilline indole diterpenes. Journal of the American Chemical Society. 2015;137(49):15410-15413
  106. 106. Smith III AB, Visnick M. An expedient synthesis of substituted indoles. Tetrahedron Letters. 1985;26(32):3757-3760
  107. 107. Smith MM, Warren VA, Thomas BS, Brochu RM, Ertel EA, Rohrer S, et al. Nodulisporic acid opens insect glutamate-gated chloride channels: Identification of a new high affinity modulator. Biochemistry. 2000;39:5543-5554
  108. 108. Dowd PF, Cole RJ, Vesonder RF. Toxicity of selected tremorgenic mycotoxins and related compounds to Spodoptera frugiperda and Heliothis zea. The Journal of Antibiotics. 1988;41:1868-1872
  109. 109. Ondeyka JG, Helms GL, Hensens OD, Goetz MA, Zink DL, Tsipouras A, et al. Nodulisporic acid A, a novel and potent insecticide from a Nodulisporium sp. Isolation, structure determination and chemical transformations. Journal of the American Chemical Society. 1997;119:8809-8816
  110. 110. Shen JW, Ruan Y, Ma BJ. Diterpenoids of macromycetes. Journal of Basic Microbiology. 2009;49(3):242-255

Written By

Fozia Farhat, Arneeb Tariq, Annam Zikrea and Riffat Nasim Fatima

Submitted: 12 March 2018 Reviewed: 29 May 2018 Published: 07 November 2018