Next Article in Journal
Application of Metabolomics in the Study of Starvation-Induced Autophagy in Saccharomyces cerevisiae: A Scoping Review
Next Article in Special Issue
High Efficiency In Vitro Wound Healing of Dictyophora indusiata Extracts via Anti-Inflammatory and Collagen Stimulating (MMP-2 Inhibition) Mechanisms
Previous Article in Journal
Entomopathogenic Fungi Biodiversity in the Soil of Three Provinces Located in Southwest China and First Approach to Evaluate Their Biocontrol Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomass and Cordycepin Production by the Medicinal Mushroom Cordyceps militaris—A Review of Various Aspects and Recent Trends towards the Exploitation of a Valuable Fungus

by
Dimitrios Kontogiannatos
*,
Georgios Koutrotsios
,
Savvina Xekalaki
and
Georgios I. Zervakis
*
Laboratory of General and Agricultural Microbiology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
*
Authors to whom correspondence should be addressed.
Submission received: 31 October 2021 / Revised: 14 November 2021 / Accepted: 17 November 2021 / Published: 19 November 2021
(This article belongs to the Special Issue Mushrooms—Mycotherapy and Mycochemistry 2.0)

Abstract

:
Cordyceps militaris is an entomopathogenic ascomycete with similar pharmacological importance to that of the wild caterpillar fungus Ophiocordyceps sinensis. C. militaris has attracted significant research and commercial interest due to its content in bioactive compounds beneficial to human health and the relative ease of cultivation under laboratory conditions. However, room for improvement exists in the commercial-scale cultivation of C. militaris and concerns issues principally related to appropriate strain selection, genetic degeneration of cultures, and substrate optimization. In particular, culture degeneration—usually expressed by abnormal fruit body formation and reduced sporulation—results in important economic losses and is holding back investors and potential growers (mainly in Western countries) from further developing this highly promising sector. In the present review, the main factors that influence the generation of biomass and metabolites (with emphasis on cordycepin biosynthesis) by C. militaris are presented and evaluated in conjunction with the use of a wide range of supplements or additives towards the enhancement of fungal productivity in large-scale cultivation processes. Moreover, physiological and genetic factors that increase or reduce the manifestation of strain degeneration in C. militaris are outlined. Finally, methodologies for developing protocols to be used in C. militaris functional biology studies are discussed.

1. Introduction

Cordyceps is a large and diverse genus of the family Cordycipitaceae (Hypocreales, Ascomycota) comprising 627 species according to MycoBank (https://www.mycobank.org; 12 November 2021). They are parasitic fungi, mostly endoparasitoids of insects and other arthropods, while some of them are parasitic on other fungi [1]. Cordyceps was named after the Greek word “kordyle” meaning “club”, and the Latin stem “-ceps” meaning “head” [2]. Most species grow on larvae, pupae, or adults of insects of the orders Arachnida, Coleoptera, Lepidoptera, Hymenoptera, Hemiptera, Orthoptera, Diptera, and Isoptera [3]. Cordyceps militaris (L.) Fr. is a common endoparasitoid of insects, usually growing on pupae and less commonly found on larvae, and is widely distributed in North and South America, Europe, and Asia. Fruit bodies (ascomata) are variable in form and especially in size; this variability is associated with the type of food source(s) available for fungal growth, while it is also affected by the size of the host and the number of stromata formed [3].
Fungi of the genus Cordyceps are among the most important “traditional medicines” since they contain bioactive compounds of high pharmacological value. Fruit bodies are traditionally collected in the wild, while some species are cultivated for the production of mycelium and ascomata [2]. C. militaris is the most studied member of the genus popularly referred to as “Bei Chong Cao” in China. It ranks second (after Ophiocordyceps sinensis) among the most commercialized Cordyceps species in East Asia, where it is used as folk tonic medicine and is considered as a cheaper substitute for O. sinensis [2,4]. Moreover, C. militaris-related products are popular nutraceuticals consumed either as culinary mushrooms or sold in several other forms, e.g., extracts, fermented powder, and tinctures. As regards the medicinal properties that have been attributed to C. militaris, they include hypoglycemic, hypolipidemic, anti-inflammatory, antitumor, antibacterial, antifungal, antiviral, antimalarial, prosexual, neuroprotective, antioxidant, and immuno-protective activities [2,5,6]. Many bioactive molecules of nutraceutical interest, such as cordycepin, ergosterol, trehalose, mannitol and several polysaccharides, nucleosides, and amino acids have been detected in (or isolated from) C. militaris [2,5,7]. Among them, cordycepin is considered as the most valuable and economically important compound produced by this fungus. Particularly noteworthy is the recent discovery of a ProTide modified form of cordycepin named NUC-7738, which presents up to 40 times greater potency for killing cancer cells than the original compound [8]. Such findings will most certainly have to be accompanied/supported by the necessary technological advancements in large-scale C. militaris biomass production to meet increased demand in cordycepin.
C. militaris mycelia and ascomata can be generated ex situ with or without the addition of alive and/or dead insect tissues. A wide range of methodologies to produce fruit bodies and mycelia of C. militaris are reported in pertinent literature including: (a) solid-state fermentation [9,10,11,12], (b) submerged static fermentation [11,13], (c) repeated batch or one-step non-static fermentation in liquid media [14,15,16], and (d) cultivation following larval or pupal infection [17,18]. However, commercial exploitation of C. militaris is still in need of improvement in several aspects related to fruit body production, metabolite biosynthesis, and fungal pathogenicity to target insects. Strain degeneration is the most essential since it significantly influences C. militaris productivity, causing considerable economic losses at the industrial level [19,20]. Several factors are involved in C. militaris degeneration that are either intrinsic/genetic or cultivation-related [20], as presented in the next section.
Furthermore, in order to optimize C. militaris productivity, many parameters have already been examined through numerous pertinent studies that will be suitably outlined and discussed in the present review. Most of them focused on developing suitable methodologies and interventions to avoid (or minimize) culture degeneration [21,22,23,24,25,26,27,28,29,30,31], on improving ascomata and mycelia production [12,15,32,33,34,35,36], and/or on enhancing cordycepin output by adopting various approaches and the use of both solid and liquid substrates [12,15,23,32,37,38,39,40,41,42,43,44,45,46,47]. In addition, the application of genetic engineering breakthrough technologies has resulted in a considerable improvement in C. militaris biomass content in various metabolites, including cordycepin [37,48,49,50,51].
In this review, we present data related to key factors influencing biomass and metabolite production in C. militaris by placing emphasis on culture degeneration, substrate composition, and the use of supplements. Moreover, we discuss genetic and cultivation aspects that affect cordycepin biosynthesis and ascomata formation in C. militaris as well as functional genomics and genetic engineering processes that could substantially enhance the exploitation of this fungus and the generation of value-added products.

2. The Effect of Culture Degeneration on the Yield and Quality of Fruit Bodies Produced by C. militaris

It is widely known that fungi lose virulence and that their morphology is altered when they are successively subcultured on artificial media. Various terms have been used to describe this phenomenon, including phenotypic degeneration, phenotypic instability or deterioration, dual phenomenon, saltation, and attenuation [52,53,54,55,56]. Morphological changes include modifications in color and growth form as well as reduced sporulation [53]. As regards C. militaris in particular, degenerated cultures demonstrate slower mycelial growth, a lighter color of hyphae (probably due to decrease in pigment content), decrease in the number of primordia formed, reduced (or no) ability to produce fruit bodies, longer growth cycle, abnormal fruit body formation, decrease in conidia production, decrease in secondary metabolites content, lower dehydrogenase activity, and reduced cellulase and amylase activities, as well as lower extracellular and higher intracellular polysaccharide content [20,21,22,24,57,58].
He et al. [25] evaluated the effect of mineral elements on colony types of C. militaris and concluded that K+, Ca2+, and Zn2+ (at concentrations of 1.0 g L−1, 0.02 g L−1, and 250–375 μg L−1, respectively) delayed, whereas Mn2+ and Mg2+ (at trace concentrations) promoted the degeneration of C. militaris (Table 1), In addition, degeneration of C. militaris was affected by oxidative stress [20,22], and it was promoted by continuous subculturing, which became particularly evident in the fourth and the fifth generations [20,58]. Furthermore, homokaryosis seems to promote degeneration of C. militaris [1,20,26]; however, degenerated strains could be rejuvenated by cross-mating of their single ascospore isolates [20,24] (Table 1).
In general, few data exist about the effect of the type of substrate (and the nutrients it contains) on degeneration of C. militaris. However, it is expected that this entomopathogenic fungus could benefit by being grown on substrates containing insect cells or tissues since such media emulate better the natural host’s environment. For example, when tussah pupae were inoculated by degenerated cultures of C. militaris, the rate of mycelial growth and the yield of fruit bodies were improved [59]. Although such results indicate that the use of insect tissues could enhance biomass production by C. militaris, further studies are needed to better understand their effect on the cultivation process.
A rather limited amount of information is available about molecular mechanisms affecting C. militaris culture degeneration. Apparently mating-type (MAT) loci allele segregation—through continuous culture—is closely related to the manifestation of this phenomenon [37,58,60,61,62,63,64]. In addition, gene mutation [31] and DNA methylation [27,30] also seem to be involved by inducing strain degeneration. Yin et al. [58] performed a transcriptome-wide analysis during C. militaris subculturing and found that genes implicated in detoxification and stress response mechanisms (such as those involving the production of streptothricinacetyltransferase, gamma-glutamyltranspeptidase, MFS multidrug transporter, glutathione S-transferase, alcohol dehydrogenase, and the 30 kDa heat shock protein) were up-regulated during strain degeneration. Similarly, expression of genes related to the metabolism of carbohydrates, lipids, proteins, amino acids, nucleic acids, and nucleotides were significantly up-regulated; these included genes encoding for the mitochondrial hypoxia responsive protein, the mitochondrial co-chaperone GrpE, the glycoside hydrolase, the trypsin-like serine protease, the metalloprotease 1, the acetate transporter, the SGT1 and CS proteins, the formyltetrahydrofolate deformylase, the nucleoside triphosphate hydrolases, and uracil phosphoribosyltransferase [58]. On the basis of these findings, the strain degeneration mechanism in C. militaris is associated with genes involved in energy metabolism, toxin biosynthesis, DNA methylation, and chromosome remodeling [58].
DNA methylation refers to the transfer of methyl groups to a specific base by DNA methyltransferase, where S-adenosylmethionine acts as the methyl donor [65]. DNA methylation plays a key role in eukaryotic gene silencing and expression, phylogeny, cell differentiation, and other processes in eucaryotic organisms [66]. As regards fungi in particular, DNA methylation levels are relatively low, but of importance in degeneration phenomena observed after repeated subculturing, as is the case with C. militaris. Xin et al. [27] used a comparative bisulfite sequencing (BS-Seq) approach between a normal wild-type C. militaris strain and a degenerated one (incapable of producing fruit bodies) to establish that DNA the methylation level in the wild-type C. militaris was 0.48%; this value is lower than this of Neurospora crassa (1.5%) [67] but higher than that of Magnaporthe grisea (0.22%) [68]. Moreover, the methylation level of the degenerated type was higher than that of the wild-type (0.56% vs. 0.48%), while the degeneration of C. militaris cultures was associated with modifications in the pyruvate and glycerophospholipid metabolic, ubiquitin-mediated proteolysis, and N-glycan biosynthetic pathways [27].

3. Substrates, Nutrient Requirements and Treatments Related with the Cultivation of C. militaris

Despite the huge medicinal and economic value of C. militaris [2,69,70] there are relatively few studies addressing cultivation performance under various conditions. In addition, most of them focused on the low-scale laboratory production of C. militaris in order to examine factors affecting cordycepin biosynthesis but did not focus on large-scale industrial production, where parameters such as cost-effectiveness, substrate efficiency, cultivation time, and scalability should be also taken into consideration.
Growth requirements under various cultivation conditions of C. militaris were recently assessed by using a genome-scale modeling (GEM) approach, which revealed interesting data about the core biochemical pathways and metabolic routes for this fungus [32]. C. militaris retains a conserved metabolism for efficiently producing ATP by mitochondrial oxidative phosphorylation in response to high-altitude environments. Moreover, it was found that C. militaris contains genes converting various hormones (e.g., serotonin, adrenaline, and dopamine) into amino acids; this is in accordance with the fact that in nature, C. militaris assimilates nitrogen and carbon from the larval corpus with high content in juvenile and neuronal hormones such as serotonin and dopamine (Table 2). Raethong et al. [32] concluded that C. militaris: (a) exhibit the highest growth rate on sucrose-based media, (b) could utilize a wide range of carbon and nitrogen sources, e.g., chitin, glucosamine, and gamma-aminobutyrate (GABA) under CN-limited conditions, (c) could not use cellulose as sole carbon source, (d) could utilize various nutrients and redirect them through the ammonia metabolism, while nitrogen content has a significant effect on the production of extracellular cordycepin, and (e) has an optimal C/N ratio for growth that is 12.7:1 (Table 2). It is worth noting that chitin and its derivatives are major components of insect exoskeletons, while rice grain is rich in GABA [32]. In line with the aforementioned findings, it was observed that chitosan (a chemical homologue of chitin) could significantly increase carotenoid content in a novel two-stage C. militaris cultivation process [71] (Table 2). In addition, low-nutrient growth media supplemented with insect cuticles increased the pathogenicity of Beauveria bassiana, Cordyceps javanica, and Metarhizium robertsii [33]. These effects are yet to be evaluated for C. militaris, but existing results have already proven a positive impact of insect-based compounds on its growth and productivity.
Considering fruit bodies production, a cost-effective substrate is mandatory in the development of relevant commercial applications. By-products of the agricultural and agro-industrial sectors are cheap, abundant, and suitable resources for the cultivation of edible and medicinal mushrooms [72,73,74,75]. However, relatively few studies have dealt with the production of C. militaris ascomata and bioactive compounds on such substrates. For example, the use of cottonseed shells or corn cobs at ratios of 8:1:1 (w/w/w) to wheat bran and rice resulted in higher fruit bodies yield compared with the use of conventional rice media, while corn cobs produced fruit bodies with the highest cordycepin content [12] (Table 2). In addition, it was observed that the addition of vegetable oils—such as those derived from soybean, peanut, rapeseed, olive, and palm, corn, and sunflower seeds—in static cultures of C. militaris significantly promoted mycelium growth; furthermore, peanut oil significantly increased cordycepin content as well [15] (Table 2). Starch-processing waste also proved to be suitable for mycelium growth of C. militaris under solid-state and submerged cultivation, but no information was provided regarding fruit bodies production in this substrate [11] (Table 2).
Hence, it could be concluded that C. militaris benefits from substrates that emulate (or relate to) its insect hosts in nature. Therefore, a question arises as to whether plant-based media can be used alone for C. militaris cultivation or if insect-based supplements should be also added. Guo et al. [17] showed that there was no significant difference in yields of C. militaris following cultivation in wheat-based substrates and by injecting live Bombyx mori pupae. On the other hand, Wang et al. [34] observed significant variability in C. militaris mycelia produced on various liquid media and in fruit bodies cultivated on B. mori or rice-based substrates. The same authors detected the highest cordycepin content (13.43 mg g−1) in C. militaris fruit bodies produced after injection of fifth instar silkworm larvae 33 days post-infection. Furthermore, the outcome of ongoing studies revealed the beneficial effect of using rice-based substrates supplemented with insect tissues in the production of C. militaris ascomata (Figure 1) [76].
Other factors that influence C. militaris productivity include the use of low concentrations of fluoride (0.01 mM), which promoted fungal growth, increased superoxide dismutase-like activity, and enhanced biomass content in bioactive substances, including carotenoids [35] (Table 2). Moreover, extracts from C. militaris fruit bodies produced with the addition of fluoride exhibited a stronger anti-proliferation effect on U2OS cancer cells [35] (Table 2).
Higher yields and biological efficiencies together with shorter fruit body maturation periods were observed in two degenerated C. militaris strains when their mycelia growing on the substrates surface were mechanically scratched by a metal scraper [36]. This scratching technique was used at various mycelium growth stages: (a) when mycelia entirely colonized the substrates eight days post inoculation in the dark, (b) when mycelia turned light yellow three days after the onset of illumination, and (c) when mycelia turned yellow and became twisted six days after the onset of illumination. Liu et al. [36] showed that scratching C. militaris-degenerated mycelia at these three vegetative stages and before stromata differentiation succeeded in shortening the period needed for growth of C. militaris fruit bodies by at least five days (Table 2). In addition, it was suggested that fruit bodies production by C. militaris-degenerated strains may be related to the Rhf1 gene and the active oxygen-scavenging genes [36].

4. Genetics, Genomics, and Genetic Engineering of C. militaris

Genetic improvement of C. militaris is of high importance in order to create new strains with high commercial potential. As stated before, a breeder has to overcome several obstacles when attempting to improve C. militaris cultivation aspects, e.g., cultures degeneration, optimization of biomass and cordycepin production, reduction in time needed for fruit body maturation, determination of nutritional and environmental requirements, and assessment of fruit body composition. So far, C. militaris can be selectively improved by using traditional breeding techniques, and these are mainly restricted to reversing degenerative phenotypes to ascomata-producing strains. Fungal sexual reproductive systems are conservatively controlled by mating-type (MAT) loci [37,60,61]. Alternative sequences occupying the same chromosomal location at the MAT loci vary and have thereby been called idiomorphs or mini sex chromosomes [37,61,77,78]. It was suggested that the MAT genes play both convergent and divergent roles in mediating stroma development and fertility in C. militaris compared with the functions of MAT orthologs in other fungi [37]. Determination and comparison of cordycepin and adenosine production in fertile and sterile fruit bodies suggested that the fungal sexual process utilizes a significant amount of energy, thereby reducing the quality of the mushroom [37]. The mating types of C. militaris are controlled by a pair of alleles: MAT1–1 (MAT1–1–1, MAT1–1–2) and MAT1–2 (MAT1–2–1) [58,62,63]. Parent strains containing both MAT1–1 and MAT1–2 are considered heterokaryons, while parent strains containing only MAT1–1 or MAT1–2 are considered homokaryons [58,63]. Li et al. [64] by using Random Amplified Polymorphic DNA (RAPD) Polymerase Chain Reaction (PCR) analyses found that all strains incapable of developing normal fruit bodies were homokaryons, while all strains with normally developed fruit bodies were heterokaryons. Moreover, genotyping of degenerated C. militaris strains revealed that these strains contained a completely deleted MAT1–2–1 region, which resulted in CmMAT1–2–1 knockdown, base substitutions in the MAT1–1–1 and MAT1–2–1 regions, and CmMAT1–1–1 and CmMAT1–1–2 reduced expression [58]. Mating-based strain improvement has been achieved by crossing hyphae from spores of MAT1–1 and MAT1–2 idiomorphs, resulting in strains with high cordycepin content [79]. Moreover cross-mating with different mating types (rather than self-mating or crossing with the same mating types) decreases heterothallic limitation and increases the efficiency of industrial-scale C. militaris production [80].
Functional genomic studies may shed more light on factors affecting the development of C. militaris ascomata. Polyethylene glycol (PEG)-mediated transformation of C. militaris mononuclear protoplasts has been performed in order to knock-down a terpenoid synthase (Tns) gene using a glufosinate ammonium selection marker [48,49]. Using the same knockdown technique, it was revealed that the flavohemoprotein-like Cmfhp gene of C. militaris is involved in fruit body and conidia production, while it also affects nitric oxide (NO) and carotenoid contents [50]. Moreover, Agrobacterium tumefaciens-mediated transformation of C. militaris has been used as a tool for insertional mutagenesis [81] and complementation of ΔCmfhp mutants with the wild-type Cmfhp gene [50]. Agrobacterium-mediated transformation of C. militaris has also been used as a technique for deleting MAT genes in single mating-type isolates of this fungus [37]. Lu et al. [37] showed that MAT1–1 and MAT1–2–1 null mutants were sterile and lost the ability to produce stromata in outcrosses with the opposite mating-type partner, while MAT1–1–1 produced barren stromata in outcrosses. Additionally, MAT1–1–2-generated fruit bodies were morphologically similar to that of the parent strain but with sterile perithecia [37]. The homothallic-like transformants MAT1–2:MAT1–1–1 (haploidic MAT1–2 isolate transformed with the MAT1–1–1 gene) produced sterile stromata, whereas the MAT1–1:MAT1–2–1 (haploidic MAT1–1 isolate transformed with the MAT1–2–1 gene) mutant lost the ability to produce ascomata [37]. The findings relating to the fully fertile gene-complementation mutants suggested that the genomic location is not essential for the MAT genes to fulfill their functions in C. militaris [37].
The clustered regularly interspaced short palindromic repeats (CRISPR) system is an innovative and efficient genome-editing tool that allows gene knockout, insertion, and replacement in almost all eukaryotic systems. Efficient CRISPR in C. militaris was achieved by creating a Cas9-stably transformed strain via Agrobacterium-mediated transformation and then by delivering of a presynthesized sgRNA targeting the ura3 gene by PEG-mediated protoplast transformation [51]. In the same study it was reported that this method was superior to co-transformation of a single vector expressing a sgRNA-cmcas9 cassette, providing encouraging results for future interventions (genetic engineering) in the cordycepin biosynthesis pathway. A schematic overview of genetic engineering approaches used in C. militaris is provided in Figure 2.
C. militaris produces water-soluble carotenoid-containing pigments that are responsible for the characteristic yellow or orange color of the fruit bodies formed [82,83]. Although C. militaris pigments are secondary metabolites whose production is induced by light, not all light-induced genes are implicated in carotenoid biosynthesis. In a comparative transcriptome analysis approach using C. militaris mycelia exposed to light or darkness, Lou et al. [84] found a total of 1722 differentially expressed DEGs between these two conditions; therefore, only two genes, i.e., CCM_06728 (CAO-2) and CCM_09155 (YLO-1), could be associated with the biosynthesis of carotenoids. Unexpectedly, the expression levels of these genes were not significantly different between mycelia exposed to light and darkness [84]. The terpenoid synthase gene Cmtns seems to be involved in carotenoid biosynthesis of this fungus. Moreover, ΔCmtns C. militaris strains [48] showed a decrease in carotenoid content which was reversed by an Agrobacterium-mediated Cmtns complementation approach [84]; the reduced size of fruit bodies produced suggests that Cmtns might be a multifunctional gene.
C. militaris represents a valuable resource for isolating new natural compounds as products of its biosynthetic activity. The cyclodepsipeptide beauverolides were initially isolated and characterized from the entomopathogenic fungus Beauveria tenella in 1975 [85]; among them, beauveriolide I showed moderate insecticidal activities [86], while beauveriolide I and III showed specific inhibition of lipid droplet formation in mouse macrophages [87,88]. Moreover, beauveriolides inhibited acyl-CoA:cholesterol acyltransferases (ACATs) selectively, leading to a block in cholesteryl esters biosynthesis. Moreover, they led to a decrease in cholesterol concentration with no visible cytotoxic activities, making them candidate compounds for the production of antiatherosclerotic agents [89]. By using a genome mining approach on beauveriolides from C. militaris CM01, Wang et al. [90] identified one four-gene cluster (named cm3) related to cyclodepsipeptide biosynthesis in this fungus. After heterologous expression of the entire cm3 cluster into Aspergillus nidulans through the use of a protoplast transformation approach, beauveriolides I and III were detected at a concentration of 13 and 18 mg/L, respectively [90]. In the same study, the presence of both beauveriolides was evidenced in commercially produced fruit bodies of C. militaris.

5. Molecular, Nutritional, and Environmental Aspects of Cordycepin Biosynthesis and Ascomata Formation

As already mentioned, C. militaris produces a large number of bioactive metabolites, including ergothioneine, ergosterol, adenosine, polysaccharides, and cordycepin [14,91,92,93,94]. Among them, cordycepin exhibits a large range of health beneficial effects, such as broad-spectrum antibiotic activity and the ability to inhibit cell proliferation and to induce cell apoptosis, as well as having antioxidant, anticancer, and anti-inflammatory properties [95,96,97,98,99]. It is currently considered to be one of the most promising fungal compounds with respect to its pharmacological and therapeutic potential; therefore, the study of aspects related to cordycepin biosynthesis and ascomata production is essential to the commercial exploitation of this species.
The basic metabolic route for cordycepin biosynthesis has been recently explored using a combination of in silico analyses and functional genomics approaches [100]. Four genes for cordycepin synthetase were identified in C. militaris genome and were designated as cns1cns4; these genes contain different conserved domains, such as the oxidoreductase/dehydrogenase domain in Cns1, the HDc family of metal-dependent phosphohydrolase domain in Cns2, and the N-terminal nucleoside/nucleotide kinase (NK) and C-terminal HisG domains in Cns3, while Cns4 is a putative ATP-binding cassette type of transporter. Using Agrobacterium-mediated single and joint gene deletion mutants of the aforementioned genes, Xia et al. [100] demonstrated that Cns1 and Cns2 are required for cordycepin biosynthesis, while Cns3 is implicated in the biosynthesis of an additional “safeguard” molecule, i.e., pentostatin. In addition, the same authors reported that cordycepin biosynthesis (starting from adenosine and proceeding through the stepwise reactions of phosphorylation, dephosphorylation, and reduction) is catalyzed by the Cns1/Cns2 complex in parallel with the biosynthesis of pentostatin. This dual production is similar to the bacterial “protector-protégé strategy” of purine metabolism in which the safeguarded cordycepin can be deaminated to 30-deoxyinosine once the former reaches a self-toxic level in fungal cells [100].
Sensing light is considered a signal for morphogenesis and metabolite production in fungi [82]. In the heterothallic filamentous fungus Neurospora crassa, the white collar (WC) complex consisting of the WC-1 and WC-2 proteins is the sensor for blue light [101]. WC-1 is a transcription factor implicated in all known blue light responses, including mycelial carotenogenesis, perithecial beak phototropism, circadian rhythms of conidiation, sexual development, and circadian clock resetting [82,102,103,104,105,106,107]. WC-1 contains a zinc finger DNA-binding domain, glutamine-rich putative transcription activation and protein–protein interaction domains, a nuclear localization signal, and a chromophore-binding domain [82,101]. WC-1 and WC-2 interact through the protein–protein interaction domains to form the functional white collar complex that binds to the promoters of light-regulated genes to rapidly activate transcription in response to light [82,104]. In C. militaris, Cmwc-1 deletion resulted in disordered fruit body development and a decrease in conidial production, while it also led to a significant reduction in pigmentation as well as in cordycepin production; in addition, it affected spore formation and secondary metabolite production [82]. Moreover, in Cmwc-1-deletion mutants, 166 common genes were differentially expressed in response to light compared with the wild-type Cmwc-1. C. militaris was also found to contain homologs of the CmWC-2, VVD, PHY, and CRY (CRY-1, CRY-2, and CPD photolyase) genes, which confirms the importance of light perception in fruit body development [82]. Additionally, disruption of Cmcry-DASH resulted in completely different phenotypes than those of ΔCmwc-1, while Cmcry-DASH expression was strongly induced by light in a CmWC-1-dependent manner [108].
Albinism has also been implicated in the productivity of C. militaris. In a spontaneous albino mutation, with no color formation in either mycelia or ascomata, it was reported that albino mutants presented reduced conidial production and deformed fruit bodies of whitish color [109]. Comparative transcriptome analysis under light stress response showed that many more genes were expressed in the albino strain to reduce light impairment and that the significantly overexpressed pathways in the albino mutant were mainly involved in replication and repair mechanisms [109]. In addition, the expression levels of some secondary metabolite backbone genes were found to be differentially expressed by over 2-fold in the albino compared with the normal strains, e.g., the Zn2Cys6 transcription factor and post-modification enzymes.
As previously mentioned with light stress, heat stress also influences cordycepin biosynthesis in C. militaris. It was suggested that during the late maturation stage of ascomata, heat and light stresses lead to a significant increase in cordycepin biosynthesis without affecting biological efficiency and that heat stress significantly promotes carotenoid production [110]. Moreover, it was observed that the optimal growth temperature for C. militaris is 20 °C on agar medium, while growth at 25 °C is compromised.
miRNA-like RNAs (milRNAs) have been discovered in various species of fungi with very little information with respect to their functions. RNA-dependent RNA polymerase (RDRP), argonaute (AGO), and Dicer are differentially expressed at different developmental stages of C. militaris, supporting the idea that the miRNAs pathways are present in this species [111]. High-throughput sequencing revealed the expression of 38 novel milRNAs; 19 are exclusively expressed in the sexual developmental stage, while the other 19 are expressed in both asexual and sexual developmental stages [111]. Two of them—the milR4 and milR16—were knocked out and over-expressed in C. militaris, and only the wild type and the overexpressed milR16 produced normal primordia and mature fruit bodies. Interestingly, yellow mycelia fully colonized pupae inoculated with the knocked miR-4 at 21 days post-injection, but this mutant was unable to form normal primordia.
A great variability in cordycepin content in ascomata and mycelia of C. militaris was assessed when pertinent literature was examined. Despite intense efforts to improve cordycepin production by C. militaris by modifying cultivation conditions and/or supplementing substrates with various nutrients, no widely accepted methodology has yet been established that could lead to consistently high cordycepin levels in the biomass obtained. Consequently, large fluctuations are commonly observed in cordycepin values measured either in mycelia generated from liquid fermentation (i.e., from 30 mg L−1 up to 8570 mg L−1) or in cultivated ascomata (i.e., from 0.6 mg g−1 up to 77.4 mg g−1) (Table 3). Such variability is attributed to various factors, one of the most important being the strain used. As it was previously presented, the genetic makeup of C. militaris seems to have a tremendous impact on both the ability to form ascomata and—most importantly—on the cordycepin biosynthetic capability of each individual strain [37,79,109] (Table 3.). Moreover, the analytical methodology adopted and the cordycepin extraction protocols employed to quantify cordycepin also confer highly diverse values that are difficult to interpret and associate with the fungus and the cultivation conditions used [38,39,112,113,114] (Table 3). Last, substrate composition could also exert a significant effect on cordycepin production, as evidenced by the relevant cases reported in Table 3 [40,41,42].

6. Conclusions

The ethnopharmacological importance of C. militaris has been widely analyzed in the past by many researchers [2,10,13,70,122,123,124] since this fungus is considered to be a valuable source of metabolites that act directly on various human metabolic pathways. For example, the methanolic extract of C. militaris presented antioxidant, antibacterial, antifungal, and antiproliferative properties in different human tumor cell lines [122], while cordycepin, the major bioactive compound of C. militaris, presented potent anti-inflammatory, anticancer, anti-metastatic, and immune-modulatory activities [123,124,125,126,127]. Although C. militaris can be easily cultivated in rice-based media, particular requirements related to the physiology and genetic makeup of this fungus impose serious obstacles in commercial applications. Hence, strain degeneration can completely hinder fruit body production at a commercial-scale, while improper cultivation techniques can downgrade metabolite production. Because of the entomopathogenic nature of C. militaris, the use of insect-based substrates in large-scale cultivation projects must be taken into consideration. During the past few years, insect mass production systems have become a contemporary trend in organic waste treatment and biotransformation [128], while the insect-based pet feed industry is growing exponentially. Considering the increasing availability of such by-products, the cultivation practices of C. militaris could exploit this type of waste material in the frame of the circular economy concept.

Author Contributions

Conceptualization, D.K. and G.I.Z.; investigation, D.K., S.X. and G.K.; resources, D.K., G.K. and G.I.Z.; writing—original draft preparation, D.K. and G.K.; writing—review and editing, D.K. and G.I.Z.; project administration, G.I.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was co-financed by Greece and the European Union (European Social Fund–ESF) through the Operational Programme “Human Resources Development, Education and Lifelong Learning” in the context of the project “Reinforcement of Postdoctoral Researchers–2nd Cycle” (MIS-5033021), implemented by the State Scholarships Foundation (ΙΚΥ).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nikoh, N.; Fukatsu, T. Interkingdom Host Jumping Underground: Phylogenetic Analysis of Entomoparasitic Fungi of the Genus Cordyceps. Mol. Biol. Evol. 2000, 17, 629–638. [Google Scholar] [CrossRef] [PubMed]
  2. Olatunji, O.J.; Tang, J.; Tola, A.; Auberon, F.; Oluwaniyi, O.; Ouyang, Z. The Genus Cordyceps: An Extensive Review of Its Traditional Uses, Phytochemistry and Pharmacology. Fitoterapia 2018, 129, 293–316. [Google Scholar] [CrossRef] [PubMed]
  3. Mains, E.B. North American Entomogenous Species of Cordyceps. Mycologia 1958, 50, 169–222. [Google Scholar] [CrossRef]
  4. Wang, M.; Meng, X.Y.; Le Yang, R.; Qin, T.; Wang, X.Y.; Zhang, K.Y.; Fei, C.Z.; Li, Y.; Hu, Y.L.; Xue, F.Q. Cordyceps militaris Polysaccharides Can Enhance the Immunity and Antioxidation Activity in Immunosuppressed Mice. Carbohydr. Polym. 2012, 89, 461–466. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, J.; Wen, C.; Duan, Y.; Zhang, H.; Ma, H. Advance in Cordyceps militaris (Linn) Link Polysaccharides: Isolation, Structure, and Bioactivities: A Review. Int. J. Biol. Macromol. 2019, 132, 906–914. [Google Scholar] [CrossRef]
  6. Das, G.; Shin, H.-S.; Leyva-Gómez, G.; Prado-Audelo, M.L.D.; Cortes, H.; Singh, Y.D.; Panda, M.K.; Mishra, A.P.; Nigam, M.; Saklani, S. Cordyceps spp.: A Review on Its Immune-Stimulatory and Other Biological Potentials. Front. Pharmacol. 2021, 11, 2250. [Google Scholar] [CrossRef] [PubMed]
  7. Zhong, X.; Gu, L.; Xiong, W.-T.; Wang, H.-Z.; Lian, D.-H.; Zheng, Y.-M.; Zhou, S.; Zhou, W.; Gu, J.-L.; Shen, J.-H. 1H NMR Spectroscopy-Based Metabolic Profiling of Ophiocordyceps sinensis and Cordyceps militaris in Water-Boiled and 50% Ethanol-Soaked Extracts. J. Pharm. Biomed. Anal. 2020, 180, 113038. [Google Scholar] [CrossRef]
  8. Schwenzer, H.; De Zan, E.; Elshani, M.; Van Stiphout, R.; Kudsy, M.; Morris, J.; Ferrari, V.; Um, I.H.; Chettle, J.; Kazmi, F. The Novel Nucleoside Analogue ProTide NUC-7738 Overcomes Cancer Resistance Mechanisms in Vitro and in a First-in-Human Phase 1 Clinical Trial. Clin. Cancer Res. 2021. [Google Scholar] [CrossRef]
  9. Rao, Y.K.; Fang, S.-H.; Wu, W.-S.; Tzeng, Y.-M. Constituents Isolated from Cordyceps militaris Suppress Enhanced Inflammatory Mediator’s Production and Human Cancer Cell Proliferation. J. Ethnopharmacol. 2010, 131, 363–367. [Google Scholar] [CrossRef]
  10. Chiu, C.-P.; Liu, S.-C.; Tang, C.-H.; Chan, Y.; El-Shazly, M.; Lee, C.-L.; Du, Y.-C.; Wu, T.-Y.; Chang, F.-R.; Wu, Y.-C. Anti-Inflammatory Cerebrosides from Cultivated Cordyceps militaris. J. Agric. Food Chem. 2016, 64, 1540–1548. [Google Scholar] [CrossRef]
  11. Lee, J.; Cho, K.; Shin, S.G.; Bae, H.; Koo, T.; Han, G.; Hwang, S. Nutrient Recovery of Starch Processing Waste to Cordyceps militaris: Solid State Cultivation and Submerged Liquid Cultivation. Appl. Biochem. Biotechnol. 2016, 180, 274–288. [Google Scholar] [CrossRef]
  12. Lin, Q.; Long, L.; Wu, L.; Zhang, F.; Wu, S.; Zhang, W.; Sun, X. Evaluation of Different Agricultural Wastes for the Production of Fruiting Bodies and Bioactive Compounds by Medicinal Mushroom Cordyceps militaris. J. Sci. Food Agric. 2017, 97, 3476–3480. [Google Scholar] [CrossRef]
  13. Cui, J.D. Biotechnological Production and Applications of Cordyceps militaris, a Valued Traditional Chinese Medicine. Crit. Rev. Biotechnol. 2015, 35, 475–484. [Google Scholar] [CrossRef] [PubMed]
  14. Zeng, Y.; Han, Z.; Qiu, P.; Zhou, Z.; Tang, Y.; Zhao, Y.; Zheng, S.; Xu, C.; Zhang, X.; Yin, P. Salinity-Induced Anti-Angiogenesis Activities and Structural Changes of the Polysaccharides from Cultured Cordyceps militaris. PLoS ONE 2014, 9, e103880. [Google Scholar]
  15. Tang, J.; Qian, Z.; Wu, H. Enhancing Cordycepin Production in Liquid Static Cultivation of Cordyceps militaris by Adding Vegetable Oils as the Secondary Carbon Source. Bioresour. Technol. 2018, 268, 60–67. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, C.-C.; Wu, J.-Y.; Chang, C.-Y.; Yu, S.-T.; Liu, Y.-C. Enhanced Exopolysaccharide Production by Cordyceps militaris Using Repeated Batch Cultivation. J. Biosci. Bioeng. 2019, 127, 499–505. [Google Scholar] [CrossRef]
  17. Guo, M.; Guo, S.; Huaijun, Y.; Bu, N.; Dong, C. Comparison of Major Bioactive Compounds of the Caterpillar Medicinal Mushroom, Cordyceps militaris (Ascomycetes), Fruiting Bodies Cultured on Wheat Substrate and Pupae. Int. J. Med. Mushrooms 2016, 18, 327–336. [Google Scholar] [CrossRef]
  18. Shi, K.; Yang, G.; He, L.; Yang, B.; Li, Q.; Yi, S. Purification, Characterization, Antioxidant, and Antitumor Activity of Polysaccharides Isolated from Silkworm Cordyceps. J. Food Biochem. 2020, 44, e13482. [Google Scholar] [CrossRef]
  19. Zheng, Z.; Qiu, X.; Han, R. Identification of the Genes Involved in the Fruiting Body Production and Cordycepin Formation of Cordyceps militaris Fungus. Mycobiology 2015, 43, 37–42. [Google Scholar] [CrossRef] [Green Version]
  20. Lou, H.; Lin, J.; Guo, L.; Wang, X.; Tian, S.; Liu, C.; Zhao, Y.; Zhao, R. Advances in Research on Cordyceps militaris Degeneration. Appl. Microbiol. Biotechnol. 2019, 103, 7835–7841. [Google Scholar] [CrossRef]
  21. Lin, Q.; Qiu, X.; Zheng, Z.; Xie, C.; Xu, Z.; Han, R. Characteristics of the Degenerate Strains of Cordyceps militaris. Mycosystema 2010, 29, 670–677. [Google Scholar]
  22. Xiong, C.; Xia, Y.; Zheng, P.; Wang, C. Increasing Oxidative Stress Tolerance and Subculturing Stability of Cordyceps militaris by Overexpression of a Glutathione Peroxidase Gene. Appl. Microbiol. Biotechnol. 2013, 97, 2009–2015. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, B.-X.; Wei, T.; Xue, L.-N.; Zheng, Q.-W.; Ye, Z.-W.; Zou, Y.; Yang, Y.; Yun, F.; Guo, L.-Q.; Lin, J.-F. Transcriptome Analysis Reveals the Flexibility of Cordycepin Network in Cordyceps militaris Activated by L-Alanine Addition. Front. Microbiol. 2020, 11, 577. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, A.; Wang, Y.; Shao, Y.; Huang, B. A Novel Technique for Rejuvenation of Degenerated Caterpillar Medicinal Mushroom, Cordyceps militaris (Ascomycetes), a Valued Traditional Chinese Medicine. Int. J. Med. Mushrooms 2017, 19, 87–91. [Google Scholar] [CrossRef]
  25. He, L.; Han, C.; Li, P.; Chen, Y.; Liu, D.; Geng, L. Effect of Mineral Elements on Colony Types of Cordyceps militaris in Subculturing. J. Shenyang Agric. Univ. 2009, 40, 672–677. [Google Scholar]
  26. Wang, H.; Wei, J.; Lin, N.; Feng, A.; Chen, M.; Bao, D. Distribution of Mating-Type Genes in Fruiting and Non-Fruiting Forms of Cordyceps militaris. Acta Edulis Fungi 2010, 17, 1–4. [Google Scholar]
  27. Xin, X.; Yin, J.; Zhang, B.; Li, Z.; Zhao, S.; Gui, Z. Genome-Wide Analysis of DNA Methylation in Subcultured Cordyceps militaris. Arch. Microbiol. 2019, 201, 369–375. [Google Scholar] [CrossRef]
  28. Sun, H.; Hu, T.; Guo, Y.; Liang, Y. Preservation Affects the Vgetative Growth and Fruiting Body Production of Cordyceps militaris. World J. Microbiol. Biotechnol. 2018, 34, 166. [Google Scholar] [CrossRef]
  29. Lee, H.-H.; Kang, N.; Park, I.; Park, J.; Kim, I.; Kim, J.; Kim, N.; Lee, J.-Y.; Seo, Y.-S. Characterization of Newly Bred Cordyceps militaris Strains for Higher Production of Cordycepin through HPLC and URP-PCR Analysis. J. Microbiol. Biotechnol. 2017, 27, 1223–1232. [Google Scholar] [CrossRef] [Green Version]
  30. Wang, Y.; Wang, Z.; Liu, C.; Wang, S.; Huang, B. Genome-Wide Analysis of DNA Methylation in the Sexual Stage of the Insect Pathogenic Fungus Cordyceps militaris. Fungal Biol. 2015, 119, 1246–1254. [Google Scholar] [CrossRef]
  31. Meina, L.; Chunyan, L.; Bosen, F.; Qingwei, L. Molecular Analysis of Degeneration of Artificial Flanted Cordyceps militaris. Jun Wu Xi Tong Mycosystema 2003, 22, 277–282. [Google Scholar]
  32. Raethong, N.; Wang, H.; Nielsen, J.; Vongsangnak, W. Optimizing Cultivation of Cordyceps militaris for Fast Growth and Cordycepin Overproduction Using Rational Design of Synthetic Media. Comput. Struct. Biotechnol. J. 2020, 18, 1–8. [Google Scholar] [CrossRef] [PubMed]
  33. Montesinos-Matías, R.; Ordaz-Hernández, A.; Angel-Cuapio, A.; Colin-Bonifacio, Y.; Garcia-Garcia, R.E.; Ángel-Sahagún, C.A.; Arredondo-Bernal, H.C. Principal Component Analysis of the Biological Characteristics of Entomopathogenic Fungi in Nutrient-limited and Cuticle-based Media. J. Basic Microbiol. 2021, 61, 147–156. [Google Scholar] [CrossRef]
  34. Wang, L.-Y.; Liang, X.; Zhao, J.; Wang, Y.; Li, S.-P. Dynamic Analysis of Nucleosides and Carbohydrates during Developmental Stages of Cordyceps militaris in Silkworm (Bombyx mori). J. AOAC Int. 2019, 102, 741–747. [Google Scholar] [CrossRef]
  35. Li, X.; Wang, J.; Zhang, H.; Xiao, L.; Lei, Z.; Kaul, S.C.; Wadhwa, R.; Zhang, Z. Low Dose of Fluoride in the Culture Medium of Cordyceps militaris Promotes Its Growth and Enhances Bioactives with Antioxidant and Anticancer Properties. J. Fungi 2021, 7, 342. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, G.-Q.; Qiu, X.-H.; Cao, L.; Han, R.-C. Scratching Stimuli of Mycelia Influence Fruiting Body Production and ROS-Scavenging Gene Expression of Cordyceps militaris. Mycobiology 2018, 46, 382–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Lu, Y.; Xia, Y.; Luo, F.; Dong, C.; Wang, C. Functional Convergence and Divergence of Mating-Type Genes Fulfilling in Cordyceps militaris. Fungal Genet. Biol. 2016, 88, 35–43. [Google Scholar] [CrossRef]
  38. Li, W.; Zhao, S.; Chen, H.; Yuan, H.; Wang, T.; Huang, X. High-Yielding Cordycepin in Cordyceps militaris Modified by Low-Energy Ion Beam. Sheng Wu Gong Cheng Xue Bao Chin. J. Biotechnol. 2009, 25, 1725–1731. [Google Scholar]
  39. Singpoonga, N.; Rittiron, R.; Seang-On, B.; Chaiprasart, P.; Bantadjan, Y. Determination of Adenosine and Cordycepin Concentrations in Cordyceps militaris Fruiting Bodies Using Near-Infrared Spectroscopy. ACS Omega 2020, 5, 27235–27244. [Google Scholar] [CrossRef]
  40. Fan, D.; Wang, W.; Zhong, J.-J. Enhancement of Cordycepin Production in Submerged Cultures of Cordyceps militaris by Addition of Ferrous Sulfate. Biochem. Eng. J. 2012, 60, 30–35. [Google Scholar] [CrossRef]
  41. Lee, S.K.; Lee, J.H.; Kim, H.R.; Chun, Y.; Lee, J.H.; Yoo, H.Y.; Park, C.; Kim, S.W. Improved Cordycepin Production by Cordyceps militaris KYL05 Using Casein Hydrolysate in Submerged Conditions. Biomolecules 2019, 9, 461. [Google Scholar] [CrossRef] [Green Version]
  42. Mao, X.-B.; Eksriwong, T.; Chauvatcharin, S.; Zhong, J.-J. Optimization of Carbon Source and Carbon/Nitrogen Ratio for Cordycepin Production by Submerged Cultivation of Medicinal Mushroom Cordyceps militaris. Process Biochem. 2005, 40, 1667–1672. [Google Scholar] [CrossRef]
  43. Masuda, M.; Urabe, E.; Honda, H.; Sakurai, A.; Sakakibara, M. Enhanced Production of Cordycepin by Surface Culture Using the Medicinal Mushroom Cordyceps militaris. Enzyme Microb. Technol. 2007, 40, 1199–1205. [Google Scholar] [CrossRef]
  44. Masuda, M.; Das, S.K.; Hatashita, M.; Fujihara, S.; Sakurai, A. Efficient Production of Cordycepin by the Cordyceps militaris Mutant G81-3 for Practical Use. Process. Biochem. 2014, 49, 181–187. [Google Scholar] [CrossRef]
  45. Hu, T.; Liang, Y.; Zhao, G.; Wu, W.; Li, H.; Guo, Y. Selenium Biofortification and Antioxidant Activity in Cordyceps militaris Supplied with Selenate, Selenite, or Selenomethionine. Biol. Trace Elem. Res. 2019, 187, 553–561. [Google Scholar] [CrossRef] [PubMed]
  46. Shih, L.; Tsai, K.-L.; Hsieh, C. Effects of Culture Conditions on the Mycelial Growth and Bioactive Metabolite Production in Submerged Culture of Cordyceps militaris. Biochem. Eng. J. 2007, 33, 193–201. [Google Scholar] [CrossRef]
  47. Jiapeng, T.; Yiting, L.; Li, Z. Optimization of Fermentation Conditions and Purification of Cordycepin from Cordyceps militaris. Prep. Biochem. Biotechnol. 2014, 44, 90–106. [Google Scholar] [CrossRef]
  48. Lou, H.; Ye, Z.; Yun, F.; Lin, J.; Guo, L.; Chen, B.; Mu, Z. Targeted Gene Deletion in Cordyceps militaris Using the Split-Marker Approach. Mol. Biotechnol. 2018, 60, 380–385. [Google Scholar] [CrossRef]
  49. Lou, H.-W.; Ye, Z.-W.; Yu, Y.-H.; Lin, J.-F.; Guo, L.-Q.; Chen, B.-X.; Tang, H.-B.; Wei, T.; Chen, L.-T.; Yun, F. The Efficient Genetic Transformation of Cordyceps militaris by Using Mononuclear Protoplasts. Sci. Hortic. 2019, 243, 307–313. [Google Scholar] [CrossRef]
  50. Lou, H.-W.; Zhao, Y.; Chen, B.-X.; Yu, Y.-H.; Tang, H.-B.; Ye, Z.-W.; Lin, J.-F.; Guo, L.-Q. Cmfhp Gene Mediates Fruiting Body Development and Carotenoid Production in Cordyceps militaris. Biomolecules 2020, 10, 410. [Google Scholar] [CrossRef] [Green Version]
  51. Chen, B.-X.; Wei, T.; Ye, Z.-W.; Yun, F.; Kang, L.-Z.; Tang, H.-B.; Guo, L.-Q.; Lin, J.-F. Efficient CRISPR-Cas9 Gene Disruption System in Edible-Medicinal Mushroom Cordyceps militaris. Front. Microbiol. 2018, 9, 1157. [Google Scholar] [CrossRef]
  52. Butt, T.M.; Goettel, M.S. Bioassays of Entomogenous Fungi. In Bioassays of Entomopathogenic Microbes and Nematodes; Centre for Agriculture and Bioscience International: Wallingford, UK, 2000; pp. 141–195. [Google Scholar]
  53. Butt, T.M.; Wang, C.; Shah, F.A.; Hall, R. Degeneration of Entomogenous Fungi. In An Ecological and Societal Approach to Biological Control; Springer: Berlin/Heidelberg, Germany, 2006; pp. 213–226. [Google Scholar]
  54. Kawakami, K. On the Changes of Characteristics of the Silkworm Muscardines through Successive Cultures. Bull. Seric. Exp. Stn. Jpn. 1960, 16, 83–99. [Google Scholar]
  55. Nagaich, B.B. Verticillium sp. Pathogenic on Aphids. Indian Phytopathol. 1973, 26, 163–165. [Google Scholar]
  56. Ibrahim, L.; Jenkinson, P. Effect of Artificial Culture Media on Germination, Growth, Virulence and Surface Properties of the Entomopathogenic Hyphomycete Metarhizium anisopliae. Mycol. Res. 2002, 106, 705–715. [Google Scholar] [CrossRef]
  57. Sun, S.-J.; Deng, C.-H.; Zhang, L.-Y.; Hu, K.-H. Molecular Analysis and Biochemical Characteristics of Degenerated Strains of Cordyceps Militaris. Arch. Microbiol. 2017, 199, 939–944. [Google Scholar] [CrossRef] [PubMed]
  58. Yin, J.; Xin, X.; Weng, Y.; Gui, Z. Transcriptome-Wide Analysis Reveals the Progress of Cordyceps militaris Subculture Degeneration. PLoS ONE 2017, 12, e0186279. [Google Scholar] [CrossRef] [Green Version]
  59. Wang, D.; He, X.; Zhao, R.; Ren, G.; Liu, Y. Comparison of Cordyceps militaris Spawn Rejuvenation Method. North. Hortic. 2016, 40, 137–139. [Google Scholar]
  60. Grognet, P.; Bidard, F.; Kuchly, C.; Tong, L.C.H.; Coppin, E.; Benkhali, J.A.; Couloux, A.; Wincker, P.; Debuchy, R.; Silar, P. Maintaining Two Mating Types: Structure of the Mating Type Locus and Its Role in Heterokaryosis in Podospora anserina. Genetics 2014, 197, 421–432. [Google Scholar] [CrossRef] [Green Version]
  61. Ni, M.; Feretzaki, M.; Sun, S.; Wang, X.; Heitman, J. Sex in Fungi. Annu. Rev. Genet. 2011, 45, 405–430. [Google Scholar] [CrossRef]
  62. Yin, J.; Xin, X.-D.; Weng, Y.-J.; Li, S.-H.; Jia, J.-Q.; Gui, Z.-Z. Genotypic Analysis of Degenerative Cordyceps militaris Cultured in the Pupa of Bombyx mori. Entomol. Res. 2018, 48, 137–144. [Google Scholar] [CrossRef]
  63. Yokoyama, E.; Yamagishi, K.; Hara, A. Development of a PCR-Based Mating-Type Assay for Clavicipitaceae. FEMS Microbiol. Lett. 2004, 237, 205–212. [Google Scholar] [CrossRef] [Green Version]
  64. Li, M.F.; He, J.; Ding, L.; Kang, J.; Zhang, Q.; Zheng, Q. Single Spore Strains without Producing Fruit Body Isolated from Cordyceps militaris and Their RAPD Analysis. Southwest China J. Agric. Sci. 2007, 20, 547–550. [Google Scholar]
  65. Sulewska, A.; Niklinska, W.; Kozlowski, M.; Minarowski, L.; Naumnik, W.; Niklinski, J.; Dabrowska, K.; Chyczewski, L. DNA Methylation in States of Cell Physiology and Pathology. Folia Histochem. Cytobiol. 2007, 45, 149–158. [Google Scholar]
  66. Lee, T.; Zhai, J.; Meyers, B.C. Conservation and Divergence in Eukaryotic DNA Methylation. Proc. Natl. Acad. Sci. USA 2010, 107, 9027–9028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Selker, E.U.; Tountas, N.A.; Cross, S.H.; Margolin, B.S.; Murphy, J.G.; Bird, A.P.; Freitag, M. The Methylated Component of the Neurospora crassa Genome. Nature 2003, 422, 893–897. [Google Scholar] [CrossRef] [PubMed]
  68. Jeon, J.; Choi, J.; Lee, G.-W.; Park, S.-Y.; Huh, A.; Dean, R.A.; Lee, Y.-H. Genome-Wide Profiling of DNA Methylation Provides Insights into Epigenetic Regulation of Fungal Development in a Plant Pathogenic Fungus, Magnaporthe oryzae. Sci. Rep. 2015, 5, 8567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Ashraf, S.A.; Elkhalifa, A.E.O.; Siddiqui, A.J.; Patel, M.; Awadelkareem, A.M.; Snoussi, M.; Ashraf, M.S.; Adnan, M.; Hadi, S. Cordycepin for Health and Wellbeing: A Potent Bioactive Metabolite of an Entomopathogenic Medicinal Fungus Cordyceps with Its Nutraceutical and Therapeutic Potential. Molecules 2020, 25, 2735. [Google Scholar] [CrossRef] [PubMed]
  70. Das, S.K.; Masuda, M.; Sakurai, A.; Sakakibara, M. Medicinal Uses of the Mushroom Cordyceps militaris: Current State and Prospects. Fitoterapia 2010, 81, 961–968. [Google Scholar] [CrossRef]
  71. Zheng, Q.; Wei, T.; Lin, Y.; Ye, Z.-W.; Lin, J.-F.; Guo, L.-Q.; Yun, F.; Kang, L. Developing a Novel Two-Stage Process for Carotenoid Production by Cordyceps militaris (Ascomycetes). Int. J. Med. Mushrooms 2019, 21, 47–57. [Google Scholar] [CrossRef]
  72. Koutrotsios, G.; Mountzouris, K.C.; Chatzipavlidis, I.; Zervakis, G.I. Bioconversion of Lignocellulosic Residues by Agrocybe Cylindracea and Pleurotus Ostreatus Mushroom Fungi–Assessment of Their Effect on the Final Product and Spent Substrate Properties. Food Chem. 2014, 161, 127–135. [Google Scholar] [CrossRef]
  73. Koutrotsios, G.; Larou, E.; Mountzouris, K.C.; Zervakis, G.I. Detoxification of Olive Mill Wastewater and Bioconversion of Olive Crop Residues into High-Value-Added Biomass by the Choice Edible Mushroom Hericium erinaceus. Appl. Biochem. Biotechnol. 2016, 180, 195–209. [Google Scholar] [CrossRef]
  74. Koutrotsios, G.; Kalogeropoulos, N.; Kaliora, A.C.; Zervakis, G.I. Toward an Increased Functionality in Oyster (Pleurotus) Mushrooms Produced on Grape Marc or Olive Mill Wastes Serving as Sources of Bioactive Compounds. J. Agric. Food Chem. 2018, 66, 5971–5983. [Google Scholar] [CrossRef] [PubMed]
  75. Zervakis, G.I.; Koutrotsios, G.; Katsaris, P. Composted versus Raw Olive Mill Waste as Substrates for the Production of Medicinal Mushrooms: An Assessment of Selected Cultivation and Quality Parameters. BioMed Res. Int. 2013, 2013, 546830. [Google Scholar] [CrossRef] [PubMed]
  76. Kontogiannatos, D.; Koutrotsios, G.; Xekalaki, S.; Tarantilis, P.; Zervakis, G.I. Cultivation of the Entomopathogenic Fungus Cordyceps militaris in Insect-Enriched Substrates. In Proceedings of the ISMS e-Congress, Virtual, 14–17 September 2021. [Google Scholar]
  77. Czaja, W.; Miller, K.Y.; Skinner, M.K.; Miller, B.L. Structural and Functional Conservation of Fungal MatA and Human SRY Sex-Determining Proteins. Nat. Commun. 2014, 5, 5434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Metzenberg, R.L.; Glass, N.L. Mating Type and Mating Strategies in Neurospora. Bioessays 1990, 12, 53–59. [Google Scholar] [CrossRef]
  79. Kang, N.; Lee, H.-H.; Park, I.; Seo, Y.-S. Development of High Cordycepin-Producing Cordyceps militaris Strains. Mycobiology 2017, 45, 31–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Zhang, G.; Liang, Y. Improvement of Fruiting Body Production in Cordyceps militaris by Molecular Assessment. Arch. Microbiol. 2013, 195, 579–585. [Google Scholar] [CrossRef] [PubMed]
  81. Zheng, Z.; Huang, C.; Cao, L.; Xie, C.; Han, R. Agrobacterium tumefaciens-Mediated Transformation as a Tool for Insertional Mutagenesis in Medicinal Fungus Cordyceps militaris. Fungal Biol. 2011, 115, 265–274. [Google Scholar] [CrossRef]
  82. Yang, T.; Guo, M.; Yang, H.; Guo, S.; Dong, C. The Blue-Light Receptor CmWC-1 Mediates Fruit Body Development and Secondary Metabolism in Cordyceps militaris. Appl. Microbiol. Biotechnol. 2016, 100, 743–755. [Google Scholar] [CrossRef]
  83. Dong, J.Z.; Wang, S.H.; Ai, X.R.; Yao, L.; Sun, Z.W.; Lei, C.; Wang, Y.; Wang, Q. Composition and Characterization of Cordyxanthins from Cordyceps militaris Fruit Bodies. J. Funct. Foods 2013, 5, 1450–1455. [Google Scholar] [CrossRef]
  84. Lou, H.-W.; Zhao, Y.; Tang, H.-B.; Ye, Z.-W.; Wei, T.; Lin, J.-F.; Guo, L.-Q. Transcriptome Analysis of Cordyceps militaris Reveals Genes Associated with Carotenoid Synthesis and Identification of the Function of the Cmtns Gene. Front. Microbiol. 2019, 10, 2105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Frappier, F.; Pais, M.; Elsworth, J.F.; Grove, J.F. Revised Structure of Beauvellide, a Cyclodepsipeptide from Beauveria tenella. Phytochemistry 1978, 17, 545–546. [Google Scholar] [CrossRef]
  86. Mochizuki, K.; Ohmori, K.; Tamura, H.; Shizuri, Y.; Nishiyama, S.; Miyoshi, E.; Yamamura, S. The Structures of Bioactive Cyclodepsipeptides, Beauveriolides I and II, Metabolites of Entomopathogenic Fungi Beauveria sp. Bull. Chem. Soc. Jpn. 1993, 66, 3041–3046. [Google Scholar] [CrossRef]
  87. Namatame, I.; Tomoda, H.; Tabata, N.; SI, S.; Omura, S. Structure Elucidation of Fungal Beauveriolide III, a Novel Inhibitor of Lipid Droplet Formation in Mouse Macrophages. J. Antibiot. 1999, 52, 7–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Namatame, I.; Tomoda, H.; Si, S.; Yamaguchi, Y.; Masuma, R.; Omura, S. Beauveriolides, Specific Inhibitors of Lipid Droplet Formation in Mouse Macrophages, Produced by Beauveria sp. FO-6979. J. Antibiot. 1999, 52, 1–6. [Google Scholar] [CrossRef] [Green Version]
  89. Namatame, I.; Tomoda, H.; Ishibashi, S.; Omura, S. Antiatherogenic Activity of Fungal Beauveriolides, Inhibitors of Lipid Droplet Accumulation in Macrophages. Proc. Natl. Acad. Sci. USA 2004, 101, 737–742. [Google Scholar] [CrossRef] [Green Version]
  90. Wang, X.; Gao, Y.-L.; Zhang, M.-L.; Zhang, H.-D.; Huang, J.-Z.; Li, L. Genome Mining and Biosynthesis of the Acyl-CoA: Cholesterol Acyltransferase Inhibitor Beauveriolide I and III in Cordyceps militaris. J. Biotechnol. 2020, 309, 85–91. [Google Scholar] [CrossRef]
  91. Cohen, N.; Cohen, J.; Asatiani, M.D.; Varshney, V.K.; Yu, H.-T.; Yang, Y.-C.; Li, Y.-H.; Mau, J.-L.; Wasser, S.P. Chemical Composition and Nutritional and Medicinal Value of Fruit Bodies and Submerged Cultured Mycelia of Culinary-Medicinal Higher Basidiomycetes Mushrooms. Int. J. Med. Mushrooms 2014, 16, 273–291. [Google Scholar] [CrossRef]
  92. Cunningham, K.G.; Manson, W.; Spring, F.S.; Hutchinson, S.A. Cordycepin, a Metabolic Product Isolated from Cultures of Cordyceps militaris (Linn.) Link. Nature 1950, 166, 949. [Google Scholar] [CrossRef]
  93. Paterson, R.R.M. Cordyceps—A Traditional Chinese Medicine and Another Fungal Therapeutic Biofactory? Phytochemistry 2008, 69, 1469–1495. [Google Scholar] [CrossRef] [Green Version]
  94. Zhang, Q.; Liu, Y. The Strategies for Increasing Cordycepin Production of Cordyceps militaris by Liquid Fermentation. Fungal Genom. Biol. 2016, 6, 134. [Google Scholar] [CrossRef]
  95. Kim, H.G.; Shrestha, B.; Lim, S.Y.; Yoon, D.H.; Chang, W.C.; Shin, D.-J.; Han, S.K.; Park, S.M.; Park, J.H.; Park, H.I. Cordycepin Inhibits Lipopolysaccharide-Induced Inflammation by the Suppression of NF-ΚB through Akt and P38 Inhibition in RAW 264.7 Macrophage Cells. Eur. J. Pharmacol. 2006, 545, 192–199. [Google Scholar] [CrossRef]
  96. Noh, E.-M.; Kim, J.-S.; Hur, H.; Park, B.-H.; Song, E.-K.; Han, M.-K.; Kwon, K.-B.; Yoo, W.-H.; Shim, I.-K.; Lee, S.J. Cordycepin Inhibits IL-1β-Induced MMP-1 and MMP-3 Expression in Rheumatoid Arthritis Synovial Fibroblasts. Rheumatology 2009, 48, 45–48. [Google Scholar] [CrossRef] [Green Version]
  97. Tian, X.; Li, Y.; Shen, Y.; Li, Q.; Wang, Q.; Feng, L. Apoptosis and Inhibition of Proliferation of Cancer Cells Induced by Cordycepin. Oncol. Lett. 2015, 10, 595–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Tuli, H.S.; Sandhu, S.S.; Sharma, A.K. Pharmacological and Therapeutic Potential of Cordyceps with Special Reference to Cordycepin. 3 Biotech 2014, 4, 1–12. [Google Scholar] [CrossRef] [Green Version]
  99. Zhou, X.; Cai, G.; He, Y.I.; Tong, G. Separation of Cordycepin from Cordyceps militaris Fermentation Supernatant Using Preparative HPLC and Evaluation of Its Antibacterial Activity as an NAD+-Dependent DNA Ligase Inhibitor. Exp. Ther. Med. 2016, 12, 1812–1816. [Google Scholar] [CrossRef] [PubMed]
  100. Xia, Y.; Luo, F.; Shang, Y.; Chen, P.; Lu, Y.; Wang, C. Fungal Cordycepin Biosynthesis Is Coupled with the Production of the Safeguard Molecule Pentostatin. Cell Chem. Biol. 2017, 24, 1479–1489.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Ballario, P.; Vittorioso, P.; Magrelli, A.; Talora, C.; Cabibbo, A.; Macino, G. White Collar-1, a Central Regulator of Blue Light Responses in Neurospora, Is a Zinc Finger Protein. EMBO J. 1996, 15, 1650–1657. [Google Scholar] [CrossRef]
  102. Dunlap, J.C. Proteins in the Neurospora Circadian Clockworks. J. Biol. Chem. 2006, 281, 28489–28493. [Google Scholar] [CrossRef] [Green Version]
  103. Chen, C.-H.; Dunlap, J.C.; Loros, J.J. Neurospora Illuminates Fungal Photoreception. Fungal Genet. Biol. 2010, 47, 922–929. [Google Scholar] [CrossRef] [Green Version]
  104. Corrochano, L.M. Fungal Photoreceptors: Sensory Molecules for Fungal Development and Behaviour. Photochem. Photobiol. Sci. 2007, 6, 725–736. [Google Scholar] [CrossRef]
  105. Linden, H.; Ballario, P.; Macino, G. Blue Light Regulation In Neurospora crassa. Fungal Genet. Biol. 1997, 22, 141–150. [Google Scholar] [CrossRef]
  106. Linden, H. A White Collar Protein Senses Blue Light. Science 2002, 297, 777–778. [Google Scholar] [CrossRef]
  107. Idnurm, A.; Verma, S.; Corrochano, L.M. A Glimpse into the Basis of Vision in the Kingdom Mycota. Fungal Genet. Biol. 2010, 47, 881–892. [Google Scholar] [CrossRef] [Green Version]
  108. Wang, F.; Song, X.; Dong, X.; Zhang, J.; Dong, C. DASH-Type Cryptochromes Regulate Fruiting Body Development and Secondary Metabolism Differently than CmWC-1 in the Fungus Cordyceps militaris. Appl. Microbiol. Biotechnol. 2017, 101, 4645–4657. [Google Scholar] [CrossRef]
  109. Wang, F.; Liu, Q.; Zhang, J.; Liu, K.; Li, K.; Liu, G.; Dong, C. Comparative Transcriptome Analysis between a Spontaneous Albino Mutant and Its Sibling Strain of Cordyceps militaris in Response to Light Stress. Front. Microbiol. 2018, 9, 1237. [Google Scholar] [CrossRef]
  110. Jiaojiao, Z.; Fen, W.; Kuanbo, L.; Qing, L.; Ying, Y.; Caihong, D. Heat and Light Stresses Affect Metabolite Production in the Fruit Body of the Medicinal Mushroom Cordyceps militaris. Appl. Microbiol. Biotechnol. 2018, 102, 4523–4533. [Google Scholar] [CrossRef] [PubMed]
  111. Shao, Y.; Tang, J.; Chen, S.; Wu, Y.; Wang, K.; Ma, B.; Zhou, Q.; Chen, A.; Wang, Y. MilR4 and MilR16 Mediated Fruiting Body Development in the Medicinal Fungus Cordyceps militaris. Front. Microbiol. 2019, 10, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Luo, Q.-Y.; Cao, H.-F.; Liu, S.-K.; Wu, M.; Li, S.-S.; Zhang, Z.-Q.; Chen, A.-J.; Shen, G.-H.; Wu, H.-J.; Li, M.-L. Novel Liquid Fermentation Medium of Cordyceps militaris and Optimization of Hydrothermal Reflux Extraction of Cordycepin. J. Asian Nat. Prod. Res. 2020, 22, 167–178. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, H.-J.; Pan, M.-C.; Chang, C.-K.; Chang, S.-W.; Hsieh, C.-W. Optimization of Ultrasonic-Assisted Extraction of Cordycepin from Cordyceps militaris Using Orthogonal Experimental Design. Molecules 2014, 19, 20808–20820. [Google Scholar] [CrossRef] [Green Version]
  114. Wu, H.-C.; Chen, S.-T.; Chang, J.-C.; Hsu, T.-Y.; Cheng, C.-C.; Chang, H.-S.; Liu, C.-S.; Wu, Y.-C.; Liang, Z.-C. Radical Scavenging and Antiproliferative Effects of Cordycepin-Rich Ethanol Extract from Brown Rice—Cultivated Cordyceps militaris (Ascomycetes) Mycelium on Breast Cancer Cell Lines. Int. J. Med. Mushrooms 2019, 21, 657–669. [Google Scholar] [CrossRef] [PubMed]
  115. Dong, J.Z.; Lei, C.; Zheng, X.J.; Ai, X.R.; Wang, Y.; Wang, Q. Light Wavelengths Regulate Growth and Active Components of Cordyceps militaris Fruit Bodies. J. Food Biochem. 2013, 37, 578–584. [Google Scholar] [CrossRef]
  116. Dong, J.Z.; Lei, C.; Ai, X.R.; Wang, Y. Selenium Enrichment on Cordyceps militaris Link and Analysis on Its Main Active Components. Appl. Biochem. Biotechnol. 2012, 166, 1215–1224. [Google Scholar] [CrossRef] [PubMed]
  117. Wen, T.C.; Li, G.R.; Kang, J.C.; Kang, C.; Hyde, K.D. Optimization of Solid-State Fermentation for Fruiting Body Growth and Cordycepin Production by Cordyceps militaris. Chiang Mai J. Sci. 2014, 41, 858–872. [Google Scholar]
  118. Kang, C.; Wen, T.-C.; Kang, J.-C.; Meng, Z.-B.; Li, G.-R.; Hyde, K.D. Optimization of Large-Scale Culture Conditions for the Production of Cordycepin with Cordyceps militaris by Liquid Static Culture. Sci. World J. 2014, 2014, 510627. [Google Scholar] [CrossRef] [Green Version]
  119. Chou, T.-Y.; Kuo, H.-P.; Tsai, S.-F.; Huang, S.-T.; Yang, M.-J.; Lee, S.-S.; Chang, C.-C. Doubled Production of Cordycepin Analogs in Cultured Cordyceps militaris by Addition of Andrea Droppings. Nat. Prod. Res. 2020, 1–6. [Google Scholar] [CrossRef]
  120. Suparmin, A.; Kato, T.; Dohra, H.; Park, E.Y. Insight into Cordycepin Biosynthesis of Cordyceps militaris: Comparison between a Liquid Surface Culture and a Submerged Culture through Transcriptomic Analysis. PLoS ONE 2017, 12, e0187052. [Google Scholar] [CrossRef] [Green Version]
  121. Wen, T.C.; Kang, J.C.; Lei, B.X.; Li, G.R.; He, J. Enhanced Production of Cordycepin by Submerged Culture Using Additives in Cordyceps militaris. Food Sci. 2010, 31, 175–179. [Google Scholar]
  122. Reis, F.S.; Barros, L.; Calhelha, R.C.; Ćirić, A.; Van Griensven, L.J.; Soković, M.; Ferreira, I.C. The Methanolic Extract of Cordyceps militaris (L.) Link Fruiting Body Shows Antioxidant, Antibacterial, Antifungal and Antihuman Tumor Cell Lines Properties. Food Chem. Toxicol. 2013, 62, 91–98. [Google Scholar] [CrossRef] [Green Version]
  123. Chen, Y.-C.; Chen, Y.-H.; Pan, B.-S.; Chang, M.-M.; Huang, B.-M. Functional Study of Cordyceps sinensis and Cordycepin in Male Reproduction: A Review. J. Food Drug Anal. 2017, 25, 197–205. [Google Scholar] [CrossRef] [Green Version]
  124. Kunhorm, P.; Chaicharoenaudomrung, N.; Noisa, P. Enrichment of Cordycepin for Cosmeceutical Applications: Culture Systems and Strategies. Appl. Microbiol. Biotechnol. 2019, 103, 1681–1691. [Google Scholar] [CrossRef]
  125. Lee, C.-T.; Huang, K.-S.; Shaw, J.-F.; Chen, J.-R.; Kuo, W.-S.; Shen, G.; Grumezescu, A.M.; Holban, A.M.; Wang, Y.-T.; Wang, J.-S. Trends in the Immunomodulatory Effects of Cordyceps militaris: Total Extracts, Polysaccharides and Cordycepin. Front. Pharmacol. 2020, 11, 1824. [Google Scholar] [CrossRef] [PubMed]
  126. Nakamura, K.; Shinozuka, K.; Yoshikawa, N. Anticancer and Antimetastatic Effects of Cordycepin, an Active Component of Cordyceps sinensis. J. Pharmacol. Sci. 2015, 127, 53–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Yoon, S.Y.; Park, S.J.; Park, Y.J. The Anticancer Properties of Cordycepin and Their Underlying Mechanisms. Int. J. Mol. Sci. 2018, 19, 3027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Wang, Y.-S.; Shelomi, M. Review of Black Soldier Fly (Hermetia illucens) as Animal Feed and Human Food. Foods 2017, 6, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. C. militaris strains producing ascomata following cultivation on rice-based substrates supplemented by insect tissues (Laboratory of General and Agricultural Microbiology, Agricultural University of Athens, Greece); (A) strain CmDK1, (B) strain CmDK2 [76].
Figure 1. C. militaris strains producing ascomata following cultivation on rice-based substrates supplemented by insect tissues (Laboratory of General and Agricultural Microbiology, Agricultural University of Athens, Greece); (A) strain CmDK1, (B) strain CmDK2 [76].
Jof 07 00986 g001
Figure 2. Schematic overview of genetic engineering approaches commonly used in C. militaris.
Figure 2. Schematic overview of genetic engineering approaches commonly used in C. militaris.
Jof 07 00986 g002
Table 1. Factors that affect culture degeneration in C. militaris, their effect, and potential treatment.
Table 1. Factors that affect culture degeneration in C. militaris, their effect, and potential treatment.
FactorsEffects on Culture Degeneration: P: Promotes; H: HindersTreatmentReferences
K+, Ca2+, Zn2+HAddition in culture[25]
Mn2+, Mg2+PAvoid usage[25]
PreservationHPreservation at 4 °C[28]
Cellular accumulation of reactive oxygen species (ROS)P [22]
SubculturingPAvoid subculturing after the third generation
Rejuvenation by mating single ascospore isolates and producing new heterokaryons
[58]
Culture temperature-mediumPDegenerated strains could be rejuvenated by using an insect host[59]
HomokaryosisPRejuvenation every 6 months
Appropriate light regime during preservation
[21,24,26,29]
DNA methylationP [27,30]
Genetic mutationsP [27,31]
Table 2. Basic requirements and cultivation practices affecting ascomata production or mycelium growth in C. militaris.
Table 2. Basic requirements and cultivation practices affecting ascomata production or mycelium growth in C. militaris.
Substrates, Nutrients, and Treatments in the Cultivation of C. militarisCultivation-Related PracticesEffects on Ascomata Production or Mycelium GrowthReferences
Serotonin, adrenaline, and dopamine catabolism to amino acidsAddition of insect-based supplements in cultureInduces amino acids production[32]
Chitin, glucosamine, and GABA catabolism in low C/N availabilityAddition of insect-based supplements
Addition of rice
Addition of chitosan
Highest growth rate and improved cordycepin production[32,33,34]
Sucrose utilizationAddition (instead of glucose)Highest growth rate[32]
C/N ratio12.7:1Optimal[32]
Cottonseed shells, corn cobs particles at ratios of 8:1:1 (w/w/w) with rice and wheat-Increased fruit body yield and improved cordycepin levels[12]
Vegetable oilsAddition in liquid cultureCell growth and cordycepin production enhancement[15]
FluorideAddition of 0.01 mMGrowth promotion and bioactive substance enhancement; stronger anti-proliferation effects of ascomata extracts on U2OS cancer cells[35]
Aerial mycelia scratchingScratching C. militaris degenerated mycelia before stromata differentiationShortening growth periods of C. militaris fruit bodies by at least 5 days[36]
Table 3. Cordycepin production by C. militaris strains as measured in mycelia (M), fruit bodies (F), and liquid media (extracellular, L) under various cultivation conditions/treatments.
Table 3. Cordycepin production by C. militaris strains as measured in mycelia (M), fruit bodies (F), and liquid media (extracellular, L) under various cultivation conditions/treatments.
Strain Code
(or N/A When No Strain Code Appears)
Cultivation ConditionsType of Biomass and/or Substrate MeasuredCordycepin ConcentrationReferences
Ν/AEffects of various light wavelengths (darkness, red, pink blue, and daylight)M1.15–1.98 mg g−1[115]
NBRC 9787Addition of 1 g L−1 adenine plus glycine or l-glutamine in batch operation
Addition of 1 g L−1 adenine plus glycine or l-glutamine repeated batch operation
M19.7–33.7 mg L−1
62.4–98.2 mg L−1
[43]
NBRC 9787
G81-3
Various strains and autoclaving techniquesM76–237 mg g−1[44]
Ν/AOptimized carbon and nitrogen sources, i.e., 42.0 g L−1 glucose and 15.8 g L−1 peptoneMUp to 345.4 mg L−1[42]
KYL05pH 6, 25 °C, 150 rpm, culture period of 6 days, carbon source: casein hydrolysate at 2%MUp to 445 mg L−1[41]
Ν/ApH 6, 25 °C, 110 rpm, culture period of 15–20 days, addition of 1 g L−1 FeSO4 at day 0MUp to 596.6 mg L−1[40]
CICC 14014Addition of 30 g L−1 peanut oil in standard mediumMUp to 5290 mg L−1[15]
Ν/AEffects of sodium selenite (0–18 ppm)F0–0.6 mg g−1[116]
CGMCC2459Effect of various mineral salts: K2HPO4, KH2PO4, Ca(NO3)2, CaCl2, KCl, MgSO47H2O, FeSO4F0.95–5.72 mg g−1[117]
CGMCC33.16322Wheat standard substrate and pupal (B. mori) injectionF~1 mg g−1
~1.2 mg g−1
[17]
CGMCC2459Effect of different nitrogen sources (wheat bran, soybean oil meal, beef extract, peptone, yeast extract, silkworm pupa, NH4NO3)F1.78–10.90 mg g−1[117]
CGMCC245920 g of brown rice, millet, sorghum, corn, wheat, and glutinous rice as fruiting medium supplemented with 32 mL of nutrient solutionF2.42–5.62 mg g−1[117]
CGMCC2459Effect of various growth factors (vitamins B1, B9, α-naphthyl acetic acid, 2,4-dichlorophenoxyacetic acid, indole-3-butytric acidF2.92–6.21 mg g−1[117]
CGMCC3.16321Generation of 498 (CGMCC 5.2190) sibling normal strain, generation of 505 (CGMCC 5.2191) albino strain by spontaneous mutationF3.09 mg g−1 for the normal sibling strain
6.70 mg g−1 for the albino strain
[109]
CGMCC2459Effect of various carbon sources (glucose, sucrose, amidulin, lactose, maltose, mannose)F3.77–6.50 mg g−1[117]
CGMCC2459Effect of initial pH (5.0–8.0)F4.39–7.40 mg g−1[117]
CGMCC 3.16321Heat stress treatment at 25 °C, light at 1700 lxFUp to 5.56 mg g−1[110]
KSP8Single spore mating of SPNU 1006xKACC44455FUp to 6.63 mg g−1[118]
N/AAddition of tea leaves or Andraca theae droppings in basal mediaF8.35–12.85 mg g−1[119]
Cm09Generation of ΔMAT1-1-2; injection of 107 ΔMAT1-1-2xΜAΤ1-2 spores/mL into the Chinese Tussah silkworm pupaeFUp to 16.77 mg g−1[37]
NO. 20130508Corn cob particles/wheat bran/rice bran (8:1:1) + 20 g L−1 glucose, and 5 g L−1 peptone
Cottonseed shells/wheat bran/rice (8:1:1) + 20 g L−1 glucose, and 5 g L−1 peptone; 20 g rice+ 20 g L−1 glucose, and 5 g L−1 peptone
F26.9 mg g−1
23.4 mg g−1
34.5 mg g−1
[12]
Ν/A20 g rice and 20 mL potato dextrose medium + selenate or selenite or selenomethionine at a concentration of 40 μg g−1 in riceF43.3 mg g−1
69.3 mg g−1
77.4 mg g−1
[45]
CM10Basal medium: 20 g L−1 peptone, 24.7 g L−1 sucrose, 1.11 g L−1 K2HPO4⋅3H2O, 0.90 g L−1 MgSO4⋅7H2O, 0.01 g L−1 vitamin B1 plus 8 g L−1 L-alanine, and overexpression of CCM_02568 and CCM_01481 transcription factorsL30.04–99.83 mg L−1[23]
Ν/ALiquid fermentation in basal media containing silkworm pupae powder, wheat, or silkworm pupae powder, plus wheat by applying different extraction methodologies (heat, frequency, solvents, and resins)LUp to 39.40 mg L−1[112]
CCRC 32219Multifactorial analysis (four factors): pH 4 to 7, various nitrogen sources, varying yeast extract content, shake vs. static conditionsL44.4–1375.6 mg L−1[46]
TBRC6039Rational design of synthetic mediaL65.7–377 mg L−1[32]
GACP08Y5
GACP08Y1
GACP0746
20 g L−1 sucrose, 20 g L−1 peptone, 1 g L−1 KH2PO4, and 0.5 g L−1 MgSO4·7H2O in static liquid bioreactors of different culture volumesL271–4376 mg L−1[118]
NBRC 9787Addition of 1 g L−1 adenine plus glycine or l-glutamine in batch operation
Addition of 1 g L−1 adenine plus glycine or l-glutamine repeated batch operation
L542.4–2500 mg L−1
3400–14,100 mg L−1
[43]
CGMCC2459Optimized medium (20 g L−1 peptone, 24.7 g L−1 sucrose, 1.11 g L−1 K2HPO4·3H2O, 0.90 g L−1 MgSO4·7H2O, 10 mg L−1 vitamin B1, 5.45 g L−1 hypoxanthine, and 12.23 g L−1 L-alanine)LUp to 2008 mg L−1[118]
NBRC 9787
G81-3
Various strains and autoclaving conditionsL2400–10,900 mg L−1[44]
BCRC34380Effect of porcine liver extracts (0.5 g L−1, 1 g L−1, 5 g L−1, 7.5 g L−1, and 10 g L−1)LUp to 2452 mg L−1[12]
BCRC34380Effect of blue light irradiation
(0, 8, 16, and 24 h d−1)
LUp to 3483 mg L−1[12]
NBRC 10375272.5 g L−1 yeast extract, 62.6 g L−1 glucose (pH 5.6), and Vogel’s medium at 1:10 concentrationLUp to ~5000 mg L−1[120]
CM1401460 g L−1 glucose, 0.7 g L−1 KH2PO4, 0.7 g L−1 MgSO4 7H2O, 9.00 g L−1 yeast extract, and 17.10 g L−1 tryptone at 27.1 °C; seed age, 3 days; inoculum size, 10%LUp to ~7350 mg L−1[47]
NBRC 9787Surface liquid culture and mutagenesis by ion beam irradiationLUp to 8570 mg L−1[38]
CM016Solid state fermentation on rice-based medium containing 20 g L−1 sucrose, 10 g L−1 peptone, 0.1 g L−1 MgSO4 7H2O, and 0.1 g L−1 KH2PO4 using a four-factor, five-leveled central composite against glucose, peptone, adenine, histidineF1.92–20.86 mg g−1[121]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kontogiannatos, D.; Koutrotsios, G.; Xekalaki, S.; Zervakis, G.I. Biomass and Cordycepin Production by the Medicinal Mushroom Cordyceps militaris—A Review of Various Aspects and Recent Trends towards the Exploitation of a Valuable Fungus. J. Fungi 2021, 7, 986. https://0-doi-org.brum.beds.ac.uk/10.3390/jof7110986

AMA Style

Kontogiannatos D, Koutrotsios G, Xekalaki S, Zervakis GI. Biomass and Cordycepin Production by the Medicinal Mushroom Cordyceps militaris—A Review of Various Aspects and Recent Trends towards the Exploitation of a Valuable Fungus. Journal of Fungi. 2021; 7(11):986. https://0-doi-org.brum.beds.ac.uk/10.3390/jof7110986

Chicago/Turabian Style

Kontogiannatos, Dimitrios, Georgios Koutrotsios, Savvina Xekalaki, and Georgios I. Zervakis. 2021. "Biomass and Cordycepin Production by the Medicinal Mushroom Cordyceps militaris—A Review of Various Aspects and Recent Trends towards the Exploitation of a Valuable Fungus" Journal of Fungi 7, no. 11: 986. https://0-doi-org.brum.beds.ac.uk/10.3390/jof7110986

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop