Next Article in Journal
Strategies for the Enhancement of Secondary Metabolite Production via Biosynthesis Gene Cluster Regulation in Aspergillus oryzae
Previous Article in Journal
Age-Related Conservation in Plant–Soil Feedback Accompanied by Ectomycorrhizal Domination in Temperate Forests in Northeast China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Advances in the Functional Genes of Edible and Medicinal Fungi: Research Techniques, Functional Analysis, and Prospects

by
Wenyun Li
1,2,
Gen Zou
1,
Dapeng Bao
1,2,* and
Yingying Wu
1,2,*
1
National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization, Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
2
College of Food Sciences and Technology, Shanghai Ocean University, Shanghai 201306, China
*
Authors to whom correspondence should be addressed.
J. Fungi 2024, 10(5), 311; https://doi.org/10.3390/jof10050311
Submission received: 8 March 2024 / Revised: 2 April 2024 / Accepted: 22 April 2024 / Published: 25 April 2024
(This article belongs to the Section Fungal Genomics, Genetics and Molecular Biology)

Abstract

:
Functional genes encode various biological functions required for the life activities of organisms. By analyzing the functional genes of edible and medicinal fungi, varieties of edible and medicinal fungi can be improved to enhance their agronomic traits, growth rates, and ability to withstand adversity, thereby increasing yield and quality and promoting industrial development. With the rapid development of functional gene research technology and the publication of many whole-genome sequences of edible and medicinal fungi, genes related to important biological traits have been mined, located, and functionally analyzed. This paper summarizes the advantages and disadvantages of different functional gene research techniques and application examples for edible and medicinal fungi; systematically reviews the research progress of functional genes of edible and medicinal fungi in biological processes such as mating type, mycelium and fruit growth and development, substrate utilization and nutrient transport, environmental response, and the synthesis and regulation of important active substances; and proposes future research directions for functional gene research for edible and medicinal fungi. The overall aim of this study was to provide a valuable reference for further promoting the molecular breeding of edible and medicinal fungi with high yield and quality and to promote the wide application of edible and medicinal fungi products in food, medicine, and industry.

1. Introduction

The term “edible and medicinal fungi” broadly refers to macrofungi that are highly beneficial for human consumption or medicinal purposes and are characterized by their nutrient richness, delightful flavor, and a range of substances that are advantageous for human health. In the dry matter of edible and medicinal fungi, proteins account for 15–35%, carbohydrates account for 51.3–62.5%, and dietary fibers account for 8–10.4% [1,2], with a sufficient content of essential amino acids to complement vegetables, legumes, and dairy products [3]. Edible and medicinal fungi contain polysaccharides, antifungal proteins [4], terpenoids, steroids, adenosine, and other bioactive substances, offering antioxidant, anti-aging, anti-tumor, and antivirus benefits and reducing blood lipids, lowering blood pressure and cholesterol, liver-protective detoxification, immune control, and other vital roles [5,6,7,8]. Almost 150,000 species of fungi have been described, of which more than 2000 are edible species prevalent in Asia, Europe, America, Africa, and Oceania [9]. Currently, more than 100 types of edible and medicinal fungi have been cultivated artificially, among which Lentinula edodes, Pleurotus spp., Auricularia spp., Agaricus bisporus, and Flammulina filiformis are the main cultivars in the world [10]. In addition to their food and medicinal value, edible and medicinal fungi can utilize agricultural and forestry wastes as a source of cultivation substrates, compost, feed, or biofuel [11], playing an important role as a core link in the circular agricultural economy [12], can simultaneously realize the ultimate goals of economic development and environmental protection [13].
Despite the rapid development of the edible and medicinal fungi industry worldwide, problems, such as a single genetic background of factory-produced varieties, a high degree of product homogenization, and easy degradation of strains, have seriously constrained the sustainable and healthy development of the industry. Currently, the cultivation of high-yield and high-quality edible and medicinal fungi varieties and the development of high-value products are the most economical and effective ways to solve the problems of the edible and medicinal fungi industry. Basic scientific research on edible and medicinal fungi is closely related to their industrial development, and these industrial issues provide important directions for scientific research on edible and medicinal fungi. The growth cycle, agronomic traits, intrinsic quality, and other important traits related to the edible and medicinal fungi industry are subject to complex genetic regulation, and the screening and identification of relevant functional genes is a key step in the study of the molecular mechanisms of the biological phenogenesis of various edible and medicinal fungi and is one of the important aspects of the construction of the discipline of edible and medicinal fungi genetics [14]. However, constrained by complex genetic backgrounds and small research populations, the established technological system for conventional functional gene research in plants and other microorganisms cannot be directly applied to most edible and medicinal fungi species, particularly Basidiomycota. In recent years, with the rapid development of molecular sequencing technology and the successful application of modern biological techniques, such as gene editing, important functional genes in edible and medicinal fungi, including genes related to the development of mycelia and fruiting bodies, those regulating the synthesis of active substances, those for mating type, and those for environmental response have been analyzed step-by-step. With an increasing number of research teams focusing on the research field of edible and medicinal fungi functional genes and achieving many scientific advances, a body of work has been gradually formed based on edible and medicinal fungi germplasm resources, multi-omics data, and the evaluation of important agronomic traits.
Using the technologies of multi-omics, genetic transformation, gene-editing, chassis cell as a platform, and adopting synthetic biology strategies through the development and application of important functional genes will ultimately realize a circular economy and a variety of innovations (Figure 1).
This paper summarizes and compares the main techniques applied by global researchers in recent years in the field of functional gene research in edible and medicinal fungi and analyzes the important identified gene functions to lay a scientific foundation for further elucidation of the intrinsic mechanism of the life activities of edible and medicinal fungi. This paper aims to provide a theoretical reference for future germplasm innovation and cross-disciplinary application of edible and medicinal fungi products.

2. Advances in Functional Gene Research Techniques in Edible and Medicinal Fungi

The screening and characterization of functional genes are the basis for studying how genes regulate important phenotypes in edible and medicinal fungi. The successful completion of this work relies heavily on the development and application of relevant bioinformatics and biological experimental techniques. In edible and medicinal fungi research, to isolate a certain functional gene for a certain purpose, we usually start from a specific phenotypic trait and use omics technology methods, such as genetic map construction, genome-wide association analysis, and expression profiling difference analysis, to mine and screen the associated genes and further use genetic transformation, gene editing, and chassis cell series technology to functionally resolve and validate the candidate genes.

2.1. Multi-Omics Technology

Evolving omics technologies are effective aids in resolving important phenotype-associated functional genes in edible and medicinal fungi, including genomics, pan-genomics, transcriptomics, proteomics, and metabolomics. Zhao et al. [15] sequenced and annotated the whole genome of Morchella esculenta MT1, revealing that its functional genes were focused on metabolic processes, cellular processes, and binding and catalytic activity processes, and it was hypothesized that this mushroom has stronger low-temperature and cold-resistant properties. With the development of genomics, the construction of pan-genomic data accelerated the resolution of the genetic mechanisms underlying the formation of agronomic traits in edible and medicinal fungi. Drott et al. [16] studied the pan-genomes of the macrofungi Amanita phalloides and Agaricales and reported that the differential expression of MSDIN, a gene encoding a toxic secondary metabolite, leads to different phenotypes. They further revealed the dynamics of the toxin gene in natural evolution, providing an effective pathway for exploring secondary metabolites in other Basidiomycota. The pangenome of Auricularia heimuer constructed by Guo et al. [17] included 14,089 gene families, 67.56% of which were core gene families and 31.88% of which were minor gene families. The genome was screened for the chitinase gene ahchi1 and a carbohydrate-binding module (CBM)-related gene, which provided a reference for genes related to substrate utilization in edible and medicinal fungi.
Transcriptomics focuses on the overall study of gene transcription and regulation in cells, thereby revealing the molecular mechanisms of a particular biological process. Light plays a specific role in the regulation of secondary metabolites in edible and medicinal fungi, but the molecular mechanisms and light-induced genes are still unclear. Kim et al. [18] compared the transcriptome data of mushrooms under blue light and darkness; screened 221 upregulated genes and 541 downregulated genes; and reported that the DDR48-heat-shock proteins, 12 kD heat-shock proteins, and conserved fungal proteins were highly expressed under blue-light irradiation. This provides important information for the understanding of blue-light sensing and receptor mechanisms in fungi. Hao et al. [19] analyzed the transcriptome at three developmental stages—the mycelium, primordium, and fruiting body stages—and identified 9690 differentially differentiated genes mainly involved in oxidoreductase, biodegradation, energy metabolism, and hydrolase activity, among which CO-sensing-related genes (CA-2, CA-3, PKA-1, and PKA-20), heat-shock protein genes (HSP60 and HSP90), and transcription factors (steA, MYB, nosA, HAP1, and GATA-4/5/6) play key roles in protoplast formation, supporting the further study of substrate development. Shen et al. [20] sequenced the transcriptomes of L. edodes substrates (cap and stipe) at different developmental stages and showed that genes encoding carbonic anhydrase, chitinase, GH5, and cytochrome P450s may function in stipe elongation.
Proteomics is a pathway for the comprehensive understanding of molecular mechanisms, and this type of research brings “life” to genomics [21]. To date, more than 800 edible and medicinal fungi have been reported to possess important pharmacological properties, such as anti-tumor and immunomodulatory activities [22]. The use of proteomics technology allows the identification of a large number of differentially regulated proteins, providing an important platform for the discovery of potential therapeutic targets. Lin et al. [23] determined changes in the proteomic profiles of LLC1 cells carrying tumor lesions in mice treated with Ganoderma lucidum fungal immunomodulatory proteins (LZ-8) and reported that HSP60 and HSP70 inhibitors effectively inhibited cell migration and reduced cell viability, suggesting that HSPs may be associated with anti-lung cancer activity. To gain insight into the mechanism of action of different light sources on the growth and development of edible and medicinal fungi, Zhu et al. [24] compared the protein-expression profiles of comparative proteomic analysis, revealing differential protein expression of Hypsizigus marmoreus in response to different light qualities under different colors of light (white, red, green, blue, and black) based on proteomics technology; KEGG enrichment was shown to be related to the pathways of MAPK, light stress, proteasome, glycolysis, and carbohydrate-active enzymes. Coprinus comatus is a perishable and difficult-to-preserve edible and medicinal fungi owing to its high moisture content and high respiration rate. Qu et al. [25] used a tandem mass tags-based quantitative proteomic technique to analyze the proteomic response of post-harvested C. comatus stored for 16 d at 8 ± 1 °C, and the results showed that differential abundance proteins (DAPs) were mainly involved in ribosome, citrate cycle, glycolysis, and glutamate metabolism.
The secondary metabolites of edible and medicinal fungi have important applications as food additives, pharmaceutical intermediates, bio-based materials, and dietary amino acids [26]. Metabolomics technology can effectively analyze the metabolite species, content, and differences in edible and medicinal fungi. Jiang et al. [27] analyzed the metabolomics of G. lucidum after treatment with methyl jasmonate (MeJA) for 24 h using gas chromatography–mass spectrometry (Agilent, Palo Alto, CA, USA) and liquid chromatography–mass spectrometry (Thermo Fisher Scientific, Waltham, MA, USA) and reported that its cellular energy metabolism shifted from basal metabolism to secondary metabolism, confirming that MeJA was involved in the promotion of ganoderic acid synthesis. G. lucidum extract can be used as a fermentation medium to promote the growth of probiotics. Liu et al. [28] analyzed the effect of G. lucidum aqueous extract (GLE) on key metabolites of Lactobacillus rhamnosus GG (LGG) based on non-targeted metabolomics and identified different differential metabolites, such as pyrotechnic acid, taurocholic acid, and estradiol, which are mainly involved in metabolic pathways such as tyrosine metabolism, pyrimidine metabolism, and alanine and aspartate enrichment, providing theoretical support for using GLE to improve probiotic production. Zeng et al. [29] analyzed the metabolite changes of Dictyophora indusiate under three drying methods (vacuum freeze-drying FD, vacuum drying VD, and hot-air drying HD) based on quantitative metabolomics. They reported that acidity was provided by organic acids produced in the tricarboxylic acid cycle, and browning was caused by the melted reaction, the oxidative degradation of ascorbic acid, and the metabolic pathways mainly catalyzed by endogenous enzymes mainly in the phenylalanine metabolic pathway.
The results of the multi-omics joint analysis can be corroborated with each other, which can help improve the scientificity and credibility of the study and will be more widely and deeply applied to the analysis of edible and medicinal fungi functional genes in the future. Sun et al. [30] utilized the genome and transcriptome to gain insight into the molecular mechanism of nucleus formation in Pleurotus tuber-regium and reported that the genome size was 35.82 Mb and encoded 12,173 putative proteins. Of these, 1146 and 1249 genes were upregulated and downregulated, respectively, and it was hypothesized that these differential genes were related to substrate catabolism, oxidation–reduction process, cell wall synthesis, and other biological processes. Cai et al. [31] combined metabolomics and transcriptomics technology to study the browning of A. bisporus, which revealed that the genes AbPPO2, AbPPO3, and AbPPO4 were involved in the browning-related pathway and the resistance to browning could be judged by the expression level of AbPPO. Furthermore, metabolomics revealed that the metabolites of organic acids in vivo further inhibited the activity of PPO by lowering the pH value. The results of this study are significant for the adoption of appropriate measures to mitigate mushroom browning. Functional-omics technologies are the key to link gene expression and phenotype. Marian et al. [32] performed comparative genomics and transcriptomics analysis of two wild strains of Schizophyllum commune and identified roc1 as a key regulator of cellulose degradation in basidiomycetes for the first time. This model species can provide insights into cellulose degradation in basidiomycetes. In addition, advanced functional-omics technologies can be used to analyze light-responsive regulatory genes. To provide a reference for light conditions in the process of factory cultivation, Liu et al. [33] used gene expression profiling and Chip-Seq to study the molecular mechanism of GATA transcription factor CcNsdD2 from Coprinopsis cinerea in hypha kinking and light morphogenesis. The phenomenon of low-temperature autolysis in Volvariella volvacea seriously hinders the postharvest storage and marketing of commercial edible and medicinal fungi; therefore, our team analyzed the gene and metabolite expression profiles of V. volvacea after treatment with the L345-0044 inhibitor at 4 °C using absolute quantitative transcriptome and metabolome analysis [34]. The two key enzymes, tyrosinase and β-glucosidase, were screened out. The inhibitor treatment revealed that the activities of these enzymes were reduced by 27.25% and 73.20%, respectively, which is favorable for slowing down low-temperature autolysis.
In summary, whole-genome sequences can be used to gain insight into genetic data, transcriptomics can be used to analyze differentially expressed genes in different phenotypes of the same edible and medicinal fungi, and proteomics can be used to explore the biosynthetic pathways of amino acids and enzymes. Metabolomics technology can be used to predict substance metabolic pathways and identify metabolomic markers in large edible and medicinal fungi. Despite the many benefits, there are still certain limitations to genomics technologies. For example, genomics can only provide information about metabolic potential, metabolomics and proteomics show poor reproducibility, there are limitations in multi-omics technology in annotating and interpreting the functions of individual genes and molecules, and omics technologies are relatively homogenously applied [35]. Most of the genomes of edible and medicinal fungi that have been sequenced thus far are single genomes, which do not cover a wide range of genetic diversity and are difficult to study. In the future, pan-genomic studies of edible and medicinal fungi should be strengthened and combined with the joint application of multi-omics technology to facilitate a more comprehensive and precise identification of genes involved in important biological processes at the bioinformatics level. Additionally, there is a need to focus on combining wet and dry experiments to provide more reliable evidence of gene function supported by molecular genetic techniques.

2.2. Molecular Genetic Techniques in Edible and Medicinal Fungi

2.2.1. Genetic Transformation Technology

Genetic transformation technology for edible and medicinal fungi is used to obtain strains with targeted changes in genetic traits by integrating target fragments into vectors using molecular biology techniques and genetic engineering, followed by transformation into recipient cells [36]. Compared to unicellular microorganisms, such as Escherichia coli, edible and medicinal fungi have a complex genetic background, specific morphology, and thicker cell walls, which makes it more difficult to introduce exogenous gene fragments. Therefore, the development of efficient genetic transformation techniques is an effective way to explore the functional genes in edible and medicinal fungi. Genetic transformation techniques for edible and medicinal fungi include Agrobacterium tumefaciens-mediated transformation (ATMT), protoplast-mediated transformation (PMT), liposome-mediated transformation (LMT), and electroporation transformation (EP) [37]. The advantages, disadvantages, and applications of these approaches are summarized in Table 1. ATMT and PMT are more commonly used in laboratories because they combine conversion efficiency, stability, ease of operation, and equipment requirements.
ATMT technology has been used in combination with target gene overexpression or RNA interference methods to study gene functions related to growth and development, environmental stress responses, and the metabolism of functional, active ingredients in a variety of edible and medicinal fungi (Table 2). Overexpression involves placing the target gene downstream of a strong promoter or an inducible promoter to enhance the expression of the target gene, whereas RNA interference technology silences the target gene by constructing a hairpin or reverse double promoter structure. As can be seen from Table 2, many studies have been conducted on F. filiformis, L. edodes, and G. lucidum. For example, fvhom1, Ste12-like, FIP-fve, and FfGal in F. filiformis were overexpressed by ATMT, and RNA interference was performed on FIP, FvHmg1, Fv-ada, and FfGal. These genes are involved in the growth, development, and environmental stress of F. filiformis. Hsp20 and LeHD1 were overexpressed in L. edodes, and RNA interference was performed on LeHD1, LELCRP1, LeDnaJ, TrpE, YUCCA, and LetrpB. These genes are involved in the growth, development, biodegradation, and environmental stress responses of L. edodes. Based on the overexpression of D9desA, Gl-aact, and HMGR in G. lucidum, these genes participate in the biosynthesis of functionally active substances.
Conversion efficiency is an important index for measuring ATMT technology, and studies have shown that the promoter, receptor material, and Agrobacterium tumefaciens type are important influencing factors [64]. Promoters are important cis-acting elements that control gene transcription, and there may be significant differences in the levels of exogenous gene expression driven by different promoters. Lv et al. [65] utilized a binary vector consisting of polyubiquitin and the GPD promoter in conjunction with the ATMT method to successfully achieve the genetic transformation of M. esculenta. Our team constructed two plasmids with CaMV35s and H. marmoreus GPD as promoters and reported that the latter increased the expression levels of exogenous genes by 22–36 times [66]. The genome of Pleurotus eryngii contains two gpd genes, of which gpd1 is significantly more highly expressed than gpd2 at different developmental stages and drives the expression of exogenous genes more efficiently [67]. Receptor materials commonly used for ATMT in edible and medicinal fungi include mycelia, mononucleate spores, and protoplasts, with varying conversion efficiencies. Liu et al. [68] used the mycelia of A. bisporus as the receptor material and a colonized millet grain method for transformation by Agrobacterium, which resulted in a transformation efficiency of up to 53.85% with more than 85% of the transformants remaining mitotically stable for five consecutive rounds of culture. Liu et al. [69] used the mycelial fragments and protoplasts of mononuclear dans and binucleate G1 as conversion receptors and reported that the mycelial fragments of the binucleate had the highest conversion efficiency, up to 40.31%.
Differences in the efficiency of genetic transformation mediated by A. tumefaciens were observed. In P. ostreatus, the conversion efficiencies from highest to lowest are LBA4404, GV3101, EHA101, and EHA105 [70]; therefore, selecting suitable strains is conducive to genetic transformation. In addition, the mycelium medium and receptor genotype play key roles in conversion efficiency. Yan et al. [71] established a genetic transformation system for the mononuclear and binuclear bodies of L. edodes and reported that the conversion efficiency of the grain medium was the highest, up to 75.48%, and the conversion efficiency of binuclear bodies was higher than that of mononuclear bodies, providing a new method for the genetic transformation of edible and medicinal fungi. Since the first successful transformation in yeast in 1978, the PMT method has gradually become a common cell fusion technology based on the principle that the formation of precipitates on the cell surface by PEG and divalent cations, such as Ca2+, Mn2+, and Mg2+, with exogenous DNA alters the permeability of the membrane and facilitates the entry of exogenous DNA. The green fluorescent protein reporter gene was transferred into Agrocybe aegerita using the PMT method. The results showed that the expression of the GFP gene was independent of introns, laying a foundation for subsequent research on functional genes [72]. Antroquinonol (AQ) is an important biologically active constituent of Antrodia cinnamomea with antiviral, nephroprotective, and neuroprotective effects [73]. Liu et al. [74] transferred and overexpressed ubiA and CoQ2 genes, which enhance antroquinonol production via the overexpression of 4-hydroxybenzoate polyprenyltransferase biosynthesis-related genes in A. cinnamomea; the concentration of AQ was 2- and 2.61-times higher than that of the control after fermentation, respectively. Pleurotus ferulae, a unique edible fungus from Xinjiang, China, can effectively degrade lignin. However, its low conversion efficiency has limited the systematic study of its molecular mechanisms and metabolic control. Therefore, Zhang et al. [75] established the PMT conversion method in P. ferulae for the first time and reported that adding 50 μg SS-DNA, 70 μg λDNA, and 0.4 μmol spermidine could maximize the conversion efficiency. An overexpression system containing three different endogenous promoters (PPFGPD1, PPFGPD2, and PPFSAR1) was established to drive the expression of different genes, promoting the expression of functional genes and novel secondary metabolic pathways.
Currently, the main screening markers used in ATMT technology are antibiotics, metabolites, nutritional deficiencies, herbicides, and fungicide-resistance genes, which may pose threats to environmental and food safety. Therefore, the selection of biosafety genes as screening markers is a focus for the optimization of edible and medicinal fungi genetic transformation technologies in the future. Our team used Pesdi1, an autosomal mutated gene in P. eryngii, as a safety-selective marker and achieved ATMT in P. eryngii, which provided a reference for the development of biosafety markers in other edible and medicinal fungi [76]. However, the molecular mechanism of T-DNA transfer and integration in fungal receptors is different from that of plants, which needs to be further explored to determine its signaling and interaction patterns and to improve the expression of exogenous genes in edible and medicinal fungi [77]. In addition, the key to genetic transformation is the preparation and regeneration of protoplasts; the age of the bacteria, enzyme system, osmotic stabilizers, enzyme digestion time, and temperature will affect the quantity and quality of protoplasts [78], and conditions should be optimized and explored for different edible and medicinal fungi for genetic transformation to achieve the best efficiency.

2.2.2. Gene-Editing Technology

Gene-editing technology uses artificially designed or modified nucleases to make precisely targeted modifications to the genome. Double-stranded DNA repair is performed by cutting a specific site using homology-directed repair (HDR) or non-homologous end joining (NHEJ) [79]. CRISPR/Cas9, a third-generation gene-editing technology with the advantages of simple operation, high precision, and low cost, has been widely used for gene editing in edible and medicinal fungi in the past few years, such as in A. bisporus, G. lucidum, C. cinerea, and C. militaris.
A. bisporus was the first edible fungus to undergo gene editing using the CRISPR/Cas9 system. In 2016, Yang et al. [80] used this system to knock out one of six genes encoding polyphenol oxidase (PPO) in A. bisporus, thereby reducing PPO enzyme activity by 30%, effectively slowing down postharvest browning and extending shelf life. The announcement by the United States Department of Agriculture (USDA) in the same period confirmed the feasibility of gene editing in crop applications, and since then, it has been progressing toward an era of sophisticated molecular breeding, gradually paving the way for the era of synthetic biology.
G. lucidum is an important medicinal fungus, and the development of a suitable gene-editing system can provide an efficient and feasible pathway for the efficient synthesis of secondary metabolites. Professor Zhong Jianjiang’s group at Shanghai Jiaotong University successfully established a CRISPR/Cas9-mediated gene-editing system in G. lucidum for the first time [81], successfully controlled the expression of Cas9 using the G. lucidum gpd promoter and Trichoderma reesei pdc terminator, adopted the T7 promoter for in vitro transcription of sgRNA, successfully transcribed them into Cas9 protein-expressing strains, and ultimately succeeded in obtaining uracil nutritionally deficient strains. Wang et al. [79] studied cyp515018, a gene encoding cytochrome P450 monooxygenase, using the CRISPR/Cas9 system and reported that the production of ganoderic acid was significantly reduced in the mutant compared to the wild type. Liu et al. [82] added introns upstream of the Cas9 gene to reach 14–18 ura3 mutants, whereas the use of double sgRNA in G. lucidum resulted in the deletion of ura3 and GL17624 gene fragments, effectively expanding the library of mutants in G. lucidum. Using the PMT of G. lucidum, Zhang et al. [83] obtained a strain containing a complete cas9 expression box and optimized this using 0.006% Tratone X-100, increasing the number of ura3 mutants from 4 to 18 and providing a more efficient method for future studies of gene function and molecular breeding.
C. cinerea is a model organism that can help researchers understand the growth, development, and reproductive mechanisms of other edible and medicinal fungi. Sugano et al. [84] successfully applied CRISPR/Cas9 gene-editing technology in this species using the CcDED1 promoter to express codon-optimized Cas9 genes from humans, Candida, Arabidopsis, and Basidiomycota and ligated the same vector with sgRNA expression elements. Only 19 transformants from C. cinerea were observed to have reduced green fluorescent protein. The attenuation of green fluorescent proteins was detected in the two transformed strains, with an editing efficiency of 10.5%. Yu et al. [85] transformed in vitro-expressed Cas9 and sgRNAs into C. cinerea via protoplasts and did not detect pyrG editing, which was hypothesized to be due to the degradation of Cas9 proteins and sgRNAs during the transformation process or because the buffer used in vitro cleavage experiments was unfavorable for the assembly and zymography activities.
C. militaris is an edible medicinal fungus, and the successful application of gene-editing technology has accelerated the development and utilization of its active substances. Chen et al. [86] used PMT to introduce fragments containing Cas9 and GFP fusion genes into Cas9 protein-expressing strains and successfully mutated the target site with an editing efficiency of 11%. In the same year, Meng et al. [87] constructed an efficient gene-editing system based on autonomously replicating plasmid with an AMA1 sequence for the first time in C. militaris and, through homologous directed repair, the efficiency of Cmwc-55 and Cmvvd reached 1.89% and 1%, respectively. The simultaneous editing efficiency of dual genes was 10%. This technique can be used for the simultaneous modification of multiple genes in functional gene research.
Subsequently, researchers developed a pre-assembled Cas9-sgRNA ribonucleoprotein (RNP) technology based on the CRISPR/Cas9 gene-editing technology. These two principles are similar, but RNPs can be incubated with purified Cas9 protein and synthesized sgRNA in vitro to form the RNP complex without plasmid construction during the experiment, which brings the technical advantages of simplicity and high universality [88]. In 2017, Al Abdallah et al. [89] successfully edited pksp, a gene related to melanin synthesis in Aspergillus fumigatus, using an RNP system. In 2019, Jan et al. [90] constructed an RNP system for S. commune, a model organism of edible and medicinal fungi, and successfully edited the effective editing of transcription factor hom2. In 2021, our team optimized the RNP system by adding the surfactant Triton X-100, which increases the proportion of homonuclear transformants in filamentous fungi [91]. Subsequently, in cooperation with other teams, the RNP complex-mediated HR system effectively knocked out three target genes of alkaloid synthesis in Claviceps purpurea, with editing efficiencies ranging from 50% to 100%, which greatly facilitated the study of the ergot alkaloid synthesis pathway [92]. In 2022, Liu et al. [93] optimized a CRISPR/Cas9 gene-editing method for the in vitro assembly of ribonucleoprotein complexes in F. filiformis with the addition of Triton X-100. This resulted in editing efficiencies of up to 5% and the more rapid growth of transformants compared to common gene-editing techniques. In 2023, Boontawon et al. [94] established a system of RNPs in P. ostreatus without exogenous DNA genes, and double-gene-edited mutants were obtained by screening RNP-added strains for 9-FOX resistance.
The genetic transformation of edible and medicinal fungi is relatively difficult, and with the advent of the post-genomic era, many predicted and annotated functional genes have been successfully validated using gene-editing techniques. However, the CRISPR/Cas9 system faces several challenges, including low editing efficiency, poor accuracy, off-target effects, and difficulties in screening mutants [95]. In the future, we can improve the editing efficiency by optimizing the codons of target species and selecting appropriate promoters to express Cas9 genes; improve the accuracy by inhibiting NHEJ repair or enhancing HDR repair; detect off-target effects on time using chromatin immunoprecipitation sequencing (ChIP-seq) and genome-wide, unbiased identification of DSBs enabled by sequencing (GUIDE-seq) methods, optimized sgRNA sequences, and the modification of Cas9 proteins to reduce off-target effects; and the addition of chemical reagents to increase the proportion of mononuclear protoplasts to improve the efficiency of obtaining pure transformants, thus reducing the difficulty of screening [96].

2.3. Chassis Cell Technology

Chassis cells are the host cells where metabolic reactions occur and are the reaction platforms into which systems of natural or synthetic functionalized components, circuits, and pathways are placed for rational design purposes [97]. Introducing target genes/gene clusters obtained from screening or localization into adapted chassis cells to achieve heterologous expression is an important method of verifying gene functions [98]. To date, chassis cell systems used in functional gene studies of edible and medicinal fungi include E. coli, Saccharomyces cerevisiae, Pichia pastoris, and T. reesei.
For a long time, E. coli has been used as a model strain for genetic and metabolic engineering because of its simple culture conditions, short growth cycle, and clear genetic background, making it an ideal host for heterologous expression. Adams et al. [99] cloned psilocybin synthesis genes (psiD, psiK, and psiM) and heterologously expressed them in E. coli, which increased the yield of psilocybin to 1.16 g/L at the fermenter level and improved the feasibility of industrial production. To address the problem of unclear key enzymes in the sugar donor synthesis pathway of G. lucidum polysaccharides, Li et al. [100] cloned three key enzyme genes of G. lucidum polysaccharides (gl-pgm, gl-ugpg, and gl-pmi) and induced expression and mass preparation in E. coli with the specific enzyme activities of 4.75, 6.26, and 13.68 U/mg after purification, which further provides theoretical bases for the development of efficient fermentation regulation strategies. Fungal glucans play key roles in providing energy and maintaining cellular structures; however, their biosynthetic mechanisms need to be elucidated. Liang et al. [101] optimized the (UDP)-glycosyltransferase GFUGT88A1 in Griflola frondosa and successfully heterologously expressed it in E. coli. The molecular weight of the purified protein was 51.7 kDa, which provides a reference for the resolution of the glucan biosynthesis pathway in edible and medicinal fungi.
Yeast and edible and medicinal fungi, both eukaryotes, are widely used because their secretory pathways are capable of protein processing and post-translational modification without endotoxins. Wang et al. [102] screened cytochrome P450 monooxygenase (CYP450) candidate genes in G. lucidum using S. cerevisiae as a host and reported, for the first time, that one of them, cyp5150l8, could produce anti-tumor ganoderic acid in this host at a concentration of 5.120 mg/L after 5 days of liquid fermentation. This provides an effective strategy for optimizing heterologous cell factories for ganoderic acid production. Bai et al. [103] cloned CDA3, the coding gene for a novel chitin deacetylase in C. cinerea, and introduced it into P. pastoris GS3 for successful expression. The affinity chromatography-purified CDA3 protein yielded 0.067 mg/mL, with a molecular weight of approximately 27 kDa, which has the potential to be further applied to the large-scale production of chitosan. Yang et al. [104] cloned the glutamine transaminase (TGase)-coding gene tgM from C. militaris and expressed it in P. pastoris GS115, with an enzyme activity of 100 U/L in the supernatant of recombinant yeast fermentation. This provides a reference for the heterologous expression of TGase and its potential industrial application. Lee et al. [105] cloned cml, the lipase-coding gene of C. militaris, and introduced it into P. pastoris X-33 for heterologous expression and reported that the specific activity of the purified protein was 6.52 ± 0.26 U/mg, which showed a stronger catalytic efficiency compared with that of the unpurified protein and is expected to be developed as a biocatalyst.
T. reesei is the main industrial production strain of cellulase and hemicellulose, with strong protein-secretion ability, and is one of the most widely researched industrial fungi, with more than 80 years of research and application development. Dong et al. [106] expressed the optimized laccase-encoding gene pox1 from P. ostreatus in T. reesei and reported that the exogenous laccase gene could be efficiently secreted and expressed with an enzyme activity as high as 237.1 IU/mL. Zhou et al. established a gene-editing system in T. reesei via the in vitro transcription of codon-optimized Cas9 and gRNA, and multiple genome edits with three targets were obtained based on PMT [107].
Chassis cell technology has many benefits, but there are still challenges in the heterologous synthesis of a few active secondary metabolites, such as alkaloids and phenols, including a lack of precise regulation, complex gene structure, and antibacterial properties of bioactive natural products that are toxic to chassis cells [12]. Therefore, the development of chassis cells with better compatibility for edible fungal metabolites can better realize the large-scale production of complex active metabolites from edible and medicinal fungi using synthetic biology technology.

2.4. Synthetic Biology

Synthetic biology is a discipline that combines observational and analytical methods in life sciences with design thinking in engineering to create new biological components, devices, and systems or redesign and adapt existing living systems in nature. Compared to other microorganisms, edible and medicinal fungi have large genomes and complex metabolic regulatory networks, and high-throughput screening of biosynthetic gene clusters to promote the production of active substances is extremely important.
The use of edible and medicinal fungi, such as G. lucidum, C. militaris, and H. erinaceus, can synthesize a variety of drugs and develop high-value-added cell factories based on the active substances of edible and medicinal fungi. Ganoderic acid is a series of highly oxidized secondary metabolites naturally produced by the edible and medicinal fungi Ganoderma. It is difficult to elucidate the biosynthetic process of ganoderic acid by analyzing the function of the P450 gene, which is responsible for the multi-step oxidation. Zhong Jianjiang’s group at Shanghai Jiao Tong University and the Tianjin Institute of Industrial Biotechnology cooperated in establishing a high-throughput automated screening platform [108], which utilized a tunable expression strategy, functionally characterized hundreds of cytochromes P450 from G. lucidum in S. cerevisiae, excavated and discovered key genes of the type-II ganoderic acid biosynthesis pathway, and efficiently biosynthesized the genes in heterologous form in S. cerevisiae. The highest content of ganoderic acid reached 56.44 mg/L after liquid fermentation for 144 h [109]. This work shows the potential and application value of synthetic biology in the discovery of novel natural products. Chen et al. [110] developed a marker-free CRISPR-Cas9-TRAMA genome-editing system for C. militaris to achieve the precise editing of multiple genes and the deletion of large clusters of genes, which can be used for mining and utilizing medicinal metabolic pathways. Aspergillus oryzae has a complete translational modification system; strong protein production and secretion capacity; secretes amylase, protease, pectinase, and saccharification enzymes; and is widely used in cell factories to produce a variety of enzyme preparations. Han et al. [111] identified a gene, herA, encoding orsellinic acid (OA) polyketide synthase in H. erinaceus and successfully heterologously expressed it in A. oryzae; the rate of OA production reached 57.68 mg/L by liquid fermentation when maltose was used as the carbon source, which could provide a valuable reference for the biosynthesis of important pharmaceutical intermediates.
In addition to promoting the precise breeding of edible and medicinal fungi and optimizing their growth, synthetic biology strategies can be used to mine a variety of biological components in large fungi, including edible species. Based on microbial chassis cells with excellent performance, an efficient green biofuturing technology has been developed to synthesize high-value natural drug-active molecules, which has gradually become a research hotspot in the field. Several studies have shown that biosynthetic gene clusters can be randomly activated by modifying the expression of signaling pathways, cell wall structures, and stress response elements using synthetic biology strategies. In the future, based on a deeper understanding of the gene-splicing mechanism, we can study and develop synthetic components that can be effectively used in multi-gene expression systems, construct efficient chassis that can be easily modified and screened in high throughput [112], and realize the synthesis of large quantities of target active molecules and their derivatives from edible fungus sources. The efficient degradation of SMS could be realized to significantly contribute to the healthy development of the edible fungus industry under the goal of “carbon neutrality” and the creation of a high-economic-value industry.

3. Advances in the Analysis of Important Functional Genes of Edible and Medicinal Fungi

Through the continuous development and optimization of various functional gene research techniques in edible and medicinal fungi, the functions of a few important genes have been elucidated, including genes related to mating type, mycelial growth, substrate utilization, nutrient transport, environmental response, and the synthesis and regulation of important active substances. The expression of these genes directly or indirectly affects important traits, such as the growth cycle, yield, quality, and active ingredients of edible and medicinal fungi. Compared with the traditional methods of edible and medicinal fungi breeding, the exploration and analysis of functional genes will provide new ideas for modernized molecular and design breeding.

3.1. Mating-Type Genes

In edible and medicinal fungi, mating-type genes control the mating behavior between amenable monokaryosomes and the expression of downstream genes that ultimately lead to the production of sexual spores. The mating-type genes of edible and medicinal fungi such as C. militaris, M. esculenta, F. filiformis, L. edodes, and P. eryngii are now known.
The sexual reproduction of Ascomycota is controlled by mating-type loci that contain two allogeneic genes—MAT1-1 and MAT1-2. The degradation of C. militaris strains cannot be ignored and can lead to a slow growth rate, low conidial production, reduced number of primordia, a prolonged growth cycle, and reduced secondary metabolism, resulting in significant economic losses [63]. Feng et al. [113] pre-screened the excellent traits of artificially cultivated C. militaris and isolated two different mating-type genes, Mat-1 and Mat1-2, from 72 monospore strains. They conducted cultivation experiments to show that only strains with different mating-type genes can develop into fruition bodies, which provides a theoretical basis for preventing the degradation of C. militaris during large-scale cultivation. To increase the chance of success of single-spore cross-breeding of M. esculenta, Guo et al. [114] screened MAT1-1 and MAT1-2 mating types, a total of 20 single mating-type hybrid parents with good culture traits, and hybrid strains of mating-type genes. They reported that the fungus cap is large, and the stipe is small, non-friable, and has good commercial properties, which can help promote the selection of good varieties of M. esculenta and provide a theoretical basis for their cross-breeding.
Basidiomycota comprise the majority of edible and medicinal fungi and are complex mating-type systems composed of multiple loci and multiple alleles, which are usually divided into dipolar and tetrapolar mating systems, accounting for 25% [115] and 55–65% [116], respectively. The most typical of these is the latter, which contains two unlinked A mating-type and B mating-type loci. According to edible and medicinal fungi genetics, the A mating-type loci contain HD1 and HD2 genes encoding homology domain transcription factor proteins, which control behaviors such as the production of clamp connections, nucleus pairing, and division of the nucleus [117,118], whereas the B mating-type loci encode pheromone receptors and pheromone precursors, which control behaviors such as the migration of the nucleus and ablation of septa between cells [119]. The structure of the mating-type loci of common edible and medicinal fungi is shown in Figure 2 [120,121,122,123,124,125,126], which shows that the A mating-type loci contain a pair of mating-type genes that encode a pair of dissimilar homozygous proteins—HD1 and HD2; only at different alleles can they be paired to form heterodimers, whereas the B loci contain a pheromone receptor-coding gene and one to several pheromone-coding genes and originate from different alleles before they can recognize each other. From the perspective of collinearity, most of the A mating-type loci follow the typical mip-HD-β-fg structure, but L. edodes and F. filiformis do not, and the order and number of genes present at the loci vary considerably in different species, which possesses the characteristics of mating-type locus polymorphism. With the continuous evolution of nature, certain mating genes will be added or lost; separate HD genes will exist at the A locus of F. filiformis, A. bisporus, C. cinerea, S. commune, P. ostreatus, etc.; and a few pheromone precursor coding genes will exist outside the B locus. The mating of compatible monokaryons carrying mating genes A and B is necessary for the formation of fruiting bodies. The A mating genes HD1 and HD2 and the B mating genes PEphb3.1 and PEphb3.3 from the monokaryon of P. eryngii were knocked into compatible monokaryotic strains by our team using the PMT technique [120]; the obtained transformants had clamp connections and distinct dikaryotic mycelium cells, which is conducive to the further exploration of mating-type gene function. Subsequently, the team knocked A and B mating genes into the mononucleosomes of P. eryngii and reported that they could promote the development of the fruit body but could not form basidiospores. Transcriptomic analyses showed that the transformants activated the expression of endogenous homology domains, pheromone receptor genes, and mating-type signaling pathways, providing new avenues for the study of fruiting body development [127].
Mating-type genes are important functional genes that regulate the development of mycelia and fruiting bodies in dikaryotes and have a complex mating-type system that should be widely considered. A comprehensive understanding of the mating loci and genetic polymorphisms of strains is of great benefit for species maintenance, variety innovation, strain identification, and molecular breeding. However, there are still significant gaps in the field of mating-type genes, such as insufficient knowledge of the HD genes alone in the A locus of edible and medicinal fungi and the pheromone precursor coding genes outside the B locus, which need to be further studied in depth by resolving methods such as gene insertion and knockout. The evolutionary process and causes of the alteration of chromosome structure as a result of the covariation of mating-type genes still have to be investigated in detail, and it is necessary to identify the effect of synthetic pheromone peptides or products secreted through the autoregulation of clamp cell formation [128].

3.2. Research Progress on Genes Related to Mycelium and Fruiting Body Development

Since the middle of the 20th century, the mechanisms of fruiting body growth and development have become the focus of research on edible and medicinal fungi. As the most complex structure in fungi, the growth and development of fruiting bodies are synergistically regulated by environmental factors such as temperature, humidity, light, and CO2 concentration and intrinsic factors dominated by genes. The growth and development stages involve the regulation of a variety of pathways, including signaling, nutrient synthesis and transport, metabolism, hormones, and transcription.
Currently, the functions of more than 40 genes related to mycelial growth and fruiting body development of edible and medicinal fungi have been analyzed and verified, mainly in commonly cultivated species such as F. filiformis, G. lucidum, P. ostreatus, C. militaris, and V. volvacea (Table 3). Among them, regulatory genes can be mainly divided into coding genes, such as hydrophobin, lectin, transcription factor, and laccase, which play crucial roles in the different growth and developmental stages of edible and medicinal fungi during their complete life histories.
Hydrophobic proteins are involved in multiple developmental processes in edible and medicinal fungi and play an important role in spore spread, pathogenesis, and the formation of ectomycorrhiza. Tao et al. [136] used interference methods to identify the function of Hyd9 in F. filiformis and reported that the surfaces of the aerated mycelia were dense after overexpression, while the surfaces of the silenced mycelia were sparse, suggesting that the hydrophobic protein could help fungi form aerated mycelia. Han et al. [129] knocked out the vmh2 and vmh3 genes in P. ostreatus and reported that the strain grew slower than the wild type under the stressed conditions of SDS and H2O2. However, for C. militaris, the class II hydrophobic protein-coding gene Cmhyd1 and the class I hydrophobic protein-coding gene Cmhyd4 played opposite genetic functions. Li et al. [151] constructed a Cmhyd4 knockout vector, and the deletion of this gene increased the density, height, and conidium production of the strain compared with the wild type and negatively regulated the development of the fruiting body of C. militaris.
Lectins, as non-immunoglobulins, play an important role in the growth and development, morphogenesis, and molecular recognition of edible and medicinal fungi during the early stages of mycorrhizalization and nutrient storage. Su et al. [130] cloned polectin2 from P. ostreatus, successfully constructed a recombinant expression vector, conducted low-temperature stress experiments on a positive transformer, and reported that the gene has a low-temperature tolerance function. Lu et al. [137] used RNA interference and the overexpression of Fv-JRL1 in F. filiformis and reported that this gene plays a positive role in the formation of aerogenic mycelia and fruiting bodies. Our team cloned the CMlec3 gene in C. militaris, successfully expressed it heterologously in E. coli, and reported that the expression level of this gene in the primordium was 5.19 times that in the mycelium and 1.35 times that in the fruition body, indicating that this gene is related to fruiting body development [152].
Transcription factors are important protein molecules involved in transcription initiation and play important roles in mycelial growth, fruiting, spore formation, sexual reproduction, and sclerotium formation in edible and medicinal fungi [168]. Lyu et al. [138] conducted knockout and overexpression experiments on Fvclp1 in F. filiformis and reported that the knockout strain produced fewer fruiting bodies than the wild type, and the overexpressed strain had improved fruiting ability. This indicates that Fvclp1 can effectively promote sexual reproduction and fruiting body development. Wu et al. [140] verified the function of lfc1 in F. filiformis and reported that the strains overexpressing lfc1 had reduced buttons and folds on the edge of the cap, and the stalk length of the strains with low lfc1 increased by more than 27%. These results suggest that the goals of shortening cultivation times and increasing yields could be achieved by reducing the expression of lfc1. Zhang et al. [149] silenced GlSwi6 in G. lucidum and observed that the mutant strains exhibited multiple developmental defects, such as reduced mycelial growth and the formation of fruitless bodies. Chen et al. [160] overexpressed the Vvrin1 gene of V. volvacea, and the transformed hyphae obtained were thicker. The surface color of the colony was darker, indicating that the gene regulates the mycelial growth rate and pigment synthesis or accumulation in this species.
Laccase is a copper-containing polyphenol oxidase that has a strong ability to degrade lignin and catalyze the oxidation of various substrates. Most edible and medicinal fungi are capable of secreting laccases during growth and development, thus effectively utilizing lignin in the substrate. Armas-Tizapantzi et al. [134] silenced lacc2 in P. ostreatus and reported that laccase activity decreased by 30–55%; mycelium development was delayed in PDA medium compared to the wild type; and air mycelia, primary bases, and fruity bodies could not be formed in wheat straw cultures. Zhang et al. [155] cloned and overexpressed lcc1 in H. marmoreus and reported that the transformed mycelia grew faster, the fruit body matured approximately five days earlier than the wild type, and the laccase activity was higher during development. These results suggest that lcc1 plays a positive role in regulating the growth and development of mycelia and fruit bodies.
Currently, there are many studies on the regulatory genes of mycelial and fruiting body growth and development in edible and medicinal fungi, but the similarity of the same family of genes from different species is quite different. Most studies have focused only on whether and how many genes are expressed at different stages of fruiting body growth and development, and there are few studies on the response mechanisms of various genes and environmental factors. The development and maturation of edible and medicinal fungi are adaptations to matrix carbon and nitrogen sources, pH, humidity, temperature, and other environmental factors. Therefore, combined with the study of the regulatory mechanisms, we can better understand the functions of genes related to the development of edible fungal mycelia and fruiting bodies.

3.3. Research Progress on Genes Related to Substrate Utilization and Nutrient Transport

Large amounts of rice, wheat, corn straw, and other agricultural by-products are produced worldwide every year, which are rich in cellulose and hemicellulose for resource utilization. Further enhancing the hydrolysis ability and saccharification efficiency and solving the inhibition of complex products has become an important issue in the field of substrate utilization by edible and medicinal fungi. Li et al. [169,170] expressed LeXyn and LeCel7A, the xylanase-encoding genes of L. edodes in P. pastoris, and reported that the two recombinant proteins enhanced the hydrolyzing ability of rice straw, wheat straw, and corn straw. The team further reported that L. edodes xylanase could effectively hydrolyze wheat and simultaneously resist pepsin degradation at the same time, which could improve the utilization rate of wheat in animal feed [171]. To improve the cold adaptability of mesophilic cellulase, our team isolated a novel cold-active cellobiohydrolase-coding gene vvcbh1-1 from V. volvacea and successfully expressed it heteroglyphically in T. reesei after low-temperature induction, which is beneficial for the efficient low-temperature storage of straw [172]. Li et al. [173] successfully expressed the new lytic polysaccharide monooxygenase coding gene PdLPM09A from Pleurotus djamor in P. pastoris, and when the dosage of rPdLPM09A was 0.66 mg/g and hydrolyzed for 48 h, the greatest improvement in cellulase-mediated saccharification of corn stalk was achieved. The product inhibition effect of the cellulose hydrolysis process is an important problem to be solved in cellulase preparations. Our team codon optimized and heterologously expressed the endoglucanase-encoding gene pacel3a from Polyporus arcularius in T. reesei; when compared with the natural PaCel3A, the recombinant protein CMCase had higher activity and the glycosylation efficiency was increased by 17%, which effectively mitigated the product inhibition of the cellulase complex of T. reesei [174].
Forest ecosystems play an important role in the global carbon cycle, where edible and medicinal fungi degrade persistent organic compounds in soil litter and humus layers. Heneghan et al. [175] silenced the serine protease-coding gene SPR1 in A. bisporus and reported that SPR1 contributed to nutrient acquisition in compost and was a key enzyme in the biomass degradation process. Jiang et al. [176] analyzed the high-quality genome of G. lucidum and reported that salh, phea, cyp53a1, cyp102a, glpk, and amie in G. lucidum are enriched in functions such as plant–pathogen interactions and the benzoate degradation pathway, which provides valuable genomic resources for the utilization of conifer cultivation substrates.
Edible and medicinal fungi are rich in the high-quality proteins and amino acids required by humans, and their content and composition are important indicators for evaluating the nutrition and flavor of edible and medicinal fungi. Lian et al. [177] silenced the alkaline leucine-transcription-factor-coding gene Gcn4 in G. lucidum and reported that the amino acid content of the mutant strain was reduced by 54.2–88.3% compared with the wild type. They hypothesized that GCN4 could modulate the expression of amino-acid-related synthesis genes and transporter genes, providing precursors for amino acid biosynthesis by enhancing the TCA cycle and glycolytic pathway. Carbon Catabolite Repressor (CCR) directly affects the selective utilization of complex carbon sources by fungi. Pareek et al. [178] used the RNPs system to knock out cre1, an important transcription-factor-encoding gene of CCR in C. cinerea and reported that the gene expression profile of the mutant showed dysregulation of transcription-factor-encoding genes, such as carbohydrate metabolism, plant-cell-wall-degrading enzymes, and plasma membrane transporters, which provided gene recognition targets for related transcription factors in the process of wood degradation by edible and medicinal fungi.

3.4. Genes Related to Environmental Response

In addition to the genetic background and culture medium, the growth environment is a key factor affecting the fruiting body development of edible and medicinal fungi, having impacts on important traits such as shape, yield, flavor, and stress resistance. Exploring the response mechanisms of edible and medicinal fungi to environmental factors, such as temperature, humidity, light, and carbon dioxide concentration, can help enhance cultivation environment control and, thus, improve the yield and quality of germplasm. As shown in Table 4, the functions of environmental-response-related genes in edible and medicinal fungi are mainly related to temperature, light, and metal stress.
Temperature plays an important role in fruiting body differentiation in edible and medicinal fungi. Yang et al. [179,181] conducted heat and cold stress treatments on V. volvacea, analyzed the enzyme activities and growth of the strains, and reported that VvCAT1 had a defense response function under low-temperature stress and Mn-SOD could tolerate heat, cold, and salt stress, providing a theoretical basis for the anticlimactic-directed breeding and preservation of V. volvacea. In addition, researchers have reported that LeMnP1, LeDnaJ, YUCCA8, TrpB, and LetrpE in L. edodes are beneficial for enhancing the heat tolerance of mycelia and improving the defense response to heat stress. Over the course of long-term evolution, edible and medicinal fungi have developed a perfect photosensory system to better adapt to the environment. For example, the blue light receptor coding gene Cmwc-1 in C. militaris contributes to pigment precipitation and promotes conidiation [182], the blue light complex transcription factor coding genes Wc-1 and Wc-2 in S. commune inhibit vegetative growth and protect against phototoxicity [183], and blue light receptor coding gene Icwc-1 in Isaria cicadae has the function of regulating genes related to pigment and enzyme synthesis [184]. Similarly, the blue light and its receptor white collar complex coding genes FfWc1 and FfWc2 in F. filiformis can regulate the role of morphogenesis [185]. In addition, Gf.BMR1 from G. frondosa contributes to fruiting body development and pigment accumulation [186], abl-D from L. edodes promotes the formation of brown film in mycelia [187], and Pabs from A. bisporus [188] enhances UV tolerance. These studies can provide references for light control in the production of edible and medicinal fungi in factories. Since edible and medicinal fungi can accumulate various trace elements in the cultivation matrix, excessive heavy metals have become a food safety issue for consumers. M. esculenta is a rare edible fungus that needs to be cultivated in soil. However, heavy metal pollution in the soil poses a potential threat to its mycelial growth and the quality and safety of fruiting body products. Chen et al. [189] reported that ATX1, a keto chaperone gene, may play an important role in response to Cd stress in M. esculenta, which is of great significance for revealing the molecular mechanisms of Cd metabolism and the reduction of Cd in edible and medicinal fungi. Bian Yinbing’s team analyzed the molecular mechanism of cadmium tolerance differentiation in L. edodes using combined mRNA and milRNA analysis and verified the role of α amylase gene LeAmy in enhancing the cadmium tolerance of L. edodes [190]. This lays an important theoretical foundation for the selection of excellent L. edodes varieties that are suitable for heavy metal remediation.

3.5. Research Progress on Genes Related to the Synthesis and Regulation of Important Active Substances in Edible and Medicinal Fungi

Over the course of long-term evolution and environmental adaptation, edible and medicinal fungi have developed efficient chemical defense mechanisms and produced metabolites with novel structures and diverse activities. Based on their chemical structures, these can be divided into polysaccharides, terpenes, phenols, and nitrogenous heterocyclic compounds and their derivatives. Many of these compounds have been proven to have immunomodulatory, anti-tumor, antibacterial, antiviral, antioxidant, antihypertensive, and antilipemic properties [191]. The analysis of the biosynthetic regulatory genes in the metabolic pathways of the active metabolites from edible and medicinal fungi helps to achieve the targeted synthesis of target molecules using synthetic biology technologies and provides a new method for the cultivation of new varieties and processed products with special nutrition, flavor, and health effects in the edible and medicinal fungi industry.

3.5.1. Ganoderma Triterpenoids

G. lucidum is an important medicinal and food-homologous fungus. Various triterpenoids have been isolated from G. lucidum spore powder, fruiting bodies, mycelia, and fermentation broth extracts, which have a wide range of pharmacological activities, such as anti-tumor [192], antivirus [193], anti-inflammatory [194], liver protection, and detoxification activities [195], and ameliorating the psychological effects of anxiety and depression [196].
The biosynthesis of Ganoderma triterpenoids can be divided into two parts—the synthesis of lanosterol and the biosynthesis of different ganoderic acid monomers (Figure 3) [197,198,199]. Lanosterol is synthesized via the mevalonate pathway (MVA), which is followed by a series of reactions to produce lanosterol-containing triterpenes and ganoderic acid monomers with different structures. Currently, Gl-acat, Gl-hmgs, Gl-hmgr, Gl-mvd, Gl-fps, Gl-sqs, and Gl-ls are the key genes cloned in the triterpene biosynthesis pathway of G. lucidum, and their gene expression levels are consistent with triterpene content [197]. In addition, the biological regulatory factors of Ganoderma triterpenoids include Ca2+ signaling and plant hormones, such as salicylic acid (SA) and methyl jasmonate (MeJA), which regulate the biosynthesis of Ganoderma triterpenoids and CYP450 inducers [200].
Ganoderic acid is a highly oxidized secondary metabolite of lanosterane-type tetracyclic triterpenoids—classified into type I and type II ganoderic acid—and is an important indicator for controlling the quality of G. lucidum. Thus, the study of ganoderic acid synthesis genes can help the breeding and creation of high-quality G. lucidum varieties. The structural genes that have been cloned and reported in the process of ganoderic acid biosynthesis include ACAT [201], HMGS [202], HMGR [60], FPS [203], and MVD [204]. The high and low expression levels of these genes are consistent with the trends of ganoderic acid content, and they are directly involved in the process of ganoderic acid synthesis. Jiang et al. [205] screened cytochrome P450 genes related to ganoderic acid biosynthesis and reported that cyp512v2 played an important role in ganoderic acid T biosynthesis. In addition to the structural genes, regulatory genes in the ganoderic acid biosynthesis pathway directly regulate catalase (Table 5). From Table 5, it can be seen that the regulation of ROS signaling and Ca2+ signaling affects the synthesis of ganoderic acid, and the changes in ROS content caused by silencing AOX, GCN4, and GlSnf1 are consistent with the changes in ganoderic acid content, while the silencing of GlTert causes an increase in the ROS content but a decrease in ganoderic acid content. These specific synthetic regulatory mechanisms need to be further explored.
The biosynthesis of Ganoderma triterpenoids and their regulatory mechanisms have a profound impact on further improving the medicinal value of G. lucidum. The simple genetic operation systems and gene-editing technologies applied to G. lucidum provide a good platform for the synthesis of important active substances. For example, a high-throughput automated screening platform can identify key genes in the triterpenoid biosynthesis pathway of G. lucidum to achieve efficient heterologous biosynthesis [109], increase yield by adjusting the copy number of plasmids [213], and solve the problem of poor fitness of homologous gene expression and low yield by a closely related host [214].

3.5.2. Cordycepin

The structural formula of cordycepin, C10H13N5O3, chemically known as 3′-deoxyadenosine, was first isolated from the fermentation broth of C. militaris by the German scientist Cunningham in 1950 [215]. As an important secondary metabolite in C. militaris, it has anti-tumor [216], neuroprotective [217], antibacterial, and anti-inflammatory effects [218], and alongside other functions, improves the body’s comprehensive resistance to disease [219].
With the completion of whole-genome sequencing and data analysis, the biosynthetic pathway of cordycepin was gradually clarified; it is first converted from the pentose phosphate pathway to AMP and then synthesized through the nucleoside/nucleotide biosynthetic pathway and single gene cluster biosynthetic pathway (Figure 4) [220,221,222]. AMP generates ADP under the action of ADEK and then generates 3′-dADP and 2′-dADP under the action of ribonucleotide reductases. 3′-dADP generates 3′-dAMP under the action of ADEK, followed by cordycepin under the action of NT5E, while 2′-dADP finally generates 2′-deoxyadenosine to participate in nucleotide metabolism. For single gene cluster synthesis, 3′-AMP is the precursor of cordycepin synthesis, which is mainly converted by the nucleotide domain of Cns3-NK and is produced by the phosphorylation of 2′,3′-cAMP, which is subsequently converted via Cns2 and Cns1 redox to cordycepin.
Xia et al. [223] identified four highly homologous protein-coding genes (Cns1, Cns2, Cns3, and Cns4) in C. militaris. Protein function analysis revealed that Cns1 encodes oxidoreductase, Cns2 encodes ion-dependent hydrolase, Cns3 encodes ATP-dependent phosphotransferase, and Cns4 encodes an ABC-type transporter protein. To further determine the role of these functional genes, Cns1Cns4 knockout and heterologous expression in S. cerevisiae showed that Cns1 and Cns2 are key genes for cordycepin biosynthesis, Cns3 is involved in the biosynthesis of pentostatin, and Cns4 may encode the pentostatin transporter. However, pentostatin can prevent cordycepin deamination by inhibiting ADA activity, which is a key factor in regulating cordycepin biosynthesis. SNF1/AMPK are important regulators of fungal development and metabolism [224]. Our team increased the expression level of the cordycepin biosynthesis gene cluster by knocking out SNF1/AMPK in C. militaris, which resulted in more than a seven-fold increase [225]. In the same year, Chen et al. [226] overexpressed two transcription factors, CmTf1 and CmTf2, in C. militaris, which resulted in a three-fold increase in cordycepin content, up to 99 mg/L after liquid fermentation, compared to that in the wild-type. Studies have shown that improving the hypoxic adaptability of the fungus contributes to the synthesis of the active substance. For example, our team overexpressed the predicted bHLH transcription factors of sterol regulatory element binding proteins (SREBPs) in vivo, which increased the cordycepin content more than two-fold, providing a scientific basis for optimizing the cultivation and fermentation gas supply strategy [227].
Synthetic biology has developed rapidly in recent years, with prokaryotic and eukaryotic chassis cells being used to express exogenous genes, realizing the efficient production of a wide range of active substances. During the long-term evolution of C. militaris, through the introduction of efficient cellular-degrading enzymes, this species has acquired the ability to degrade waste bacterial residues, and the genes related to the cordycepin synthesis gene cluster have been knocked out. This has continuously improved the production of pentastatin anti-cancer drugs and finally realized environmentally friendly and sustainable green bio-manufacturing [228]. In addition, the long biosynthetic cycle of natural cordycepin and competition with other intracellular secondary metabolites for substrates are the main factors restricting the industrial production of cordycepin. Therefore, it is necessary to focus on the transcription factors, cofactors, and key enzymes involved in metabolic processes. To continuously expand the scale of cordycepin production, optimizing the metabolic flux of the chassis, reducing the cytotoxicity of the product, and improving the efficiency of extracellular transport are required.

3.5.3. Polysaccharides

The polysaccharides of edible and medicinal fungi are natural macromolecular active substances that have a variety of effects, such as antibacterial, antiviral, anti-cancer, antioxidant, and anti-diabetic properties. For example, the polysaccharide–protein complex of Phellinus igniarius can be used as an anti-inflammatory agent [229], L. edodes polysaccharides can reduce the proliferation of leukemia cells [230], and M. esculenta polysaccharides can regulate the activity of antioxidant enzymes [231]. Due to their good biological activity, polysaccharides from edible and medicinal fungi can be used as food additives, biodegradable films, functional foods, nutritional supplements, and animal feeds after processing. Furthermore, owing to their anti-aging and antioxidant effects, they can be used in functional cosmetics, such as L. edodes polysaccharides, which can be used as a facial cream ingredient with excellent moisture absorption, good water solubility, and viscosity [232].
Functional analysis of polysaccharide synthesis genes in edible and medicinal fungi will help to enhance the content of target products and promote the application of edible fungal polysaccharides in food, cosmetics, and environmental protection. Polysaccharides from edible and medicinal fungi can be divided into intracellular polysaccharides (IPS), extracellular polysaccharides (EPS), and cell wall polysaccharides (CWP). Currently, IPS and EPS are primarily observed in C. cinerea, C. militaris, G. frondosa, G. lucidum, and Hericium erinaceus. Zhou et al. [233] overexpressed phosphoglucomutase (PGM) and UDP-glucose pyrophosphorylase (UGP) genes in C. cinerea singly or with co-overexpression and reported that the overexpression of PGM and UGP increased the EPS content by 30% and 16%, respectively, and the co-overexpression of PGM and UGP increased the EPS content by 75%. These results suggest that compared to a single pathway, multiple-gene co-overexpression increases polysaccharide production compared to single pathways. Similarly, the homologous overexpression of C. militaris PGM showed that the engineered strain produced 78.13% more EPS than the wild-type strain [234]. Cui et al. [235] silenced the glucan synthase gene GFGLS in G. frondosa and reduced EPS production by 0.38 g/L. Subsequently, our team silenced the two glucan synthase genes, GFGLS and GFGLS2, in G. frondosa and reported that the silencing of GFGLS2 synthesized less EPS than that of GFGLS and that their co-silencing had a synergistic downregulation effect that slowed down the production of glucan [236]. Zan et al. [237] constructed an RNA interference vector of the reverse-double promoter system of gfugp in G. frondose, in which the EPS was mainly composed of glucose (98.25%) and a small amount of arabinose (0.86%); silencing gfugp reduced the glucose content by 62.79–66.79%, and it was speculated that the silencing of gfugp affects the synthesis of EPS. Xu et al. [238] overexpressed PGM in G. lucidum, and IPS and EPS increased by 40.5% and 44.3%, respectively, compared to the wild type. Li et al. [239] introduced the Vitreoscilla hemoglobin coding gene VHb into G. lucidum and reported that the coding genes of polysaccharide biosynthesis enzymes, such as PGM, UGP, and GLS, were affected, which increased the IPS and EPS by 30.5% and 88.2%, respectively, compared with the wild type. This could be beneficial for large-scale fermentation by improving the potential application of G. lucidum polysaccharides. Xu et al. [240] co-overexpressed β-1,3-glucan synthase (gls) and UDP-glucose pyrophosphorylase in G. lucidum, and IPS and EPS were 1.31 times and 1.50 times higher than in the wild type, which is helpful for the ultra-production and application of G. lucidum. Our team used E. coli and S. cerevisiae to validate the function and activity of A6180, the proposed gene encoding UDP-Glucose 4-Epimerase (UGE) in the highly active polysaccharides of H. erinaceus, and reported that the recombinant protein converted UDP-α-D-glucose to UDP-α-D-galactose under optimal conditions and enhanced macrophage activity in vitro [241].
Currently, polysaccharides from edible and medicinal fungi are mainly extracted from cultivated and wild-picked mushrooms; however, the production of polysaccharides through genetic or metabolic engineering has certain limitations, including unstable strains, unknown side effects, complex downstream extraction processes, and unclear assembly and polymerization processes of synthetic precursor nucleotide glycosaminoglycan. The polysaccharides of some edible and medicinal fungi, such as G. frondose protein binding β-glucose [242], Coriolus versicolor protein binding polysaccharide PSK [243], and S. commune neutral polysaccharide SPG [244], can be used as anti-tumor, anti-cancer and immunomodulatory drugs [245]. In addition, other bioactive polysaccharides still need to be further studied, and the positive impact of genetic engineering on polysaccharide production of edible and medicinal fungi will greatly promote the potential application of in vitro synthesis-related research in industrial production.

3.5.4. Immunomodulatory Proteins of Edible and Medicinal Fungi

Fungal immunomodulatory proteins (FIPs) are active compounds isolated from edible and medicinal fungi. To date, more than 30 types of FIPs have been identified in edible and medicinal fungi, such as F. filiformis, A. camphorata, M. esculenta, G. lucidum, and H. marmoreus, with coagulation, immune-system-regulation [246], and anti-inflammatory [247], anti-allergic, and anti-tumor effects [248]. With the development of biotechnology, at least 26 recombinant FIPs (rFIPs) related to edible and medicinal fungi have been identified using genome mining, homologous cloning, and protein purification to form a unique protein family [249].
Lin et al. [250] cloned FIP-fve from F. filiformis and expressed it in P. pastoris GS115; the results showed that the purified FIP-fve protein had immunomodulatory and anti-tumor activities in vitro activity assay. Li et al. [251] efficiently expressed the immunomodulatory protein ACA from A. camphorata through E. coli with a yield of up to 187.122 μg/mL, and the purified rACA significantly promoted the proliferation and phagocytic activity of mouse macrophages, which can be used as a food additive and immunomodulator. Wu et al. [252] identified a new FIP-mAb in M. esculenta and successfully expressed this in P. pastoris followed by apoptosis experiments. They showed that rFIP-mco reduced inflammatory cytokines by inhibiting NF-κB signaling, which has broad application prospects in cancer treatment and medical research. Zhou et al. [253] cloned FIP-gap1 and FIP-gap2 from G. lucidum and successfully expressed them in P. pastoris, and the recombinant contents reached 247.4 mg/L and 197.5 mg/L, respectively, and both rFIPs were able to coagulate blood and enhance IL-2 and IFN-γ in vitro bioactivity assay. Our team conducted considerable research in this field. Seven FIPs were identified and their physiological and biochemical characteristics were predicted in V. volvacea, and it is speculated that the new FIP-vvo82 had efficient immunomodulatory activity for inducing spleen lymphocytes [254]; two immunomodulatory proteins, LZ-8 and LZ-9, in G. lucidum, were predicted and analyzed comparatively using bioinformatics and the immunomodulatory activity of LZ-8 was analyzed from the perspective of molecular structure [255], and the amino acids closely related to high activity were observed for subsequent research [256]; two immunomodulatory proteins, FIP-lti1 and FIP-lti2, derived from L. tigrinus, were identified, and their expression and purification revealed that they could protect the liver from Concanavalin A-induced hepatic oxidative damage through the Nrf2/NF-κB/NLRP3/IL-1β pathway [257]; and a novel FIP-hma was observed in H. marmoreus and its expression in E. coli resulted in the upregulation of iNOS, IL-264, IL-7β, and TNF-α levels by rFIP-hma in macrophages, which is hypothesized to activate immune responses by regulating central cytokines and provides new ideas for the study of the physiological functions of FIPs and their applications in the field of medicine [258].
Recombinantly expressed FIPs obtained by genetic engineering have similar structures and biological functions to those obtained by natural isolation and purification and have better heat and acid resistance and dehydration stability, which can potentially be exploited as anti-cancer drugs. However, further research is required on the mechanisms and reasons for the differences in the activities of FIPs to improve their pharmacological activities and optimize recombinant expression conditions to obtain the highest yields, which will continue to support application in the fields of food and medicine.

4. Conclusions and Prospects

Edible and medicinal fungi can synthesize a variety of natural products, such as antibiotics, pigments, enzyme inhibitors, and hormones, and are widely used in medicine, the chemical industry, agriculture, and basic biological research. However, their complex genetic backgrounds hinder further development and utilization. In recent years, researchers have continuously strengthened and developed efficient chassis cells and multi-omics technology platforms, constructed efficient genetic expression platforms and developed bidirectional selection marker systems. With the combination of high-precision gene-editing technology, remarkable achievements have been made in the study of the mating type, fruiting body development, substrate utilization, nutrient transport, environmental responses, and important active substance synthesis regulatory genes in edible and medicinal fungi. However, scientific problems still urgently need to be resolved.
First, regarding functional analysis technologies for edible and medicinal fungi, the lack of high-quality reference genomes is an important reason for the slow pace of research. Therefore, it is necessary to strengthen standardized, versatile, and accurate genotype and phenotype identification platforms and bioinformatics analysis platforms, conduct in-depth pan-genome sequencing and analysis, continuously explore new genes, and find excellent alleles. In this process, due to the spread of edible and medicinal fungi all over the world, the corresponding strain and species names have not been unified. We hope that the unification of database nomenclature can be strengthened in the future.
Second, the structure and genes of mating-type loci in edible and medicinal fungi are polymorphic, and the collinearity of mating-type loci in different species varies greatly; therefore, the evolutionary process needs to be further studied. Further studies on photoreceptor proteins, signaling pathways, fruiting-body-induced temperature sensor systems, and environmental response mechanisms are needed. Through the further analysis of functional gene regulatory networks and related gene clusters, omics technology can be combined with metabolic network research to promote further developments in bioengineering [259].
Finally, with the continuous development of synthetic biology, the application of functional gene research in various fields should be strengthened. Future studies should reveal whether SMS can be transformed into a pure mycelic material, which can then be used to produce materials with both compressive strength and flexibility for the packaging and construction industries. In addition, mating-type genes can be used for precise and efficient breeding, and stress resistance genes can be used to improve fruiting body yield, thus moving towards an era of precision molecular breeding. Established genetic transformation and gene-editing technologies can be used to study the mechanisms of degeneration, and high-quality engineered strains can be constructed by crossing genes containing opposite mating types. Furthermore, edible and medicinal fungi have great medicinal potential and are valuable to human health. In the future, the development of the edible and medicinal fungi industry will be upgraded to the field of intensive processing and health, such as the expression of key genes in the active substance synthesis pathways of uncultivable edible and medicinal bacteria in heterologous hosts and their application to large-scale production in cell factories.

Author Contributions

Conceptualization, G.Z., D.B. and Y.W.; writing—original draft preparation, W.L.; visualization, G.Z.; supervision, D.B.; project administration, Y.W.; funding acquisition, Y.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Shanghai Agriculture Applied Technology Development Program, China (Grant No. X2022-02-08-00-12-F01137) and the Excellent Team Plan of Shanghai Academy of Agricultural Sciences (2022)014.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cheung, P.C.K. The nutritional and health benefits of mushrooms. Nutr. Bull. 2010, 35, 292–299. [Google Scholar] [CrossRef]
  2. Das, S.; Prakash, B. Chapter 11—Edible mushrooms: Nutritional composition and medicinal benefits for improvement in quality life. In Research and Technological Advances in Food Science; Academic Press: Cambridge, MA, USA, 2022; pp. 269–300. [Google Scholar]
  3. Mingyi, Y.; Belwal, T.; Devkota, H.P.; Li, L.; Luo, Z.S. Trends of utilizing mushroom polysaccharides (MPs) as potent nutraceutical components in food and medicine: A comprehensive review. Trends Food Sci. Technol. 2019, 92, 94–110. [Google Scholar] [CrossRef]
  4. Wong, J.H.; Ng, T.B.; Cheung, R.C.; Ye, X.J.; Wang, H.X.; Lam, S.K.; Lin, P.; Chan, Y.S.; Fang, E.F.; Ngai, P.H.; et al. Proteins with antifungal properties and other medicinal applications from plants and mushrooms. Appl. Microbiol. Biotechnol. 2010, 87, 1221–1235. [Google Scholar] [CrossRef] [PubMed]
  5. Yin, Z.H.; Sun-Waterhouse, D.; Wang, J.M.; Ma, C.Y.; Waterhouse, G.I.N.; Kang, W.Y. Polysaccharides from edible fungi Pleurotus spp.: Advances and perspectives. J. Future Foods 2021, 1, 128–140. [Google Scholar] [CrossRef]
  6. Zhang, Y.R.; Wang, D.W.; Chen, Y.T.; Liu, T.T.; Zhang, S.S.; Fan, H.X.; Liu, H.C.; Li, Y. Healthy function and high valued utilization of edible fungi. Food Sci. Hum. Well 2021, 10, 408–420. [Google Scholar] [CrossRef]
  7. Zhao, J.H.; He, R.J.; Zhong, H.; Liu, S.Z.; Liu, X.F.; Hussain, M.; Sun, P.L. A cold-water extracted polysaccharide-protein complex from Grifola frondosa exhibited anti-tumor activity via TLR4-NF-κB signaling activation and gut microbiota modification in H22 tumor-bearing mice. Int. J. Biol. Macromol. 2023, 239, 124291. [Google Scholar] [CrossRef] [PubMed]
  8. Wasser, S.P. Current findings, future trends, and unsolved problems in studies of medicinal mushrooms. Appl. Microbiol. Biotechnol. 2011, 89, 1323–1332. [Google Scholar] [CrossRef] [PubMed]
  9. Li, H.; Tian, Y.; Menolli, N., Jr.; Ye, L.; Karunarathna, S.C.; Perez-Moreno, J.; Rahman, M.M.; Rashid, M.H.; Phengsintham, P.; Rizal, L.; et al. Reviewing the world’s edible mushroom species: A new evidence-based classification system. Compr. Rev. Food Sci. Food Saf. 2021, 20, 1982–2014. [Google Scholar] [CrossRef]
  10. Royse, D.J.; Baars, J.; Tan, Q. Current Overview of Mushroom Production in the World: Technology and Applications. In Edible and Medicinal Mushrooms; John Wiley and Sons Ltd.: Hoboken, NJ, USA, 2017; pp. 5–13. [Google Scholar]
  11. Grimm, D.; Wösten, H.A.B. Mushroom cultivation in the circular economy. Appl. Microbiol. Biotechnol. 2018, 102, 7795–7803. [Google Scholar] [CrossRef]
  12. Zou, G.; Nielsen, J.B.; Wei, Y.J. Harnessing synthetic biology for mushroom farming. Trends Biotechnol. 2023, 41, 480–483. [Google Scholar] [CrossRef]
  13. Ye, D.; Hu, Q.X.; Bai, X.; Zhang, W.J.; Guo, H.W. Increasing the value of Phragmites australis straw in a sustainable integrated agriculture model (SIAM) comprising edible mushroom cultivation and spent mushroom substrate compost. Sci. Total Environ. 2023, 869, 161807. [Google Scholar] [CrossRef]
  14. Bao, D.P. Development and prospect of genetics of edible mushrooms in China. Mycosystema 2021, 40, 806–821. [Google Scholar]
  15. Zhao, Z.M.; Fan, D.Y.; Yao, L.; Li, C.T.; Li, Y. Sequencing and Analysis of the Whole Genome of Morchella importuna MT1. J. Fungal Res. 2021, 19, 271–276. [Google Scholar]
  16. Drott, M.T.; Park, S.C.; Wang, Y.W.; Harrow, L.; Keller, N.P.; Pringle, A. Pangenomics of the death cap mushroom Amanita phalloides, and of Agaricales, reveals dynamic evolution of toxin genes in an invasive range. ISME J. 2023, 17, 1236–1246. [Google Scholar] [CrossRef] [PubMed]
  17. Guo, Y.X.; Liu, Z.H.; Fu, Y.P.; Li, Y.; Dai, Y.T.; Xiao, S.J. Pan-Genomes Provide Insights into the Genetic Basis of Auricularia heimuer Domestication. J. Fungi 2022, 8, 581. [Google Scholar] [CrossRef] [PubMed]
  18. Kim, J.Y.; Kim, D.Y.; Park, Y.J.; Jang, M.J. Transcriptome analysis of the edible mushroom Lentinula edodes in response to blue light. PLoS ONE 2020, 15, e0230680. [Google Scholar] [CrossRef] [PubMed]
  19. Hao, H.B.; Zhang, J.J.; Wang, Q.; Huang, J.C.; Juan, J.X.; Kuai, B.K.; Feng, Z.Y.; Chen, H. Transcriptome and Differentially Expressed Gene Profiles in Mycelium, Primordium and Fruiting Body Development in Stropharia rugosoannulata. Genes 2022, 13, 1080. [Google Scholar] [CrossRef] [PubMed]
  20. Shen, N.; Zhang, J.C.; Wang, C.C.; Bian, Y.B.; Xiao, Y. Studies on Transcriptome During Fruiting Body Development of Lentinula edodes. Acta Hortic. Sin. 2022, 49, 801–815. [Google Scholar]
  21. Cao, L.P.; Zhang, Q.; Miao, R.Y.; Lin, J.B.; Feng, R.C.; Ni, Y.Q.; Li, W.S.; Yang, D.L.; Zhao, X. Application of omics technology in the research on edible fungi. Curr. Res. Food Sci. 2023, 6, 100430. [Google Scholar] [CrossRef]
  22. Jakopovic, B.; Oršolić, N.; Jakopovich, I. Proteomic Research on the Antitumor Properties of Medicinal Mushrooms. Molecules 2021, 26, 6708. [Google Scholar] [CrossRef]
  23. Lin, T.Y.; Hua, W.J.; Yeh, H.; Tseng, A.J. Functional proteomic analysis reveals that fungal immunomodulatory protein reduced expressions of heat shock proteins correlates to apoptosis in lung cancer cells. Phytomed. Int. J. Phytother. Phytopharm. 2021, 80, 153384. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, L.P.; Su, Y.; Ma, Z.H.; Guo, L.Z.; Yang, S.; Yu, H. Comparative proteomic analysis reveals differential protein expression of Hypsizygus marmoreus in response to different light qualities. Int. J. Biol. Macromol. 2022, 223, 1320–1334. [Google Scholar] [CrossRef] [PubMed]
  25. Qu, H.; Zhou, H.B.; Ma, T.; Zheng, Z.H.; Zheng, E.P.; Yang, H.L.; Gao, H.Y. TMT-based quantitative proteomic analysis of postharvest Coprinus comatus fruiting body during storage. Postharvest Biol. Technol. 2022, 185, 111786. [Google Scholar] [CrossRef]
  26. Jo, C.; Zhang, J.; Tam, J.M.; Church, G.M.; Khalil, A.S.; Segrè, D.; Tang, T.C. Unlocking the magic in mycelium: Using synthetic biology to optimize filamentous fungi for biomanufacturing and sustainability. Mater. Today Bio 2023, 19, 100560. [Google Scholar] [CrossRef] [PubMed]
  27. Jiang, A.L.; Liu, Y.N.; Liu, R.; Ren, A.; Ma, H.Y.; Shu, L.B.; Shi, L.; Zhu, J.; Zhao, M.W. Integrated Proteomics and Metabolomics Analysis Provides Insights into Ganoderic Acid Biosynthesis in Response to Methyl Jasmonate in Ganoderma Lucidum. Int. J. Mol. Sci. 2019, 20, 6116. [Google Scholar] [CrossRef]
  28. Liu, Y.C.; Zhang, D.; Ning, Q.y.; Wang, J.H. Growth characteristics and metabonomics analysis of Lactobacillus rhamnosus GG in Ganoderma lucidum aqueous extract medium. Food Biosci. 2023, 53, 102486. [Google Scholar] [CrossRef]
  29. Zeng, Z.; Wang, J.Q.; Wen, X.F.; Wang, Y.; Li, X.; Liu, D.Y.; Geng, F. Metabolomic analysis provides insights into the mechanism of color and taste changes in Dictyophora indusiata fruiting bodies under different drying processes. Food Res. Int. 2022, 162, 112090. [Google Scholar] [CrossRef]
  30. Sun, X.Y.; Wu, J.Y.; Zhang, S.H.; Luo, L.; Mo, C.Y.; Sheng, L.; Ma, A.M. Genome and Comparative Transcriptome Dissection Provide Insights into Molecular Mechanisms of Sclerotium Formation in Culinary-Medicinal Mushroom Pleurotus tuber-regium. Front. Microbiol. 2021, 12, 815954. [Google Scholar] [CrossRef]
  31. Cai, Z.X.; Chen, M.Y.; Lu, Y.P.; Guo, Z.J.; Zeng, Z.H.; Liao, J.H.; Zeng, H. Metabolomics and transcriptomics unravel the mechanism of browning resistance in Agaricus bisporus. PLoS ONE 2022, 17, e0255765. [Google Scholar] [CrossRef]
  32. Marian, I.M.; Vonk, P.J.; Valdes, I.D.; Barry, K.; Bostock, B.; Carver, A.; Daum, C.; Lerner, H.; Lipzen, A.; Park, H.; et al. The Transcription Factor Roc1 Is a Key Regulator of Cellulose Degradation in the Wood-Decaying Mushroom Schizophyllum commune. mBio 2022, 13, e0062822. [Google Scholar] [CrossRef]
  33. Liu, C.C.; Kang, L.Q.; Lin, M.; Bi, J.J.; Liu, Z.H.; Yuan, S. Molecular Mechanism by Which the GATA Transcription Factor CcNsdD2 Regulates the Developmental Fate of Coprinopsis cinerea under Dark or Light Conditions. mBio 2021, 13, e0362621. [Google Scholar] [CrossRef]
  34. Gong, M.; Wang, Y.; Su, E.; Zhang, J.G.; Tang, L.H.; Li, Z.P.; Zhang, L.L.; Zou, G.; Wan, J.N.; Bao, D.P. The promising application of a β-glucosidase inhibitor in the postharvest management of Volvariella volvacea. Postharvest Biol. Technol. 2022, 185, 111784. [Google Scholar] [CrossRef]
  35. Wijayawardene, N.N.; Boonyuen, N.; Ranaweera, C.B.; de Zoysa, H.K.S.; Padmathilake, R.E.; Nifla, F.; Dai, D.Q.; Liu, Y.; Suwannarach, N.; Kumla, J.; et al. OMICS and Other Advanced Technologies in Mycological Applications. J. Fungi 2023, 9, 688. [Google Scholar] [CrossRef]
  36. Shangguan, J.L.; Zhu, J.; Shi, L.; Ren, A.; ZHao, M.W. Research on Edible Fungi Genetic System. J. Fungal Res. 2019, 17, 224–228. [Google Scholar]
  37. Li, J.G.; Liu, Q.; Liu, D.F.; Wu, M.; Tian, C.G. Advances in metabolic engineering of filamentous fungi. Chin. J. Biotechnol. 2021, 37, 1637–1658. [Google Scholar]
  38. Bao, D.P.; Huang, Z.H.; Li, Y.; Zhou, C.L.; Li, Y.; Wan, J.N.; Tang, L.H.; Mao, W.J.; Wang, Y.; Gong, M.; et al. Agrobacterium-mediated transformation of arthroconidia obtained from the edible mushroom Hypsizygus marmoreus. J. Microbiol. Method 2020, 171, 105878. [Google Scholar] [CrossRef]
  39. Liu, L.; Xiao, Z.Y.; Guo, L.Q.; Lin, J.F.; You, L.F.; Liao, J.W. Establishment of genetic transformation system of Hericium erinaceus using PEG mediated method. Mycosystema 2014, 33, 121–128. [Google Scholar]
  40. 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]
  41. Xu, C. The Construction of Liposome-Mediated Transformation Method and the Role of Laccases in the Development of Flammulina velutipes. Master’s Thesis, Nanjing Agricultural University, Nanjing, China, 2016. [Google Scholar]
  42. Zhang, Y.; Zhang, C.H.; Liu, D.R.; Wang, X.F.; Wang, J.M.; Qiu, L.Y. Liposome-mediated Fruit Body Gill Tissue Transformation of Agaricus bisporus and Function Exploration of 1-aminocyclopropane-1-carboxylic Acid Oxidase Gene (ACO). J. Agric. Biotechnol. 2015, 23, 1112–1120. [Google Scholar]
  43. Kim, J.K.; Park, Y.J.; Kong, W.S.; Kang, H.W. Highly Efficient Electroporation-mediated Transformation into Edible Mushroom Flammulina velutipes. Mycobiology 2010, 38, 331–335. [Google Scholar] [CrossRef]
  44. Shi, L.L.; Peer, A.V.; Guo, L.X.; Chen, R.L.; Wang, W.; Yan, J.J.; Deng, Y.J.; Xie, B.G. Agrobacterium-mediated Transformation of an Endogenous HMG-box Transcription Factor fvhom1 in Flammulina velutipes. Genom. Appl. Biol. 2014, 33, 1268–1274. [Google Scholar]
  45. Lyu, X.M.; Wang, Q.J.; Liu, A.; Liu, F.; Meng, L.; Wang, P.M.; Zhang, Y.; Wang, L.; Li, Z.; Wang, W. The transcription factor Ste12-like increases the mycelial abiotic stress tolerance and regulates the fruiting body development of Flammulina filiformis. Front. Microbiol. 2023, 14, 1139679. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, H.; Fan, T.N.; Bai, Y.D.; Guo, Q.Q.; Wang, H.A.; Chen, G.; Gao, Y.X.; Lin, J.W. Native bio-function of FIP-fve towards Flammulina filiformis. J. Huazhong Agric. Univ. 2023, 42, 131–138. [Google Scholar]
  47. Zhang, X.; Xia, X.; Duan, Z.W.; Guan, S.Y.; Li, H.G.; Chen, L.J.; Zhang, L.; Fan, W.L.; Lin, J.W. Construction of Silencing Vector for FIP-fve Gene and Genetic Transformation of Enoki Mushroom (Flammulina velutipes). Mol. Plant Breed. 2017, 15, 4491–4497. [Google Scholar]
  48. Meng, L.; Lyu, X.M.; Shi, L.L.; Wang, Q.J.; Wang, L.; Zhu, M.J.; Mukhtar, I.; Xie, B.G.; Wang, W. The transcription factor FvHmg1 negatively regulates fruiting body development in Winter Mushroom Flammulina velutipes. Gene 2021, 785, 145618. [Google Scholar] [CrossRef] [PubMed]
  49. Sekiya, S.; Yamada, M.; Shibata, K.; Okuhara, T.; Yoshida, M.; Inatomi, S.; Taguchi, G.; Shimosaka, M. Characterization of a gene coding for a putative adenosine deaminase-related growth factor by RNA interference in the Basidiomycete Flammulina velutipes. J. Biosci. Bioeng. 2013, 115, 360–365. [Google Scholar] [CrossRef] [PubMed]
  50. Du, M.Y.; Xie, Y.B.; Wang, M.; Yang, H.; Hu, B.H.; Mukhtar, I.; Liu, Y.Y.; Tao, Y.X.; Liu, F.; Xie, B.G. FFGA1 Protein Is Essential for Regulating Vegetative Growth, Cell Wall Integrity, and Protection against Stress in Flammunina filiformis. J. Fungi 2022, 8, 401. [Google Scholar] [CrossRef] [PubMed]
  51. Ling, Y.Y.; Ling, Z.L.; Zhao, R.L. Construction of a heat-resistant strain of Lentinus edodes by fungal Hsp20 protein overexpression and genetic transformation. Front. Microbiol. 2022, 13, 1009885. [Google Scholar] [CrossRef] [PubMed]
  52. Gong, W.B.; Liu, K.F.; Li, X.R.; Zhang, L.; Shen, N.; Bian, Y.B.; Xiao, Y. QTL mapping reveals mating type gene LeHD1 regulating mycelial growth in shiitake mushroom, Lentinula edodes. Sci. Hortic. 2022, 305, 111417. [Google Scholar] [CrossRef]
  53. Hu, Y.; Cai, Y.L.; Yu, J.J.; Zhou, Y.; Li, J.P.; Bian, Y.B.; Gong, Y.H. Function analysis of HMG-box transcription factor lelcrp1 in Lentinula edodes. Acta Microbiol. Sin. 2018, 58, 2100–2109. [Google Scholar]
  54. Wang, G.Z.; Zhou, S.S.; Luo, Y.; Ma, C.J.; Gong, Y.H.; Zhou, Y.; Gao, S.S.; Huang, Z.C.; Yan, L.L.; Hu, Y.; et al. The heat shock protein 40 LeDnaJ regulates stress resistance and indole-3-acetic acid biosynthesis in Lentinula edodes. Fungal Genet. Biol. 2018, 118, 37–44. [Google Scholar] [CrossRef] [PubMed]
  55. Ma, C.J.; Wang, G.Z.; Zhou, S.S.; Luo, Y.; Gong, Y.H.; Bian, Y.B. Functional analyses of anthranilate synthase gene LetrpE in Lentinula edodes by RNAi mediated gene knockdown. Mycosystema 2018, 37, 576–583. [Google Scholar]
  56. Zhong, H.Y.; Luo, Y.; Gong, Y.H.; Xu, R.P.; Bian, Y.B. Functional Analysis of Indole-3-pyruvate Monooxygenase Gene YUCCA8 Involved in Heat Resistance of Lentinula edodes by RNAi. Acta Edulis Fungi 2021, 28, 11–18. [Google Scholar]
  57. Wang, C.; Luo, Y.; Wang, G.Z.; Xu, R.P.; Zhou, Y.; Gong, Y.H.; Bian, Y.B. Functional Analyses of Tryptophan Synthase Gene LetrpB in Lentinula edodes by RNAi Method. Acta Edulis Fungi 2019, 26, 161. [Google Scholar]
  58. Liu, Y.N.; Zhang, T.J.; Lu, X.X.; Ma, B.L.; Ren, A.; Shi, L.; Jiang, A.L.; Yu, H.S.; Zhao, M.W. Membrane fluidity is involved in the regulation of heat stress induced secondary metabolism in Ganoderma lucidum. Environ. Microbiol. 2017, 19, 1653–1668. [Google Scholar] [CrossRef] [PubMed]
  59. Fang, X.; Shi, L.; Ren, A.; Jiang, A.-L.; Wu, F.-L.; Zhao, M.-W. The cloning, characterization and functional analysis of a gene encoding an acetyl-CoA acetyltransferase involved in triterpene biosynthesis in Ganoderma lucidum. Mycoscience 2013, 54, 100–105. [Google Scholar] [CrossRef]
  60. Xu, J.W.; Xu, Y.N.; Zhong, J.J. Enhancement of ganoderic acid accumulation by overexpression of an N-terminally truncated 3-hydroxy-3-methylglutaryl coenzyme A reductase gene in the basidiomycete Ganoderma lucidum. Appl. Environ. Microbiol. 2012, 78, 7968–7976. [Google Scholar] [CrossRef]
  61. Lin, Y.L.; Lee, Y.R.; Tsao, N.W.; Wang, S.Y.; Shaw, J.F.; Chu, F.H. Characterization of the 2,3-Oxidosqualene Cyclase Gene from Antrodia cinnamomea and Enhancement of Cytotoxic Triterpenoid Compound Production. J. Nat. Prod. 2015, 78, 1556–1562. [Google Scholar] [CrossRef]
  62. Qi, Y.C.; Chen, H.J.; Zhang, M.K.; Wen, Q.; Qiu, L.Y.; Shen, J.W. Identification and expression analysis of Pofst3 suggests a role during Pleurotus ostreatus primordia formation. Fungal Biol. 2019, 123, 200–208. [Google Scholar] [CrossRef]
  63. Lou, H.W.; Lin, J.F.; Guo, L.Q.; Wang, X.W.; Tian, S.Q.; Liu, C.X.; Zhao, Y.; Zhao, R.Y. Advances in research on Cordyceps militaris degeneration. Appl. Microbiol. Biotechnol. 2019, 103, 7835–7841. [Google Scholar] [CrossRef]
  64. Jiang, N.; Song, C.Y.; Liu, J.Y.; Tan, Q.; Zhang, L.J.; Shang, X.D. Agrobacterium-mediated transformation of the Lentinula edodes mycelia. Mycosystema 2017, 36, 1514–1523. [Google Scholar]
  65. Lv, S.Y.; Chen, X.; Mou, C.Y.; Dai, S.H.; Bian, Y.B.; Kang, H. Agrobacterium-mediated transformation of the ascomycete mushroom Morchella importuna using polyubiquitin and glyceraldehyde-3-phosphate dehydrogenase promoter-based binary vectors. World J. Microb. Biot. 2018, 34, 148. [Google Scholar] [CrossRef]
  66. Du, S.S.; Bao, D.P.; Ma, L.J.; Li, X.L.; Shang, J.J. Expression analyses of exogenous genes controlled by different promoters in Hypsizygus marmoreus. Mycosystema 2021, 40, 2065–2073. [Google Scholar]
  67. Shang, J.J.; Yang, R.H.; Tang, L.H.; Li, Y.; Li, Y.; Mao, W.J.; Gong, M.; Wang, Y.; Honda, Y.C.; Bao, D.P. Differential expression of two gpd genes in the cultivated mushroom Pleurotus eryngii using RNA sequencing analysis. Mycoscience 2019, 60, 351–353. [Google Scholar] [CrossRef]
  68. Liu, J.Y.; Song, C.Y.; Li, Q.Z.; Xu, Z.; Zhang, D.; Zhang, M.Y.; Tan, Q.; Shang, X.D. A colonized millet grain method for Agrobacterium-mediated transformation of the button mushroom Agaricus bisporus. J. Microbiol. Method 2018, 152, 148–153. [Google Scholar] [CrossRef]
  69. Liu, J.Y.; Xu, Z.; Zhang, D.; Wang, R.J.; Tan, Q.; Shang, X.D. Efficiency of Flammulina velutipes Transformation by Agrobacterium tumefaciens-mediated Transformation Using Different Receptors. Acta Edulis Fungi 2015, 22, 7–12. [Google Scholar]
  70. Lei, M.; Wu, X.L.; Zhang, J.X.; Wang, H.X.; Huang, C.Y. Establishment of an efficient transformation system for Pleurotus ostreatus. World J. Microbiol. Biotechnol. 2017, 33, 214. [Google Scholar] [CrossRef]
  71. Yan, L.L.; Xu, R.P.; Zhou, Y.; Gong, Y.H.; Dai, S.H.; Liu, H.Y.; Bian, Y.B. Effects of Medium Composition and Genetic Background on Agrobacterium-Mediated Transformation Efficiency of Lentinula edodes. Genes 2019, 10, 467. [Google Scholar] [CrossRef]
  72. Herzog, R.; Solovyeva, I.; Bölker, M.; Lugones, L.G.; Hennicke, F. Exploring molecular tools for transformation and gene expression in the cultivated edible mushroom Agrocybe aegerita. Mol. Genet. Genom. 2019, 294, 663–677. [Google Scholar] [CrossRef]
  73. Wang, C.C.; Zhang, W.W.; Wong, J.H.; Ng, T.; Ye, X.J. Diversity of potentially exploitable pharmacological activities of the highly prized edible medicinal fungus Antrodia camphorata. Appl. Microbiol. Biotechnol. 2019, 103, 7843–7867. [Google Scholar] [CrossRef]
  74. Liu, X.F.; Xia, Y.J.; Zhang, Y.; Liang, L.H.; Xiong, Z.Q.; Wang, G.Q.; Song, X.; Ai, L.Z. Enhancement of antroquinonol production via the overexpression of 4-hydroxybenzoate polyprenyltransferase biosynthesis-related genes in Antrodia cinnamomea. Phytochemistry 2021, 184, 112677. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, Q.; Zhao, L.T.; Shen, M.Y.; Liu, J.Y.; Li, Y.R.; Xu, S.; Chen, L.; Shi, G.Y.; Ding, Z.Y. Establishment of an Efficient Polyethylene Glycol (PEG)-Mediated Transformation System in Pleurotus eryngii var. ferulae Using Comprehensive Optimization and Multiple Endogenous Promoters. J. Fungi 2022, 8, 186. [Google Scholar] [CrossRef] [PubMed]
  76. Shang, J.J.; Li, Y.; Yang, R.H.; Wang, Y.; Mao, W.J.; Tang, L.H.; Wu, Y.Y.; Nakazawa, T.; Honda, Y.C.; Li, Y.; et al. Efficient transformation of Pleurotus eryngii with a safe selective marker mutated from the Pesdi1 gene. J. Microbiol. Methods 2018, 152, 7–9. [Google Scholar] [CrossRef] [PubMed]
  77. Zha, L.Y.; Zhou, W.J.; Wang, Y.; Zhang, Z.W.; Wen, H.S.; Mo, M.H. Research Progress on Agrobacterium mediated Transformation of Edible Fungi. Acta Edulis Fungi 2020, 27, 105–118. [Google Scholar]
  78. Shen, H.M.; Li, C.; Gao, L.; Liu, T.G.; Liu, B.; Chen, W.Q. Research progress in transformation of fungi mediated by protoplasts. Plant Prot. 2017, 43, 25–28. [Google Scholar]
  79. Wang, P.A.; Xiao, H.; Zhong, J.J. CRISPR-Cas9 assisted functional gene editing in the mushroom Ganoderma lucidum. Appl. Microbiol. Biotechnol. 2020, 104, 1661–1671. [Google Scholar] [CrossRef] [PubMed]
  80. Waltz, E. Gene-edited CRISPR mushroom escapes US regulation. Nature 2016, 532, 293. [Google Scholar] [CrossRef] [PubMed]
  81. Qin, H.; Xiao, H.; Zou, G.; Zhou, Z.; Zhong, J.J. CRISPR-Cas9 assisted gene disruption in the higher fungus Ganoderma species. Process Biochem. 2017, 56, 57–61. [Google Scholar] [CrossRef]
  82. Liu, K.; Sun, B.; You, H.; Tu, J.L.; Yu, X.Y.; Zhao, P.; Xu, J.W. Dual sgRNA-directed gene deletion in basidiomycete Ganoderma lucidum using the CRISPR/Cas9 system. Microb. Biotechnol. 2020, 13, 386–396. [Google Scholar] [CrossRef]
  83. Zhang, Z.G.; Zhang, J.S.; Zou, G.; Tang, C.H.; Feng, J.; Bao, D.P.; Chen, J.B.; Tan, Y. Construction of a CRISPR/Cas9-Based Genome Editing System in Ganoderma lucidum “Hunong No.1’Cultivar. Acta Edulis Fungi 2023, 30, 9–18. [Google Scholar]
  84. Sugano, S.S.; Suzuki, H.; Shimokita, E.; Chiba, H.; Noji, S.; Osakabe, Y.; Osakabe, K. Genome editing in the mushroom-forming basidiomycete Coprinopsis cinerea, optimized by a high-throughput transformation system. Sci. Rep. 2017, 7, 1260. [Google Scholar] [CrossRef] [PubMed]
  85. Yu, H. Research on the application of CRISPR-Cas9 gene editing system in Coprinus cinereus. Master’s Thesis, Nanjing Normal University, Nanjing, China, 2018. [Google Scholar]
  86. Chen, B.X.; Wei, T.; Ye, Z.W.; Yun, F.; Kang, L.Z.; Tang, H.B.; Guo, L.i.Q.; Lin, J.F. Efficient CRISPR-Cas9 Gene Disruption System in Edible-Medicinal Mushroom Cordyceps militaris. Front. Microbiol. 2018, 9, 1157. [Google Scholar] [CrossRef] [PubMed]
  87. Meng, G.L.; Wang, X.P.; Liu, M.Q.; Wang, F.; Liu, Q.Z.; Dong, C.H. Efficient CRISPR/Cas9 system based on autonomously replicating plasmid with an AMA1 sequence and precisely targeted gene deletion in the edible fungus, Cordyceps militaris. Microb. Biotechnol. 2022, 15, 2594–2606. [Google Scholar] [CrossRef]
  88. Yu, X.; Liang, X.Q.; Xie, H.M.; Kumar, S.; Ravinder, N.; Potter, J.; de Mollerat du Jeu, X.; Chesnut, J.D. Improved delivery of Cas9 protein/gRNA complexes using lipofectamine CRISPRMAX. Biotechnol. Lett. 2016, 38, 919–929. [Google Scholar] [CrossRef]
  89. Al Abdallah, Q.; Ge, W.; Fortwendel, J.R. A Simple and Universal System for Gene Manipulation in Aspergillus fumigatus: In Vitro-Assembled Cas9-Guide RNA Ribonucleoproteins Coupled with Microhomology Repair Templates. mSphere 2017, 2, e00446-00417. [Google Scholar] [CrossRef] [PubMed]
  90. Jan Vonk, P.; Escobar, N.; Wösten, H.A.B.; Lugones, L.G.; Ohm, R.A. High-throughput targeted gene deletion in the model mushroom Schizophyllum commune using pre-assembled Cas9 ribonucleoproteins. Sci. Rep. 2019, 9, 7632. [Google Scholar] [CrossRef] [PubMed]
  91. Zou, G.; Xiao, M.L.; Chai, S.X.; Zhu, Z.H.; Wang, Y.; Zhou, Z.H. Efficient genome editing in filamentous fungi via an improved CRISPR-Cas9 ribonucleoprotein method facilitated by chemical reagents. Microb. Biotechnol. 2021, 14, 2343–2355. [Google Scholar] [CrossRef]
  92. Yu, L.; Xiao, M.L.; Zhu, Z.H.; Wang, Y.M.; Zhou, Z.H.; Wang, P.P.; Zou, G. Efficient genome editing in Claviceps purpurea using a CRISPR/Cas9 ribonucleoprotein method. Synth. Syst. Biotechnol. 2022, 7, 664–670. [Google Scholar] [CrossRef]
  93. Liu, J.Y.; Cui, H.Y.; Wang, R.J.; Xu, Z.; Yu, H.L.; Song, C.Y.; Lu, H.; Li, Q.Z.; Xing, D.R.; Tan, Q.; et al. A Simple and Efficient CRISPR/Cas9 System Using a Ribonucleoprotein Method for Flammulina filiformis. J. Fungi 2022, 8, 1000. [Google Scholar] [CrossRef]
  94. Boontawon, T.; Nakazawa, T.; Choi, Y.J.; Ro, H.S.; Oh, M.J.; Kawauchi, M.; Sakamoto, M.; Honda, Y.C. Double-gene targeting with preassembled Cas9 ribonucleoprotein for safe genome editing in the edible mushroom Pleurotus ostreatus. FEMS Microbiol. Lett. 2023, 370, 370. [Google Scholar] [CrossRef]
  95. Xu, R.P.; Zhou, Y.; Bian, Y.B. Application of CRISPR/Cas9 Technology in Macrofungi Research. Acta Edulis Fungi 2019, 26, 119–127. [Google Scholar]
  96. Zhang, Y.; Chen, S.T.; Yang, L.; Zhang, Q. Application progress of CRISPR/Cas9 genome-editing technology in edible fungi. Front. Microbiol. 2023, 14, 1169884. [Google Scholar] [CrossRef] [PubMed]
  97. Ding, Q.; Ye, C. Microbial cell factories based on filamentous bacteria, yeasts, and fungi. Microb. Cell Fact. 2023, 22, 20. [Google Scholar] [CrossRef] [PubMed]
  98. Yang, Y.F.; Geng, B.N.; Song, H.Y.; Hu, M.M.; He, Q.N.; Chen, S.W.; Bai, F.W.; Yang, S.H. Progress and perspective on development of non-model industrial bacteria as chassis cells for biochemical productionin the synthetic biology era. Chin. J. Biotechnol. 2021, 37, 874–910. [Google Scholar]
  99. Adams, A.M.; Kaplan, N.A.; Wei, Z.; Brinton, J.D.; Monnier, C.S.; Enacopol, A.L.; Ramelot, T.A.; Jones, J.A. In vivo production of psilocybin in E. coli. Metab. Eng. 2019, 56, 111–119. [Google Scholar] [CrossRef] [PubMed]
  100. Li, Y.; Zhao, L.T.; Gu, Z.H.; Li, Y.R.; Shi, G.Y.; Ding, Z.Y. Heterologous expression and characterization of the key enzymes involved in sugar donor synthesis of polysaccharide in Ganoderma lucidum. Microbiol. China 2019, 46, 3233–3247. [Google Scholar]
  101. Liang, Y.Y.; Zan, X.Y.; Sun, L.; Fu, X.; Cui, F.J.; Tan, M.; Shao, Z.Y.; Sun, W.J. A uridine diphosphate-glycosyltransferase GFUGT88A1 derived from edible mushroom Grifola frondosa extends oligosaccharide chains. Process Biochem. 2022, 112, 80–91. [Google Scholar] [CrossRef]
  102. Wang, W.F.; Xiao, H.; Zhong, J.J. Biosynthesis of a ganoderic acid in Saccharomyces cerevisiae by expressing a cytochrome P450 gene from Ganoderma lucidum. Biotechnol. Bioeng. 2018, 115, 1842–1854. [Google Scholar] [CrossRef] [PubMed]
  103. Bai, Y.; Wang, Y.X.; Liu, X.; Zhao, J.; Kang, L.Q.; Liu, Z.H.; Yuan, S. Heterologous expression and characterization of a novel chitin deacetylase, CDA3, from the mushroom Coprinopsis cinerea. Int. J. Biol. Macromol. 2020, 150, 536–545. [Google Scholar] [CrossRef] [PubMed]
  104. Yang, C.; Guo, L.q.; Ye, Z.w.; Zou, Y.; Yu, Y.H.; Lin, J.F. Cloning of the Transglutaminase Gene from Cordyceps militaris and Its Expressionin Pichia pastoris. J. Chin. Inst. Food Sci. Technol. 2022, 22, 107–113. [Google Scholar]
  105. Lee, J.; Lee, H.; Lee, J.; Chang, P.S. Heterologous expression, purification, and characterization of a recombinant Cordyceps militaris lipase from Candida rugosa-like family in Pichia pastoris. Enzyme Microb. Technol. 2023, 168, 110254. [Google Scholar] [CrossRef] [PubMed]
  106. Dong, X.R.; Qin, L.N.; Tao, Y.; Huang, J.Z.; Dong, Z.Y. Overexpression and characterization of a laccase gene from Pleurotus ostreatus in Trichoderma reesei. Acta Microbiol. Sin. 2012, 52, 850–856. [Google Scholar]
  107. Zou, G.; Zhou, Z.H. CRISPR/Cas9-Mediated Genome Editing of Trichoderma reesei. Methods Mol. Biol. 2021, 2234, 87–98. [Google Scholar] [PubMed]
  108. Lan, X.T.; Yuan, W.; Wang, M.; Xiao, H. Efficient biosynthesis of antitumor ganoderic acid HLDOA using a dual tunable system for optimizing the expression of CYP5150L8 and a Ganoderma P450 reductase. Biotechnol. Bioeng. 2019, 116, 3301–3311. [Google Scholar] [CrossRef]
  109. Yuan, W.; Jiang, C.J.; Wang, Q.; Fang, Y.B.; Wang, J.; Wang, M.; Xiao, H. Biosynthesis of mushroom-derived type II ganoderic acids by engineered yeast. Nat. Commun. 2022, 13, 7740. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, B.X.; Xue, L.N.; Wei, T.; Wang, N.; Zhong, J.R.; Ye, Z.W.; Guo, L.Q.; Lin, J.F. Multiplex gene precise editing and large DNA fragment deletion by the CRISPR-Cas9-TRAMA system in edible mushroom Cordyceps militaris. Microb. Biotechnol. 2022, 15, 2982–2991. [Google Scholar] [CrossRef] [PubMed]
  111. Han, H.Y.; Yu, C.Y.; Qi, J.Z.; Wang, P.C.; Zhao, P.P.; Gong, W.B.; Xie, C.L.; Xia, X.K.; Liu, C.W. High-efficient production of mushroom polyketide compounds in a platform host Aspergillus oryzae. Microb. Cell Fact. 2023, 22, 60. [Google Scholar] [CrossRef] [PubMed]
  112. Valiante, V. Advances in Synthetic Biology of Fungi and Contributions to the Discovery of New Molecules. Chembiochem. 2023, 24, e202300008. [Google Scholar] [CrossRef]
  113. Feng, D.L.; Hu, H.P.; Yong, T.Q.; Liu, Y.C.; Xiao, C.; Huang, L.H.; Xie, Y.Z.; WU, Q.P. Induction of sexual fruiting-body formation by pairing the opposite mating-type isolates of Cordyceps militaris. Mycosystema 2023, 42, 344–352. [Google Scholar]
  114. Guo, Y.X.; Chen, Q.J.; Zhang, G.Q.; Wang, H.X.; Qin, Y. Application of Mating Type Gene Molecular Markers in Morchella Single-spore Cross Breeding. Mol. Plant Breed. 2022, 20, 4067–4074. [Google Scholar]
  115. Whitehouse, H.L.K. Multiple-Allelomorph Heterothallism in the Fungi. New Phytol. 1949, 48, 212–244. [Google Scholar] [CrossRef]
  116. Davis, R.H. Genetics of Sexuality in Higher Fungi. BioScience 1967, 17, 200. [Google Scholar] [CrossRef]
  117. Casselton, L.A. Fungal sex genes-searching for the ancestors. BioEssays News Rev. Mol. Cell. Dev. Biol. 2008, 30, 711–714. [Google Scholar] [CrossRef] [PubMed]
  118. Raudaskoski, M.; Kothe, E. Basidiomycete mating type genes and pheromone signaling. Eukaryot. Cell 2010, 9, 847–859. [Google Scholar] [CrossRef]
  119. Casselton, L.A.; Olesnicky, N.S. Molecular genetics of mating recognition in basidiomycete fungi. Microbiol. Mol. Biol. R. 1998, 62, 55–70. [Google Scholar] [CrossRef] [PubMed]
  120. Xu, S.J.; Wan, J.N.; Li, Y.; Li, Y.; Shang, J.J.; Bao, D.P. Characterization of clamps and nuclear phases in the monokaryons with transformed mating genes in Pleurotus eryngii. Mycosystema 2020, 39, 1130–1138. [Google Scholar]
  121. Bao, D.P. Research progress on the mating-typing locus structures of basidiomycete mushrooms. Mycosystema 2019, 38, 2061–2077. [Google Scholar]
  122. Sun, Y.; Zhang, Y.J.; Cao, X.Y.; Zhou, J.S.; Jiang, J.H. Characteristics related to sexual reproduction and mating type locus of Inonotus obliquus. Mycosystema 2023, 42, 520–529. [Google Scholar]
  123. Ma, L.J.; Li, X.L.; Bao, D.P.; Shang, J.J.; Zhou, C.L.; Yang, R.H. Genetic Structure and Polymorphism of Mating Type Loci in Different Hypsizygus marmoreus Strains. Acta Edulis Fungi 2022, 29, 23–30. [Google Scholar]
  124. Shang, J.J.; Hou, D.; Li, Y.; Zhou, C.L.; Guo, T.; Tang, L.H.; Mao, W.J.; Chen, Q.; Bao, D.P.; Yang, R.H. Analyses of mating systems in Stropharia rugosoannulata based on genomic data. Mycosystema 2020, 39, 1152–1161. [Google Scholar]
  125. Qian, R.; Chen, W.M.; Chai, H.M.; Tao, N.; Zhao, Y.C.; Chen, Y.H. B Mating Locus from Agrocybe aegerita: Structural Analysis. Chin. Agric. Sci. Bull. 2021, 37, 14–19. [Google Scholar]
  126. Zhang, S.S.; Li, X.; Li, G.J.; Huang, Q.; Tian, J.H.; Wang, J.L.; Li, M.; Li, S.M. Genetic and Molecular Evidence of a Tetrapolar Mating System in the Edible Mushroom Grifola frondosa. J. Fungi 2023, 9, 959. [Google Scholar] [CrossRef] [PubMed]
  127. Shang, J.J.; Xu, S.J.; Tang, L.H.; Yang, R.H.; Gong, M.; Li, Y.; Wang, Y.; Zou, G.; Wan, J.N.; Bao, D.P. Transformation of Compatible Mating-Type Genes in Monokaryons Triggers Fruiting Body Development by Activating Mating Pathways in Pleurotus eryngii. Microbiol. Spectr. 2023, 11, e0527222. [Google Scholar] [CrossRef] [PubMed]
  128. Riffiani, R.; Chen, F.C.; Zhang, W.; Wada, T.; Shimomura, N.; Yamaguchi, T.; Aimi, T. Identification, characterization and expression of A-mating type genes in monokaryons and dikaryons of the edible mushroom Mycoleptodonoides aitchisonii (Bunaharitake). Mycoscience 2021, 62, 106–114. [Google Scholar] [CrossRef] [PubMed]
  129. Han, J.X.; Kawauchi, M.; Schiphof, K.; Terauchi, Y.; Yoshimi, A.; Tanaka, C.; Nakazawa, T.; Honda, Y.C. Features of disruption mutants of genes encoding for hydrophobin Vmh2 and Vmh3 in mycelial formation and resistance to environmental stress in Pleurotus ostreatus. FEMS Microbiol. Lett. 2023, 370, 370. [Google Scholar] [CrossRef] [PubMed]
  130. Su, W.Y.; Sheng, L.Z.; Dai, Y.T.; Song, B.; Fu, Y.P.; Li, Y. Cloning and function analysis of fruiting body lectin gene from Pleurotus ostreatus. Mycosystema 2017, 36, 1111–1120. [Google Scholar]
  131. Shen, J.W.; Liang, S.S.; Qiu, L.Y.; Qi, Y.C. Cloning and Expression of a fst3 Gene from Pleurotus ostreatus. Acta Edulis Fungi 2015, 22, 15–20. [Google Scholar]
  132. Zhao, C.; Wang, X.; Yan, Z.; Hao, J.B.; Wang, F.; Liu, J.Y. Analysis of HMG-box Transcription Factors in Pleurotus ostreatus and Cloning of a Candidate Gene. Acta Edulis Fungi 2023, 30, 1–11. [Google Scholar]
  133. Nakazawa, T.; Morimoto, R.; Wu, H.L.; Kodera, R.; Sakamoto, M.; Honda, Y.C. Dominant effects of gat1 mutations on the ligninolytic activity of the white-rot fungus Pleurotus ostreatus. Fungal Biol. 2019, 123, 209–217. [Google Scholar] [CrossRef] [PubMed]
  134. Armas-Tizapantzi, A.; Martínez, Y.P.J.L.; Fernández, F.J.; Mata, G.; Hernández-Cuevas, L.V.; Ortiz Ortiz, E.; García Nieto, E.; Tomasini, A.; Sierra-Palacios, E.; Marcial-Quino, J.; et al. Silencing of the Laccase (lacc2) Gene from Pleurotus ostreatus Causes Important Effects on the Formation of Toxocyst-like Structures and Fruiting Body. Int. J. Mol. Sci. 2023, 24, 8143. [Google Scholar] [CrossRef]
  135. Hou, L.D.; Liu, Z.Q.; Yan, K.X.; Xu, L.J.; Chang, M.C.; Meng, J.L. Mnsod1 promotes the development of Pleurotus ostreatus and enhances the tolerance of mycelia to heat stress. Microb. Cell Fact. 2022, 21, 155. [Google Scholar] [CrossRef]
  136. Tao, Y.X.; Chen, R.L.; Yan, J.J.; Long, Y.; Tong, Z.J.; Song, H.B.; Xie, B.G. A hydrophobin gene, Hyd9, plays an important role in the formation of aerial hyphae and primordia in Flammulina filiformis. Gene 2019, 706, 84–90. [Google Scholar] [CrossRef] [PubMed]
  137. Lu, Y.P.; Chen, R.L.; Long, Y.; Li, X.; Jiang, Y.J.; Xie, B.G. A Jacalin-Related Lectin Regulated the Formation of Aerial Mycelium and Fruiting Body in Flammulina velutipes. Int. J. Mol. Sci. 2016, 17, 1884. [Google Scholar] [CrossRef] [PubMed]
  138. Lyu, X.M.; Jiang, S.Y.; Wang, L.; Chou, T.S.; Wang, Q.J.; Meng, L.; Mukhtar, I.; Xie, B.G.; Wang, W. The Fvclp1 gene regulates mycelial growth and fruiting body development in edible mushroom Flammulina velutipes. Arch. Microbiol. 2021, 203, 5373–5380. [Google Scholar] [CrossRef] [PubMed]
  139. Wu, T.J.; Hu, C.C.; Xie, B.G.; Zhang, L.; Yan, S.J.; Wang, W.; Tao, Y.X.; Li, S.J. A Single Transcription Factor (PDD1) Determines Development and Yield of Winter Mushroom (Flammulina velutipes). Appl. Environ. Microbiol. 2019, 85, e01735-01719. [Google Scholar] [CrossRef] [PubMed]
  140. Wu, T.J.; Hu, C.C.; Xie, B.G.; Wei, S.L.; Zhang, L.; Zhu, Z.X.; Zhang, Z.Y.; Li, S.J. A putative transcription factor LFC1 negatively regulates development and yield of winter mushroom. Appl. Microbiol. Biotechnol. 2020, 104, 5827–5844. [Google Scholar] [CrossRef] [PubMed]
  141. Han, X.; Wei, Z.Y.; Zhang, Y.L.; Huang, R.M.; Huang, Q.H.; Xie, B.G. Identification and Expression Analysis of Two Oligopeptide Transporter Encoding Genes fvoptl and fvopt2 from Flammulina velutipes. Acta Edulis Fungi 2018, 25, 17–23. [Google Scholar]
  142. Wu, T.J.; Zhang, Z.Y.; Hu, C.C.; Zhang, L.; Wei, S.L.; Li, S.J. A WD40 Protein Encoding Gene Fvcpc2 Positively Regulates Mushroom Development and Yield in Flammulina velutipes. Front. Microbiol. 2020, 11, 498. [Google Scholar] [CrossRef] [PubMed]
  143. Long, Y.; Yan, J.J.; Tong, Z.J.; Liu, Y.Y.; Miao, J.; Tao, Y.X.; Jiang, Y.J.; Xie, B.G. fv-gs6: A glucan synthase gene associated with stipe elongation of Flammulina velutipes. J. Fujian Agric. For. Univ. (Nat. Sci. Ed.) 2018, 47, 698–704. [Google Scholar]
  144. Yun, Y.H.; Koo, J.S.; Kim, S.H.; Kong, W.S. Cloning and Expression Analysis of Phenylalanine Ammonia-Lyase Gene in the Mycelium and Fruit Body of the Edible Mushroom Flammulina velutipes. Mycobiology 2015, 43, 327–332. [Google Scholar] [CrossRef]
  145. Liu, A.; Lyu, X.M.; Wang, Q.J.; Wang, X.F.; Li, Z.; ZHang, J.Y.; Wang, W. Differential expression of key genes in pheromone signaling pathway in fruiting body development of Flammulina filiformis. Microbiol. China 2022, 49, 2563–2574. [Google Scholar]
  146. Wang, R.Q.; Yan, J.J.; Li, Y.N.; Yang, H.; Ma, X.B.; Wang, M.; Tao, Y.X.; Xie, B.G. Cytochrome c peroxidase gene (ffccp) and its differential expression during stipe elongation in Flammulina filiformis. Mycosystema 2020, 39, 993–1005. [Google Scholar]
  147. Tu, J.L.; Xu, J.W. Subcellular Localization of Ganoderma lucidum CRZl and Transcription Levels of crzl at Different Developmental Stages. Acta Edulis Fungi 2021, 28, 1–9. [Google Scholar]
  148. Wu, F.L.; Zhang, G.; Ren, A.; Dang, Z.H.; Shi, L.; Jiang, A.L.; Zhao, M.W. The pH-responsive transcription factor PacC regulates mycelial growth, fruiting body development, and ganoderic acid biosynthesis in Ganoderma lucidum. Mycologia 2016, 108, 1104–1113. [Google Scholar]
  149. Zhang, G.; Ren, A.; Shi, L.; Zhu, J.; Jiang, A.L.; Shi, D.K.; Zhao, M.W. Functional analysis of an APSES transcription factor (GlSwi6) involved in fungal growth, fruiting body development and ganoderic-acid biosynthesis in Ganoderma lucidum. Microbiol. Res. 2018, 207, 280–288. [Google Scholar] [CrossRef] [PubMed]
  150. Zhang, G.; Sun, Z.H.; Ren, A.; Shi, L.; Shi, D.K.; Li, X.B.; Zhao, M.W. The mitogen-activated protein kinase GlSlt2 regulates fungal growth, fruiting body development, cell wall integrity, oxidative stress and ganoderic acid biosynthesis in Ganoderma lucidum. Fungal Genet. Biol. 2017, 104, 6–15. [Google Scholar] [CrossRef]
  151. Li, X.; Liu, M.Q.; Dong, C.H. Hydrophobin Gene Cmhyd4 Negatively Regulates Fruiting Body Development in Edible Fungi Cordyceps militaris. Int. J. Mol. Sci. 2023, 24, 4586. [Google Scholar] [CrossRef] [PubMed]
  152. Bao, D.P.; Ma, Y.W.; Gong, M.; Li, Y.; Gao, Y.N.; Yang, R.H.; Yang, R.F.; Mao, W.J.; Wang, Y. Sequence analysis and heterologous expression of lectin-like gene CMLec3 from the medicinal fungus Cordyceps militaris. Mycoscience 2018, 59, 119–123. [Google Scholar] [CrossRef]
  153. Zhang, Z.J.; Yin, Y.Y.; Cui, Y.; Zhang, Y.X.; Liu, B.Y.; Ma, Y.C.; Liu, Y.N.; Liu, G.Q. Chitinase Is Involved in the Fruiting Body Development of Medicinal Fungus Cordyceps militaris. Life 2023, 13, 764. [Google Scholar] [CrossRef]
  154. Zhang, J.J.; Hao, H.B.; Liu, H.; Wang, Q.; Chen, M.J.; Feng, Z.Y.; Chen, H. Genetic and functional analysis of the Zn(II)(2)Cys(6) transcription factor HADA-1 in Hypsizygus marmoreus. Appl. Microbiol. Biotechnol. 2021, 105, 2815–2829. [Google Scholar] [CrossRef]
  155. Zhang, J.J.; Chen, H.; Chen, M.J.; Ren, A.; Huang, J.C.; Wang, H.; Zhao, M.W.; Feng, Z.Y. Cloning and functional analysis of a laccase gene during fruiting body formation in Hypsizygus marmoreus. Microbiol. Res. 2015, 179, 54–63. [Google Scholar] [CrossRef]
  156. Shioya, T.; Nakamura, H.; Ishii, N.; Takahashi, N.; Sakamoto, Y.; Ozaki, N.; Kobayashi, M.; Okano, K.; Kamada, T.; Muraguchi, H. The Coprinopsis cinerea septin Cc.Cdc3 is involved in stipe cell elongation. Fungal Genet. Biol. 2013, 58–59, 80–90. [Google Scholar] [CrossRef] [PubMed]
  157. Pelkmans, J.F.; Vos, A.M.; Scholtmeijer, K.; Hendrix, E.; Baars, J.J.P.; Gehrmann, T.; Reinders, M.J.T.; Lugones, L.G.; Wösten, H.A.B. The transcriptional regulator c2h2 accelerates mushroom formation in Agaricus bisporus. Appl. Microbiol. Biot. 2016, 100, 7151–7159. [Google Scholar] [CrossRef] [PubMed]
  158. Zhang, C.H.; Zhang, G.; Wen, Y.M.; Li, T.; Gao, Y.Q.; Meng, F.M.; Qiu, L.Y.; Ai, Y.C. Pseudomonas sp. UW4 acdS gene promotes primordium initiation and fruiting body development of Agaricus bisporus. World J. Microb. Biot. 2019, 35, 163. [Google Scholar] [CrossRef] [PubMed]
  159. Zhou, S.H.; Shen, Y.Y.; Cai, W.M.; Jin, Q.L.; Fang, L.J.; Song, T.T.; Feng, W.L.; Zhang, X.H. Cloning and functional prediction of the Ppcsl-1 related to change-temperature fruiting of Pleurotus pulmonarius. Mycosystema 2016, 35, 946–955. [Google Scholar]
  160. Chen, B.Z.; Li, L.; Chen, T.C.; Meng, Q.M.; Wu, M.W.; Xie, B.G.; Jiang, Y.J. Construction of the overexpression transformant strains with MADS-box transcription factor Vvrin1 gene and analysis on differential growth rate of Volvariella volvacea. Mycosystema 2020, 39, 1049–1055. [Google Scholar]
  161. Chen, B.Z.; Li, L.; Zhong, Q.G.; Qiu, M.M.; Pan, X.Y.; Xie, B.G.; Jiang, Y.J. Expression analyses of Vvrin1 gene encoding MADS-box transcription factors in different development stages of Volvariella volvacea. Mycosystema 2018, 37, 1628–1634. [Google Scholar]
  162. Wang, W.; Wang, L.; Chen, B.Z.; Mukhtar, I.; Xie, B.G.; Li, Z.; Meng, L. Characterization and expression pattern of homeobox transcription factors in fruiting body development of straw mushroom Volvariella volvacea. Fungal. Biol. 2019, 123, 95–102. [Google Scholar] [CrossRef] [PubMed]
  163. Yan, J.J.; Tong, Z.J.; Liu, Y.Y.; Zhang, L.; Zhang, Y.; Xie, B.G. Sequence Characterization and Differential Expression Analysis of a Aryl Alcohol Oxidase Gene vvaao1 from Volvariella volvacea. Biotechnol. Bull. 2018, 34, 107–114. [Google Scholar]
  164. Sato, T.; Suzuki, Y.; Naito, M.; Minami, A.; Suzuki, N.; Yaegashi, K.; Hirano, T. Overexpression of the laccase gene, lcc1, in Lentinula edodes using the pChG vector. Mycoscience 2019, 60, 246–249. [Google Scholar] [CrossRef]
  165. Ma, X.X.; Lu, L.X.; Zhang, Y.M.; Fang, M.; Shao, K.S.; Sun, X.; Yao, F.J.; Wang, P. Velvet Family Members Regulate Pigment Synthesis of the Fruiting Bodies of Auricularia cornea. J. Fungi 2023, 9, 412. [Google Scholar] [CrossRef]
  166. Zhu, G.; Hayashi, M.; Shimomura, N.; Yamaguchi, T.; Aimi, T. Expression of α-glucosidase during morphological differentiation in the basidiomycetous fungus Pholiota microspora. J. Basic. Microb. 2016, 56, 1036–1045. [Google Scholar] [CrossRef]
  167. Zan, X.Y.; Zhu, H.A.; Jiang, L.H.; Liang, Y.Y.; Sun, W.J.; Tao, T.L.; Cui, F.J. The role of Rho1 gene in the cell wall integrity and polysaccharides biosynthesis of the edible mushroom Grifola frondosa. Int. J. Biol. Macromol. 2020, 165, 1593–1603. [Google Scholar] [CrossRef] [PubMed]
  168. Sun, X.Y.; Liu, D.M.; Zhao, X.H. Transcription factors: Switches for regulating growth and development in macrofungi. Appl. Microbiol. Biot. 2023, 107, 6179–6191. [Google Scholar] [CrossRef]
  169. Li, L.Z.; Qu, M.R.; Liu, C.J.; Xu, L.J.; Pan, K.; Song, X.Z.; OuYang, K.H.; Li, Y.J.; Zhao, X.H. Expression of a Recombinant Lentinula edodes Xylanase by Pichia pastoris and Its Effects on Ruminal Fermentation and Microbial Community in in vitro Incubation of Agricultural Straws. Front. Microbiol. 2018, 9, 2944. [Google Scholar] [CrossRef]
  170. Li, L.z.; Qu, M.R.; Liu, C.J.; Pan, K.; Xu, L.J.; OuYang, K.H.; Song, X.Z.; Li, Y.J.; Zhao, X.H. Expression of a recombinant Lentinula edodes cellobiohydrolase by Pichia pastoris and its effects on in vitro ruminal fermentation of agricultural straws. Int. J. Biol. Macromol. 2019, 134, 146–155. [Google Scholar] [CrossRef]
  171. Liu, C.J.; Zhang, W.J.; Li, Y.J.; Pan, K.; OuYang, K.H.; Song, X.Z.; Xiong, X.W.; Zang, Y.T.; Wang, L.; Qu, M.E.; et al. Characterization of yeast cell surface displayed Lentinula edodes xylanase and its effects on the hydrolysis of wheat. Int. J. Biol. Macromol. 2022, 199, 341–347. [Google Scholar] [CrossRef]
  172. Gong, M.; Wang, Y.; Bao, D.P.; Jiang, S.; Chen, H.Y.; Shang, J.J.; Wang, X.J.; Yu, H.H.; Zou, G. Improving cold-adaptability of mesophilic cellulase complex with a novel mushroom cellobiohydrolase for efficient low-temperature ensiling. Bioresour Technol. 2023, 376, 128888. [Google Scholar] [CrossRef] [PubMed]
  173. Li, Y.L.; Li, T.B.; Guo, J.T.; Wang, G.; Chen, G. Expression and characterization of a novel lytic polysaccharide monooxygenase, PdLPMO9A, from the edible fungus Pleurotus djamor and its synergistic interactions with cellulase in corn straw biomass saccharification. Bioresource Technol. 2022, 348, 126792. [Google Scholar] [CrossRef] [PubMed]
  174. Zou, G.; Bao, D.P.; Wang, Y.; Zhou, S.C.; Xiao, M.L.; Yang, Z.S.; Wang, Y.M.; Zhou, Z.H. Alleviating product inhibition of Trichoderma reesei cellulase complex with a product-activated mushroom endoglucanase. Bioresour Technol. 2021, 319, 124119. [Google Scholar] [CrossRef]
  175. Heneghan, M.N.; Burns, C.; Costa, A.M.S.B.; Burton, K.S.; Challen, M.P.; Bailey, A.M.; Foster, G.D. Functional analysis of Agaricus bisporus serine proteinase 1 reveals roles in utilization of humic rich substrates and adaptation to the leaf-litter ecological niche. Environ. Microbiol. 2016, 18, 4687–4696. [Google Scholar] [CrossRef]
  176. Jiang, N.; Hu, S.; Peng, B.; Li, Z.H.; Yuan, X.H.; Xiao, S.J.; Fu, Y.P. Genome of Ganoderma Species Provides Insights Into the Evolution, Conifers Substrate Utilization, and Terpene Synthesis for Ganoderma tsugae. Front. Microbiol. 2021, 12, 724451. [Google Scholar] [CrossRef] [PubMed]
  177. Lian, L.D.; Qiao, J.J.; Guo, X.Y.; Xing, Z.Z.; Ren, A.; Zhao, M.W.; Zhu, J. The transcription factor GCN4 contributes to maintaining intracellular amino acid contents under nitrogen-limiting conditions in the mushroom Ganoderma lucidum. Microb. Cell Factories 2023, 22, 205. [Google Scholar] [CrossRef]
  178. Pareek, M.; Hegedüs, B.; Hou, Z.H.; Csernetics, Á.; Wu, H.L.; Virágh, M.; Sahu, N.; Liu, X.B.; Nagy, L. Preassembled Cas9 Ribonucleoprotein-Mediated Gene Deletion Identifies the Carbon Catabolite Repressor and Its Target Genes in Coprinopsis cinerea. Appl. Environ. Microbiol. 2022, 88, e0094022. [Google Scholar] [CrossRef]
  179. Yang, H.L.; Chang, T.T.; Zhao, Y.; You, H.F.; Zha, L.; Li, Z.P.; Tong, Z.J.; Chen, M.J. Expression of catalase gene (VvCAT1) from Volvariella volvacea in Escherichia coli and its temperature tolerance. Microbiol. China 2021, 48, 4054–4060. [Google Scholar]
  180. Yang, H.L.; You, H.F.; Zhao, Y.; Pattana, K.; Chang, T.T.; Yu, C.X.; Cheng, M.J. Heterologous Expression of Manganese Superoxide Dismutase from Volvariella volvacea in E. coli and Characterization of ts Stress Tolerance. Acta Edulis Fungi 2020, 27, 51–58. [Google Scholar]
  181. Yang, H.L.; Tong, Z.J.; Zhao, Y.; Jiang, J.; Zha, L.; Cheng, M.J. Bioinformatics analysis of manganese peroxidase gene (LeMnP1) in Lentinula edodes and effects of high temperature stress on its expression. Mycosystema 2020, 39, 1056–1064. [Google Scholar]
  182. Yang, T.; Dong, C.H. Photo morphogenesis and photo response of the blue-light receptor gene Cmwc-1 in different strains of Cordyceps militaris. FEMS Microbiol. Lett. 2014, 352, 190–197. [Google Scholar] [CrossRef]
  183. Pelkmans, J.F.; Patil, M.B.; Gehrmann, T.; Reinders, M.J.T.; Wösten, H.A.B.; Lugones, L.G. Transcription factors of Schizophyllum commune involved in mushroom formation and modulation of vegetative growth. Sci. Rep. 2017, 7, 310. [Google Scholar] [CrossRef]
  184. Song, L.H.; Shrivastava, N.; Gai, Y.P.; Li, D.; Cai, W.M.; Shen, Y.Y.; Lin, F.C.; Liu, J.Y.; Wang, H.K. Role of the blue light receptor gene Icwc-1 in mycelium growth and fruiting body formation of Isaria cicadae. Front. Microbiol. 2022, 13, 1038034. [Google Scholar] [CrossRef] [PubMed]
  185. Li, J.; Xu, C.; Jing, Z.H.; Li, X.Y.; Li, H.; Chen, Y.Z.; Shao, Y.P.; Cai, J.F.; Wang, B.; Xie, B.G.; et al. Blue light and its receptor white collar complex (FfWCC) regulates mycelial growth and fruiting body development in Flammulina filiformis. Sci. Hortic. 2023, 309, 111623. [Google Scholar] [CrossRef]
  186. Kurahashi, A.; Shimoda, T.; Sato, M.; Fujimori, F.; Hirama, J.; Nishibori, K. A putative transcription factor Gf.BMR1 in Grifola frondosa, the homolog of BMR1 in Bipolaris oryzae, was strongly induced by near-ultraviolet light and blue light. Mycoscience 2015, 56, 177–182. [Google Scholar] [CrossRef]
  187. Hong, C.P.; Moon, S.; Yoo, S.I.; Noh, J.H.; Ko, H.G.; Kim, H.A.; Ro, H.S.; Cho, H.; Chung, J.W.; Lee, H.Y.; et al. Functional Analysis of a Novel ABL (Abnormal Browning Related to Light) Gene in Mycelial Brown Film Formation of Lentinula edodes. J. Fungi 2020, 6, 272. [Google Scholar] [CrossRef] [PubMed]
  188. Hu, X.; Lu, Z.L.; Shen, Y.M.; Tao, Y.; Song, S.Y. Para-aminobenzoic acid synthase from mushroom Agaricus bisporus enhances UV-C tolerance in Arabidopsis by reducing oxidative DNA damage. Acta Physiol. Plant. 2019, 41, 160. [Google Scholar] [CrossRef]
  189. Chen, X.; Lv, S.Y.; Mou, C.Y.; Bian, Y.B.; Kang, H. Functions of gene ATX1 under cadmium stress in Morchella importuna. Mycosystema 2020, 39, 827–838. [Google Scholar]
  190. Shen, N.; Xu, C.J.; Zhang, J.C.; Liu, K.F.; Liu, G.L.; He, S.F.; Wang, L.; Bian, Y.B.; Xiao, Y. Molecular mechanism underlying cadmium tolerance differentiation in Lentinula edodes as revealed by mRNA and milRNA analyses. J. Hazard. Mater. 2022, 440, 129841. [Google Scholar] [CrossRef]
  191. Elsayed, E.A.; El Enshasy, H.; Wadaan, M.A.; Aziz, R. Mushrooms: A potential natural source of anti-inflammatory compounds for medical applications. Mediat. Inflamm. 2014, 2014, 805841. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, J.J.; Pu, J.F.; Zhang, Z.X.; Feng, Z.; Han, J.; Su, X.J.; Shi, L.S. Triterpenoids of Ganoderma lucidum inhibited S180 sarcoma and H22 hepatoma in mice by regulating gut microbiota. Heliyon 2023, 9, e16682. [Google Scholar] [CrossRef] [PubMed]
  193. Ahmad, M.F.; Ahmad, F.A.; Khan, M.I.; Alsayegh, A.A.; Wahab, S.; Alam, M.I.; Ahmed, F. Ganoderma lucidum: A potential source to surmount viral infections through β-glucans immunomodulatory and triterpenoids antiviral properties. Int. J. Biol. Macromol. 2021, 187, 769–779. [Google Scholar] [CrossRef] [PubMed]
  194. Zhao, Z.Z.; Ji, B.Y.; Wang, Z.Z.; Si, Y.Y.; Sun, Y.J.; Chen, H.; Feng, W.S.; Zheng, X.K.; Liu, J.K. Lanostane triterpenoids with anti-proliferative and anti-inflammatory activities from medicinal mushroom Ganoderma lingzhi. Phytochemistry 2023, 213, 113791. [Google Scholar] [CrossRef] [PubMed]
  195. Zhang, X.Q.; Gao, X.X.; Long, G.Q.; Yang, Y.C.; Chen, G.; Hou, G.L.; Huo, X.T.; Jia, J.M.; Wang, A.H.; Hu, G.S. Lanostane-type triterpenoids from the mycelial mat of Ganoderma lucidum and their hepatoprotective activities. Phytochemistry 2022, 198, 113131. [Google Scholar] [CrossRef]
  196. Mi, X.; Zeng, G.R.; Liu, J.Q.; Luo, Z.S.; Zhang, L.; Dai, X.M.; Fang, W.T.; Zhang, J.; Chen, X.C. Ganoderma Lucidum Triterpenoids Improve Maternal Separation-Induced Anxiety- and Depression-like Behaviors in Mice by Mitigating Inflammation in the Periphery and Brain. Nutrients 2022, 14, 2268. [Google Scholar] [CrossRef]
  197. Tan, Y.; Tang, C.H.; Feng, J.; Feng, N.; Wu, Y.Y.; Bao, D.P.; Yang, Y.; Zhang, J.S. Current Progress in the Study on Biosynthesisand Regulation of Ganoderma Triterpenoids. Acta Edulis Fungi 2019, 26, 125–140. [Google Scholar]
  198. Gu, L.; Zhong, X.; Lian, D.H.; Zheng, Y.M.; Wang, H.Z.; Liu, X. Triterpenoid biosynthesis and the transcriptional response elicited by nitric oxide in submerged fermenting Ganoderma lucidum. Process Biochem. 2017, 60, 19–26. [Google Scholar] [CrossRef]
  199. He, X.Y.; Chen, Y.W.; Li, Z.H.; Fang, L.; Chen, H.M.; Liang, Z.S.; Abozeid, A.; Yang, Z.Q.; Yang, D.F. Germplasm resources and secondary metabolism regulation in Reishi mushroom (Ganoderma lucidum). Chin. Herb. Med. 2023, 15, 376–382. [Google Scholar] [CrossRef] [PubMed]
  200. Xu, J.; Wang, Y.Y.; Zhang, Y.; Xiong, K.H.; Yan, X.Y.; Ruan, S.Y.; Wu, X.Q. Identification of a Novel Metabolic Target for Bioactive Triterpenoids Biosynthesis in Ganoderma lucidum. Front. Microbiol. 2022, 13, 878110. [Google Scholar] [CrossRef] [PubMed]
  201. Wu, F.L.; Shi, L.; Yao, J.; Ren, A.; Zhou, C.; Mu, D.S.; ZHao, M.W. The cloning, characterization, and functional analysis of a gene encoding an isopentenyl diphosphate isomerase involved in triterpene biosynthesis in the Lingzhi or reishi medicinal mushroom Ganoderma lucidum (higher Basidiomycetes). Int. J. Med. Mushrooms 2013, 15, 223–232. [Google Scholar] [CrossRef] [PubMed]
  202. Ren, A.; Ouyang, X.; Shi, L.; Jiang, A.L.; Mu, D.S.; Li, M.J.; Han, Q.; Zhao, M.W. Molecular characterization and expression analysis of GlHMGS, a gene encoding hydroxymethylglutaryl-CoA synthase from Ganoderma lucidum (Ling-zhi) in ganoderic acid biosynthesis pathway. World J. Microb. Biot. 2013, 29, 523–531. [Google Scholar] [CrossRef] [PubMed]
  203. Fei, Y.; Li, N.; Zhang, D.H.; Xu, J.W. Increased production of ganoderic acids by overexpression of homologous farnesyl diphosphate synthase and kinetic modeling of ganoderic acid production in Ganoderma lucidum. Microb. Cell Fact. 2019, 18, 115. [Google Scholar] [CrossRef] [PubMed]
  204. Shi, L.; Qin, L.; Xu, Y.J.; Ren, A.; Fang, X.; Mu, D.S.; Tan, Q.; ZHao, M.W. Molecular cloning, characterization, and function analysis of a mevalonate pyrophosphate decarboxylase gene from Ganoderma lucidum. Mol. Biol. Rep. 2012, 39, 6149–6159. [Google Scholar] [CrossRef]
  205. Jiang, L.X.; Wang, J.J.; Li, N.; Xu, J.W. Identification and Analysis of Candidate Cytochrome P450 Genes Involved in the Biosynthesis of Ganoderic Acid in Ganoderma lingzhi. Acta Edulis Fungi 2018, 25, 35–41. [Google Scholar]
  206. Shi, D.K.; Zhu, J.; Sun, Z.H.; Zhang, G.; Liu, R.; Zhang, T.J.; Wang, S.L.; Ren, A.; ZHao, M.W. Alternative oxidase impacts ganoderic acid biosynthesis by regulating intracellular ROS levels in Ganoderma lucidum. Microbiology 2017, 163, 1466–1476. [Google Scholar] [CrossRef]
  207. Lian, L.D.; Wang, L.S.; Song, S.Q.; Zhu, J.; Liu, R.; Shi, L.; Ren, A.; ZHao, M.W. GCN4 Regulates Secondary Metabolism through Activation of Antioxidant Gene Expression under Nitrogen Limitation Conditions in Ganoderma lucidum. Appl. Environ. Microbiol. 2021, 87, e0015621. [Google Scholar] [CrossRef] [PubMed]
  208. Hu, Y.R.; Xu, W.Z.; Hu, S.S.; Lian, L.D.; Zhu, J.; Shi, L.; Ren, A.; ZHao, M.W. In Ganoderma lucidum, Glsnf1 regulates cellulose degradation by inhibiting GlCreA during the utilization of cellulose. Environ. Microbiol. 2020, 22, 107–121. [Google Scholar] [CrossRef] [PubMed]
  209. Zhang, G.; Zhang, C.H.; Leng, D.D.; Yan, P.; Wang, Z.H.; Zhang, M.X.; Wu, Z.W. The non-canonical functions of telomerase reverse transcriptase gene GlTert on regulating fungal growth, oxidative stress, and ganoderic acid biosynthesis in Ganoderma lucidum. Appl. Microbiol. Biotechnol. 2021, 105, 7353–7365. [Google Scholar] [CrossRef] [PubMed]
  210. Zhang, X.; Ren, A.; Li, M.J.; Cao, P.F.; Chen, T.X.; Zhang, G.; Shi, L.; Jiang, A.L.; ZHao, M.W. Heat Stress Modulates Mycelium Growth, Heat Shock Protein Expression, Ganoderic Acid Biosynthesis, and Hyphal Branching of Ganoderma lucidum via Cytosolic Ca2. Appl. Environ. Microb. 2016, 82, 4112–4125. [Google Scholar] [CrossRef]
  211. Shi, L.; Yue, S.N.; Gao, T.; Zhu, J.; Ren, A.; Yu, H.S.; Wang, H.; ZHao, M.W. Nitrate reductase-dependent nitric oxide plays a key role on MeJA-induced ganoderic acid biosynthesis in Ganoderma lucidum. Appl. Microbiol. Biotechnol. 2020, 104, 10737–10753. [Google Scholar] [CrossRef] [PubMed]
  212. Zhu, J.; Song, S.Q.; Sun, Z.H.; Lian, L.D.; Shi, L.; Ren, A.; ZHao, M.W. Regulation of glutamine synthetase activity by transcriptional and posttranslational modifications negatively influences ganoderic acid biosynthesis in Ganoderma lucidum. Environ. Microbiol. 2021, 23, 1286–1297. [Google Scholar] [CrossRef] [PubMed]
  213. Wang, W.F.; Xiao, H.; Zhong, J.J. Biosynthesis of a novel ganoderic acid by expressing CYP genes from Ganoderma lucidum in Saccharomyces cerevisiae. Appl. Microbiol. Biot. 2022, 106, 523–534. [Google Scholar] [CrossRef] [PubMed]
  214. Yuan, Y.J.; Cheng, S.; Bian, G.K.; Yan, P.; Ma, Z.N.; Dai, W.; Chen, R.; Fu, S.; Huang, H.W.; Chi, H.M.; et al. Efficient exploration of terpenoid biosynthetic gene clusters in filamentous fungi. Nat. Catal. 2022, 5, 277–287. [Google Scholar] [CrossRef]
  215. 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]
  216. Deng, Q.F.; Li, X.R.; Fang, C.Q.; Li, X.; Zhang, J.; Xi, Q.; Li, Y.; Zhang, R.X. Cordycepin enhances anti-tumor immunity in colon cancer by inhibiting phagocytosis immune checkpoint CD47 expression. Int. Immunopharmacol. 2022, 107, 108695. [Google Scholar] [CrossRef]
  217. Wei, P.J.; Wang, K.; Luo, C.; Huang, Y.C.; Misilimu, D.; Wen, H.M.; Jin, P.; Li, C.H.; Gong, Y.; Gao, Y.Q. Cordycepin confers long-term neuroprotection via inhibiting neutrophil infiltration and neuroinflammation after traumatic brain injury. J. Neuroinflamm. 2021, 18, 137. [Google Scholar] [CrossRef] [PubMed]
  218. Zhou, M.H.; Zha, Z.B.; Zheng, Z.; Pan, Y.M. Cordycepin suppresses vascular inflammation, apoptosis and oxidative stress of arterial smooth muscle cell in thoracic aortic aneurysm with VEGF inhibition. Int. Immunopharmacol. 2023, 116, 109759. [Google Scholar] [CrossRef] [PubMed]
  219. Sharma, S.; Madaan, K.; Kaur, R. Cordycepin as a Metabolite with Pharmacological Potential: A Review. Int. J. Med. Mushrooms 2022, 24, 1–20. [Google Scholar] [CrossRef]
  220. Zhao, X.Y.; Li, Q.; Liu, W.J.; Guan, H.Q.; Li, C.; Wang, J.H.; Wang, L. Advances in biosynthesis of cordycepin from Cordyceps militaris. Chin. J. Biotechnol. 2020, 36, 1293–1304. [Google Scholar]
  221. Duan, X.Y.; Yang, H.; Wang, C.; Liu, H.H.; Lu, X.Y.; Tian, Y. Microbial synthesis of cordycepin, current systems and future perspectives. Trends Food Sci. Technol. 2023, 132, 162–170. [Google Scholar] [CrossRef]
  222. Tan, H.P.; Wang, L.; Wang, H.G.; Cheng, Y.H.; Li, X.; Wan, H.H.; Liu, C.G.; Liu, T.; Li, Q. Engineering Komagataella phaffii to biosynthesize cordycepin from methanol which drives global metabolic alterations at the transcription level. Synth. Syst. Biotechnol. 2023, 8, 242–252. [Google Scholar] [CrossRef] [PubMed]
  223. Xia, Y.L.; Luo, F.F.; Shang, Y.F.; Chen, P.L.; Lu, Y.Z.; Wang, C.S. Fungal Cordycepin Biosynthesis Is Coupled with the Production of the Safeguard Molecule Pentostatin. Cell Chem. Biol. 2017, 24, 1479–1489.e1474. [Google Scholar] [CrossRef] [PubMed]
  224. Li, Y.J.; Yan, P.F.; Lu, X.J.; Qiu, Y.L.; Liang, S.; Liu, G.; Li, S.F.; Mou, L.; Xie, N. Involvement of PaSNF1 in Fungal Development, Sterigmatocystin Biosynthesis, and Lignocellulosic Degradation in the Filamentous Fungus Podospora anserina. Front. Microbiol. 2020, 11, 1038. [Google Scholar] [CrossRef] [PubMed]
  225. Wāng, Y.; Wang, R.; Wáng, Y.; Li, Y.; Yang, R.H.; Gong, M.; Shang, J.J.; Zhang, J.S.; Mao, W.J.; Zou, G.; et al. Diverse function and regulation of CmSnf1 in entomopathogenic fungus Cordyceps militaris. Fungal Genet. Biol. 2020, 142, 103415. [Google Scholar] [CrossRef]
  226. 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]
  227. Wang, Y.; Yang, Z.S.; Bao, D.P.; Li, B.; Yin, X.; Wu, Y.Y.; Chen, H.Y.; Tang, G.R.; Li, N.Y.; Zou, G. Improving Hypoxia Adaption Causes Distinct Effects on Growth and Bioactive Compounds Synthesis in an Entomopathogenic Fungus Cordyceps militaris. Front. Microbiol. 2021, 12, 698436. [Google Scholar] [CrossRef]
  228. Zou, G.; Li, B.; Wang, Y.; Yin, X.; Gong, M.; Shang, J.; Wei, Y.; Li, X.; Bao, D. Efficient conversion of spent mushroom substrate into a high value-added anticancer drug pentostatin with engineered Cordyceps militaris. Green Chem. 2021, 23, 10030–10038. [Google Scholar] [CrossRef]
  229. Hu, T.; Lin, Q.L.; Guo, T.; Yang, T.; Zhou, W.H.; Deng, X.F.; Yan, J.K.; Luo, Y.; Ju, M.M.; Luo, F.J. Polysaccharide isolated from Phellinus linteus mycelia exerts anti-inflammatory effects via MAPK and PPAR signaling pathways. Carbohydr. Polym. 2018, 200, 487–497. [Google Scholar] [CrossRef]
  230. Rao, Z.L.; Dong, Y.T.; Zheng, X.J.; Tang, K.Y.; Liu, J. Extraction, purification, bioactivities and prospect of lentinan: A review. Biocatal. Agric. BioTechnol. 2021, 37, 102163. [Google Scholar] [CrossRef]
  231. Yang, Y.X.; Chen, J.L.; Lei, L.; Li, F.H.; Tang, Y.; Yuan, Y.; Zhang, Y.Q.; Wu, S.R.; Yin, R.; Ming, J. Acetylation of polysaccharide from Morchella angusticeps peck enhances its immune activation and anti-inflammatory activities in macrophage RAW264.7 cells. Food Chem. Toxicol. 2019, 125, 38–45. [Google Scholar] [CrossRef]
  232. Bisen, P.S.; Baghel, R.K.; Sanodiya, B.S.; Thakur, G.S.; Prasad, G.B. Lentinus edodes: A macrofungus with pharmacological activities. Curr. Med. Chem. 2010, 17, 2419–2430. [Google Scholar] [CrossRef]
  233. Zhou, J.S.; Bai, Y.; Dai, R.J.; Guo, X.L.; Liu, Z.H.; Yuan, S. Improved Polysaccharide Production by Homologous Co-overexpression of Phosphoglucomutase and UDP Glucose Pyrophosphorylase Genes in the Mushroom Coprinopsis cinerea. J. Agric. Food Chem. 2018, 66, 4702–4709. [Google Scholar] [CrossRef] [PubMed]
  234. Wang, Y.F.; Yang, X.; Chen, P.; Yang, S.L.; Zhang, H. Homologous overexpression of genes in Cordyceps militaris improves the production of polysaccharides. Food Res. Int. 2021, 147, 110452. [Google Scholar] [CrossRef] [PubMed]
  235. Cui, F.J.; Wu, X.H.; Tao, T.L.; Zan, X.Y.; Sun, W.J.; Mu, D.S.; Yang, Y.; Wu, D. Functions of a Glucan Synthase Gene GFGLS in Mycelial Growth and Polysaccharide Production of Grifola frondosa. J. Agric. Food Chem. 2019, 67, 8875–8883. [Google Scholar] [CrossRef]
  236. Jiang, L.H.; Li, X.F.; Zan, X.Y.; Fu, X.; Cui, F.J.; Zhu, H.A.; Sun, W.J.; Tao, T.L. The β-1,3-glucan synthase gene GFGLS2 plays major roles in mycelial growth and polysaccharide synthesis in Grifola frondosa. Appl. Microbiol. Biot. 2022, 106, 563–578. [Google Scholar] [CrossRef]
  237. Zan, X.Y.; Wu, X.H.; Cui, F.J.; Zhu, H.A.; Sun, W.J.; Jiang, L.H.; Tao, T.L.; Zhao, X. UDP-glucose pyrophosphorylase gene affects mycelia growth and polysaccharide synthesis of Grifola frondosa. Int. J. Biol. Macromol. 2020, 161, 1161–1170. [Google Scholar] [CrossRef]
  238. Xu, J.W.; Ji, S.L.; Li, H.J.; Zhou, J.S.; Duan, Y.Q.; Dang, L.Z.; Mo, M.H. Increased polysaccharide production and biosynthetic gene expressions in a submerged culture of Ganoderma lucidum by the overexpression of the homologous α-phosphoglucomutase gene. Bioproc. Biosyst. Eng. 2015, 38, 399–405. [Google Scholar] [CrossRef]
  239. Li, H.J.; Zhang, D.H.; Yue, T.H.; Jiang, L.X.; Yu, X.; Zhao, P.; Li, T.; Xu, J.W. Improved polysaccharide production in a submerged culture of Ganoderma lucidum by the heterologous expression of Vitreoscilla hemoglobin gene. J. Biotechnol. 2016, 217, 132–137. [Google Scholar] [CrossRef]
  240. Xu, Y.L.; Yuan, H.; Li, N.; Xiao, J.H.; Xu, J.W. Increased production and anti-senescence activity of exopolysaccharides in Ganoderma lingzhi by co-overexpression of β-1,3-glucan synthase and UDP-glucose pyrophosphorylase. Int. J. Biol. Macromol. 2023, 253, 126778. [Google Scholar] [CrossRef]
  241. Zou, G.; Ren, J.B.; Wu, D.; Zhang, H.N.; Gong, M.; Li, W.; Zhang, J.S.; Yang, Y. Characterization and Heterologous Expression of UDP-Glucose 4-Epimerase From a Hericium erinaceus Mutant with High Polysaccharide Production. Front. Bioeng. BioTechnol. 2021, 9, 796278. [Google Scholar] [CrossRef]
  242. Wu, J.Y.; Siu, K.C.; Geng, P. Bioactive Ingredients and Medicinal Values of Grifola frondosa (Maitake). Foods 2021, 10, 95. [Google Scholar] [CrossRef]
  243. Jędrzejewski, T.; Pawlikowska, M.; Sobocińska, J.; Wrotek, S. COVID-19 and Cancer Diseases-The Potential of Coriolus versicolor Mushroom to Combat Global Health Challenges. Int. J. Mol. Sci. 2023, 24, 4864. [Google Scholar] [CrossRef]
  244. Zhang, Y.F.; Kong, H.L.; Fang, Y.P.; Nishinari, K.; Phillips, G.O. Schizophyllan: A review on its structure, properties, bioactivities and recent developments. Bioact. Carbohydr. Diet. Fibre 2013, 1, 53–71. [Google Scholar] [CrossRef]
  245. Sivanesan, I.; Muthu, M.; Gopal, J.; Oh, J.W. Mushroom Polysaccharide-Assisted Anticarcinogenic Mycotherapy: Reviewing Its Clinical Trials. Molecules 2022, 27, 4090. [Google Scholar] [CrossRef]
  246. Xu, J.; Xu, D.Z.; Hu, Q.H.; Ma, N.; Pei, F.; Su, A.X.; Ma, G.X. Immune regulatory functions of biologically active proteins from edible fungi. Front. Immunol. 2022, 13, 1034545. [Google Scholar] [CrossRef]
  247. Lin, Z.H.; Yeh, H.; Lo, H.C.; Hua, W.J.; Ni, M.Y.; Wang, L.K.; Chang, T.T.; Yang, M.H.; Lin, T.Y. GMI, a fungal immunomodulatory protein, ameliorates SARS-CoV-2 envelope protein-induced inflammation in macrophages via inhibition of MAPK pathway. Int. J. Biol. Macromol. 2023, 241, 124648. [Google Scholar] [CrossRef] [PubMed]
  248. Li, Q.Z.; Zheng, Y.Z.; Zhou, X.W. Fungal immunomodulatory proteins: Characteristic, potential antitumor activities and their molecular mechanisms. Drug Discov. Today 2019, 24, 307–314. [Google Scholar] [CrossRef] [PubMed]
  249. Rezvani, V.; Pourianfar, H.R.; Mohammadnejad, S.; Madjid Ansari, A.; Farahmand, L. Anticancer potentiality and mode of action of low-carbohydrate proteins and peptides from mushrooms. Appl. Microbiol. Biot. 2020, 104, 6855–6871. [Google Scholar] [CrossRef] [PubMed]
  250. Lin, J.W.; Jia, J.; Shen, Y.H.; Zhong, M.; Chen, L.J.; Li, H.G.; Ma, H.; Guo, Z.F.; Qi, M.F.; Liu, L.X.; et al. Functional expression of FIP-fve, a fungal immunomodulatory protein from the edible mushroom Flammulina velutipes in Pichia pastoris GS115. J. Biotechnol. 2013, 168, 527–533. [Google Scholar] [CrossRef] [PubMed]
  251. Li, C.C.; Zhu, C.; Yang, G.D.; Wu, J.F.; Wang, J.J.; Qiao, Y.; Fu, M.R.; Cui, X.X.; Dong, B.X. Overexpression and activity analysis of Antrodia camphorata immunomodulatory protein. Acta Microbiol. Sin. 2021, 61, 2397–2412. [Google Scholar]
  252. Wu, G.G.; Sun, Y.; Deng, T.S.; Song, L.L.; Li, P.; Zeng, H.J.; Tang, X.M. Identification and Functional Characterization of a Novel Immunomodulatory Protein From Morchella conica SH. Front. Immunol. 2020, 11, 559770. [Google Scholar] [CrossRef] [PubMed]
  253. Zhou, S.Y.; Guan, S.X.; Duan, Z.W.; Han, X.; Zhang, X.; Fan, W.L.; Li, H.G.; Chen, L.J.; Ma, H.; Liu, H.M.; et al. Molecular cloning, codon-optimized gene expression, and bioactivity assessment of two novel fungal immunomodulatory proteins from Ganoderma applanatum in Pichia. Appl. Microbiol. Biotechnol. 2018, 102, 5483–5494. [Google Scholar] [CrossRef] [PubMed]
  254. Wang, Y.; Wang, Y.; Gao, Y.N.; Li, Y.; Wan, J.N.; Yang, R.H.; Mao, W.J.; Zhou, C.L.; Tang, L.H.; Gong, M.; et al. Discovery and Characterization of the Highly Active Fungal Immunomodulatory Protein Fip-vvo82. J. Chem. Inf. Model. 2016, 56, 2103–2114. [Google Scholar] [CrossRef]
  255. Bao, D.P.; Bai, R.; Gao, Y.N.; Li, Y.; Wang, Y. Comparative analysis of two fungal immunomodulatory proteins LZ-8 and LZ-9 of Ganoderma lucidum. Acta Agric. Shanghai 2017, 33, 1–5. [Google Scholar]
  256. Bao, D.P.; Bai, R.; Gao, Y.N.; Wu, Y.Y.; Wang, Y. Computational Insights into the Molecular Mechanism of the High Immunomodulatory Activity of LZ-8 Protein Isolated from the Lingzhi or Reishi Medicinal Mushroom Ganoderma lucidum (Agaricomycetes). Int. J. Med. Mushrooms 2018, 20, 537–548. [Google Scholar] [CrossRef]
  257. Gao, Y.N.; Wáng, Y.; Wāng, Y.; Wu, Y.Y.; Chen, H.Y.; Yang, R.H.; Bao, D.P. Protective Function of Novel Fungal Immunomodulatory Proteins Fip-lti1 and Fip-lti2 from Lentinus tigrinus in Concanavalin A-Induced Liver Oxidative Injury. Oxid. Med. Cell. Longev. 2019, 2019, 3139689. [Google Scholar] [CrossRef] [PubMed]
  258. Yu, S.H.; Wang, Y.; Wu, Y.Y.; Bao, D.P.; Bing, W.; Li, Y.; Chen, H.Y. Characterization, Recombinant Production, and Bioactivity of a Novel Immunomodulatory Protein from Hypsizygus marmoreus. Molecules 2023, 28, 4796. [Google Scholar] [CrossRef] [PubMed]
  259. Ye, C.; Wei, X.Y.; Shi, T.Q.; Sun, X.M.; Xu, N.; Gao, C.; Zou, W. Genome-scale metabolic network models: From first-generation to next-generation. Appl. Microbiol. Biotechnol. 2022, 106, 4907–4920. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The ideas and applications of functional gene analysis technology in edible and medicinal fungi research.
Figure 1. The ideas and applications of functional gene analysis technology in edible and medicinal fungi research.
Jof 10 00311 g001
Figure 2. Schematic diagram showing the molecular genetic structure of mating loci in common mushrooms [120,121,122,123,124,125,126]. 1. Lentinula edodes ‘L54A’ (A locus) and ‘No. 939P42’ (B locus); 2. Flammulina filiformis ‘KACC42780’; 3. Agaricus bisporus ‘H97’; 4. Volvariella volvacea ‘V23-1’; 5. Coprinopsis cinerea ‘Okayama 7 #130’; 6. Schizophyllum commune ‘H4-8’; 7. Pleurotus ostreatus ‘PC15’; 8. Pleurotus tuoliensis ‘489P1’; 9. Pleurotus eryngii ‘181’ (A locus) and ‘SDF55’ (B locus); 10. Ganoderma lucidum ‘G.260125-1’; 11. Lentinus tigrinus ‘Lenti7’; 12. Inonotus obliquus ‘MDJCBS88’; 13. Hypsizygus marmoreus ‘H3-1’; 14. Stropharia rugosoannulata ‘SR1’; 15. Grifola frondosa ‘y59’; 16. Agrocybe aegerita ‘YS8’. The mating loci and the mating genes in the figure are named according to the literature.
Figure 2. Schematic diagram showing the molecular genetic structure of mating loci in common mushrooms [120,121,122,123,124,125,126]. 1. Lentinula edodes ‘L54A’ (A locus) and ‘No. 939P42’ (B locus); 2. Flammulina filiformis ‘KACC42780’; 3. Agaricus bisporus ‘H97’; 4. Volvariella volvacea ‘V23-1’; 5. Coprinopsis cinerea ‘Okayama 7 #130’; 6. Schizophyllum commune ‘H4-8’; 7. Pleurotus ostreatus ‘PC15’; 8. Pleurotus tuoliensis ‘489P1’; 9. Pleurotus eryngii ‘181’ (A locus) and ‘SDF55’ (B locus); 10. Ganoderma lucidum ‘G.260125-1’; 11. Lentinus tigrinus ‘Lenti7’; 12. Inonotus obliquus ‘MDJCBS88’; 13. Hypsizygus marmoreus ‘H3-1’; 14. Stropharia rugosoannulata ‘SR1’; 15. Grifola frondosa ‘y59’; 16. Agrocybe aegerita ‘YS8’. The mating loci and the mating genes in the figure are named according to the literature.
Jof 10 00311 g002
Figure 3. Triterpenoid biosynthesis pathway in Ganoderma [197,198,199]. ACAT: Acelyl-CoA acelyltransferase; HMGS: 3-hydroxy-3-methylglutaryl-CoA synthase; HMGR: 3-hydroxy-3-methylglutaryl-CoA reductase; MVK: mevalonate kinase; MVD: Pyrophosphomevalonate decarboxylase; IDI: isopentenyl diphosphate isomerase; FPS: farnesyl pyrophosphate synthase; GPS: Geranyl pyrophosphate synthase; SQS: Squalene synthase; SE: Squalene epoxidase; LS: lanosterol synthase; CYP51: sterol 14a-demethylase.
Figure 3. Triterpenoid biosynthesis pathway in Ganoderma [197,198,199]. ACAT: Acelyl-CoA acelyltransferase; HMGS: 3-hydroxy-3-methylglutaryl-CoA synthase; HMGR: 3-hydroxy-3-methylglutaryl-CoA reductase; MVK: mevalonate kinase; MVD: Pyrophosphomevalonate decarboxylase; IDI: isopentenyl diphosphate isomerase; FPS: farnesyl pyrophosphate synthase; GPS: Geranyl pyrophosphate synthase; SQS: Squalene synthase; SE: Squalene epoxidase; LS: lanosterol synthase; CYP51: sterol 14a-demethylase.
Jof 10 00311 g003
Figure 4. Map of the cordycepin biosynthesis pathway [220,221,222]. IMP: inosine monophosphate; 3′-AMP: adenosine-3′-monophosphate; 2′-C-3′-dA: 2′-carbonyl-3′-deoxy adenosine; 3′-dI: 3′-deoxyinosine; COR: cordycepin; AMP: adenosine monophosphate; ADP: adenosine diphosphate; dAMP: deoxyadenosine monophosphate; dADP: deoxyadenosine diphosphate; RNRs: ribonucleotide reductases; ADK: adenosine kinase; ADEK: adenylate kinase; NT5E: 5′-nucleotidase; ADA: adenosine deaminase; NK: an N-terminal nucleoside kinase; HisG: a C-terminal HisG family of ATP phosphoribosyltransferases.
Figure 4. Map of the cordycepin biosynthesis pathway [220,221,222]. IMP: inosine monophosphate; 3′-AMP: adenosine-3′-monophosphate; 2′-C-3′-dA: 2′-carbonyl-3′-deoxy adenosine; 3′-dI: 3′-deoxyinosine; COR: cordycepin; AMP: adenosine monophosphate; ADP: adenosine diphosphate; dAMP: deoxyadenosine monophosphate; dADP: deoxyadenosine diphosphate; RNRs: ribonucleotide reductases; ADK: adenosine kinase; ADEK: adenylate kinase; NT5E: 5′-nucleotidase; ADA: adenosine deaminase; NK: an N-terminal nucleoside kinase; HisG: a C-terminal HisG family of ATP phosphoribosyltransferases.
Jof 10 00311 g004
Table 1. General methods for genetic transformation of edible and medicinal fungi.
Table 1. General methods for genetic transformation of edible and medicinal fungi.
MethodAdvantageDisadvantageApplication CaseRefs.
ATMTEfficient; copy number of DNA is inserted low; high efficiency of homologous recombinationTime-consuming; partially sensitive to acetosyringoneConstruction of ATMT in H. marmoreus, with CaMV35s as a promoter to efficiently drive the expression of GUS genes[37,38]
PMTEfficient; high copy number of inserted DNA; suitable for most edible and medicinal fungiRequires special lytic enzyme; difficult to prepare and not easy to store; low regeneration efficiencySuccessful PMT of linear DNA fragments containing the bar gene into C. militaris protoplasts yielded 87 positive mutants containing the bar gene[39,40]
LMTEasy operation; transient transfection; wide range of cell typesCombining other methods for cell wall treatment; only a single copy of the gene is integrated into the chromosomeThrough LMT of A. bisporus, the ACO gene was verified, and the ethylene biosynthesis pathway was explored[41,42]
EPEasy to operate; high copy number of inserted DNA;
suitable for a variety of cell types
The cells are highly damaged and inefficient; requires special equipmentThe pFTXHg plasmid was constructed from the ftx gene of F. filiformis and was transformed by EP, and high conversion efficiency was obtained[37,43]
ATMT: Agrobacterium tumefaciens-mediated transformation; PMT: protoplast-mediated transformation; LMT: liposome-mediated transformation; EP: electroporation transformation.
Table 2. A case study of gene function in edible and medicinal fungi using the ATMT technique.
Table 2. A case study of gene function in edible and medicinal fungi using the ATMT technique.
SpeciesGeneFunctionMethodVectorRefs.
PlasmidPromoter
Flammulina filiformisHMG-box transcription factor (fvhom1)GOEpBHG-fvhom1A. bisporus gpd[44]
Transcription factor (Ste12-like)EOESte12-likeOEPgpd[45]
Fungal immunomodulatory proteins (FIP-fve)GOEpCAMBIA1301-pGPD-FIP-fvepGPD[46]
Fungal immunomodulatory proteins (FIP)GRNAipTCK303-fve (F)-fve (R)Ubi-1[47]
Transcription factor (FvHmg1)GRNAiFvhmg1-RNAiPgpd[48]
Adenosine deaminase (Fv-ada)GRNAipFungiway-Fv-adaPgpd[49]
Heterotrimeric G protein α subunits (FfGa1)EOE
RNAi
FfGa1-OE
FfGa1-RNAi
Pgpd[50]
Lentinus edodesFungal Hsp20 protein (hsp20)EOEpEHg-gdp-hsp20L. edodes gpd[51]
Mating-type gene (LeHD1)GOE
RNAi
LeHD1-OE
LeHD1-RNAi
L. edodes gpd[52]
HMG-box transcription factor (Lelcrp1)BRNAipCAMBIA1300-g-lelcrplL. edodes gpd[53]
Heat shock protein 40 (LeDnaJ)ERNAipCAMBIA1300-g-dualL. edodes gpd[54]
Anthranilate synthase (TrpE)ERNAipCAMBIA1300-g-trpEL. edodes gpd[55]
Flavin-containing monooxygenases (YUCCA)ERNAipCAMBIA1300-g-YUCCA8L. edodes gpd[56]
Tryptophan synthase (LetrpB)ERNAipCAMBIA1300-g-LetrpBL. edodes gpd[57]
Ganoderma lucidumDelta 9 fatty acid desaturase (D9desA)AOEOE::D9desAG. lucidum gpd[58]
Acelyl-CoA acelyltransferase (Gl-aact)AOEpGl-aactG. lucidum gpd[59]
3-hydroxy-3-methylglutaryl-CoA reductase (HMGR)AOEpJW-tHMGRPgpd[60]
Antrodia
cinnamomea
2,3-Oxidosqualene Cyclase (OSC)AOEpCAM-AcOSCCaMV35s[61]
Pleurotus
ostreatus
Transcription factor (Pofst3)GOE
RNAi
pPo-GPD-Pofst3+
pPo-GPD-Pofst3
P. ostreatus gpd[62]
Cordyceps militarisGlutathione peroxidase (gpxA)EOEpDHt-gpd-barA. nidulans gpdA[63]
G: growth and development; E: environmental stress response; B: biodegradation; A: biosynthesis of functionally active substances; OE, overexpression; RNAi: RNA interference.
Table 3. A list of genes that regulate the development of mycelia and fruiting bodies in edible and medicinal fungi.
Table 3. A list of genes that regulate the development of mycelia and fruiting bodies in edible and medicinal fungi.
SpeciesNameCategoryFunctionRefs.
Pleurotus ostreatusVmh2, Vmh3HydrophobinRegulate the fungus to form aerial hyphae[129]
polectin2LectinPromote low-temperature resistance of fruiting body[130]
fst3Transcription factorRegulation of primordium and fruiting body development[131]
PoHMG11Transcription factorRegulation of fruiting body maturation[132]
PoGat1Transcription factorPromote lignin decomposition and fruiting body formation[133]
Pofst3Transcription factorPromote the growth and development of fruiting bodies[62]
Lacc2LaccasePromote the growth of aerial hyphae, primordia, and fruiting bodies[134]
Mnsod1Manganese superoxide dismutasePromote the development of fruiting bodies and enhance mycelia tolerance to heat stress[135]
Flammulina filiformisHyd9HydrophobinPromote mycelia and fruiting body development[136]
Fv-JRL1LectinIncrease aerial hyphae and promote fruiting body development[137]
Fvclp1Transcription factorInvolved in sexual reproduction and fruiting body development[138]
ste12-likeTranscription factorIncrease aerial hyphae and promote fruiting body development[45]
pdd1Transcription factorPromote fruiting body development and increase yield[139]
FvHmg1Negative transcription factorNegatively regulate the fruiting body development[48]
LFC1Negative transcription factorNegatively regulates fruiting body development and yield[140]
fvopt1, fvopt2Oligopeptide transporterRegulation of primordium and fruiting body development[141]
Fvcpc2WD40 ProteinPositively regulates the development and yield of fruiting bodies[142]
fv-gs6β-1,6-glucan synthasePromote cell wall synthesis and stalk elongation process[143]
FvpalPhenylalanine Ammonia-LyaseInvolved in fruiting body growth and development[144]
FfPkacAdenylate-dependent protein kinase A pathwayPromote mycelia and fruiting body development[145]
ffccpCytochrome C peroxidasePromote elongation of fruiting body stalk[146]
Ganoderma lucidumcrzlTranscription factorInvolved in fruiting body growth and development[147]
GlPacCTranscription factorInvolved in mycelium growth and fruiting body development[148]
GlSwi6Transcription factorPromote mycelium growth and fruiting body development[149]
GlSlt2Mitogen-activated protein kinasesRegulates mycelium growth, fruiting body development, and cell wall integrity[150]
Cordyceps militarisCmhyd4HydrophobinNegative regulation of fruiting body growth and development[151]
CMLec3LectinInvolved in fruiting body growth and development[152]
Chi1, Chi4ChitinasePromote fruiting body growth and development[153]
Hypsizygus marmoreusHADA-1Transcription factorPromote mycelium growth and fruiting body development[154]
lcc1LaccasePromote fruiting body growth and development[155]
Coprinopsis cinereaCcNsdD2Transcription factorPromote secondary mycelia and fruiting body growth and development[33]
Cc.Cdc3a homolog of S. cerevisiae CDC3 septinPromote the elongation of fruiting body stalk cells[156]
Agaricus bisporusc2h2Transcription factorShorten fruiting body development period[157]
acdS1-aminocyclopropane-1-carboxylic acid (ACC) deaminase (AcdS)Promote primordium initiation and fruiting body development[158]
Pleurotus pulmonariusPpcsl-1Transcription factorRegulates fruiting body development[159]
Volvariella volvaceaVvrin1Transcription factorPromote mycelium growth and pigment accumulation, elongation of fruiting stalk, and opening of the lid[160,161]
VvHox1-VvHox8Transcription factorAffect the elongation of fruiting bodies and the formation of primordia[162]
vvaao1Aryl Alcohol OxidasePromote the formation and development of fruiting bodies[163]
Lentinula edodeslcc1LaccaseInvolved in fruiting body growth and development[164]
Auricularia corneaAcveAVelvet factor family proteinRegulation of fruiting body pigment synthesis[165]
Pholiota microsporaPnGcsα-glucosidaseInvolved in fruiting body development[166]
Griflola frondosaRho1β-1,3-glucan synthasePromote mycelium growth[167]
Table 4. A list of genes related to the environmental response of edible and medicinal fungi.
Table 4. A list of genes related to the environmental response of edible and medicinal fungi.
TypeSpeciesNameCategoryFunctionRefs.
TemperatureV. volvaceaVvCAT1CatalaseLow-temperature stress defense response[179]
V. volvaceaMn-SODMn-superoxide dismutaseHeat, cold, and salt stress tolerance[180]
L. edodesLeMnP1Manganese peroxidaseRapid growth recovery under high-temperature stress[181]
L. edodesLeDnaJHeat shock protein 40Enhance the heat resistance of mycelium[54]
L. edodesYUCCA8Flavin-containing monooxygenasesEnhance the heat resistance of mycelium[56]
L. edodesTrpBTrytophan synthaseDefense response to heat stress[57]
L. edodesLetrpEAnthranilate synthaseEnhance the heat resistance of mycelium[55]
F. filiformisFfGa1Heterotrimeric G protein α subunitsStrong tolerance to heat stress and maintenance of cell wall integrity[50]
LightC. militarisCmwc-1Blue-light receptorColony pigmentation and promote conidia production[182]
S. communeWc-1
Wc-2
Blue light complex transcription factorInhibit vegetative growth
Phototoxic protection
[183]
Isaria
cicadae
Icwc-1Blue light receptorRegulation of fruiting body, pigment, and enzyme synthesis[184]
F. filiformisFfWc1 FfWc2Blue light and its receptor white collar complexRegulatory morphogenesis[185]
G. frondosaGf.BMR1Transcription factorFruiting body development and pigment accumulation[186]
L. edodesabl-DAbnormal browning related to lightBrown film formation of mycelial tissue[187]
A. bisporusPabsPara-aminobenzoic acid synthaseEnhance UV tolerance[188]
Metal stressM. esculentaATX1Copper chaperonesRegulation of cadmium metabolism[189]
L. edodesLeAmyα amylaseEnhance cadmium tolerance[190]
Table 5. Genes affecting the biosynthesis of ganoderma acids.
Table 5. Genes affecting the biosynthesis of ganoderma acids.
GeneMode of Action and Change of GA ContentRefs.
Alternative oxidase (AOX)Silent AOX, ROS content ↑, GA content ↑[206]
Transcription factor (GCN4)Silent GCN4, ROS content ↑, GA content ↑[207]
AMP-activated protein kinase homolog (GlSnf1)Silent GlSnf1, ROS content ↑, GA content ↑[208]
Telomerase reverse transcriptase (GlTert)Silent GlTert, ROS content ↑, GA content ↓[209]
Calcium-permeable channel (Cch)Silent Cch, Ca2+ content ↓, GA content ↓[210]
Nitrate reductase (NR)Silent NR, GA content ↑[211]
Glutamine synthetase (GS)Silent GS, GA content ↓[212]
Note: ↑: increase; ↓: decrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, W.; Zou, G.; Bao, D.; Wu, Y. Current Advances in the Functional Genes of Edible and Medicinal Fungi: Research Techniques, Functional Analysis, and Prospects. J. Fungi 2024, 10, 311. https://doi.org/10.3390/jof10050311

AMA Style

Li W, Zou G, Bao D, Wu Y. Current Advances in the Functional Genes of Edible and Medicinal Fungi: Research Techniques, Functional Analysis, and Prospects. Journal of Fungi. 2024; 10(5):311. https://doi.org/10.3390/jof10050311

Chicago/Turabian Style

Li, Wenyun, Gen Zou, Dapeng Bao, and Yingying Wu. 2024. "Current Advances in the Functional Genes of Edible and Medicinal Fungi: Research Techniques, Functional Analysis, and Prospects" Journal of Fungi 10, no. 5: 311. https://doi.org/10.3390/jof10050311

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