814
Views
0
CrossRef citations to date
0
Altmetric
Review Article

Paecilomyces cicadae: a systematic overview of the biological activities and potential mechanisms of its active metabolites

, , , , &
Article: 2243059 | Received 22 Mar 2023, Accepted 19 Jul 2023, Published online: 11 Aug 2023

ABSTRACT

The microbe Paecilomyces cicadae (Miquel) Samson (P. cicadae) is an asexual strain of Cordyceps cicadae (C. cicadae) used in traditional Chinese medicine. Due to the abundance of secondary metabolites found in cultured metabolites, including polysaccharides, hyaluronic acid, ergosterol peroxide, and active nucleosides, P. cicadae and/or C. cicadae exhibit several therapeutic activities, including immunoregulatory, antioxidant, and anti-diabetic effects. Given the great attention for health care and the desirable medical value of C. cicadae and/or P. cicadae, they are becoming potential alternatives of Cordyceps sinensis (C. sinensis). Although there are many studies on P. cicadae/C. cicadae, most of them are experimental research articles. There are limited reviews to summarize the latest researches on P. cicadae/C. cicadae. This review mainly focuses on the metabolites, physiological activities and mechanisms of action of P. cicadae and/or C. cicadae. This review can serve as a theoretical basis for the more efficient use of this medicinal fungus.

1. Introduction

Paecilomyces cicadae (Miquel) Samson (P. cicadae), as the anamorph stage of Cordyceps cicadae (C. cicadae), is an entomogenous and medicinal fungus. C. cicadae is also known as Isaria cicadae Miquel (I. cicadae), Cordyceps zhejianggensis (C. zhejianggensis). Similar to the well-known Cordyceps sinensis (C. sinensis) that selectively infects Hepialus larvae (Zhu et al., Citation1998a), P. cicadae selectively infects bamboo cicada larvae (Ahn et al., Citation2000; Chandrashekar, Citation2006; Li et al., Citation2021a; Zeng et al., Citation2014). After the infection, its lifecycle prolonged. The complex of insects and fungi is also known as the cicada flower because its top branches “sprout” like flowers. The formation process of cicada flower has been reported widely. According to the reports (Ke & Lee, Citation2019; Yan et al., Citation2014), C. cicadae forms in the manner described below. P. cicadae infect the larvae in the late summer or early autumn, then the bamboo cicada larvae ingest nutrients and proliferate to infect many mycelia. The infected larvae become stiff worms in the winter and eventually branch and blossom at the top of the cicada larvae to form C. cicadae the next spring or summer.

As a traditional Chinese medicine (TCM), C. cicadae has many polysaccharides, cordycepin, cordycepic acid, and other active metabolites (Hur, Citation2008; Li et al., Citation2021a), which have effects on anti-oxidation (Olatunji et al., Citation2016), anti-inflammation (Wang et al., Citation2019a), anti-diabetes (Yang et al., Citation2020b), anticancer (Xie et al., Citation2019), immune regulation (Kuo et al., Citation2003), and other effects. For centuries, it has been used as tonic and folk medicine to treat malaria, cancer, fever, diabetes, chronic renal diseases, heart palpitations, infantile convulsions, and dizziness (Ke & Lee, Citation2019; Citation2015; Kuo et al., Citation2003; Olatunji et al., Citation2016; Xie et al., Citation2019; Yang et al., Citation2020b). Although C. cicadae has been used for over 1600 years, it is not the most popular amongst the Cordyceps family. C. sinensis is the most explored and widely used, followed by Cordyceps militaris (C. militaris) (Chen et al., Citation2018; Olatunji et al., Citation2018; Patterson, Citation2008; Zhao et al., Citation2014). Commercial products have been developed from C. sinensis and C. militaris, such as Cobrin, Ningxinbao, Golden Sun Cordyceps, and C. militaris mycelia powder and capsule (Dong et al., Citation2015). However, the market for C. cicadae derived products in China is still very small (Dong et al., Citation2015).

C. cicadae related researches done over the past two decades have shown that it poses similar biological properties and bioactive compounds as C. sinensis and C. militaris, and suggested that it can be used as an alternative source of Cordyceps (Nxumalo et al., Citation2020).

The permission of C. cicadae to be used or sold as food has obtained in China. The fruiting body of C. cicadae (artificial cultivation) has been recognized by the People's Republic of China's National Health Commission as a new type of food raw material since 7 January 2021. In addition to Cordyceps guangdongensis and Cordyceps militaris, it is the third new food raw material belong to genus Cordyceps. It means that C. cicadae can be added into the ordinary food and sells commonly without the permission of health food or drugs.

However, due to the scarcity of naturally occurring C. cicadae and the rise in C. cicadae demand in recent years, the competitive cultivation and fermentation methods are urgently needed. It is possible to culture C. cicadae fruiting bodies artificially, and it has been discovered that P. cicadae, the species’ anamorph, has good and controllable quality. A foundation for the effective use of this medicinal fungus is provided by reports that the chemical constituents of these artificially cultured P. cicadae are essentially the same as those of natural C. cicadae (Ge et al., Citation2007).

The safety assessment has been drawn close attention by the researchers. The insecticidal and nematicidal beauvericin, a kind of cyclodepsipeptides found in C. cicadae, can induce cytotoxicity and cell apoptosis in a dose-dependent manner (Mallebrera et al., Citation2016). Traditionally, instead of using pure compounds, C. cicadae mycelium or fruiting bodies samples have been tested for safety assessments in rat, rabbit, pig, mouse, and human models (Chen et al., Citation2015; Chen et al., Citation2017a; Fu et al., Citation2021; Horng et al., Citation2021; Hsu & Chen, Citation2021; Li et al., Citation2017a; Tsai et al., Citation2021). Excellent safety was demonstrated by the acute toxicity test, subacute toxicity test, and clinical measurement (Chen, Citation1993; Tsai et al., Citation2021). These data support the applications of cicada fermentation products in functional foods, medicines, or cosmetics.

Although there are many studies on P. cicadae/C. cicadae, most of them are experimental research articles. There are limited reviews to summarize the latest researches on P. cicadae/C. cicadae. This review mainly focuses on the metabolites, physiological activities and mechanisms of action of P. cicadae and/or C. cicadae. We hope to this paper could provide theoretical basis for future researchers. In addition, cicada flowers were also included in the study.

2. Metabolic actives

The studies of fungal metabolic actives have gathered a lot of attention ever since Byerrum first reported the antitumor activity of polysaccharides in mushrooms in the 1960s (Byerrum et al., Citation1957). Numerous pieces of literatures have revealed recently that the metabolic actives of some fungi have hypoglycemic (Dai et al., Citation2015), immunomodulatory (Yu et al., Citation2018), antiviral (Li et al., Citation2021c; Verma, Citation2022; Zhu et al., Citation2020a), anti-inflammatory (Qi et al., Citation2021; Xu & Du, Citation2020), antioxidant (Zhu et al., Citation2018), and antitumor effects (Tian et al., Citation2019; Zhang et al., Citation2021c). The active ingredients of various medicinal fungi are also being used increasingly in medicine, food, biology, cosmetics, and other fields. We summarized some typical outcomes for various polysaccharides, proteins, extracts, and other active substances in fungi ().

Table 1. Functions and mechanisms of active substances in some fungi.

Numerous studies on P. cicadae and/or C. cicadae have revealed that the mycelia, fermentation broths, and fruiting bodies contain polysaccharides, adenosine, cordycepin, ergosterol, essential amino acids, and alkaloids with biological activities such as immune-enhancing, antitumor, anti-inflammatory, antibacterial, and antiviral effects (Chai et al., Citation2007; Kim et al., Citation2011; Kim et al., Citation2013; Li et al., Citation2018; Shen et al., Citation2007; Sun et al., Citation2017; Tian et al., Citation2021a; Wang et al., Citation2012b; Zeng et al., Citation2014; Zhang et al., Citation2017a).

Some typical compounds obtained from P. cicadae or C. cicadae are shown in . We mentioned the following ingredients in this section to thoroughly define the characteristics, structures, functions, and mechanisms.

Figure 1. Chemical structures of compounds extracted from P. cicadae and/or C. cicadae. The active metabolites of P. cicadae/C. cicadae are classified into six major categories, namely polysaccharides, nucleoside compounds, ergosterol and its peroxides, cordycepin acid, myriocin, and other compounds.

2.1. Polysaccharides

A class of polymeric carbohydrates known as polysaccharides is composed of at least 10 monosaccharides that are linked by glycosidic bonds. Polysaccharides have the general formula Cx(H2O)y, where x is often a significant value between 200 and 2500. The general formula can also be written as (C6H10O5) n, where n is usually between 40 and 3000. The repeating units in the polymer backbone are usually six-carbon monosaccharides.

Polysaccharides are one kind of the main active components in mushrooms (Won et al., Citation2011). They are structurally diverse macromolecules that serve a variety of roles in many biological processes. The distinct advantages of polysaccharides are their excellent safety and easy availability. Furthermore, growing evidences have suggested that natural polysaccharides can directly scavenge reactive oxidative free radicals (ROS) (Asker et al., Citation2009), increase cell viabilities (Vinderola et al., Citation2006), and boost immunity in cells (Yamada et al., Citation1997).

In 1982, researchers discovered for the first time the structure of P. cicadae polysaccharides. The galactomannan in P. cicadae was isolated by Japanese researchers (Ukai et al., Citation1983). Its relative molecular weight is 27 000, making it a kind of water-soluble polysaccharides. It is composed of D-mannose and D-galactose in a 4:3 ratio. The P. cicadae polysaccharides were subsequently isolated using a chromatographic column by Ren et al. (Ren et al., Citation2014), who discovered that mannose (43.2%), rhamnose (32.1%), xylose (14.5%), and arabinose (10.2%) made up the majority of its structure. According to Li et al. (Li, He, et al., Citation2016a), the primary structural characteristics of P. cicadae polysaccharides were rhamnose, arabinose, fucose, xylose, mannose, glucose, and galactose with a molar ratio of 0.63:13.1:1.30:21.3:46.5:6.0:10.22. Although different structures were identified, there are three kinds of monosaccharides, mannose, galactose, and arabinose, are usually existed.

Recently, it has been discovered that a significant amount of hyaluronic acid was detected in the fermentation broth of P. cicadae (Tan et al., Citation2007), which has gathered people's attention. It may be another source to obtain hyaluronic acid economically, and conveniently. Hyaluronic acid ((C14H21NO11)n, , 1) is a polymer, non-protein, acidic mucopolysaccharide, whose structural units are acetylglucosamine and glucuronic acid (Chandrashekar, Citation2006; Hedén et al., Citation2020). Hyaluronic hacid exhibits substantial health benefits for the body, such as strong water retention properties. It plays roles in cell proliferation, wound healing, migration, and differentiation, which makes it an important biomedical raw material (Donejko et al., Citation2015; Mandal et al., Citation2018). According to Tan et al. (Citation2007), an optimal fermentation medium composition for hyaluronic acid was obtained, those were 6% lactose, 0.5% beef extract, and yeast extract in a 1:1 and 0.1% magnesium sulfate. The yield of hyaluronic acid could reach up to 11.35 mg/mL. Furthermore, the process of extracting hyaluronic acid from P. cicadae has also been studied (Huang & Tian, Citation2009).

The physical and biological properties of polysaccharides are influenced by many important factors, one of which is molecular weight. It is reported that high-molecular-weight polysaccharides are necessary for their immunomodulatory and antitumor activities (Ren et al., Citation2012; Suarez et al., Citation2006). However, some low-weight molecular polysaccharides can also exhibit strong biological functions (He et al., Citation2020; Li et al., Citation2021b; Yuan et al., Citation2020). C. cicadae polysaccharides have strong antioxidant and hypoglycemic properties with a molecular weight of 60.7 kDa (Tang et al., Citation2022). JCH-3, a relatively low-weight molecular polysaccharide isolated from C. cicadae, had excellent immunoregulatory activities, compared to JCH-1 and JCH-2, according to Xu et al (Xu et al., Citation2020). A similar result was observed when we discussed the anti-inflammatory properties of Ganoderma lucidum (G. lucidum) polysaccharides of low weight (Liu et al., Citation2018).

The biological activities may be affected by the high proportion of monosaccharides in polysaccharides. According to Yang et al. (Citation2020d), The antioxidant activity of the polysaccharides was significantly influenced by the content of uronic acid. In general, neutral polysaccharides are thought to have a higher free radical scavenging capacity than acidic polysaccharides. Another literature reported that C. cicadae polysaccharides with high ratios of uronic acid, arabinose, and galactose were considered to have significant contributions to antioxidant activity (Wang et al., Citation2022).

Furthermore, the composition of monosaccharides in polysaccharides were also closely related to the biological activities (Su & Li, Citation2020; Xu et al., Citation2019; Yiyong & Yiting, Citation2018). Mannose, glucose, and galactose were found to positively impact on the antioxidant activity of cicada polysaccharides (Tang et al., Citation2022). Additionally, polysaccharide conformations are another element that can influence biological activity. According to Surenjav et al., the antitumor biological activity of polysaccharides with a single flexible chain was significantly lower than that of natural triple helix lentinan (Surenjava et al., Citation2006).

It is intriguing and important to study the chemical structure of polysaccharides, the composition of monosaccharides, molar ratio of parent compounds and their biological activities. The other parameters that influence the activities are also eager to be found out.

2.2. Nucleosides

The major nucleosides of P. cicadae are uracil (C4H4N2O2, , 2), uridine (C9H12N2O6, , 3), thymidine (C10H14N2O5, , 5), N6-(2-hydroxyethyl) adenosine (HEA) (C12H17N5O5, , 6), adenosine (C10H13N5O4, , 7) and cordycepin (C10H13N5O3, , 8). Nucleoside analogs share adenosine and cordycepin's structural similarities. The absence of 3’ -hydroxy in cordycepin is the only structural difference between it and adenosine. provides detailed information.

Among them, adenosine is frequently used as a typical compound for determining nucleoside levels since it has a base that contains nitrogen. When adenosine is used as a raw material or precursor in synthesizing cordycepin, it serves as a key quality index for measuring cicada flowers. Here, we discussed the typical nucleosides existed in P. cicadae and/or C. cicadae.

2.2.1. Cordycepin

Cordycepin (C10H13N5O3, , 8) as received a lot of attention for its great therapeutic potential. It is also called 3’-deoxyadenosine (Frederiksen et al., Citation1965), and is an important secondary metabolite in Cordyceps species such as Cordyceps militaris (C. militaris) (Cunningham et al., Citation1950) and Cordyceps sinensis (C. sinensis). Additionally, it is present in the broth and mycelium of P. cicadae (Li et al., Citation2007).

Cordycepin combines with certain adenosine receptor and activates downstream biomarkers to perform biological activities subsequently. According to the reports, Cordycepin can interact with A1, A2A, A2B and A3 adenosine receptors subtypes to exert benefits such as anti-inflammation, anti-virus, cell repair, and protection of liver, kidney, heart, and brain (Cao et al., Citation2017; Chen et al., Citation2014; Ravani et al., Citation2017; Sheth et al., Citation2014). It can also inhibit protein synthesis and cell adhesion by altering intracellular signal transduction (Varani et al., Citation2011).

Previously, the fruiting body of C. militaris was mostly used to extract natural cordycepin. However, the needs of the industry could not be met by this production process because it had complicated cultural conditions and a low cordycepin yield. The chemical production of cordycepin has been reported since the 1960s, giving it a preliminary development in the field (Hansske & Robins, Citation1985; Kwon et al., Citation2003; Ohno et al., Citation1984). Adenosine is typically used as a raw material to undergo a sequence of reactions in acidic or alkaline conditions. However, several chemical methods have limitations that make them unsuitable for industrial production, including high costs, environmental pollution, complex processes, and poor yields. Later studies of biological cordycepin synthesis methods, such as enzymatic production and fermentation, have been reported.

Researchers have made great contributions to improving cordycepin yield in C. militaris, such as spore hybridization and mutagenesis technology (Kang et al., Citation2017; Masuda et al., Citation2014; Wongsa et al., Citation2020). P. cicadae can also be found to obtain cordycepin from the mycelium, with a relatively high content (Li et al., Citation2007). The medium components were closely related to cordycepin yield.

Although the health benefits of cordycepin have been recognized but the commercial products of cordycepin is rare. The fruit bodies or the mycelia of C. militaris have been successfully commercialized for medicinal and health products, and even for direct consumption as edible mushroom in supermarket. Up to 2023, there are 43 health foods made from Cordyceps approved by the Ministry of Health of the People's Republic of China. Among them, there are at least two medicinal products made from C. militaris: C. militaris mycelia powder (Z20030034) and capsule (Z20030035) manufactured by Jilin Zhongsheng Pharmaceutical Co., Ltd. (Dong et al., Citation2015).

Many different biological activities and pharmacological effects of cordycepin, including antibacterial (Jiang et al., Citation2019), antiviral (Ryu et al., Citation2014), antitumor (Khan & Tania, Citation2020), and anti-inflammatory effects (Ko et al., Citation2013; Yoou et al., Citation2017a), have been demonstrated in many investigations. In , we summarized some typical underlying mechanisms of antitumor, antiviral, anti-inflammatory and antioxidant activities reported in literatures.

Figure 2. The antioxidant, antitumor, antiviral and anti-inflammatory effects and their mechanisms of cordycepin (COR). Different models or diseases are listed in each aspect. Some biomarkers with the up arrow are accelerated, while, biomarkers with the down arrow are inhibited. Abbreviations: transforming growth factor-β: TGFβ; Superoxide dismutase: SOD; Catalase: CAT; Glutathione Peroxidase: GPX; glutathione reductase: GR; Glutathione S-transferase: GST; Malondialdehyde: MDA; Aspartate Transaminase: AST; Alanine transaminase: ALT; C/EBP homologous protein: CHOP; Reactive oxygen species: ROS; Nuclear factor-κB: NF-κB; Nuclear factor erythroid 2–related factor 2: Nrf2; Heme oxygenase-1: HO-1; Interleukin-1β: (IL)-1β, - 13, -6; Mouse doubleminute 2 homolog: MDM2; B-cell lymphoma-2: Bcl2.

Figure 2. The antioxidant, antitumor, antiviral and anti-inflammatory effects and their mechanisms of cordycepin (COR). Different models or diseases are listed in each aspect. Some biomarkers with the up arrow are accelerated, while, biomarkers with the down arrow are inhibited. Abbreviations: transforming growth factor-β: TGFβ; Superoxide dismutase: SOD; Catalase: CAT; Glutathione Peroxidase: GPX; glutathione reductase: GR; Glutathione S-transferase: GST; Malondialdehyde: MDA; Aspartate Transaminase: AST; Alanine transaminase: ALT; C/EBP homologous protein: CHOP; Reactive oxygen species: ROS; Nuclear factor-κB: NF-κB; Nuclear factor erythroid 2–related factor 2: Nrf2; Heme oxygenase-1: HO-1; Interleukin-1β: (IL)-1β, - 13, -6; Mouse doubleminute 2 homolog: MDM2; B-cell lymphoma-2: Bcl2.

2.2.2. N6-(2-hydroxyethyl)–adenosine (HEA)

N6-(2-hydroxyethyl)–adenosine (HEA, C12H17N5O5, ) is another significant nucleoside compound that can be found in C. cicadae (Ke & Lee, Citation2016; Ke & Lee, Citation2019) and C. sinensis (Zhang et al., Citation2016).

HEA is a potent antioxidant, anti-inflammatory, and hypoglycemic compound with various therapeutic properties, including anti-diabetes, renal protection, anti-inflammatory, antitumor, and sedative effects (Li et al., Citation2019a; Lu et al., Citation2015; Meng et al., Citation2015; Zheng et al., Citation2018). Studies show that HEA has anti-inflammatory benefits by increasing antioxidant enzyme activity, inhibiting lipid peroxidation, and lowering pro-inflammatory factors like IL-6, IL-1β, TNFα, and NF-kB (Wang et al., Citation2019a). Additionally, HEA can inhibit H2O2-induced cell damage by lowering ROS production to restore or preserve mitochondrial function (Zhang, Wu, et al., Citation2019a). Studies demonstrated that cicada HEA has significant hypoglycemic and hypolipidemic effects on type 2 diabetic rats (Sun et al., Citation2021), and can be used as a supplement to diabetes management and reduce the many diabetic complications in genetically obese mice (Li et al., Citation2019a).

2.3. Ergosterol and its peroxide

The secondary metabolites ergosterol (C28H44O, , 9) and its peroxide, ergosterol peroxide (EP, a C28 sterol, , 10), are both significant secondary metabolites in numerous medicinal mushrooms, apart from P. cicadae and C. cicadae.

Ergosterol is a precursor to vitamin D2 (), which can aid in promoting calcium absorption and digestion in humans (Banlangsawan & Sanoamuang, Citation2016; Chen et al., Citation2022; Jasinghe & Perera, Citation2005). In 1889, ergosterol was first isolated from Xylaria avenae (Weete, Citation1989). Soon after, scientists extracted ergosterol from P. cicadae/C. cicadae and other fungi (Ge et al., Citation2007; Kuo et al., Citation2002a; Kuo et al., Citation2003; Toh Choon et al., Citation2012). Ge et al. reported the yield of ergosterol (0.6256 ± 0.0309 mg/g) from P. cicadae, which was higher than that in natural C. cicadae (0.5743 ± 0.018 mg/g) (Ge et al., Citation2007).

Figure 3. Conversion of ergosterol to vitamin D2.

Figure 3. Conversion of ergosterol to vitamin D2.

Ergosterol has been the subject of many studies, and its biological functions, such as anti-inflammation, antitumor, etc., have gathered a lot of attention. The supplement of ergosterol can been used to prevent vitamin D deficiency and type 2 diabetes (Nutrition et al., Citation2022; Jasinghe & Perera, Citation2005; Banlangsawan & Sanoamuang, Citation2016; Chen et al., Citation2022). The antitumor properties have also been studied in the lung (Wu et al., Citation2018), liver (Lin et al., Citation2017; Sankaran et al., Citation2017), breast (Li et al., Citation2015), stomach (Hu et al., Citation2014b) and prostate (Muñoz-Fonseca et al., Citation2021) cell lines.

EP was reported to have been isolated from a wide variety of fungi, including C. cicadae, G. lucidum, Paecilomyces tenuipes (P. tenuipes), Trichophyton Schoenleini (T. Schoenleini), and Sarcodon aspratus (S. aspratus) (Bauslaugh et al., Citation1964; Chen et al., Citation2009; Nam et al., Citation2001; Takei et al., Citation2005). Nowadays, the fermentation broth is another resource to get EP (He et al., Citation2018).

EP demonstrates physiological activities like immune regulation (Kuo et al., Citation2003), antioxidation (Kim et al., Citation1999), anti-inflammation (Kobori et al., Citation2007), antitumor abilities (Kahlos et al., Citation1989) and antiviral effects (Lindequist et al., Citation1989). Numerous cancer cells, including breast cancer (El-Sherif et al., Citation2020a), prostate cancer (Rhee et al., Citation2012), myeloma (Li, Wu, et al., Citation2016), and renal cell carcinoma (He et al., Citation2018), can be inhibited by EP in vitro. The key antitumor mechanisms are involved in inhibiting cell growth, arresting cell cycle, downregulating oncogene expressions, and upregulating tumor suppressor gene expressions.

2.4. Cordycepic acid

Cordycepic acid (C6H14O6, , 11) is a polyol-structured isomer of quinic acid, also known as 1,3,4,5-tetrahydroxycyclohexanoic acid. Cordycepic acid was initially isolated from C. sinensis in 1957 (Chatterjee et al., Citation1957). The composition of the crystalline substance was determined to be D-mannitol (Chatterjee et al., Citation1957). The content of cordycepic acid is one of the key factors used to evaluate the quality of C. sinensis.

In addition, the fermentation broths, the mycelia, and the fruiting bodies of C. cicadae, P. cicadae, and C. militaris, all contain cordycepic acid (Dong et al., Citation2012; Dou et al., Citation2013; Raethong et al., Citation2020; Shi et al., Citation2022). Xia et al. (Citation2014) discussed the contents of cordycepic acid in the fruiting bodies of different strains, and found that the fruiting body of P. cicadae had a high concentration of cordycepic acid; it may contain up to 78.19 mg/g, which is higher than that in C. sinensis (77.27 mg/g), C. militaris (43.36 mg/g), and natural C. cicadae (53.6 ± 1.16 mg/g). Apart from the different strains, the contents in fruiting bodies also change as progress through their growth stages and are influenced by various fermentation factors, including carbon sources, nitrogen sources, and inorganic salts (Dou et al., Citation2013; Raethong et al., Citation2020; Shi et al., Citation2022). Additionally, the parameters in solid fermentation, such as the medium, light and humidity were also affected cordycepic acid contents (Curran, Citation1980).

Cordycepic acid can be used clinically to treat patients with cerebral ischemia and trauma, such as enhancing cerebral microcirculation and cerebral blood flow (Jiang, Citation1987; Mendelow et al., Citation1985). Besides, cordycepic acid shows a significant lung tumor inhibitory effect by regulating Nrf-2/HO-1/NLRP3/NF-κB signaling pathway (Wang et al., Citation2020).

2.5. Myriocin

Myriocin (C21H39NO6, , 12) has been known as either ISP-1 or thermozymocidin and has been produced from Mycelia sterilia, Isaria sinclairii and C. cicadae (Lee et al., Citation2010). It is a long-chain alpha-amino acid, with unsaturated fatty acids chains in the structure. Its chemical structure was identified as (2S, 3R, 4R, 6E)-2-amino-3,4-dihydroxy-2-hydroxy-methyl-14-oxo-6-eicosenoic acid (St-Jacques, Citation1973). Myriocin can be produced through fermentation because Yang et al. discovered that yeast extract and sucrose are best for its synthesis (Yang et al., Citation2008).

As an immunosuppressive agent, myriocin has the potential to be clinically effective. In vivo and in vitro experiments have revealed that it is 10–100 times more potent than the immunosuppressive agent cyclosporine A (CsA), with fewer adverse effects to the kidneys (Fujita et al., Citation1994). In addition, the antitumor effect of myriocin is proved by Choi et al. (Citation2014). It was shown that ISP-1 can activate DR4 pathway and induce apoptosis of lung cancer cells.

2.5. Other metabolites

Other secondary metabolites, such as amino acids, terpenoids, and flavonoids, can be found in large quantities in the fermentation broth or mycelium of P. cicadae and C. cicadae.

Some cyclodepsipeptides have been identified from P. cicadae and C. cicadae. Kuo et al. (Citation2002a) isolated cyclodepsipeptides with antitumor, anti-convulsant, anti-arrhythmic, and sedative effects from cicada flowers. Later, other cyclodepsipeptides, such as beauvericin (, 19), beauvericin A (, 20), beauvericin B (, 21), beauvericin E (, 22), beauvericin J (, 23) and cordycecin A (, 24) have also been purified (Lin et al., Citation2005; Citation2017; Citation2017; Wang et al., Citation2014a).

Moreover, furan-containing compounds are purified from the flowers of C. cicadae, those are 5-(2-hydroxyethyl) -2-fluoroacetic acid (, 15), 2-(5-(3-hydroxybutyl) furan-2-yl) acetic acid (, 16), methyl-2-(5- (3-hydroxybutyl) furan-2-yl) acetate (, 17), 2-(5-(3-hydroxybutyl) furan-2-yl) acetic acid (, 18), and a-furoic acid (, 25) (Chu et al., Citation2015; Yang et al., Citation2019c; Zhang, Hu, et al., Citation2019b).

Several antibiotic compounds such as cephalosporolide E (, 26), cephalosporolide J (, 27), 3-benzyl-6-isopropyl-2,5-piperazinedione (, 28), and 3-isobutyl-6-isobutylpiperazine-2,5-dione (, 29) were found in the mycelia of the hybridization of C. cicadae and C. militaris (Yang et al., Citation2019c). Among them, 3-benzyl-6-isopropyl-2,5-piperazinedione and alpha-furoic acid were both showed moderate inhibitory activities against Panagrellus redivivus (Yang et al., Citation2019c).

In addition to ergosterol and its peroxides, other sterols have been found, such as 9,11-dehydroergosterol peroxides (, 30), 3β, 5α, 6β- (22E, 24R) -ergosta-7,22-dien-3,5,6-triol (, 31), 3β, 5α, 6β-6-methoxyergosta-(22E, 24R)-7,22-dien-3,5-diol (, 32), 4-hydroxy-17R-methylincisterol (, 33), 5α, 6α-epoxy- (22E, 24R) -ergosta-8, 22-diene-3β, and 7α-diol (, 35), (22E, 24R) -Ergosta-5,7,trien-3-ol(ergosterol) (, 36) (Wang et al., Citation2017a).

Some aromatic compounds have also been isolated from cicadas, such as 5-methoxycinnamic acid 3- (4’-O-methyl-β-glucopyranoside) (, 37), cordycepone (, 38), oosporein (, 39), fumimycin (, 40) and stipitatonic acid (, 34) (Zhang & Xuan, Citation2007; Zhang, Hu, et al., Citation2019b). Among them, oosporein, fumimycin and stipitatonic acid have been reported to have broad-spectrum in vitro antibacterial, antioxidant and cytotoxic activities (Zhang, Hu, et al., Citation2019b).

Researches will be performed to continue to discover the unknown secondary metabolites with pharmacologic effects.

3. Biological activities

Metabolites have been reported to have a wide range of biological activities, such as immunoregulatory, antibacterial, antioxidant, and antitumor activities. In the study, we comprehensively summarized the pharmacological effects and their mechanisms of these metabolites derived from P. cicadae and/or C. cicadae (). Here, we discussed in detail based on each pharmacological effect.

3.1. Immunoregulatory activity

It has been observed that polysaccharides originating from microorganisms or plants can improve immunity by activating macrophages or enhancing the synthesis of cytokines and immunoglobulins (Huang et al., Citation2016; Li et al., Citation2017c; Shen et al., Citation2017). Polysaccharide is one of the most abundant and bioactive components found in mushroom fruit bodies, cultured mycelia, and fermentation broths (Chen et al., Citation2021). It interacts with receptors on the cell surface, affects the expression of some signaling biomarkers, and then increases the production of immune factors or antibodies, so as to play a role in regulating the immune function of the body (Jiang et al., Citation2010).

The polysaccharides from P. cicadae and/or C. cicadae have been reported to have immunomodulatory properties (Jin et al., Citation2008; Tian et al., Citation2022; Xu et al., Citation2018). They can regulate the NF-κB/MAPK signaling pathway to improve cellular immunity (Tian et al., Citation2022) and the TLR4 signaling (Kim et al., Citation2011) pathway to increase dendritic cell maturation. We are all aware that an excessive increase in inflammatory cytokines can cause inflammation, but an increase in cytokines within a reasonable range can stimulate cellular immunity. High doses of polysaccharides have been shown to significantly increase serum levels of TNF-α, IL-1β, IL-6, IL-10, NO, or immunoglobulin A levels, activating the cellular immune response and enhancing cellular immunity (Jin et al., Citation2008; Wang et al., Citation2022; Xu et al., Citation2018).

Similar to the study on the regulation of intestinal flora by polysaccharides from Grifola frondosa (Li et al., Citation2019b), polysaccharides from P. cicadae can regulate the systemic immune response by elevating the relative abundance of beneficial bacteria (Lactobacillus) and the concentration of short-chain fatty acids in the intestine (Wan et al., Citation2022). C. cicadae polysaccharides have been reported to have the abilities in improving metabolic disorders and reducing liver oxidative stress by increasing glutathione peroxidase (GSH-Px), SOD, and catalase (CAT) activities and decreasing lipid peroxidation (MDA) levels (Zhang et al., Citation2021). In addition, P. cicadae polysaccharides can affect cellular proliferative activities and their immune-enhancing activity. showed the general reported mechanisms of immune regulation of C. cicadae and/or P. cicadae in detail.

Figure 4. The general mechanism of immune regulation of C. cicadae and/or P. cicadae. The immunomodulatory effect exerts in two ways. ① C. cicadae and/or P. cicadae polysaccharides can regulate the systemic immune response by increasing the relative abundance of beneficial bacteria (e. g. Lactobacillus) and the concentration of short-chain fatty acids in the intestine; ② C. cicadae polysaccharides can alleviate metabolic disorders and reduce liver oxidative stress by changing the activities of enzymes and reducing the levels of inflammatory factors.

Figure 4. The general mechanism of immune regulation of C. cicadae and/or P. cicadae. The immunomodulatory effect exerts in two ways. ① C. cicadae and/or P. cicadae polysaccharides can regulate the systemic immune response by increasing the relative abundance of beneficial bacteria (e. g. Lactobacillus) and the concentration of short-chain fatty acids in the intestine; ② C. cicadae polysaccharides can alleviate metabolic disorders and reduce liver oxidative stress by changing the activities of enzymes and reducing the levels of inflammatory factors.

As a source of polysaccharides, C. cicadae spores are also being researched. The active ingredients found in C. cicadae spores are typically believed to be nearly identical to those found in C. cicadae mycelia and C. cicadae fermentation broth (Sun et al., Citation2017; Wang et al., Citation2017a). Polysaccharides in spores have also been reported to have immune-enhancing properties (Fu et al., Citation2019; Sun et al., Citation2017; Wang et al., Citation2017c). Similar to lentinan, C. cicadae spore polysaccharides could protect mice given the immunosuppressive drug cyclophosphamide by increasing macrophage phagocytic activity and regulating the secretion of cytokines and immunoglobulins (Zheng et al., Citation2022).

3.2. Antitumor activities

Numerous studies have extensively explored the antitumor properties of P. cicadae and C. cicadae. Several tumor cells were used as the assessment model, including renal cancer cells (He et al., Citation2018), gastric cancer cells (Xie et al., Citation2019), liver cancer cells (Wang et al., Citation2014a), and cervical cancer cells (Xu et al., Citation2021a) to evaluate the mechanisms of the components from P. cicadae or C. cicadae. Several components have been claimed to have antitumor properties, including EP, adenine, uridine, adenosine, HEA, polysaccharides, and certain organic solvent extracts from fruiting bodies, mycelia, and fermentation broth.

How cancer cells are out of the regulation of cell proliferation was the important question to be answered by the researchers. The two most important mechanisms of antitumor action are apoptosis and cell cycle arrest. However, the fundamental driving forces are very diverse depending on the various elements or components.

EP, a secondary metabolite of P. cicadae, was discovered to have antitumor activity by He et al. (Citation2018). It was reported that EP promoted apoptosis, inhibited migration and invasion, blocked the cell cycle, and prevented DNA replication and mitosis (He et al., Citation2018). The ethanol extract of C. cicadae, with adenine, uridine, adenosine, and HEA presented, showed antitumor effect by the mechanism of cell cycle arrest, MMP depolarization, Ca2+ overload, over-expressed Bax, AIF, caspase-8, caspase-6, and caspase-3 and decreased Bcl-2 (Xie et al., Citation2019).

The polysaccharide extracts from C. cicadae spore powders have antitumor properties comparable to those of Ganoderma lucidum spores (Wang et al., Citation2012a; Zhu et al., Citation2012). These extracts can significantly lower the expression of β-catenin and N-cadherin and inhibit Wnt/β-catenin signaling and caspase-mediated mitochondrial apoptosis pathways (Sun et al., Citation2017). Additionally, C. cicadae may also have an impact on G2/M arrest to obtain the antitumor impact. Besides, C. cicadae polysaccharides could upregulate the expression of P53 (Xu et al., Citation2021a) and downregulate the expression of cyclin E, cyclin A, and CDK2, which prevented Hela cells from proliferating.

To maximize the antitumor benefits, some researchers have suggested combining natural elements from P. cicadae and C. cicadae with medicines like cisplatin, adriamycin, and cyclophosphamide (Cai et al., Citation2010; Jin et al., Citation2008). Various outcomes were obtained. Cai et al. (Citation2010) found that the combination of polysaccharides and adenosine had specific antitumor effects in vitro. While the addition of polysaccharides did not strengthen the antitumor activity of adriamycin or cisplatin. Another study (Jin et al., Citation2008) found that combining P. cicadae with cyclophosphamide synergistically enhanced antitumor activity.

These studies have demonstrated the potential for P. cicadae and C. cicadae to serve as sources of natural antitumor drugs. Exploring the synergies of the combination of natural extracts and chemical compounds is a meaningful direction.

3.3. Antioxidant and anti-aging activities

Several mechanisms, such as reducing capacity, inhibiting chain initiation, binding transition metal ion catalysts, peroxides decomposition, and radical scavenging, can be used to explain the antioxidant and anti-aging activities. The cellular assessment of antioxidant activities involves the enzyme capacities (SOD, CAT, GSH-Px, etc.), lipid peroxidation products levels (malondialdehyde, lipofuscin, etc.), and the accumulation of ROS. The antioxidant effect, which has an anti-aging impact, is attained by raising or lowering these levels.

Numerous studies have shown that P. cicadae and/or C. cicada have free radical scavenging abilities (He et al., Citation2010; Ren et al., Citation2014; Wang et al., Citation2019b). According to (Citation2019b), the polysaccharides from C. cicada scavenged hydroxyl radicals at various concentrations in a dose-dependent manner. The significant antioxidant properties of P. cicadae polysaccharides and C. cicadae fruiting body polysaccharides were also demonstrated by assessing DPPH free radical, superoxide radical, and hydroxyl radical scavenging capacities in the tube (Ren et al., Citation2014). He et al. (Citation2010) also documented the ability of P. cicadae polysaccharides to scavenge DPPH, superoxide, and hydroxyl radicals.

The protective and repairing effects on oxidative stress damage models also reflect the antioxidant and anti-aging capacities of the sample. On H2O2-induced oxidative damage in PC12 cells, the protective effect of HEA was reported (Zhang, Wu, et al., Citation2019a). Compared to the damaged model, HEA could boost intracellular antioxidant enzyme activity and cell viability, decrease dehydrogenase release, and limit the ROS production and lipid peroxidation production. In addition, the decrease in inflammatory cytokines such as IL-6, IL-1β, TNF-α, and NF-кB was observed. Animal experiments supported the antioxidant properties of polysaccharides from C. cicadae. It was reported that polysaccharides from C. cicadae spores can boost SOD, CAT, and GSH-Px activities, indicating that they have the antioxidant capacity and hepatoprotective activities (Zhang et al., Citation2021). Another study found that C. cicadae polysaccharides were crucial in prolonging the lifespan of Drosophila, probably because of the upregulated expression genes associated with antioxidants such as CAT and SOD1 (Zhu et al., Citation2020b).

Hyaluronan (HA) in cells is a natural component in the skin that helps absorb moisture from the skin surface. The lack of HA results in skin water loss, sagging, increased wrinkles and decreased elasticity. Shao et al. (Citation2023) found that C. cicadae extract promoted HA synthesis in fibroblasts, and the extract was rich in polysaccharides, and five alditols (mainly mannitol).

In addition, it is reported that C. cicadae and P. cicadae showed potential therapeutic benefits in rats with adenine-induced chronic renal failure, increasing SOD activity and decreasing MDA and GSH (Li et al., Citation2018). Additionally, the serum levels of BUN and Crea were significantly decreased, and TP, ALB, RBC, HCT, and HGB were increased. These results suggested that C. cicadae and P. cicadae can inhibit oxidative stress and enhance antioxidant capacity to exert renal protective effects.

These findings show that P. cicadae and C. cicadae may be investigated as potential natural antioxidants and used as new dietary supplements to delay aging.

3.4. Anti-inflammatory effect

There are lots of factors, such as physical damage, metabolic disorders, infection-related factors, oxidative stress, and chemical substances, can cause inflammation (Salminen et al., Citation2022; Tilg et al., Citation2020). It is also well accepted that inflammation is linked with oxidative stress. Oxidative stress refers to elevated intracellular levels of ROS that is considered to be the most potent inflammatory mediators (Shin et al., Citation2008). Antioxidants play a significant role in reducing inflammation (Waxman K. Citation1996). The increase of antioxidant enzymes in cells can prevent acute and chronic inflammatory reactions, exert anti-inflammatory effects on the eyes, protect the kidneys, and have anti-diabetic properties.

Lipopolysaccharide (LPS)-induced inflammation, in vitro cell lines and in animals, represents a standard paradigm for studying inflammation. The anti-inflammatory mechanisms of the active substances may be involved in the increased expression of antioxidant enzymes, reduced the deposition of peroxide, and the decrease of the secretion of pro-inflammatory factors (). displays the general mechanisms of the anti-inflammatory effects of P. cicadae and/or C. cicadae.

Figure 5. Anti-inflammatory effect of P. cicadae and/or C. cicadae reported in the literatures. LPS-induced damage, oxidative damage and renal inflammation can be suppressed by P. cicadae and/or C. cicadae. Pathways and key hubs are exhibited. Inflammatory factors and antioxidant related enzymes are influenced. Abbreviations: Phosphatase and tensin homolog: PTEN; phosphatidylinositol 3-kinase: P13K; protein kinase B, PKB: AKT; Mechanistic Target Of Rapamycin: mTOR; Recombinant Toll Like Receptor 4: TLR4; Type I; of transforming growth factor β receptor: TβR I; Type II of transforming growth factor β receptor: TβR II;Monoclonal Antibody to Mothers Against Decapentaplegic Homolog 2, 3, 4, 7: Smad2, 3, 4, 7; Reactive oxygen species: ROS; Malondialdehvde: MDA; Matrix metalloproteinase: MMP; Lactic dehydrogenase: LDH; Cyclooxygenase 2: COX-2; Tumor necrosis factor alpha: TNF-α; Prostaglandin E2: PGE2; Superoxide dismutase: SOD; Catalase: CAT; Glutathione: GSH.

Figure 5. Anti-inflammatory effect of P. cicadae and/or C. cicadae reported in the literatures. LPS-induced damage, oxidative damage and renal inflammation can be suppressed by P. cicadae and/or C. cicadae. Pathways and key hubs are exhibited. Inflammatory factors and antioxidant related enzymes are influenced. Abbreviations: Phosphatase and tensin homolog: PTEN; phosphatidylinositol 3-kinase: P13K; protein kinase B, PKB: AKT; Mechanistic Target Of Rapamycin: mTOR; Recombinant Toll Like Receptor 4: TLR4; Type I; of transforming growth factor β receptor: TβR I; Type II of transforming growth factor β receptor: TβR II;Monoclonal Antibody to Mothers Against Decapentaplegic Homolog 2, 3, 4, 7: Smad2, 3, 4, 7; Reactive oxygen species: ROS; Malondialdehvde: MDA; Matrix metalloproteinase: MMP; Lactic dehydrogenase: LDH; Cyclooxygenase 2: COX-2; Tumor necrosis factor alpha: TNF-α; Prostaglandin E2: PGE2; Superoxide dismutase: SOD; Catalase: CAT; Glutathione: GSH.

Table 2. Display the anti-inflammatory mechanisms and pharmacological activities of Cordycepin.

Table 3. Pharmacological effects and their mechanisms of the metabolic actives derived from P. cicadae and/or C. cicadae.

It is reported that some therapeutic anti-inflammatory compounds are derived from C. cicadae and P. cicadae. An excellent anti-inflammatory impact of HEA on human bodies is thought to exist. In vivo and in vitro studies (Lu et al., Citation2015; Wang et al., Citation2019a; Zheng et al., Citation2018) have been performed to prove its anti-inflammatory properties. HEA can increase the activity of antioxidant enzymes in kidney tissues, including glutathione (GSH), catalase (CAT), and SOD. Some pro-inflammatory mediators (TNF-α, IL-6, IL-1β, NF-κB, and TGF-β1) (Wang et al., Citation2019a) can also be decreased in a dose-dependent manner.

Other active compounds in P. cicadae or C. cicadae, such as polysaccharides (Yang et al., Citation2019a; Zhang et al., Citation2021) and cordycepin (Lu et al., Citation2015), have also been demonstrated anti-inflammatory effects. Cordycepin exerts protective effects against inflammatory injury for many diseases including acute lung injury, asthma, rheumatoid arthritis, Parkinson's disease (PD), hepatitis, atherosclerosis, and atopic dermatitis (Ashraf et al., Citation2019; Cheng et al., Citation2011; Citation2020). According to Lu et al. (Citation2015), the inhibition of pro-inflammatory cytokine production by cordycepin in C. cicadae was more efficient than adenosine and HEA in LPS-stimulated RAW 264.7 macrophages.

Additionally, hepatoprotective and renal protective activities of C. cicadae polysaccharides are also partly due to their anti-inflammatory mechanism. It can reduce renal interstitial fibrosis in rats with diabetic nephropathy by reducing inflammation and managing issues with intestinal flora (Yang et al., Citation2020b). C. cicadae polysaccharides can also play roles in the treatment of liver dysfunction by anti-inflammation and against oxidative damage in a rat model of metabolic syndrome induced by a high-sugar and high-fat diet (Zhang et al., Citation2021).

The Toll-like receptor (TLR) signaling pathway plays a key role in inflammation. TLR4 is an important transmembrane pattern-recognition receptor and has been extensively documented in a variety of inflammatory conditions. The stimulation of TLR4 initiates a series of signaling cascades that result in the activation of nuclear factor κB (NF-κB), interferon β regulatory factor 3 (IRF3), and mitogen-activated protein kinases (MAPKs) which mediates the upregulation of inflammatory cytokine gene expression leading to an inflammatory response (Shim et al., Citation2013; Sugiyama et al., Citation2016). HEA can inhibit TLR4-mediated NF-κB signaling pathway to reduce the pro-inflammatory response induced by LPS (Lu et al., Citation2015). HEA also obviously decreased LPS-induced inflammatory cytokine level in RAW 264.7 cells and TGF-β1-induced fibroblast activation in NRK-49F cells by modulating NF-κB and TGF-β1/Smad signaling (Zheng et al., Citation2018).

3.5. Anti-diabetic effect

Diabetes is a global epidemic that caused by either insufficient insulin production from the pancreas (type 1), or when the body cannot effectively utilize the insulin it produces (type 2) (World Health Organization, Citation1999). Drugs that regulate blood sugar and reduce insulin resistance are the mainstays of treatment for diabetes (Evans & Krentz, Citation1999; Guo et al., Citation2016). However, these drugs may cause several adverse reactions, such as hypoglycemia, lactic acidosis, and ketoacidosis (Li et al., Citation2004). The use of natural ingredients is considered an appropriate alternative medicine for treating diabetes and its associated complications (Day, Citation1998).

According to Day (Citation1998), C. cicadae exhibited a hypoglycemic impact in a mouse model of type 1 diabetes induced by alloxan. A recent study has shown that HEA in C. cicadae can improve glucose tolerance and exhibits hypoglycemic effects on type 2 diabetes (Li et al., Citation2019a). A study compared the anti-diabetic benefits of P. cicadae and Astragalus membranaceus, and was reported that P. cicadae was more effective at postponing the onset of diabetic nephropathy than Astragalus membranaceus, in which the PI3K/AKT/mTOR signaling pathway played important roles (Yang et al., Citation2020a).

Researches will be performed continuedly to understand the underlying mechanisms and advance the application.

3.6. Antibacterial activity

Antibiotics will be used to treat bacteria using traditional antibacterial methods, but overuse of these drugs will result in bacterial resistance (Martínez, Citation2012; Yang et al., Citation2020a). Numerous reports have indicated that natural fermentation metabolites have antibacterial activity, including crude proteins and polysaccharides obtained from fungi and traditional Chinese medicines (Keypour et al., Citation2008; Kim et al., Citation2001).

According to the report, the crude proteins in the fermentation broth of P. cicadae can destroy the cell membrane structure of E. coli and alter the composition of entire cells or membrane proteins (Cen et al., Citation2021). Both of the extracellular and intracellular polysaccharides from C. cicadae have been demonstrated to have antibacterial activities. It was discovered that pathogenic microorganisms, including Escherichia coli, Klebsiella pneumoniae, Vivrio choler, Pseudomonas aeruginosa, Vibrio alginolyticus, Staphylococcus aureus, Vibrio parahaemolyticus, and Streptococcus pneumonia were significantly inhibited by both of the extra – and intracellular polysaccharides (Sharma et al., Citation2015). In addition, the inhibition rate of intracellular polysaccharides was marginally higher than that of extracellular polysaccharides.

Cordycepin was reported to have antibacterial activity by Ahn et al. (Citation2000). It can inhibit the growth of Clostridium paraputrificum and Clostridium perfringens, but have no effect on Bifidobacterium spp. and Lactobacillus spp. (Ahn et al., Citation2000).

4. Conclusion and future perspectives

The advantages of low toxicity and simple cultivation make natural P. cicadae – the same genus as C. sinensis – a desirable species. It is expected to serve as a replacement for C. sinensis. Due to the limited natural resources and challenging artificial cultivation of P. cicadae, the use of modern biological fermentation technology to cultivate artificial P. cicadae mycelium has almost become the only and fundamental way to solve the problem.

The review primarily described the main active components, physiological functions, and mechanisms of P. cicadae and C. cicadae. We also provided a scientific summary of their typical therapeutic effects.

Recent studies have demonstrated that its extracts have positive therapeutic and health-promoting effects, particularly in inflammation-mediated diseases like diabetes, kidney disease, obesity, and cancer. We hope this will gives us a theoretical basis to use medicinal fungi more effectively.

Future research will be required to determine the exact functional mechanisms of the active ingredients from P. cicadae and/or C. cicadae. The efficacy and safety of single or combined use still need researchers’ attention. In addition, there is a need to advance the industrialization of active substance preparation.

Author contributions

Conceptualization, C.W.; Data curation and software, M.L.; Project administration and Supervision, L.L.; Investigation and Validation, W.C.; Data curation and Investigation, J.Z.; writing of the manuscript, F.D.; editing, J.Z. All authors have read and agreed to the published version of the manuscript.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data will be made available on request.

References

  • Ahn, Y.-J., Park, S.-J., Lee, S.-G., Shin, S.-C., & Choi, D.-H. (2000). Cordycepin: Selective growth inhibitor derived from liquid culture of cordyceps militaris against Clostridium spp. Journal of Agricultural and Food Chemistry, 48(7), 2744–2748. https://doi.org/10.1021/jf990862n
  • Ashraf, S., Radhi, M., Gowler, P., Burston, J. J., Gandhi, R. D., Thorn, G. J., Piccinini, A. M., Walsh, D. A., Chapman, V., & De Moor, C. H. (2019). The polyadenylation inhibitor cordycepin reduces pain, inflammation and joint pathology in rodent models of osteoarthritis. Scientific Reports, 9(1), 1–17. https://doi.org/10.1038/s41598-018-37186-2
  • Asker, M. M. S., Ahmed, Y. M., & Ramadan, M. F. (2009). Chemical characteristics and antioxidant activity of exopolysaccharide fractions from Microbacterium terregens. Carbohydrate Polymers, 77(3), 563–567. https://doi.org/10.1016/j.carbpol.2009.01.037
  • Banlangsawan, N., & Sanoamuang, N. (2016). Effect of UV-B irradiation on contents of ergosterol, vitamin D2 and other nutrients in Thai edible mushrooms. Journal of the Royal College of Physicians of Edinburgh, 34(3), 237–243.
  • Bauslaugh, G., Just, G., & Blank, F. (1964). Isolation of ergosterol peroxide from Trichophyton schoenleini. Nature, 202(4938), 1218–1218. https://doi.org/10.1038/2021218a0
  • Byerrum, R., Clarke, D. A., Lucas, E., Ringler, R., Stevens, J., & Stock, C. C. (1957). Tumor inhibitors in Boletus edulis and other holobasidiomycetes. Antibiot & Chemother, 7(1), 1–4. https://doi.org/10.1016/B978-1-4831-9801-9.50150-1
  • Cai, J. F., Jiang, Z. M., Hong-Yang, L. U., & Bo-Zheng, L. U. (2010). Research of anti-tumor effects of different purified components of cordyceps cicadae in vitro. Chinese Archives of Traditional Chinese Medicine, 28(4), 360–394. https://doi.org/10.13193/j.archtcm.2010.04.90.caijf.044
  • Cai, Y., Feng, Z., Jia, Q., Guo, J., Zhang, P., Zhao, Q., Wang, Y. X., Liu, Y. N., & Liu, W. J. (2021). Cordyceps cicadae ameliorates renal hypertensive injury and fibrosis through the regulation of SIRT1-mediated autophagy. Frontiers in Pharmacology, 12. https://doi.org/10.3389/fphar.2021.801094
  • Cao, H.-L., Liu, Z.-J., & Chang, Z. (2017). Cordycepin induces apoptosis in human bladder cancer cells via activation of A3 adenosine receptors. Tumor Biology, 39(7), 101042831770691. https://doi.org/10.1177/1010428317706915
  • Carbonero, E. R., Gracher, A. H. P., Komura, D. L., Marcon, R., Freitas, C. S., Baggio, C. H., Santos, A. R. S., Torri, G., Gorin, P. A. J., & Iacomini, M. (2008). Lentinus edodes heterogalactan: Antinociceptive and anti-inflammatory effects. Food Chemistry, 111(3), 531–537. https://doi.org/10.1016/j.foodchem.2008.04.015
  • Cen, Q.-W., Wang, Z.-Y., Tang, Z.-X., Zhang, Y., Chen, T., Xue, D.-W., Xu, M.-F., Bai, X.-L., Zhou, T., & Shi, L.-E. (2021). Initial purification of antimicrobial fermentation metabolites from Paecilomyces cicadae and its antimicrobial mechanism. LWT, 148, 111785. https://doi.org/10.1016/j.lwt.2021.111785
  • Chai, Y.-Q., Jin, Y.-W., Chen, G.-J., Liu, Y.-G., Li, X.-L., & Wang, G.-E. (2007). Separation of insecticidal ingredient of paecilomyces cicadae (miquel) samson. Agricultural Sciences in China, 6(11), 1352–1358. https://doi.org/10.1016/S1671-2927(07)60183-7
  • Chandrashekar, L. (2006). Reply: Hyaluronic acid: A unique topical vehicle for the localized delivery of drugs to the skin. Journal of the European Academy of Dermatology and Venereology, 20(10), 1348–1349. https://doi.org/10.1111/j.1468-3083.2006.01708.x
  • Chatterjee, R., Srinivasan, K., & Maiti, P. (1957). Cordyceps sinensis (Berkeley) saccardo: Structure of cordycepic acid. Journal of the American Pharmaceutical Association, 46(2), 114–118. https://doi.org/10.1002/jps.3030460211
  • Chen, C.-C., Wong, T.-C., Huang, C.-L., Jhou, B.-Y., Hsu, J.-H., Yeh, S.-H., Tsai, Y.-T., & Yang, S.-H. (2017a). Clinical evaluation of blood glucose regulation and safety of cordyceps cicadae mycelium. J Food Nutr Res, 5, 137–143. https://doi.org/10.12691/jfnr-5-2-10
  • Chen, J., Zhang, M., Mujumdar, A. S., & Phuhongsunge, P. (2022). 4D printing induced by microwave and ultrasound for mushroom mixtures: Efficient conversion of ergosterol into vitamin D2. Food Chemistry, 387, 132840. https://doi.org/10.1016/j.foodchem.2022.132840
  • Chen, L., Liu, Y., Guo, Q., Zheng, Q., & Zhang, W. (2018). Metabolomic comparison between wild ophiocordyceps sinensis and artificial cultured Cordyceps militaris. Biomedical Chromatography, 32(9), e4279. https://doi.org/10.1002/bmc.4279
  • Chen, M., Xiao, D., Liu, W., Song, Y., Zou, B., Li, L., Li, P., Cai, Y., Liu, D., & Liao, Q. (2020). Intake of Ganoderma lucidum polysaccharides reverses the disturbed gut microbiota and metabolism in type 2 diabetic rats. International Journal of Biological Macromolecules, 155, 890–902. https://doi.org/10.1016/j.ijbiomac.2019.11.047
  • Chen, S., Yong, T., Zhang, Y., Su, J., Jiao, C., & Xie, Y. (2017b). Anti-tumor and anti-angiogenic ergosterols from Ganoderma lucidum. Frontiers in Chemistry, 5, 85. https://doi.org/10.3389/fchem.2017.00085
  • Chen, Y., Wang, T., Zhang, X., Zhang, F., & Linhardt, R. J. (2021). Structural and immunological studies on the polysaccharide from spores of a medicinal entomogenous fungus paecilomyces cicadae. Carbohydrate Polymers, 254, 117462. https://doi.org/10.1016/j.carbpol.2020.117462
  • Chen, Y., Yang, S.-H., Hueng, D.-Y., Syu, J.-P., Liao, C.-C., & Wu, Y.-C. (2014). Cordycepin induces apoptosis of C6 glioma cells through the adenosine 2A receptor-p53-caspase-7-PARP pathway. Chemico-Biological Interactions, 216, 17–25. https://doi.org/10.1016/j.cbi.2014.03.010
  • Chen, Y.-K., Kuo, Y.-H., Chiang, B.-H., Lo, J.-M., & Sheen, L.-Y. (2009). Cytotoxic activities of 9, 11-dehydroergosterol peroxide and ergosterol peroxide from the fermentation mycelia of Ganoderma lucidum cultivated in the medium containing leguminous plants on Hep 3B cells. Journal of Agricultural and Food Chemistry, 57(13), 5713–5719. https://doi.org/10.1021/jf900581h
  • Chen, Y.-L., Yeh, S.-H., Lin, T.-W., Chen, C.-C., Chen, C.-S., & Kuo, C.-F. (2015). A 90-day subchronic toxicity study of submerged mycelial culture of cordyceps cicadae (ascomycetes) in rats. International Journal of Medicinal Mushrooms, 17(8), 771–781. https://doi.org/10.1615/intjmedmushrooms.v17.i8.70
  • Chen, Z. (1993). Studies on cultivation of paecilomyces cicadae and its pharmacological function. Acta Mycologica Sinica, 12(2), 138–144. https://doi.org/10.13346/j.mycosystema.1993.02.010
  • Cheng, J. W., Wang, Y. B., He, L., Qian, H., Fu, L. Z., & Li, H. B. (2012). Optimization of fermentation process for the production of intracellular polysaccharide from Paecilomyces cicadae and the immuno-stimulating activity of intracellular polysaccharide. World Journal of Microbiology and Biotechnology, 28(12), 3293–3299. https://doi.org/10.1007/s11274-012-1140-0
  • Cheng, Y., Wei, Y., Yang, W., Song, Y., Shang, H., Cai, Y., Wu, Z., & Zhao, W. (2017). Cordycepin confers neuroprotection in mice models of intracerebral hemorrhage via suppressing NLRP3 inflammasome activation. Metabolic Brain Disease, 32(4), 1133–1145. https://doi.org/10.1007/s11011-017-0003-7
  • Cheng, Z., He, W., Zhou, X., Lv, Q., Xu, X., Yang, S., Zhao, C., & Guo, L. (2011). Cordycepin protects against cerebral ischemia/reperfusion injury in vivo and in vitro. European Journal of Pharmacology, 664(1-3), 20–28. https://doi.org/10.1016/j.ejphar.2011.04.052
  • Choi, K. E., Jung, Y. S., Kim, D. H., Song, J. K., & Hong, J. T. (2014). Myriocin induces apoptotic lung cancer cell death via activation of DR4 pathway. Archives of Pharmacal Research, 37(4), 501–511. https://doi.org/10.1007/s12272-013-0315-z
  • Chu, Z. B., Chang, J., Zhu, Y., & Sun, X. (2015). Chemical constituents of cordyceps cicadae. Natural Product Communications, 10, 2145–2146. https://doi.org/10.27465/d.cnki.gzzyc.2016.000162
  • Chyau, C.-C., Wu, H.-L., Peng, C.-C., Huang, S.-H., Chen, C.-C., Chen, C.-H., & Peng, R. Y. (2021). Potential protection effect of er homeostasis of n6-(2-hydroxyethyl) adenosine isolated from cordyceps cicadae in nonsteroidal anti-inflammatory drug-stimulated human proximal tubular cells. International Journal of Molecular Sciences, 22(4), 1577. https://doi.org/10.3390/ijms22041577
  • Cunningham, K., Manson, W., Spring, F., & Hutchinson, S. (1950). Cordycepin, a metabolic product isolated from cultures of Cordyceps militaris (Linn.) Link. Nature, 166(4231), 949–949. https://doi.org/10.1038/166949a0
  • Curran, P. M. (1980). The effect of temperature, p H, light and dark on the growth of fungi from Irish coastal waters. Mycologia, 72(2), 350–358. https://doi.org/10.1080/00275514.1980.12021188
  • Dai, R., Liu, R., & Xiao, J. (2015). Research progress on hypoglycemic effect and its mechanism of action of medicinal fungal polysaccharides. Zhongguo Zhong yao za zhi = Zhongguo Zhongyao Zazhi = China Journal of Chinese Materia Medica, 40, 174–179. https://doi.org/10.4268/cjcmm20150203
  • Day, C. (1998). Traditional plant treatments for diabetes mellitus: Pharmaceutical foods. British Journal of Nutrition, 80(1), 5–6. https://doi.org/10.1017/S0007114598001718
  • Dietologica, M. (2020). Possible therapeutic role of a highly standardized mixture of active compounds derived from cultured Lentinula edodes mycelia (AHCC) in patients infected with 2019 novel coronavirus. J Minerva gastroenterologica e dietologica, 66(12). https://doi.org/10.23736/s1121-421x.20.02697-5
  • Donejko, M., Przylipiak, A., Rysiak, E., Miltyk, W., Galicka, E., Przylipiak, J., Zaręba, I., & Surazynski, A. (2015). Hyaluronic acid abrogates ethanol-dependent inhibition of collagen biosynthesis in cultured human fibroblasts. Drug Design, Development and Therapy, 9, 6225–6233. https://doi.org/10.2147/dddt.s91968
  • Dong, C., Guo, S., Wang, W., & Liu, X. (2015a). Cordyceps industry in China. Mycology, 6(2), 121–129. https://doi.org/10.1080/21501203.2015.1043967
  • Dong, C., Guo, S., Wang, W., & Liu, X. (2015b). Cordyceps industry in China. Mycology, 6(2), 121–129. DOI: 10.1080/21501203.2015.1043967
  • Dong, J. Z., Lei, C., Ai, X. R., & Wang, Y. (2012). Selenium enrichment on Cordyceps militaris link and analysis on its main active components. Applied Biochemistry and Biotechnology, 166(5), 1215–1224. https://doi.org/10.1007/s12010-011-9506-6
  • Dou, Y., Xiao, J.-H., Xia, X.-X., & Zhong, J.-J. (2013). Effect of oxygen supply on biomass and helvolic acid production in submerged fermentation of cordyceps taii. Biochemical Engineering Journal, 81, 73–79. https://doi.org/10.1016/j.bej.2013.10.005
  • El-Sherif, N. F., Ahmed, S. A., Ibrahim, A. K., Habib, E. S., El-Fallal, A. A., El-Sayed, A. K., & Wahba, A. E. (2020a). Ergosterol peroxide from the Egyptian Red lingzhi or reishi mushroom, ganoderma resinaceum (agaricomycetes), showed preferred inhibition of MCF-7 over MDA-MB-231 breast cancer cell lines. International Journal of Medicinal Mushrooms, 22(4), 389–396. https://doi.org/10.1615/IntJMedMushrooms.2020034223
  • Evans, A. J., & Krentz, A. J. (1999). Recent developments and emerging therapies for type 2 diabetes mellitus. Drugs in R & D, 2(2), 75–94. https://doi.org/10.2165/00126839-199902020-00001
  • Fei, X., Zhang, X., Zhang, G. Q., Bao, W. P., Zhang, Y. Y., Zhang, M., & Zhou, X. (2017). Cordycepin inhibits airway remodeling in a rat model of chronic asthma. Biomedicine & Pharmacotherapy, 88, 335–341. https://doi.org/10.1016/j.biopha.2017.01.025
  • Frederiksen, S., Malling, H., & Klenow, H. (1965). Isolation of 3′-deoxyadenosine (cordycepin) from the liquid medium of Cordyceps militaris (L. Ex Fr. Link. Biochimica et Biophysica Acta (BBA)-Nucleic Acids and Protein Synthesis, 95(2), 189–193. https://doi.org/10.1016/0005-2787(65)90483-1
  • Fu, H. I., Hsu, J. H., Li, T. J., Yeh, S. H., & Chen, C. C. (2021). Safety assessment of HEA-enriched cordyceps cicadae mycelia on the central nervous system (CNS), cardiovascular system, and respiratory system in ICR male mice. Food Science & Nutrition, 9(9), 4905–4915. https://doi.org/10.1002/fsn3.2440
  • Fu, Y., Shi, L., & Ding, K. (2019). Structure elucidation and anti-tumor activity in vivo of a polysaccharide from spores of Ganoderma lucidum (Fr.) Karst. International Journal of Biological Macromolecules, 141, 693–699. https://doi.org/10.1016/j.ijbiomac.2019.09.046
  • Fujita, T., Inoue, K., Yamamoto, S., Ikumoto, T., Sasaki, S., & Toyama, R. (1994). Fungal metabolites. Part 11. A potent immunosuppressive activity found in Isaria sinclairii metabolite. The Journal of antibiotics, 47(2), 208–215. https://doi.org/10.7164/antibiotics.47.208
  • Ge, F., Xia, C., Li, C., Ding, T., Shao, Y., & Fan, M. (2007). Analysis of the chemical compositions of paecilomyces cicadae fermented mycelia and cordyceps cicadae fruit body. Mycosystema, 26(1), 68–75. https://doi.org/10.3969/j.issn.1672-6472.2007.01.012
  • Guo, J., Tao, H., Cao, Y., Ho, C.-T., Jin, S., & Huang, Q. (2016). Prevention of obesity and type 2 diabetes with aged citrus peel (chenpi) extract. Journal of Agricultural and Food Chemistry, 64(10), 2053–2061. https://doi.org/10.1021/acs.jafc.5b06157
  • Habijanic, J., Berovic, M., Boh, B., Plankl, M., & Wraber, B. (2015). Submerged cultivation of Ganoderma lucidum and the effects of its polysaccharides on the production of human cytokines TNF-α, IL-12, IFN-γ, IL-2, IL-4, IL-10 and IL-17. New Biotechnology, 32(1), 85–95. https://doi.org/10.1016/j.nbt.2014.07.007
  • Han, N.-R., Moon, P.-D., Kim, H.-M., & Jeong, H.-J. (2018). Cordycepin ameliorates skin inflammation in a DNFB-challenged murine model of atopic dermatitis. Immunopharmacology and Immunotoxicology, 40(5), 401–407. https://doi.org/10.1080/08923973.2018.1510964
  • Hansske, F., & Robins, M. J. (1985). Regiospecific and stereoselective conversion of ribonucleosides to 3′-deoxynucleosides. A high yield three-stage synthesis of cordycepin from adenosine. Tetrahedron Letters, 26(36), 4295–4298. https://doi.org/10.1016/S0040-4039(00)98716-1
  • He, B.-L., Zheng, Q.-W., Guo, L.-Q., Huang, J.-Y., Yun, F., Huang, S.-S., & Lin, J.-F. (2020). Structural characterization and immune-enhancing activity of a novel high-molecular-weight polysaccharide from Cordyceps militaris. International Journal of Biological Macromolecules, 145, 11–20. https://doi.org/10.1016/j.ijbiomac.2019.12.115
  • He, L., Shi, W., Liu, X., Zhao, X., & Zhang, Z. (2018). Anticancer action and mechanism of ergosterol peroxide from paecilomyces cicadae fermentation broth. International Journal of Molecular Sciences, 19(12), 3935. https://doi.org/10.3390/ijms19123935
  • He, L., Wu, X., Cheng, J., Li, H., & Lu, X. (2010). Purification, composition analysis, and antioxidant activity of exopolysaccharides from mycelial culture of paecilomyces cicadae (Miq.) samson (ascomycetes). International Journal of Medicinal Mushrooms, 12(1), 51–62. https://doi.org/10.1615/intjmedmushr.v12.i1.50
  • Hedén, P., Sellman, G., von Wachenfeldt, M., Olenius, M., & Fagrell, D. (2020). Body shaping and volume restoration: The role of hyaluronic acid. Aesthetic Plastic Surgery, 44(4), 1286–1294. https://doi.org/10.1007/s00266-020-01826-8
  • Ho, J., Konerding, M., Gaumann, A., Groth, M., & Liu, W. (2004). Fungal polysaccharopeptide inhibits tumor angiogenesis and tumor growth in mice. Life Sciences, 75(11), 1343–1356. https://doi.org/10.1016/j.lfs.2004.02.021
  • Horng, C.-T., Yang, Y.-L., Chen, C.-C., Huang, Y.-S., Chen, C., & Chen, F.-A. (2021). Intraocular pressure-lowering effect of Cordyceps cicadae mycelia extract in a glaucoma rat model. International Journal of Medical Sciences, 18(4), 1007. https://doi.org/10.7150/ijms.47912
  • Hsu, J., & Chen, C. (2021). Intraocular pressure-lowering efficacy and safety of fermented cordyceps cicadae mycelia. Hans J. Food and Nutrition Sciences, 10(01), 33–39. https://doi.org/10.12677/hjfns.2021.101005
  • Hsu, J.-H., Jhou, B.-Y., Yeh, S.-H., Chen, Y.-L., & Chen, C.-C. (2015). Healthcare functions of cordyceps cicadae. Journal of Nutrition & Food Sciences, 5(6), 1000432. https://doi.org/10.4172/2155-9600.1000432
  • Hu, P. F., Chen, W. P., Bao, J. P., Jiang, L. F., & Wu, L. D. (2014a). Cordycepin modulates inflammatory and catabolic gene expression in interleukin-1beta-induced human chondrocytes from advanced-stage osteoarthritis: An in vitro study. International Journal of Clinical and Experimental Pathology, 7(10), 6575–6584. https://doi.org/10.1016/j.biomag.2011.04.003
  • Hu, X., Jiang, D., Li, F., Wu, Z., Huang, Y., Song, S., & Wang, Z. (2014b). Ergosterol reverses multidrug resistance in SGC7901/Adr cells. Die Pharmazie-An International Journal of Pharmaceutical Sciences, 69(5), 396–400. https://doi.org/10.1691/ph.2014.3866
  • Huang, F., Zhang, R., Liu, Y., Xiao, J., Liu, L., Wei, Z., Yi, Y., Zhang, M., & Liu, D. (2016). Dietary litchi pulp polysaccharides could enhance immunomodulatory and antioxidant effects in mice. International Journal of Biological Macromolecules, 92, 1067–1073. https://doi.org/10.1016/j.ijbiomac.2016.08.021
  • Huang, K., & Tian, A. J. (2009). Study on the crude extraction method and process conditions of hyaluronic acid production by Paecilomyces cicadae. Journal of Kerry College, 27, 33–36. https://doi.org/10.3969/j.issn.1673-9329.2009.03.014
  • Hur, H. (2008). Chemical ingredients of Cordyceps militaris. Mycobiology, 36(4), 233–235. https://doi.org/10.4489/MYCO.2008.36.4.233
  • Jasinghe, V. J., & Perera, C. O. (2005). Distribution of ergosterol in different tissues of mushrooms and its effect on the conversion of ergosterol to vitamin D2 by UV irradiation. Food Chemistry, 92(3), 541–546. https://doi.org/10.1016/j.foodchem.2004.08.022
  • Jiang, M.-H., Zhu, L., & Jiang, J.-G. (2010). Immunoregulatory actions of polysaccharides from Chinese herbal medicine. Expert Opinion on Therapeutic Targets, 14(12), 1367–1402. https://doi.org/10.1517/14728222.2010.531010
  • Jiang, P. (1987). Pharmacology constituent and function of Cordyceps sinensis. J Westnorthern Med, 2(04), 43–44.
  • Jiang, Q., Lou, Z., Wang, H., & Chen, C. (2019). Antimicrobial effect and proposed action mechanism of cordycepin against escherichia coli and bacillus subtilis. Journal of Microbiology, 57(4), 288–297. https://doi.org/10.1007/s12275-019-8113-z
  • Jin, L. Q., Xiong, Z. K., & Jian-Xin, L. (2008). Experimental studies on immunomodulatory and antitumor activity of polysaccharide from Paecilomyces cicadidae. Chinese Journal of Pathophysiology, https://doi.org/10.3321/j.issn:1000-4718.2008.03.018
  • Jin, Z., & Chen, Y. (2006). Clinical observation on cordyceps cicadae shing tang in preventing the progression of chronic renal failure. Chin Arch Tradit Chin Med, 24(08), 1457–1459. https://doi.org/10.13193/j.archtcm.2006.08.67.jinzhh.026
  • Kahlos, K., Kangas, L., & Hiltunen, R. (1989). Ergosterol peroxide, an active compound from inonotus radiatus. Planta Medica, 55((04|4)), 389–390. https://doi.org/10.1055/s-2006-962036
  • Kang, N., Lee, H.-H., Park, I., & Seo, Y.-S. (2017). Development of high cordycepin-producing Cordyceps militaris strains. Mycobiology, 45(1), 31–38. https://doi.org/10.5941/MYCO.2017.45.1.31
  • Ke, B.-J., & Lee, C.-L. (2016). Investigation on the fermentation for high adenosine and N6-(2-hydroxyethyl)-adenosine (HEA) productions of Cordyceps cicadae. New Biotechnology, 33, S206–S207. https://doi.org/10.1016/j.nbt.2016.06.1433
  • Ke, B.-J., & Lee, C.-L. (2019). Using submerged fermentation to fast increase N6-(2-hydroxyethyl)-adenosine, adenosine and polysaccharide productions of cordyceps cicadae NTTU 868. Amb Express, 9(1), 1–9. https://doi.org/10.1186/s13568-018-0728-7
  • Ke-Xue, Z., Shao-Ping, N., Le-He, T., Chuan, L., De-Ming, G., & Ming-Yong, X. (2016). A polysaccharide from Ganoderma atrum improves liver function in type 2 diabetic rats via antioxidant action and short-chain fatty acids excretion. Journal of Agricultural and Food Chemistry, 64(9), 1938–1944. https://doi.org/10.1021/acs.jafc.5b06103
  • Keypour, S., Riahi, H., Moradali, M.-F., & Rafati, H. (2008). Investigation of the antibacterial activity of a chloroform extract of Ling Zhi or Reishi medicinal mushroom. Ganoderma Lucidum (W. Curt.: Fr.) P. Karst.(Aphyllophoromycetideae), from Iran. International Journal of Medicinal Mushrooms, 10(4), 345–349.
  • Khan, M. A., & Tania, M. (2020). Cordycepin in anticancer research: Molecular mechanism of therapeutic effects. Current Medicinal Chemistry, 27(6), 983–996. https://doi.org/10.2174/0929867325666181001105749
  • Kim, D.-H., Yang, B.-K., Jeong, S.-C., Park, J.-B., Cho, S.-P., Das, S., Yun, J.-W., & Song, C.-H. (2001). Production of a hypoglycemic, extracellular polysaccharide from the submerged culture of the mushroom, Phellinus linteus. Biotechnology Letters, 23(7), 513–517. https://doi.org/10.1023/A:1010312513878
  • Kim, H. S., Kim, J. Y., Ryu, H. S., Shin, B. R., Kang, J. S., Kim, H. M., Kim, Y. O., Hong, J. T., Kim, Y., & Han, S. B. (2011). Phenotypic and functional maturation of dendritic cells induced by polysaccharide isolated from paecilomyces cicadae. Journal of Medicinal Food, 14(7-8), 847–856. https://doi.org/10.1089/jmf.2011.1575
  • Kim, H. S., Kim, Y. J., Lee, H. K., Ryu, H. S., Kim, J. S., Yoon, M. J., Kang, J. S., Hong, J. T., Kim, Y., & Han, S. B. (2012). Activation of macrophages by polysaccharide isolated from paecilomyces cicadae through toll-like receptor 4. Food and Chemical Toxicology, 50(9), 3190–3197. https://doi.org/10.1016/j.fct.2012.05.051
  • Kim, H. S., Lee, H. K., Kim, Y. J., Ryu, H. S., Kim, J. S., Kim, J., Kim, Y. G., Hong, J. T., Kim, Y., & Han, S.-B. (2013). Activation of macrophages by natural TLR4 ligand isolated from Paecilomyces cicadae (P4496). Am Assoc Immnol, 190(1), 178.2–178.2. https://doi.org/10.4049/jimmunol.190.supp.178.2
  • Kim, S. W., Park, S. S., Min, T. J., et al. (1999). Antioxidant activity of ergosterol peroxide (5,8-epidioxy-5 alpha,8 alpha-ergosta-6,22E-dien-3 beta-ol) in Armillariella mellea[J]. Bulletin- Korean Chemical Society, 20(7), 819–823. https://doi.org/10.5012/bkcs.1999.20.7.819
  • Ko, B.-S., Lu, Y.-J., Yao, W.-L., Liu, T.-A., Tzean, S.-S., Shen, T.-L., & Liou, J.-Y. (2013). Cordycepin regulates GSK-3β/β-catenin signaling in human leukemia cells. PLoS One, 8(9), e76320–e76320. https://doi.org/10.1371/journal.pone.0076320.
  • Kobori, M., Yoshida, M., Ohnishi-Kameyama, M., & Shinmoto, H. (2007). Ergosterol peroxide from an edible mushroom suppresses inflammatory responses in RAW264. 7 macrophages and growth of HT29 colon adenocarcinoma cells. British Journal of Pharmacology, 150(2), 209–219. https://doi.org/10.1038/sj.bjp.0706972
  • Kuo, Y., Weng, S., Chou, C., Chang, T., & Tsai, W. (2003). Activation and proliferation signals in primary human T lymphocytes inhibited by ergosterol peroxide isolated from cordyceps cicadae. British Journal of Pharmacology, 140(5), 895. https://doi.org/10.1038/sj.bjp.0705500
  • Kuo, Y.-C., Lin, L.-C., Don, M.-J., Liao, H.-F., Tsai, Y.-P., Lee, G.-H., & Chou, C.-J. (2002a). Cyclodesipeptide and dioxomorpholine derivatives isolated from the insect-body portion of the fungus cordyceps cicadae. J Chin Med, 13(4), 209–219.
  • Kwon, D. W., Jeon, J.-H., Kang, C., & Kim, Y. H. (2003). New synthesis of 3′-deoxypurine nucleosides using samarium (III) iodide complex. Tetrahedron Letters, 44(43), 7941–7943. https://doi.org/10.1016/j.tetlet.2003.09.001
  • Lan, T., Yu, Y., Zhang, J., Li, H., Weng, Q., Jiang, S., Tian, S., Xu, T., Hu, S., & Yang, G. (2021). Cordycepin ameliorates nonalcoholic steatohepatitis by activation of the AMP-activated protein kinase signaling pathway. Hepatology, 74(2), 686–703. https://doi.org/10.1002/hep.31749
  • Lee, Y.-S., Choi, K.-M., Choi, M.-H., Ji, S.-Y., Lee, S., Sin, D.-M., Oh, K.-W., Lee, Y.-M., Hong, J.-T., Yun, Y.-P., & Yoo, H.-S. (2011). Serine palmitoyltransferase inhibitor myriocin induces growth inhibition of b16f10 melanoma cells through g(2) /m phase arrest. Cell Proliferation, 44(4), 320–329. https://doi.org/10.1111/j.1365-2184.2011.00761.x
  • Li, C. R., Wang, Y. Q., Cheng, W. M., Chen, Z. A., Hywel-Jones, N., & Li, Z. Z. (2021a). Review on research progress and prospects of cicada flower, isaria cicadae (ascomycetes). International Journal of Medicinal Mushrooms, 23(4), 81–91. https://doi.org/10.1615/intjmedmushrooms.2021038085
  • Li, F., Wei, Y., Liang, L., Huang, L., Yu, G., & Li, Q. (2021b). A novel low-molecular-mass pumpkin polysaccharide: Structural characterization, antioxidant activity, and hypoglycemic potential. Carbohydrate Polymers, 251, 117090. https://doi.org/10.1016/j.carbpol.2020.117090
  • Li, I., Hsu, J., Lin, T., Lin, W., & Chen, C. (2017a). Prenatal developmental toxicity study of HEA-enriched cordyceps cicadae mycelia in Sprague-Dawley rats. SDRP J Food Sci Technol, 2(1), 1–7. https://doi.org/10.25177/jfst.2.1.2
  • Li, I. C., Lin, S., Tsai, Y. T., Hsu, J. H., Chen, Y. L., Lin, W. H., & Chen, C. C. (2019a). Cordyceps cicadae mycelia and its active compound HEA exert beneficial effects on blood glucose in type 2 diabetic db/db mice. Journal of the Science of Food and Agriculture, 99(2), 606–612. https://doi.org/10.1002/jsfa.9221
  • Li, J., Zhong, L., Zhu, H., & Wang, F. (2017b). The protective effect of cordycepin on D-galactosamine/lipopolysaccharide-induced acute liver injury. Mediators of Inflammation, 2017, 1–12. https://doi.org/10.1155/2017/3946706
  • Li, L., Guo, W.-L., Zhang, W., Xu, J.-X., Qian, M., Bai, W.-D., Zhang, Y.-Y., Rao, P.-F., Ni, L., & Lv, X.-C. (2019b). Grifola frondosa polysaccharides ameliorate lipid metabolic disorders and gut microbiota dysbiosis in high-fat diet fed rats. Food & Function, 10(5), 2560–2572. https://doi.org/10.1039/C9FO00075E
  • Li, L., Zhang, T., Li, C., Xie, L., Li, N., Hou, T., Wang, Y., & Wang, B. (2018). Potential therapeutic effects of cordyceps cicadae and paecilomyces cicadae on adenine-induced chronic renal failure in rats and their phytochemical analysis. Drug Design, Development and Therapy, 13, 103–117. https://doi.org/10.2147/DDDT.S180543
  • Li, R., Hu, F., Chen, A., & Fan, M. (2007). Studies on liquid culture technology of cordycepin in paecilomyces cicadae. Journal of Xuzhou Institute of Technology, 22(10), 23–30.
  • Li, R.-F., Zhou, X.-B., Zhou, H.-X., Yang, Z.-F., Jiang, H.-M., Wu, X., Li, W.-J., Qiu, J.-J., Mi, J.-N., & Chen, M. (2021c). Novel fatty acid in cordyceps suppresses influenza A (H1N1) virus-induced proinflammatory response through regulating innate signaling pathways. ACS Omega, 6(2), 1505–1515. https://doi.org/10.1021/acsomega.0c05264
  • Li, W., Zheng, H., Bukuru, J., & De Kimpe, N. (2004). Natural medicines used in the traditional Chinese medical system for therapy of diabetes mellitus. Journal of Ethnopharmacology, 92(1), 1–21. https://doi.org/10.1016/j.jep.2003.12.031
  • Li, W.-J., Li, L., Zhen, W.-Y., Wang, L.-F., Pan, M., Lv, J.-Q., Wang, F., Yao, Y.-F., Nie, S.-P., & Xie, M.-Y. (2017c). Ganoderma atrum polysaccharide ameliorates ROS generation and apoptosis in spleen and thymus of immunosuppressed mice. Food and Chemical Toxicology, 99, 199–208. https://doi.org/10.1016/j.fct.2016.11.033
  • Li, W. Q., He, L., Cheng, J. W., Han, S. F., & Liu, Y. (2016a). Structure and chain conformation of a neutral intracellular heteropolysaccharide from mycelium of Paecilomyces cicadae. Carbohydrate Polymers: Scientific and Technological Aspects of Industrially Important Polysaccharides, 136, 728–737. https://doi.org/10.1016/j.carbpol.2015.09.088
  • Li, X., Wu, Q., Bu, M., Hu, L., Du, W. W., Jiao, C., Pan, H., Sdiri, M., Wu, N., & Xie, Y. (2016). Ergosterol peroxide activates Foxo3-mediated cell death signaling by inhibiting AKT and c-Myc in human hepatocellular carcinoma cells. Oncotarget, 7(23), 33948. https://doi.org/10.18632/oncotarget.8608
  • Li, X., Wu, Q., Xie, Y., Ding, Y., Du, W. W., Sdiri, M., & Yang, B. B. (2015). Ergosterol purified from medicinal mushroom amauroderma rude inhibits cancer growth in vitro and in vivo by up-regulating multiple tumor suppressors. Oncotarget, 6(19), 17832. https://doi.org/10.18632/oncotarget.4026
  • Li, Y., Tang, J., Gao, H., Xu, Y., Han, Y., Shang, H., Lu, Y., & Qin, C. (2021d). Ganoderma lucidum triterpenoids and polysaccharides attenuate atherosclerotic plaque in high-fat diet rabbits. Nutrition, Metabolism and Cardiovascular Diseases, 31(6), 1929–1938. https://doi.org/10.1016/j.numecd.2021.03.023
  • Liao, Y. H., Ling, J. Y., Zhang, G. Y., Liu, F. J., Tao, S. C., Han, Z. G., Chen, S. J., Chen, Z., & Le, H. Y. (2015). Cordycepin induces cell cycle arrest and apoptosis by inducing DNA damage and up-regulation of p53 in leukemia cells. Cell Cycle, 14(5), 761–771. https://doi.org/10.1080/15384101.2014.1000097
  • Lin, H. I., Lee, Y. J., Chen, B. F., Tsai, M. C., Lu, J. L., Chou, C. J., & Jow, G. M. (2005). Involvement of Bcl-2 family, cytochrome c and caspase 3 in induction of apoptosis by beauvericin in human non-small cell lung cancer cells. Cancer Letters, 230(2), 248–259. https://doi.org/10.1016/j.canlet.2004.12.044
  • Lin, Y.-C., Lee, B.-H., Alagie, J., & Su, C.-H. (2017). Combination treatment of ergosterol followed by amphotericin B induces necrotic cell death in human hepatocellular carcinoma cells. Oncotarget, 8(42), 72727. https://doi.org/10.18632/oncotarget.20285
  • Lindequist, U., Lesnau, A., Teuscher, E., & Pilgrim, H. (1989). Studies of the antivirial activity of ergosterolperoxide. Pharmazie, 44(8), 579–580.
  • Liu, Y., Tang, Q., Zhang, J., Xia, Y., Yang, Y., Wu, D., Fan, H., & Cui, S. W. (2018). Triple helix conformation of β-d-glucan from Ganoderma lucidum and effect of molecular weight on its immunostimulatory activity. International Journal of Biological Macromolecules, 114, 1064–1070. https://doi.org/10.1016/j.ijbiomac.2018.03.054
  • Lu, M.-Y., Chen, C.-C., Lee, L.-Y., Lin, T.-W., & Kuo, C.-F. (2015). N 6-(2-Hydroxyethyl) adenosine in the medicinal mushroom cordyceps cicadae attenuates lipopolysaccharide-stimulated pro-inflammatory responses by suppressing TLR4-mediated NF-κB signaling pathways. Journal of Natural Products, 78(10), 2452–2460. https://doi.org/10.1021/acs.jnatprod.5b00573
  • Lu, Y. Z., Luo, F. F., Cen, K., Xiao, G. H., Yin, Y., Li, C. R., Li, Z. Z., Zhan, S., Zhang, H. Z., & Wang, C. S. (2017). Omics data reveal the unusual asexual-fruiting nature and secondary metabolic potentials of the medicinal fungus Cordyceps cicadae. BMC Genomics, 18(1), 15. https://doi.org/10.1186/s12864-016-3403-x
  • Mallebrera, B., Juan-Garcia, A., Font, G., & Ruiz, M. J. (2016). Mechanisms of beauvericin toxicity and antioxidant cellular defense. Toxicology Letters, 246, 28–34. https://doi.org/10.1016/j.toxlet.2016.01.013
  • Mandal, K., Raz-Ben Aroush, D., Graber, Z. T., Wu, B., Park, C. Y., Fredberg, J. J., Guo, W., Baumgart, T., & Janmey, P. A. (2018). Soft hyaluronic gels promote cell spreading, stress fibers, focal adhesion, and membrane tension by phosphoinositide signaling, not traction force. ACS Nano, 13(1), 203–214. https://doi.org/10.1021/acsnano.8b05286
  • Martínez, J. L. (2012). Bottlenecks in the transferability of antibiotic resistance from natural ecosystems to human bacterial pathogens. Frontiers in Microbiology, 2, 265. https://doi.org/10.3389/fmicb.2011.00265
  • Masuda, M., Das, S. K., Hatashita, M., Fujihara, S., & Sakurai, A. (2014). Efficient production of cordycepin by the Cordyceps militaris mutant G81-3 for practical use. Process Biochemistry, 49(2), 181–187. https://doi.org/10.1016/j.procbio.2013.10.017
  • Mendelow, A. D., Teasdale, G. M., Russell, T., Flood, J., Patterson, J., & Murray, G. D. (1985). Effect of mannitol on cerebral blood flow and cerebral perfusion pressure in human head injury. Journal of Neurosurgery, 63(1), 43–48. https://doi.org/10.3171/jns.1985.63.1.0043
  • Meng, Z., Kang, J., Wen, T., Lei, B., & Hyde, K. D. (2015). Cordycepin and N6-(2-hydroxyethyl)-adenosine from cordyceps pruinosa and their interaction with human serum albumin. PloS One, 10(3), e0121669. https://doi.org/10.1371/journal.pone.0121669
  • Muñoz-Fonseca, M. B., Vidal-Limon, A., Fernández-Pomares, C., Rojas-Durán, F., Hernández-Aguilar, M. E., Espinoza, C., Trigos, A., & Suárez-Medellín, J. (2021). Ergosterol exerts a differential effect on AR-dependent LNCaP and AR-independent DU-145 cancer cells. Natural Product Research, 35(22), 4857–4860. https://doi.org/10.1080/14786419.2020.1737054
  • Murphy, E. J., Masterson, C., Rezoagli, E., O’Toole, D., Major, I., Stack, G. D., Lynch, M., Laffey, J. G., & Rowan, N. J. (2020). β-Glucan extracts from the same edible shiitake mushroom lentinus edodes produce differential in-vitro immunomodulatory and pulmonary cytoprotective effects—implications for coronavirus disease (COVID-19) immunotherapies. Science of the Total Environment, 732, 139330. https://doi.org/10.1016/j.scitotenv.2020.139330
  • Nam, K. S., Jo, Y. S., Kim, Y. H., Hyun, J. W., & Kim, H. W. (2001). Cytotoxic activities of acetoxyscirpenediol and ergosterol peroxide from Paecilomyces tenuipes. Life Sciences, 69(2), 229–237. https://doi.org/10.1016/S0024-3205(01)01125-0
  • Nutrition, Efsa Panel On, N.F., Allergens, F., Turck, D., Bohn, T., Castenmiller, J., De Henauw, S., Hirsch-Ernst, K.I., Maciuk, A., Mangelsdorf, I., McArdle, H.J. and Naska, A. (2022). Safety of vitamin D2 mushroom powder as a novel food pursuant to regulation (EU) 2015/2283 (NF 2019/1471). EFSA Journal, 20, e07326.
  • Nxumalo, W., Elateeq, A. A., & Sun, Y. (2020). Can cordyceps cicadae be used as an alternative to Cordyceps militaris and cordyceps sinensis? A review. Journal of Ethnopharmacology, 257, 112877. https://doi.org/10.1016/j.jep.2020.112879
  • Ohno, M., Ito, Y., Arita, M., Shibata, T., Adachi, K., & Sawai, H. (1984). Synthetic studies on biologically active natural products by a chemicoenzymatic approach: Enantioselective synthesis of C-and N-nucleosides, showdomycin, 6-azapseudouridine and cordycepin. Tetrahedron, 40(1), 145–152. https://doi.org/10.1016/0040-4020(84)85113-3
  • Olatunji, O. J., Feng, Y., Olatunji, O. O., Tang, J., Wei, Y., Ouyang, Z., & Su, Z. (2016). Polysaccharides purified from cordyceps cicadae protects PC12 cells against glutamate-induced oxidative damage. Carbohydrate Polymers, 153, 187–195. https://doi.org/10.1016/j.carbpol.2016.06.108
  • Olatunji, O. J., Tang, J., Tola, A., Auberon, F., Oluwaniyi, O., & Ouyang, Z. (2018). The genus cordyceps: An extensive review of its traditional uses, phytochemistry and pharmacology. Fitoterapia, 129, 293–316. https://doi.org/10.1016/j.fitote.2018.05.010
  • Patterson, R. R. M. (2008). Cordyceps – a traditional Chinese medicine and another fungal therapeutic biofactory? Phytochemistry, 69(7), 1469–1495. https://doi.org/10.1016/j.phytochem.2008.01.027
  • Qi, H., Liu, Y., Wang, N., & Xiao, C. (2021). Lentinan attenuated the PM2. 5 exposure-induced inflammatory response, epithelial–mesenchymal transition and migration by inhibiting the PVT1/miR-199a-5p/caveolin1 pathway in lung cancer. DNA and Cell Biology, 40(5), 683–693. https://doi.org/10.1089/dna.2020.6338
  • Qi, M., Liu, C.Y., & Li, L. (2019). Protective effects of cordycepin on CCl4 induced acute liver injury in mice. Mycosystema, 38(09), 1510–1518. https://doi.org/10.13346/j.mycosystema.190090
  • Raethong, N., Wang, H., Nielsen, J., & Vongsangnak, W. (2020). Optimizing cultivation of cordyceps militaris for fast growth and cordycepin overproduction using rational design of synthetic media. Computational and Structural Biotechnology Journal, 18, 1–8. https://doi.org/10.1016/j.csbj.2019.11.003
  • Rahman, M. A., Rahman, M. S., Bashir, N. M. B., Mia, R., Hossain, A., Saha, S. K., Kakon, A. J., & Sarker, N. C. (2021a). Rationalization of mushroom-based preventive and therapeutic approaches to COVID-19. International Journal of Medicinal Mushrooms, 23(5), 1–11. https://doi.org/10.1615/IntJMedMushrooms.2021038285.
  • Ravani, A., Vincenzi, F., Bortoluzzi, A., Padovan, M., Pasquini, S., Gessi, S., Merighi, S., Borea, P. A., Govoni, M., & Varani, K. (2017). Role and function of A2A and A3 adenosine receptors in patients with ankylosing spondylitis, psoriatic arthritis and rheumatoid arthritis. International Journal of Molecular Sciences, 18(4), 697. https://doi.org/10.3390/ijms18040697
  • Ren, G., Xu, L., Lu, T., & Yin, J. (2018). Structural characterization and antiviral activity of lentinan from lentinus edodes mycelia against infectious hematopoietic necrosis virus. International Journal of Biological Macromolecules, 115, 1202–1210. https://doi.org/10.1016/j.ijbiomac.2018.04.132
  • Ren, L., Perera, C., & Hemar, Y. (2012). Antitumor activity of mushroom polysaccharides: A review. Food & Function, 3(11), 1118–1130. https://doi.org/10.1039/c2fo10279j
  • Ren, X., He, L., Cheng, J., & Chang, J. (2014). Optimization of the solid-state fermentation and properties of a polysaccharide from Paecilomyces cicadae (Miquel) Samson and its antioxidant activities in vitro. PLoS One, 9(2), e87578. https://doi.org/10.1371/journal.pone.0087578
  • Rhee, Y.-H., Jeong, S.-J., Lee, H.-J., Lee, H.-J., Koh, W., Jung, J. H., Kim, S.-H., & Sung-Hoon, K. (2012). Inhibition of STAT3 signaling and induction of SHP1 mediate antiangiogenic and antitumor activities of ergosterol peroxide in U266 multiple myeloma cells. BMC Cancer, 12(1), 1–11. https://doi.org/10.1186/1471-2407-12-1
  • Ryu, E., Son, M., Lee, M., Lee, K., Cho, J. Y., Cho, S., Lee, S. K., Lee, Y. M., Cho, H., Sung, G.-H., & Kang, H. (2014). Cordycepin is a novel chemical suppressor of epstein-barr virus replication. Oncoscience, 1(12), 866–881. https://doi.org/10.18632/oncoscience.110
  • Salminen, A., Kaarniranta, K., & Kauppinen, A. (2022). Photoaging: UV radiation-induced inflammation and immunosuppression accelerate the aging process in the skin. Inflammation Research, 71(7-8), 817–831. https://doi.org/10.1007/s00011-022-01598-8
  • Sang, T., Guo, C., Guo, D., Wu, J., Wang, Y., Wang, Y., Chen, J., Chen, C., Wu, K., & Na, K. (2021). Suppression of obesity and inflammation by polysaccharide from sporoderm-broken spore of Ganoderma lucidum via gut microbiota regulation. Carbohydrate Polymers, 256, 117594. https://doi.org/10.1016/j.carbpol.2020.117594
  • Sankaran, M., Isabella, S., & Amaranth, K. (2017). Anti proliferative potential of ergosterol: A unique plant sterol on Hep2 cell line. Int J Pharma Res Health Sci, 5, 1736–1742.
  • Shao, L., Jiang, S., Li, Y., Yu, L., Liu, H., Ma, L., & Yang, S. (2023). Aqueous extract of cordyceps cicadae (Miq.) promotes hyaluronan synthesis in human skin fibroblasts: A potential moisturizing and anti-aging ingredient. PLoS ONE, 18(7), e0274479. https://doi.org/10.1371/journal.pone.0274479
  • Sharma, S. K., Gautam, N., & Atri, N. S. (2015). Optimized extraction, composition, antioxidant and antimicrobial activities of exo and intracellular polysaccharides from submerged culture of cordyceps cicadae. BMC Complementary and Alternative Medicine, 15, 1–8. https://doi.org/10.1155/2015/462864
  • Shen, C.-Y., Zhang, W.-L., & Jiang, J.-G. (2017). Immune-enhancing activity of polysaccharides from hibiscus sabdariffa linn. Via MAPK and NF-kB signaling pathways in RAW264. 7 cells. Journal of Functional Foods, 34, 118–129. https://doi.org/10.1016/j.jff.2017.03.060
  • Shen, Y., Cai, M., Xia, W., Liu, J., Zhang, Q., Xie, H., Wang, C., Wang, X., & Zheng, S. (2007). FTY720, a synthetic compound from Isaria sinclairii, inhibits proliferation and induces apoptosis in pancreatic cancer cells. Cancer Letters, 254(2), 288–297. https://doi.org/10.1016/j.canlet.2007.03.013
  • Sheth, S., Brito, R., Mukherjea, D., Rybak, L. P., & Ramkumar, V. (2014). Adenosine receptors: Expression, function and regulation. International Journal of Molecular Sciences, 15(2), 2024–2052. https://doi.org/10.3390/ijms15022024
  • Shi, C., Song, W., Gao, J., Yan, S., Guo, C., & Zhang, T. (2022). Enhanced production of cordycepic acid from cordyceps cicadae isolated from a wild environment. Brazilian Journal of Microbiology, 53(2), 673–688. https://doi.org/10.1007/s42770-022-00687-4
  • Shim, D. W., Han, J. W., Sun, X., Jang, C. H., Koppula, S., Kim, T. J., Kang, T. B., & Lee, K. H. (2013). Lysimachia clethroides duby extract attenuates inflammatory response in Raw 264.7 macrophages stimulated with lipopolysaccharide and in acute lung injury mouse model. Journal of Ethnopharmacology, 150(3), 1007–1015. https://doi.org/10.1016/j.jep.2013.09.056
  • Shin, E. M., Zhou, H. Y., Guo, L. Y., Kim, J. A., Lee, S. H., Merfort, I., Kang, S. S., Kim, H. S., Kim, S., & Kim, Y. S. (2008). Anti-inflammatory effects of glycyrol isolated from Glycyrrhiza uralensis in LPS-stimulated RAW264. 7 macrophages. International Immunopharmacology, 8 (11), 1524–1532. https://doi.org/10.1016/j.intimp.2008.06.008
  • Sillapachaiyaporn, C., Chuchawankul, S., Nilkhet, S., Moungkote, N., Sarachana, T., Ung, A. T., Baek, S. J., & Tencomnao, T. (2022). Ergosterol isolated from cloud ear mushroom (Auricularia polytricha) attenuates bisphenol A-induced BV2 microglial cell inflammation. Food Research International, 157, 111433. https://doi.org/10.1016/j.foodres.2022.111433
  • Song, J., Xin, J., & Zhu, Y. (2007). Effect of cordyceps cicadae on blood sugar of mice and its hematopoietic function. Chin Arch Tradit Chin Med, 25(06), 1144–1145. https://doi.org/10.13193/j.archtcm.2007.06.57.songjm.022
  • St-Jacques, M. (1973). Elucidation of structure and stereochemistry of myriocin. A novel antifungal antibiotics. The Journal of Organic Chemistry, 38(7), 1253–1260. https://doi.org/10.1021/jo00947a001
  • Su, Y., & Li, L. (2020). Structural characterization and antioxidant activity of polysaccharide from four auriculariales. Carbohydrate Polymers, 229, 115407. https://doi.org/10.1016/j.carbpol.2019.115407
  • Suarez, E. R., Syvitski, R., Kralovec, J. A., Noseda, M. D., Barrow, C. J., Ewart, H. S., Lumsden, M. D., & Grindley, T. B. (2006). Immunostimulatory polysaccharides from Chlorella pyrenoidosa. A new galactofuranan. Measurement of molecular weight and molecular weight dispersion by DOSY NMR. Biomacromolecules, 7(8), 2368–2376. https://doi.org/10.1021/bm060365x
  • Sugiyama, K., Muroi, M., Kinoshita, M., Hamada, O., Minai, Y., Sugita-Konishi, Y., Kamata, Y., & Tanamoto, K. (2016). NF-κB activation via MyD88-dependent toll-like receptor signaling is inhibited by trichothecene mycotoxin deoxynivalenol. The Journal of Toxicological Sciences, 41(2), 273–279. https://doi.org/10.2131/jts.41.273
  • Sun, C. S., Wang, Y. Q., Zhang, Z.-L., Liou, Y. L., & Zhou, H. H. (2021). Distinctive quality control method for solid-state fermented isaria cicadae from strain Ic-17-7 and application in a rat model of type 2 diabetes. Chinese Journal of Natural Medicines, 19(12), 921–929. https://doi.org/10.1016/s1875-5364(21)60113-9
  • Sun, C. S., Wang, Y. Q., Zhang, Z. L., Liou, Y. L., & Zhou, H. H. (2021). Distinctive quality control method for solid-state fermented Isaria cicadae from stran Ic-17-7 and application in a rat model of type 2 diabetes. Chinese Journal of Natural Medicines, 19(12), 921–929. https://doi.org/10.1016/S1875-5364(21)60113-9
  • Sun, Y., Wink, M., Wang, P., Lu, H., Zhao, H., Liu, H., Wang, S., Sun, Y., & Liang, Z. (2017). Biological characteristics, bioactive components and antineoplastic properties of sporoderm-broken spores from wild cordyceps cicadae. Phytomedicine, 36, 217–228. https://doi.org/10.1016/j.phymed.2017.10.004
  • Surenjav, U., Zhang, L., Xu, X., Zhang, X., & Zeng, F. (2006). Effects of molecular structure on antitumor activities of(1 -> 3)-beta-D-glucans from different Lentinus Edodes. Carbohydrate Polymers: Scientific and Technological Aspects of Industrially Important Polysaccharides, 63(1), 97–104. https://doi.org/10.1016/j.carbpol.2005.08.011
  • Suzuki, H., Okubo, A., Yamazaki, S., Suzuki, K., Mitsuya, H., & Toda, S. (1989). Inhibition of the infectivity and cytopathic effect of human immunodeficiency virus by water-soluble lignin in an extract of the culture medium of lentinusedodes mycelia (LEM). Biochemical and Biophysical Research Communications, 160(1), 367–373. https://doi.org/10.1016/0006-291X(89)91665-3
  • Takano, F., Yahagi, N., Yahagi, R., Takada, S., Yamaguchi, M., Shoda, S., Murase, T., Fushiya, S., & Ohta, T. (2005). The liquid culture filtrates of Paecilomyces tenuipes (Peck) Samson (= Isaria japonica Yasuda) and Paecilomyces cicadae (Miquel) Samson (= Isaria sinclairii (Berk.) llond) regulate Th1 and Th2 cytokine response in murine Peyer’s patch cells in vitro and ex vivo. International Immunopharmacology, 5(5), 903–916. https://doi.org/10.1016/j.intimp.2005.01.005
  • Takei, T., Yoshida, M., Ohnishi-Kameyama, M., & Kobori, M. (2005). Ergosterol peroxide, an apoptosis-inducing component isolated from Sarcodon aspratus (Berk.) S. Ito. Bioscience, Biotechnology, and Biochemistry, 69(1), 212–215. https://doi.org/10.1271/bbb.69.212
  • Tan, A. J., Li-Kun, W. U., & Xiao-Bei, Y. U. (2007). Screen of optimum broth of Paecilomyces cicadae for producing hyaluronic acid. Food Science, (08), 294–296.
  • Tan, L., Song, X., Ren, Y., Wang, M., Guo, C., Guo, D., Gu, Y., Li, Y., Cao, Z., & Deng, Y. (2020, October 8). Anti-inflammatory effects of cordycepin: A review. Phytotherapy Research, 33(3), 1284–1297. https://doi.org/10.1002/ptr.6890
  • Tang, Z., Lin, W., Chen, Y., Feng, S., Qin, Y., Xiao, Y., Chen, H., Liu, Y., Chen, H., & Bu, T. (2022). Extraction, purification, physicochemical properties, and activity of a new polysaccharide from cordyceps cicadae. Frontiers in Nutrition, 9. https://doi.org/10.3389/fnut.2022.911310
  • Tao, X.-M., Liu, P.-F., Gu, H.-Y., Lian, D.-B., Gao, L., Tao, W.-W., Yan, D., & Zhao, B. (2020). Cordycepin alleviates anterior cruciate ligament transection (ACLT)-induced knee osteoarthritis through regulating TGF-β activity and autophagy. Drug Design, Development and Therapy, 14, 4133–4134. https://doi.org/10.2147/DDDT.S251893
  • Tian, J., Tang, C., Wang, X., Zhang, X., Xiao, L., & Li, W. (2022). Supramolecular structure features and immunomodulatory effects of exopolysaccharide from Paecilomyces cicadae TJJ1213 in RAW264. 7 cells through NF-κB/MAPK signaling pathways. International Journal of Biological Macromolecules, 207, 464–474. https://doi.org/10.1016/j.ijbiomac.2022.03.029
  • Tian, J., Wang, X., Zhang, X., Zhang, C., Chen, X., Dong, M., Rui, X., Zhang, Q., Fang, Y., & Li, W. (2021a). Isolation, structural characterization and neuroprotective activity of exopolysaccharide from paecilomyces cicada TJJ1213. International Journal of Biological Macromolecules, 183, 1034–1046. https://doi.org/10.1016/j.ijbiomac.2021.05.047
  • Tian, J. J., Wang, X. M., Zhang, X. L., Chen, X. H., Rui, X., Zhang, Q. Q., Dong, M. S., & Li, W. (2021b). Simulated digestion and fecal fermentation behaviors of exopolysaccharides from paecilomyces cicadae TJJ1213 and its effects on human gut microbiota. International Journal of Biological Macromolecules, 188, 833–843. https://doi.org/10.1016/j.ijbiomac.2021.08.052
  • Tian, Y., Yi, W., Bai, L., Zhang, P., Si, J., Hou, X., Deng, Y., & Hou, J. (2019). Lentinan in-situ coated tungsten oxide nanorods as a nanotherapeutic agent for low power density photothermal cancer therapy. International Journal of Biological Macromolecules, 137, 904–911. https://doi.org/10.1016/j.ijbiomac.2019.06.183
  • Tilg, H., Zmora, N., Adolph, T. E., & Elinav, E. (2020). The intestinal microbiota fuelling metabolic inflammation. Nature Reviews Immunology, 20(1), 40–54. https://doi.org/10.1038/s41577-019-0198-4
  • Toh Choon, R., Sariah, M., & Siti Mariam, M. (2012). Ergosterol from the soilborne fungus ganoderma boninense. Journal of Basic Microbiology, 52(5), 608–612. https://doi.org/10.1002/jobm.201100308
  • Tsai, Y.-S., Hsu, J.-H., Lin, D. P.-C., Chang, H.-H., Chang, W.-J., Chen, Y.-L., & Chen, C.-C. (2021). Safety assessment of HEA-enriched cordyceps cicadae mycelium: A randomized clinical trial. Journal of the American College of Nutrition, 40(2), 127–132. https://doi.org/10.1080/07315724.2020.1743211
  • Ukai, S., Kiho, T., Hara, C., Morita, M., Goto, A., Imaizumi, N., & Hasegawa, Y. (1983). Antitumor activity of various polysaccharides isolated from Dictyophora indusiata, Ganoderma japonicum, cordyceps cicadae, Auricularia auricula-judae, and Auricularia species. Chemical and Pharmaceutical Bulletin, 31(2), 4. https://doi.org/10.1248/cpb.31.741
  • Varani, K., Padovan, M., Vincenzi, F., Targa, M., Trotta, F., Govoni, M., & Borea, P. A. (2011). A2a and A3 adenosine receptor expression in rheumatoid arthritis: Upregulation, inverse correlation with disease activity score and suppression of inflammatory cytokine and metalloproteinase release. Arthritis Research & Therapy, 13(6), 1–13. https://doi.org/10.1186/ar3527
  • Verma, A. K. (2022). Cordycepin: A bioactive metabolite of C ordyceps militaris and polyadenylation inhibitor with therapeutic potential against COVID-19. Journal of Biomolecular Structure and Dynamics, 40(8), 3745–3752. https://doi.org/10.1080/07391102.2020.1850352
  • Vinderola, G., Perdigón, G., Duarte, J., Farnworth, E., & Matar, C. (2006). Effects of the oral administration of the exopolysaccharide produced by lactobacillus kefiranofaciens on the gut mucosal immunity. Cytokine, 36(5-6), 254–260. https://doi.org/10.1016/j.cyto.2007.01.003
  • Wan, W., Cong, X., Zhijing, L., Liya, G., Jiage, M., Juncai, H., & Zhanmei, J. (2022). Physicochemical properties and bioactivity of polysaccharides from Isaria cicadae Miquel with different extraction processes: Effects on gut microbiota and immune response in mice. Food & Function, 13(18), 9268–9284. https://doi.org/10.1039/D2FO01646J
  • Wang, H., Zhang, J., Sit, W.-H., Lee, C.-Y. J., & Wan, J. M.-F. (2014a). Cordyceps cicadae induces G2/M cell cycle arrest in MHCC97H human hepatocellular carcinoma cells: A proteomic study. Chinese Medicine, 9(1), 1–12. https://doi.org/10.1186/1749-8546-9-15
  • Wang, J., Chen, H.L., Li, W.R., & Shan, L.L. (2020). Cordyceps acid alleviates lung cancer in nude mice. Journal of Biochemical and Molecular Toxicology, 35(3). https://doi.org/10.1002/jbt.22670
  • Wang, J.-H., Zhang, Z.-L., Wang, Y.-Q., Yang, M., Wang, C.-H., Li, X.-W., & Guo, Y.-W. (2017a). Chemical constituents from mycelia and spores of fungus cordyceps cicadae. Chinese Herbal Medicines, 9(2), 188–192. https://doi.org/10.1016/S1674-6384(17)60094-7
  • Wang, L., He, Y., Li, Y., Pei, C., Olatunji, O. J., Tang, J., Famurewa, A. C., Wang, H., & Yan, B. (2020). Protective effects of nucleosides-rich extract from cordyceps cicadae against cisplatin induced testicular damage. Chemistry & Biodiversity, 17(11), e2000671. https://doi.org/10.1002/cbdv.202000671
  • Wang, P. Y., Zhu, X. L., & Lin, Z. B. (2012a). Antitumor and immunomodulatory effects of polysaccharides from broken-spore of Ganoderma lucidum. Frontiers in Pharmacology, 3, 8. https://doi.org/10.3389/fphar.2012.00135
  • Wang, W., Xu, C., Liu, Z., Gu, L., Ma, J., Hou, J., & Jiang, Z. (2022). Physicochemical properties and bioactivity of polysaccharides from Isaria cicadae Miquel with different extraction processes: Effects on gut microbiota and immune response in mice. Food & Function, 13(18), 9268–9284. https://doi.org/10.1039/D2FO01646J
  • Wang, X., Qin, A., Xiao, F., Olatunji, O. J., Zhang, S., Pan, D., Han, W., Wang, D., & Ni, Y. (2019a). N6-(2-hydroxyethyl)-adenosine from cordyceps cicadae protects against diabetic kidney disease via alleviation of oxidative stress and inflammation. Journal of Food Biochemistry, 43(2), e12727. https://doi.org/10.1111/jfbc.12727
  • Wang, Y., Guo, Y., Zhang, L., & Wu, J. (2012b). Characterizations of a new cordyceps cicadae isolate and production of adenosine and cordycepin. Brazilian Journal of Microbiology, 43(2), 449–455. https://doi.org/10.1590/S1517-83822012000200004
  • Wang, Y., He, P., He, L., Huang, Q., Cheng, J., Li, W., Liu, Y., & Wei, C. (2019b). Structural elucidation, antioxidant and immunomodulatory activities of a novel heteropolysaccharide from cultured Paecilomyces cicadae (Miquel.) Samson. Carbohydrate Polymers, 216, 270–281. https://doi.org/10.1016/j.carbpol.2019.03.104
  • Wang, Y., Liu, Y., Yu, H., Zhou, S., Zhang, Z., Wu, D., Yan, M., Tang, Q., & Zhang, J. (2017c). Structural characterization and immuno-enhancing activity of a highly branched water-soluble β-glucan from the spores of Ganoderma lucidum. Carbohydrate Polymers, 167, 337–344. https://doi.org/10.1016/j.carbpol.2017.03.016
  • Wang, Y., Mo, H., Gu, J., Chen, K., Han, Z., & Liu, Y. (2017d). Cordycepin induces apoptosis of human acute monocytic leukemia cells via downregulation of the ERK/Akt signaling pathway. Experimental and Therapeutic Medicine, 14(4), 3067–3073. https://doi.org/10.3892/etm.2017.4855
  • Waxman, K. (1996). Shock: Ischemia, reperfusion, and inflammation. New Horizons (baltimore, Md), 4(2), 153–160.
  • Weete, J. D. (1989). Structure and function of sterols in fungi. In Advances in lipid research (pp. 115–167). https://doi.org/10.1016/b978-0-12-024923-7.50007-8
  • Wen, Y. T., Jhou, B. Y., Hsu, J. H., Fu, H. I., Chen, Y. L., Shih, Y. C., Chen, C. C., & Tsai, R. K. (2022). Neuroprotective effects of cordyceps cicadae (ascomycetes) mycelium extract in the Rat model of optic nerve crush. International Journal of Medicinal Mushrooms, 24(2), 41–48. https://doi.org/10.1615/IntJMedMushrooms.2021041522
  • Weng, S. C., Chou, C. J., Lin, L. C., Tsai, W. J., & Kuo, Y. C. (2002). Immunomodulatory functions of extracts from the Chinese medicinal fungus cordyceps cicadae. Journal of Ethnopharmacology, 83(1-2), 79–85. https://doi.org/10.1016/S0378-8741(02)00212-X
  • Won, D. P., Lee, J. S., Kwon, D. S., Lee, K. E., Shin, W. C., & Hong, E. K. (2011). Immunostimulating activity by polysaccharides isolated from fruiting body of Inonotus obliquus. Molecules and Cells, 31(2), 165–173. https://doi.org/10.1007/s10059-011-0022-x
  • Wongsa, B., Raethong, N., Chumnanpuen, P., Wong-Ekkabut, J., Laoteng, K., & Vongsangnak, W. (2020). Alternative metabolic routes in channeling xylose to cordycepin production of Cordyceps militaris identified by comparative transcriptome analysis. Genomics, 112(1), 629–636. https://doi.org/10.1016/j.ygeno.2019.04.015
  • World Health Organization [WHO]. (1999). Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: Diagnosis and classification of diabetes mellitus (WHO/NCD/NCS/99.2). World Health Organization.
  • Wu, M., Huang, T., Wang, J., Chen, P., Mi, W., Ying, Y., Wang, H., Zhao, D., & Huang, S. (2018). Antilung cancer effect of ergosterol and cisplatin-loaded liposomes modified with cyclic arginine-glycine-aspartic acid and octa-arginine peptides. Medicine, 97(33), e11916. https://doi.org/10.1097/md.0000000000011916
  • Wu, Q.-P., Xie, Y.-Z., Deng, Z., Li, X.-M., Yang, W., Jiao, C.-W., Fang, L., Li, S.-Z., Pan, H.-H., & Yee, A. J. (2012). Ergosterol peroxide isolated from Ganoderma lucidum abolishes microRNA miR-378-mediated tumor cells on chemoresistance.
  • Wu, Y., Li, S., Li, H., Zhao, C., Ma, H., Zhao, X., Wu, J., Liu, K., Shan, J., & Wang, Y. (2016). Effect of a polysaccharide from poria cocos on humoral response in mice immunized by H1N1 influenza and HBsAg vaccines. International Journal of Biological Macromolecules, 91, 248–257. https://doi.org/10.1016/j.ijbiomac.2016.05.046
  • Xia, B., & Hu, C. R. (2014). Research progress of perennial flowers in China. Anhui. Agricultural Science Bulletin, 7(1), 1–4. https://doi.org/10.1016/B978-1-4831-9801-9.50150-1
  • Xie, F., Li, W., Chen, M. Z., & u J, Y. (2016). Antitumor activity and mechanism of action of wild cordyceps sobolifera polysaccharide. Food Sci, 37(13), 209–213. https://doi.org/10.7506/spkx1002-6630-201613038
  • Xie, H., Li, X., Chen, Y., Lang, M., Shen, Z., & Shi, L. (2019). Ethanolic extract of cordyceps cicadae exerts antitumor effect on human gastric cancer SGC-7901 cells by inducing apoptosis, cell cycle arrest and endoplasmic reticulum stress. Journal of Ethnopharmacology, 231, 230–240. https://doi.org/10.1016/j.jep.2018.11.028
  • Xu, J., Tan, Z.-C., Shen, Z.-Y., Shen, X.-J., & Tang, S.-M. (2021a). Cordyceps cicadae polysaccharides inhibit human cervical cancer hela cells proliferation via apoptosis and cell cycle arrest. Food and Chemical Toxicology, 148, 111971. https://doi.org/10.1016/j.fct.2021.111971
  • Xu, Y., & Du, Y. (2020). Effects of lentinan on endothelial cell activity, inflammatory response, endoplasmic reticulum stress, and apoptosis in sepsis. Advances in Polymer Technology, 2020, 1–9. https://doi.org/10.1155/2020/1640208
  • Xu, Y., Wu, Y.-J., Sun, P.-L., Zhang, F.-M., Linhardt, R. J., & Zhang, A.-Q. (2019). Chemically modified polysaccharides: Synthesis, characterization, structure activity relationships of action. International Journal of Biological Macromolecules, 132, 970–977. https://doi.org/10.1016/j.ijbiomac.2019.03.213
  • Xu, Z., Yan, X., Song, Z., Li, W., Zhao, W., Ma, H., Du, J., Li, S., & Zhang, D. (2018a). Two heteropolysaccharides from Isaria cicadae Miquel differ in composition and potentially immunomodulatory activity. International Journal of Biological Macromolecules, 117, 610–616. https://doi.org/10.1016/j.ijbiomac.2018.05.164
  • Xu, Z. C., Lin, R. Y., Hou, X. N., Wu, J., Zhao, W. B., Ma, H. H., Fan, Z. Y., Li, S. J., Zhu, Y., & Zhang, D. Y. (2020). Immunomodulatory mechanism of a purified polysaccharide isolated from isaria cicadae miquel on RAW264.7 cells via activating TLR4-MAPK-NF-kappa B signaling pathway. International Journal of Biological Macromolecules, 164, 4329–4338. https://doi.org/10.1016/j.ijbiomac.2020.09.035
  • Yamada, T., Ogamo, A., Saito, T., Watanabe, J., Uchiyama, H., & Nakagawa, Y. (1997). Preparation and anti-HIV activity of low-molecular-weight carrageenans and their sulfated derivatives. Carbohydrate Polymers, 32(1), 51–55. https://doi.org/10.1016/S0144-8617(96)00128-2
  • Yan, J. K., Wang, W. Q., & Wu, J. Y. (2014). Recent advances in cordyceps sinensis polysaccharides: Mycelial fermentation, isolation, structure, and bioactivities: A review. Journal of Functional Foods, 6, 33–47. https://doi.org/10.1016/j.jff.2013.11.024
  • Yang, C. H., Su, C. H., Liu, S. C., & Ng, L. T. (2019a). Isolation, anti-inflammatory activity and physico-chemical properties of bioactive polysaccharides from fruiting bodies of cultivated cordyceps cicadae (ascomycetes). International Journal of Medicinal Mushrooms, 21(10), 995–1006. https://doi.org/10.1615/intjmedmushrooms.2019031922
  • Yang, F., Qu, Q., Zhao, C., Liu, X., Yang, P., Li, Z., Han, L., & Shi, X. (2020a). Paecilomyces cicadae-fermented radix astragali activates podocyte autophagy by attenuating PI3K/AKT/mTOR pathways to protect against diabetic nephropathy in mice. Biomedicine & Pharmacotherapy, 71(5). https://doi.org/10.1016/j.biopha.2020.110479
  • Yang, J., Dong, H., Wang, Y., Jiang, Y., Zhang, W., Lu, Y., Chen, Y., & Chen, L. (2020b). Cordyceps cicadae polysaccharides ameliorated renal interstitial fibrosis in diabetic nephropathy rats by repressing inflammation and modulating gut microbiota dysbiosis. International Journal of Biological Macromolecules, 163, 442–456. https://doi.org/10.1016/j.ijbiomac.2020.06.153
  • Yang, J.-Z., Zhuo, J., Chen, B.-K., Jin, L.-Q., Lv, J.-X., & Li, L.-J. (2008). Regulating effects of paecilomyces cicadae polysaccharides on immunity of aged rats. Zhongguo Zhong yao za zhi = Zhongguo Zhongyao Zazhi = China Journal of Chinese Materia Medica, 33, 292–295.
  • Yang, M. J. (2008). Study on the production of immune active component ISP by cicada flower fermentation. Chongqing University. https://kns.cnki.net/KCMS/detail/detail.aspx?dbname=CMFD2009&filename=2009048473.nh.
  • Yang, N. N., Jiang, N., Ma, Q. Y., Kong, F. D., Xie, Q. Y., Zhou, L. M., Yu, Z. F., & Zhao, Y. X. (2019c). Chemical study of the strain cordyceps spp. From cell fusion between Cordyceps militaris and cordyceps cicadae. Journal of Asian Natural Products Research, 21(5), 449–455. https://doi.org/10.1080/10286020.2018.1451518
  • Yang, X. F., Li, Y. X., He, Y. H., Li, T. T., Wang, W. R., Zhang, J. Y., Wei, J. Y., Deng, Y. H., & Lin, R. (2015). Cordycepin alleviates airway hyperreactivity in a murine model of Asthma by attenuating the inflammatory process. International Immunopharmacology, 26(2), 401–408. https://doi.org/10.1016/j.intimp.2015.04.017
  • Yang, Y., Ji, J., Di, L., Li, J., Hu, L., Qiao, H., Wang, L., & Feng, Y. (2020d). Resource, chemical structure and activity of natural polysaccharides against alcoholic liver damages. Carbohydrate Polymers, 241, 116355. https://doi.org/10.1016/j.carbpol.2020.116355
  • Yao, S., Yang, X., Wu, W., Jiang, Q., Deng, S, Zheng, B., Chen, L., Chen, Y., & Xiang, X. (2023). Effect of paecilomyces cicadae polysaccharide Pc0-1 on cyclophosphamide-induced immunosuppression and regulation of intestinal flora in mice. Food Bioscience, 51. https://doi.org/10.1016/j.fbio.2022.102340
  • Ying, X., Peng, L., Chen, H., Shen, Y., Yu, K., & Cheng, S. (2014). Cordycepin prevented IL-β-induced expression of inflammatory mediators in human osteoarthritis chondrocytes. International Orthopaedics, 38(7), 1519–1526. https://doi.org/10.1007/s00264-013-2219-4
  • Yiyong, C., & Yiting, X. (2018). Purification, chemical characterization and antioxidant activities of a novel polysaccharide from auricularia polytricha. International Journal of Biological Macromolecules, 120, 1087–1092. https://doi.org/10.1016/j.ijbiomac.2018.08.160
  • Yoou, M.-S., Yoon, K. W., Choi, Y., Kim, H.-M., & Jeong, H.-J. (2017a). Cordycepin diminishes thymic stromal lymphopoietin-induced interleukin-13 production. European Journal of Pharmacology, 802, 1–6. https://doi.org/10.1016/j.ejphar.2017.02.033
  • Yu, Y., Shen, M., Song, Q., & Xie, J. (2018). Biological activities and pharmaceutical applications of polysaccharide from natural resources: A review. Carbohydrate Polymers, 183, 91–101. https://doi.org/10.1016/j.carbpol.2017.12.009
  • Yuan, Q. X., Zhang, J., Xiao, C. L., Harqin, C., Ma, M. Y., Long, T., Li, Z. H., Yang, Y. L., Liu, J. K., & Zhao, L. Y. (2020). Structural characterization of a low-molecular-weight polysaccharide from Angelica pubescens Maxim. f. biserrata Shan et Yuan root and evaluation of its antioxidant activity. Carbohydrate Polymers, 236, 116047. https://doi.org/10.1016/j.carbpol.2020.116047
  • Zeng, W. B., Yu, H., Ge, F., Yang, J. Y., Chen, Z. H., Wang, Y. B., Dai, Y. D., & Adams, A. (2014). Distribution of nucleosides in populations of cordyceps cicadae. Molecules, 19(5), 6123–6141. https://doi.org/10.3390/molecules19056123
  • Zhang, H. N., & Lin, Z. B. (2004). Hypoglycemic effect of Ganoderma lucidum polysaccharides. Acta Pharmacologica Sinica, 25(2), 65–69.
  • Zhang, L., Wu, T., Olatunji, O. J., Tang, J., Wei, Y., & Ouyang, Z. (2019a). N6-(2-hydroxyethyl)-adenosine from cordyceps cicadae attenuates hydrogen peroxide induced oxidative toxicity in PC12 cells. Metabolic Brain Disease, 34(5), 1325–1334. https://doi.org/10.1007/s11011-019-00440-1
  • Zhang, M. L., Chai, Y. Q., Fang, M., Zhang, S. S., & Han, B. Y. (2019). N6-(2-hydroxyethyl) adenosine (HEA) isolated from ophiocordyceps sobolifera: Its antihypertensive effect and interaction with human serum albumin. Mycosystema, 38(6), 907–916. https://doi.org/10.13346/j.mycosystema.180299
  • Zhang, Q., Olatunji, O. J., Chen, H., Tola, A. J., & Oluwaniyi, O. O. (2018). Evaluation of the anti-diabetic activity of polysaccharide from cordyceps cicadae in experimental diabetic rats. Chemistry & Biodiversity, 15(8), e1800219. https://doi.org/10.1002/cbdv.201800219
  • Zhang, S. W., & Xuan, L. J. (2007). Five aromatics bearing a 4-O-methylglucose unit from cordyceps cicadae. Helvetica Chimica Acta, 90(2), 404–410. https://doi.org/10.1002/hlca.200790047
  • Zhang, T. Z., Yang, S. H., & Du, J. (2015). The effects of cordycepin on ovalbumin-induced allergic inflammation by strengthening treg response and suppressing Th17 responses in ovalbumin-sensitized mice. Inflammation, 38(3), 1036–1043. https://doi.org/10.1007/s10753-014-0068-y
  • Zhang, X., Li, J., Yang, B., Leng, Q., Li, J., Wang, X., Lu, J., Olatunji, O. J., & Tang, J. (2021a). Alleviation of liver dysfunction, oxidative stress, and inflammation underlines the protective effects of polysaccharides from cordyceps cicadae on high sugar/high fat diet-induced metabolic syndrome in rats. Chemistry & Biodiversity, 18(5), e2100065. https://doi.org/10.1002/cbdv.202100065
  • Zhang, X. F., Hu, Q. B., & Weng, Q. F. (2019b). Secondary metabolites (SMs) of isaria cicadae and isaria tenuipes. RSC Advances, 9(1), 172–184. https://doi.org/10.1039/C8RA09039D
  • Zhang, Y., Liu, Y., Zhou, Y., Zheng, Z., Tang, W., Song, M., Wang, J., & Wang, K. (2021b). Lentinan inhibited colon cancer growth by inducing endoplasmic reticulum stress-mediated autophagic cell death and apoptosis. Carbohydrate Polymers, 267, 118154. https://doi.org/10.1016/j.carbpol.2021.118154
  • Zhang, Y., Wu, Y.-T., Zheng, W., Han, X.-X., Jiang, Y.-H., Hu, P.-L., Tang, Z.-X., & Shi, L.-E. (2017a). The antibacterial activity and antibacterial mechanism of a polysaccharide from cordyceps cicadae. Journal of Functional Foods, 38, 273–279. https://doi.org/10.1016/j.jff.2017.09.047
  • Zhang, Z., Tudi, T., Liu, Y., Zhou, S., Feng, N., Yang, Y., Tang, C., Tang, Q., & Zhang, J. (2016). Preparative isolation of cordycepin, N6-(2-hydroxyethyl)-adenosine and adenosine from Cordyceps militaris by macroporous resin and purification by recycling high-speed counter-current chromatography. Journal of Chromatography B, 1033, 218–225. https://doi.org/10.1016/j.jchromb.2016.08.025
  • Zhao, J., Xie, J., Wang, L. Y., & Li, S. P. (2014). Advanced development in chemical analysis of cordyceps. Journal of Pharmaceutical and Biomedical Analysis, 87, 271–289. https://doi.org/10.1016/j.jpba.2013.04.025
  • Zheng, R., Zhu, R., Li, X., Li, X., Shen, L., Chen, Y., Zhong, Y., & Deng, Y. (2018). N6-(2-Hydroxyethyl) adenosine from cordyceps cicadae ameliorates renal interstitial fibrosis and prevents inflammation via TGF-β1/smad and NF-κB signaling pathway. Frontiers in Physiology, 9, 1229. https://doi.org/10.3389/fphys.2018.01229
  • Zheng, Y., Li, S. Y., Li, C., Shao, Y., & Chen, A. H. (2022). Polysaccharides from spores of cordyceps cicadae protect against cyclophosphamide-induced immunosuppression and oxidative stress in mice. Foods (basel, Switzerland), 11(4), 515–515. https://doi.org/10.3390/foods11040515
  • Zhu, H., Hu, M., Wang, D., Xu, G., Yin, X., Liu, X., Ding, M., & Han, L. (2020a). Mixed polysaccharides derived from shiitake mushroom, poriacocos, ginger, and tangerine peel enhanced protective immune responses in mice induced by inactivated influenza vaccine. Biomedicine & Pharmacotherapy, 126, 110049. https://doi.org/10.1016/j.biopha.2020.110049
  • Zhu, H., Tian, L., Zhang, L., Bi, J., Song, Q., Yang, H., & Qiao, J. (2018). Preparation, characterization and antioxidant activity of polysaccharide from spent lentinus edodes substrate. International Journal of Biological Macromolecules, 112, 976–984. https://doi.org/10.1016/j.ijbiomac.2018.01.196
  • Zhu, J.-S., Halpern, G. M., & Jones, K. (1998a). The scientific rediscovery of a precious ancient Chinese herbal regimen: Cordyceps sinensis part II. The Journal of Alternative and Complementary Medicine, 4(4), 429–457. https://doi.org/10.1089/acm.1998.4.429
  • Zhu, M., Chang, Q., Wong, L. K., Chong, F. S., & Li, R. C. (1999). Triterpene antioxidants from Ganoderma lucidum. Phytotherapy Research: An International Journal Devoted to Pharmacological and Toxicological Evaluation of Natural Product Derivatives, 13(6), 529–531. https://doi.org/10.1002/(sici)1099-1573(199909)13:6<529::aid-ptr481>3.0.co;2-x
  • Zhu, R., Chen, Y. P., Deng, Y. Y., Zheng, R., Zhong, Y. F., Wang, L., & Du, L. P. (2011b). Cordyceps cicadae extracts ameliorate renal malfunction in a remnant kidney model. Journal of Zhejiang University SCIENCE B, 12(12), 1024–1033. https://doi.org/10.1631/jzus.B1100034
  • Zhu, R., Zheng, R., Deng, Y., Chen, Y., & Zhang, S. (2014a). Ergosterol peroxide from cordyceps cicadae ameliorates TGF-β1-induced activation of kidney fibroblasts. Phytomedicine, 21(3), 372–378. https://doi.org/10.1016/j.phymed.2013.08.022
  • Zhu, R., Zheng, R., Deng, Y., Chen, Y., & Zhang, S. (2014b). Ergosterol peroxide from cordyceps cicadae ameliorates TGF-β1-induced activation of kidney fibroblasts. Phytomedicine, 21(3), 372–378. https://doi.org/10.1016/j.phymed.2013.08.022
  • Zhu, X. Y., Chen, X., Xie, J., Wang, P., & Su, W. K. (2012). Mechanochemical-assisted extraction and antioxidant activity of polysaccharides from Ganoderma lucidum spores. International Journal of Food Science & Technology, 47(5), 927–932. https://doi.org/10.1111/j.1365-2621.2011.02923.x
  • Zhu, Y., Yu, X., Ge, Q., Li, J., Wang, D., Wei, Y., & Ouyang, Z. (2020b). Antioxidant and anti-aging activities of polysaccharides from cordyceps cicadae. International Journal of Biological Macromolecules, 157, 394–400. doi:10.1016/j.ijbiomac.2020.04.163