Next Article in Journal
Increased NADPH Supply Enhances Glycolysis Metabolic Flux and L-methionine Production in Corynebacterium glutamicum
Next Article in Special Issue
Meat Substitute Development from Fungal Protein (Aspergillus oryzae)
Previous Article in Journal
Postharvest Microwave Drying of Basil (Ocimum basilicum L.): The Influence of Treatments on the Quality of Dried Products
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutritional Profile and Health Benefits of Ganoderma lucidum “Lingzhi, Reishi, or Mannentake” as Functional Foods: Current Scenario and Future Perspectives

College of Bioscience and Bioengineering, Jiangxi Agricultural University, 1101 Zhimin Road, Nanchang 330045, China
Foods 2022, 11(7), 1030; https://doi.org/10.3390/foods11071030
Submission received: 18 February 2022 / Revised: 21 March 2022 / Accepted: 26 March 2022 / Published: 1 April 2022
(This article belongs to the Special Issue Advances in Food Mycology)

Abstract

:
Ganoderma lucidum has a long history of medicinal uses in the Far East countries of more than 2000 years due to its healing properties. Recently, G. lucidum has come under scientific scrutiny to evaluate its content of bioactive components that affect human physiology, and has been exploited for potent components in the pharmacology, nutraceuticals, and cosmetics industries. For instance, evidence is accumulating on the potential of this mushroom species as a promising antiviral medicine for treating many viral diseases, such as dengue virus, enterovirus 71, and recently coronavirus disease of 2019 (COVID-19). Still, more research studies on the biotherapeutic components of G. lucidum are needed to ensure the safety and efficiency of G. lucidum and promote the development of commercial functional foods. This paper provides an extensive overview of the nutraceutical value of Ganoderma lucidum and the development of commercial functional food. Moreover, the geo-origin tracing strategies of this mushroom and its products are discussed, a highly important parameter to ensure product quality and safety. The discussed features will open new avenues and reveal more secrets to widely utilizing this mushroom in many industrial fields; i.e., pharmaceutical and nutritional ones, which will positively reflect the global economy.

Graphical Abstract

1. Introduction

1.1. What Does History Say about Ganoderma lucidum?

“Lingzhi is a miraculous king of herbs”—Chinese people (221–206 BC).
Historically, the Romans considered mushrooms in general as the food of their gods and only served them for great feasts, while the Greeks and the Vikings believed that eating mushrooms gave them strength and enthusiasm before the war. America’s indigenous people have often used mushrooms in age-old rituals (e.g., magical hallucinogens) to cross the body and mental barrier [1]. Considered as one of the main folk medicinal mushrooms, G. lucidum was used for many centuries and reported under several names in China (Lingzhi), Japan (Reishi), and Korea (Mannentake). According to bimillennial beliefs, G. lucidum can promote health and longevity, but it was also considered a combination of spiritual force and a source of immortality [2,3,4]. Moreover, the Japanese people have regarded this mushroom as a “10,000-year” mushroom [5,6,7].
Several researchers have pointed out the long history of traditional medicinal uses of mushrooms, especially G. lucidum, mostly in Far East countries, dating back more than 4000 years [7,8,9,10,11,12,13,14]. This type of mushroom has therapeutic characteristics with medical claims that can be attributed to a well-respected pharmacopeia from the Qin dynasty (221–206 BC) called Shen Nong Ben Cao Jing, or The Divine Farmer’s Materia Medica [13,15]. The ethnomedicinal uses of G. lucidum had reflections on culture, such as the artworks beginning in the Yuan Dynasty (1280–1368 AD) [7,13]. This was not limited to artworks, but the use of G. lucidum images extended to furniture, carvings, paintings, and even women’s accessories [2].
For a long time, G. lucidum has been used as a traditional medicine for treating neurasthenia, debility of prolonged illness, insomnia, anorexia, dizziness, chronic hepatitis, hypercholesterolemia, mushroom poisoning, coronary heart disease, hypertension, prevention of acute mountain sickness, “deficiency fatigue”, carcinoma, and bronchial cough in the elderly [16,17]. Studies on medicinal mushrooms began in Western science more than 30 years ago. These studies have continued until the present via a series of exciting discoveries related to the biological activities of Ganoderma lucidum, including antitumor and anti-inflammatory effects, as well as cytotoxicity to hepatoma cells [18,19].

1.2. Ganoderma lucidum through the Glasses of Botanists, Taxonomists, Economists, and Scientometric Analysis

1.2.1. Through Botanists’ Glasses

Morphologically, lucidum is a word derived from the Latin word lucidus, which means “shiny” or “brilliant”, and describes the varnished look of the mushroom’s surface. Overall, G. lucidum is a large, dark mushroom distinctively characterized by a glossy surface (including a red-varnished and kidney-shaped cap) and a woody texture (see Figure 1). The fresh mushroom is soft, corklike, flat, lacks gills on its underside, and releases its spores via fine pores. The pore color on its underside depends on the age of the mushroom, and maybe white or brown [6,20]. Chen [21] described the nature of G. lucidum’s growth on the bases and stumps of a wide variety of deciduous trees, such as oak, maple, elm, willow, sweetgum, magnolia, and locust, and less frequently found on coniferous trees (e.g., larix, ptea, pinus) in Europe, Asia, and North and South America, especially in temperate rather than subtropical regions.

1.2.2. Through Taxonomists’ Glasses

Ganoderma lucidum (Curtis) P.Karst. was first described by Curtis [22] based on material from England, and the description was sanctioned by Fries [23]. The first scientific record of G. lucidum from China was made by Teng [24] when he incorrectly identified a Lingzhi specimen as G. lucidum. Geographically, the G. lucidum sensu stricto (Curtis) Karst mushroom is native to Europe and some parts of China [25]. According to the Index Fungorum (2016) (http://www.indexfungorum.org, accessed on 16 February 2022), Ganoderma lucidum (Curt: Fr.) Karst. belongs to Basidiomycota (phylum), Polyporales (order), and Ganodermataceae (family), as classified by the taxonomist Nahata [5]:
  • Kingdom: Fungi
  • Division: Basidiomycota
  • Class: Agaricomycetes
  • Order: Polyporales
  • Family: Ganodermataceae
  • Genus: Ganoderma
  • Species: G. lucidum

1.2.3. Through Economists’ Glasses

Ganoderma-based products attract a great deal of interest in many countries within Europe and North America, although South Asia (Malaysia, Singapore, China, Japan, and Korea) are the principal producers/providers of these food products [26]. In the past, consumption of G. lucidum was restricted to the wealthy only, and therefore there was no need to expand its cultivation, and what was grown in the wild was sufficient. Recently, however, the consumption of this mushroom has increased through multiple societal groups as an effective alternative to modern medicine or alongside it, and this is what has called for the expansion of its cultivation [27,28]. With over 110,000 ton/year, China is the biggest producer and exporter of G. lucidum [29]. Therefore, G. lucidum-based products play a pivotal role in the Chinese economy as a source of foreign-exchange flow through increasing exports and as promising products at the food and medical levels.
Generally, the mushroom’s ingredients possess a wide variety of biological properties, including pharmaceutical, nutraceutical, and cosmetic, as shown in Figure 2 [8,30,31]. As such, regarding the G. lucidum mushroom, there are three types of products that are produced from it: nutraceuticals, pharmaceuticals, and cosmetics [31]. Different parts of G. lucidum are commercially available, including mycelia, spores, and fruit body [6], and are sold as many different products, including powders, dietary supplements, and herbal tea [6,13]. Table 1 illustrates some of the commercial cosmetic products produced from G. lucidum mushrooms worldwide. Nowadays, the number of Ganoderma-based products well known commercially is estimated at over 100 brands [32]. The world trade market value of G. lucidum and its derivative products has reached approximately USD 4 billion [33].

1.2.4. Scientometric Analysis

During the last decade, the G. lucidum mushroom has attracted multiple research fields, including biochemistry, genetics and molecular biology, agricultural and biological sciences, pharmacology, toxicology, pharmaceutics, and medicine. Figure 3 illustrates the increasing interest in multidisciplinary utilization of G. lucidum based on the number of research articles in the past 10 years.

1.3. Why Should Mushrooms, including Ganoderma lucidum, Be Considered Functional Foods?

1.3.1. How to Define Functional Food?

In the early 1980s, the idiom “functional food” first appeared in Japan. Functional food is a broad term that includes several concepts [36]; for example, the definition of functional food provided by the Food and Agriculture Organization (FAO) states that “the functional food is the source that provides the human body with the necessary quantities of nutrients, i.e., proteins, carbohydrates, fats, vitamins, minerals, and others to keep it healthy. In addition, functional food can be cooked or prepared using ‘artificial intelligence technology’ [37]. In addition, the European Food Safety Authority (EFSA) defined functional food as “a food, which beneficially affects one or more target functions in the body, beyond adequate nutritional effects, in a way that is relevant to either an improved state of health and well-being and/or reduction of risk of disease” [38]. As described by the Functional Food Center (FFC) in the United States, functional foods are “real or processed foods that contain known or unknown biologically active compounds that, efficient, in defined and non-toxic quantities, recorded health benefit or provide a scientifically validated using unique biomarkers for the prevention, treatment or control of chronic disease or its symbiotic diseases” [39]. According to the definition of the Institute of Food Technologists (IFT), functional foods are those with ingredients that have health benefits in addition to basic nutrition, which is similar to the definition published by the International Life Sciences Institute (ILSI) [40,41].
Comprehensively, functional food can be defined as “a whole ingredient or a part of food that is used as food. It is part of a standard diet and is consumed on a regular basis, in normal quantities. It has proven health benefits that reduce the risk of specific chronic diseases or beneficially affect target functions beyond its basic nutritional functions” [42,43].

1.3.2. What Do the Definitions of Functional Foods Conclude?

The use of the term “ingredient” means that functional food is not only conventional food but also could be a part of other food or food ingredients. In addition, the above-mentioned definitions of functional foods allow for adaptation to cultural differences, including widely differing “standards” among cultures and countries. Moreover, the use of the term “health benefits” is not restrictive. It refers to physiological, psychological, and biological advantages [43,44].

1.3.3. Functional Foods and Their Relation with Gut Health

Among the important health effects of foods including the functional ones are those associated with gut health, a major determinant of an individual’s overall health. Several diseases are related in this context; e.g., gluten-therapy-resistant celiac, Crohn’s disease, ulcerative colitis, and irritable bowel syndrome. These adverse effects are caused by overgrowth and imbalance of intestinal bacteria linked to an individual’s food system [45]. The question that comes to mind is, what are the roles of the human gut in the body? These can be summarized as follows [45]:
  • It converts food to nutrients;
  • The human gut, via epithelial cell walls, assists in the absorption process of nutrients into the blood;
  • The human gut inhibits toxic and strange particles from entering the bloodstream.
Consequently, and directly, any gut malfunction has adverse effects on human health. In this regard, functional foods, including pre- and probiotics, have become increasingly important due to their positive role in human gut health.

1.3.4. Ganoderma lucidum as a Functional Food: How?

Historically, mushrooms, including Ganoderma lucidum, were traditionally consumed due to their nutritional and culinary values, and for their medical benefits when used in folk medicine. This historical heritage has recently been translated through molecular research to explore the present bioactive components and unlock mushrooms’ nutrition and therapeutic values [46,47]. Among these health benefits, mushrooms could help prevent diseases; e.g., hypertension, diabetes, hypercholesterolemia, and cancer, as mentioned in many reports. Hence, mushrooms can be considered a curative food [8,48]. Mushrooms are still untapped sources of bioactive substances such as glycoproteins, polysaccharides, mainly β-glucans, and secondary metabolites; i.e., nucleotide analogs, metal-chelating agents, terpenoids, polyphenols, alkaloids, lactones, and sterols. These biologically active components possess several therapeutic implications, such as antiviral, anticancer, hepatoprotective, immunopotentiating, and hypocholesterolemic agents [47,48,49,50,51].
The present paper critically discusses the benefits of G. lucidum, from nutritional value to medicinal impacts, and sheds light on its potential as a source of nutraceuticals and functional food. Moreover, this review provides answers with a critical vision to many questions, such as why the bioactive compounds of G. lucidum need to be further studied in vitro and in vivo, and what secrets are still behind them. Is it important to ensure G. lucidum’s quality and safety, as well as the best method to achieve that? With the potential of G. lucidum, will the future carry us to the possibility of commercial widescale use of G. lucidum and its products as new functional foods and medicines?
Despite that Ganoderma lucidum is not edible in its raw state due to its higher content of bitter compounds, its palatability can be increased by turning it into manufactured products such as powders, supplements, and tea [52]. The nutritional value of G. lucidum will be tackled in-depth in the following section.

2. The Nutritional Profile of Ganoderma lucidum

“Medicines and food have a common origin”—Kaul [53].
For thousands of years, mushrooms have been valued throughout the world as food and medicine [8]. Nevertheless, mushrooms are still largely untapped resources in producing effective pharmaceutical products, nutrients, and cosmetics. Indeed, only approximately 150,224 species have been described [54] out of the estimated 2.2–3.8 million fungal species worldwide [55]. About 3000 species that belong to Macrofungi are safe for human consumption, such as edible mushrooms [56].
From the nutritionist’s point of view, generally, fresh mushrooms contain both soluble and insoluble fibers; the soluble fiber is mainly β-glucanpolysaccharides and chitosans [57]. However, a question comes to mind: does G. lucidum that is grown naturally or wild differ from that grown artificially in its nutritional components? According to the research in this regard, the answer is yes, as it was found that the quantities of crude protein, carbohydrates, and crude fiber were greater in the artificially grown variety [58]. Few studies have revealed the nutritional profile of G. lucidum. Roy and others [59] reported the nutritional value and mineral composition of G. lucidum. Through an analytical view of the nutritional profile of the G. lucidum mushroom (Table 2), several important conclusions can be reached.
  • G. lucidum contains a considerable amount of water-soluble proteins (19.5 g/100 g mushroom (w/w)). Moreover, 18 kinds of amino acids have been found in G. lucidum, and the most abundant amino acid was leucine, which possessed strong hypoglycemic and antioxidant activities [66,67].
  • G. lucidum contains 3.5 g of dietary fiber per 100 g of mushroom (d/w).
  • G. lucidum contains significant amounts of major minerals (e.g., phosphorus, sulfur) and other trace mineral contents; i.e., Cu, Mg, and Fe.
  • As also mentioned in Table 2, G. lucidum is a highly rich source of vitamins such as riboflavin, niacin, thiamin, etc. Additionally, Ahmad [68] reported that several vitamins have been found in G. lucidum, such as vitamins B1, B2, B6, β-carotene, C, D, and E.
  • Based on the nutritional profile of G. lucidum, this mushroom possesses a high nutrient potential that reflects positively on its health benefits.
Through this vision, the G. lucidum mushroom is increasingly becoming one of the natural and untapped medicine resources, which should be of interest to pharmaceutical, nutraceutical, and cosmetics manufacturers and consumers worldwide [69]. Ganoderma lucidum contains myriad biologically active compounds (over 400 compounds), including polysaccharides, triterpenoids, steroids, fatty acids, amino acids, nucleosides, proteins, and alkaloids [70]. Still, how do these bioactive compounds reflect their medical properties? The following section will discuss the therapeutic impacts of these bioactive compounds.

2.1. Ganoderma lucidum Is a Factory of Biologically Active Useful Compounds

“Mushroom of immortality & symbol of traditional Chinese medicine”—Chen et al. [71].
The biologically active molecules of G. lucidum rely on their chemical composition, with polysaccharides, peptidoglycans, and triterpenes being the three major bioactive compounds [58,68,70,71,72,73]. Additionally, this mushroom contains other constituents with distinct biological functions, such as minerals (e.g., germanium), proteins, lectins, crude fibers, phenols, enzymes, sterols, and long-chain fatty acids [6,74,75,76,77]. Table 3 shows the major bioactive compounds and their biological effects. Snapshots of these bioactive compounds could be found as follows.

2.2. Polysaccharides and Peptidoglycans

Polysaccharides, such as ganoderans, represent diverse biological macromolecules with a broad range of biological properties [58]. Additionally, G. lucidum is a source of polysaccharides, glycopeptides, and polysaccharide crude extracts, as indicated by several studies [86]. In addition, these components of G. lucidum mushroom showed strong biological activities, including, for example, antioxidant, anti-tumor, and antibacterial activities due to its content of sugars, glycoproteins, and polysaccharide extracts obtained from the fruiting bodies [81,87,88,89]. Anti-inflammatory, hypoglycemic, antitumorigenic, and immunostimulating activities are among the multiple biological roles of polysaccharides extracted from G. lucidum [90,91,92,93,94,95]. Free radical scavenging abilities, reducing power, and chelating on ferrous ions are among the reported antioxidant properties [96,97]. Ospina et al. [98] reported that the isolated chitosan from G. lucidum has promising and desirable characteristics in specialized sectors such as biomedicine, pharmaceutics, and cosmetics, beyond the food industry. Regarding the peptidoglycans, G. lucidum contains a proteoglucan (GLPG) that has antiviral activity [99].

2.3. Triterpenes

Several triterpenes extracted from G. lucidum have been reported (around 100 types of triterpenes), with half of these types being novel and unique to G. lucidum [18]. Ganoderic and lucidenic acids are the major triterpenes produced by G. lucidum, while other triterpenes have been identified; e.g., ganodermic, ganoderiols, and ganoderal acids [58,100,101,102,103,104,105,106,107].

2.4. Other Bioactive Compounds

2.4.1. Germanium

The element germanium has brought some attention to G. lucidum. Germanium is one of the most prevalent elements in wild G. lucidum. With 489 μg/g, germanium occupied the fifth-highest rank among the other detected minerals in terms of concentration [108]. This element possesses significant biological activities; i.e., antimutagenic, antitumor, immune-potentiating, and antioxidant [109]. There is no rigorous proof linking germanium with the specific health benefits of G. lucidum.

2.4.2. Proteins

Some bioactive proteins purified from G. lucidum have been found to contribute to the medicinal properties of this mushroom; for example:
  • LZ-8, an immunosuppressive protein [110];
  • GLP, which possesses both antioxidant and hepatoprotective activities [111,112];
  • Ganodermin, an antifungal protein [113].
Many other bioactive compounds have been isolated from G. lucidum, including:
  • Enzymes; e.g., a metalloprotease that delays clotting time [6].

3. Ganoderma lucidum as a Functional Food

For several hundred years, G. lucidum has been used to promote human health as a functional food through traditional treatment strategies. Nowadays, many published studies have established the multiple health benefits of G. lucidum in preventing or fighting multiple gastrointestinal and extraintestinal diseases, from constipation and gastritis, to anorexia, arthritis, asthma, bronchitis, and diabetes [35,75,95]. Additional studies have reported on the anticancer [6,31,52,114,115], preventing cardiovascular disease, and tumorigenesis [116,117,118,119], antioxidant [6,120,121], cardioprotective [122], antidiabetic potency [6,123,124], and antimicrobial activity [6,35,125] of this mushroom. Altogether, Figure 4 demonstrates the nutritional and health benefits of G. lucidum, which will be spotlighted individually as follows.

3.1. Antimicrobial Activity

G. lucidum has been reported as a promising source of antimicrobial molecules (mainly polysaccharides) against various viral, bacterial, and fungal pathogens [79,83,125,126,127,128,129,130]. Table 4 summarizes the antimicrobial activities of the G. lucidum mushroom and its products.

3.2. Antiviral Potential

There have been few scientific studies (particularly on animals) that examined the antiviral effects of G. lucidum (Lingzhi); however, Zhu et al. [149] examined the anti-influenza effects of a hot water extract of Lingzhi on infected mice through intranasal and oral administration. The authors of this study concluded that short-term oral consumption of Lingzhi hot water extract had a limited effect in fighting influenza. Therefore, the authors recommended further study on the long-term anti-influenza effects that could improve the functional uses of this mushroom against influenza.

3.2.1. Ganoderma lucidum against Enterovirus 71 (EV71)

Since 1969, “the same year in which the infection of human enterovirus 71 (EV71) infection was identified for the first time”, the infection mechanism has not been fully understood [150]. However, this viral infection was associated with several clinical diseases, ranging from neurological disorders to hand–foot–mouth disease (HFMD), and is considered a serious threat to children under six years old [151]. Currently, there are no certified prophylactic or therapeutic treatments for EV71 infection [152,153]. Outbreaks of EV71 infection have been periodically reported worldwide [154,155,156]. For instance, China has recently seen increased deaths linked to EV71 infection and HFMD among the young population [131,138,157,158]. As mentioned above, there are no approved drugs for preventing or treating EV71 infection, but currently, antiviral drugs with a broad spectrum (e.g., acyclovir, ganciclovir, and ribavirin) are used to partially relieve infection symptoms, although they have high cytotoxic side effects [159]. Therefore, investigation of novel and efficient medicines is urgently needed to control this severe viral infection. The adoption of natural medicinal compounds and Chinese herbal medicines has been observed across Asian countries for centuries, and recently in Western medicine [160,161]. G. lucidum is widely used as a folk medicine for a variety of ailments [162]. Zhang et al. [79] suggested that Lanosta-7,9(11),24-trien-3-one,15;26-dihydroxy (GLTA), and ganoderic acid Y (GLTB), which are triterpenoid compounds of G. lucidum, could prevent EV71 infection by interfering with the viral particle and limiting the viral adsorption to the host cells. Additionally, the interaction dynamics of GTLA and GLTA with the EV71 virion, predicted by molecular docking, showed potent molecular binding to the viral capsid protein at a hydrophobic pocket (F site), and hence a block uncoating of EV71 (Figure 5). Furthermore, it has been shown that GLTA and GLTB notably prohibited the viral RNA (vRNA) replication of EV71 by blocking EV71 uncoating. Therefore, both GLTA and GLTB may represent two promising curative agents to control and treat EV71 infection.

3.2.2. Ganoderma lucidum against Dengue Virus (DENV)

The dengue virus (DENV), classified within the Flaviviridae family, is a fatal microbe transmitted to humans through mosquitoes (Aedes albopictus and Aedes aegypti) [163,164,165], causing both hemorrhagic fever [166,167], and shock syndrome [168,169]. A total of five different serotypes of DENV have been reported to induce both dengue fever types while potentially causing fatal infections [170,171]. Proteome analysis revealed that the translated DENV polyprotein complex comprises three structural and seven nonstructural proteins [171,172]. Of particular interest, the cofactor NS2B is required to fully activate the viral NS3 protease (NS3pro) domain that encodes a serine protease (S7 family). The NS2B–NS3pro complex of the dengue virus has been recently identified as an ideal target for developing novel anti-DENV drugs [173,174,175]. As one of the bioactive compounds extracted from G. lucidum, triterpenoids have been proposed and tested as antiviral agents against different viral pathogens; e.g., the human immunodeficiency virus. Ganodermanontriol, as a potent bioactive triterpenoid, was suggested to inhibit the DENV NS3pro protein based on in vitro studies. Thus, ganodermanontriol could act as a drug against DENV infection [176].

3.2.3. Ganoderma lucidum against the 2019 Novel Coronavirus (SARS-CoV-2)

December 2019 marked in Wuhan (Hobby Province, China) the beginning of a mysterious pneumonia outbreak [177]. A month later, the infectious agent was revealed to be a new kind of coronavirus named SARS-CoV-2 (formerly 2019-nCOV) [178]. The World Health Organization (WHO) declared the pneumonia outbreak that appeared in Wuhan a major public health crisis on 11 February 2020 and gave it the official name of Coronavirus Disease-2019 (COVID-19) [179]. Multiple symptoms were reported in the COVID-19 patients, including cough, lung damage, fever, fatigue, muscle pain, diarrhea, myalgia, and respiratory symptoms [180,181]. As of 27 April 2021, 147,539,302 cases of SARS-CoV-2 infected pneumonia and 3,116,444 deaths had been reported in China and 223 other countries, areas, or territories, of which 103,503 cases were found in China [182]. Natural products are among the most important sources for modern medication industry technology, if not the most important, due to their advantages such as abundant clinical use, and their unique diversity of chemical structures and biological activities [183,184]. In this context, traditional Chinese medicine (TCM) is one of the gold mines rich in untapped natural resources [185,186] that can be employed to treat many diseases that represent a challenge for humankind, including COVID-19. The previous studies on SARS-CoV and its homology with SARS-CoV-2 may provide avenues to natural compounds that inhibit SARS-CoV-2 [187]. For instance, the helicase domain is being investigated as a possible drug target. Yu et al. [188] reported that scutellarein and myricetin potently prevented nsP13, a SARS-CoV helicase protein, in vitro by altering its ATPase activity. The RNA-dependent RNA polymerase is another potential target for developing antiviral compounds, being an essential enzyme for RNA synthesis. Indeed, dose-dependent inhibition of this SARS-CoV enzyme was reported for the extracts of G. lucidum (IC50:41.9 µg/mL), Coriolus versicolor (IC50:108.4 µg/mL), Sinomenium acutum (IC50:198.6 µg/mL), and Kang Du Bu Fei Tang (IC50:471.3 µg/mL) [189]. Therefore, G. lucidum could serve as a novel and promising source of bioactive natural compounds with anticoronavirus activity [187].

3.3. Antioxidant and Antiaging Activity

Multiple research studies reported a close relationship between the richness of G. lucidum in “phenolic compounds, triterpenes, polysaccharides, polysaccharide peptide” and its antioxidant biological activity [83,97,190,191,192]. Clinical nutritionists have demonstrated that consuming antioxidant-rich plant-based foods may protect from cancer and many other chronic diseases [193,194]; however, this causality is still not proven yet for the antioxidants of G. lucidum [195]. Hence, one of the research priorities for the G. lucidum mushroom is to conduct more studies to close the gap in the interplay between antioxidants and the host immune system [191].
The long-term presence of free radicals and reactive oxygen species (ROS) accelerates aging and numerous age-associated illnesses [13]. Therefore, studies on scavenging free radicals and ROS are particularly important in antiaging research. G. lucidum polysaccharides (GLPs) can inhibit ROS production in fibroblasts following UVB treatment [196].

3.4. Anticancer Activity

Cancer is still one of the most fatal diseases worldwide and poses a major clinical challenge despite the notable boom in early diagnostic techniques and evolution in its treatment techniques [197]. Hundreds of plant species have been investigated as sources for new therapeutics (chemopreventive or chemotherapeutic) [198]. In this regard, mushrooms; e.g., Ganoderma species, are rich sources of many biologically active components, including antitumoral agents [199,200]. For example, polysaccharides and triterpenes are two major groups of compounds extracted from G. lucidum that were reported to possess chemopreventive and/or tumoricidal activities [6,31,52,114,115,201,202,203]. In addition, the antitumor activity exhibited by G. lucidum is achieved via induction of programmed cell death, as reported by many studies [81,204,205]. Moreover, the isolated compounds from G. lucidum have been previously described as modulators of autophagy in numerous human tumor cell lines [206,207,208,209]. In the same context, a methanolic extract (extraction at room temperature) of G. lucidum fruiting bodies prevented the growth of a human gastric tumor cell line via a mechanism that involved cellular autophagy [209]. Still, it is unknown if the extract is an inducer of autophagy or an autophagic flux inhibitor. More recently, Reis et al. [210] demonstrated that a methanolic extract of G. lucidum caused autophagy induction, rather than reducing the autophagic flux in AGS cells.

3.5. Antidiabetic Activity

G. lucidum has been proved to possess compounds responsible for hypoglycemic effects, such as polysaccharides, proteoglycans, proteins, and triterpenoids [6,78,123,124]. For instance, Wang et al. [211] reported that consuming a G. lucidum spore powder (GLSP) induced a decrease in blood glucose levels by promoting glycogen synthesis and preventing gluconeogenesis.

3.6. Cardioprotective Effects

How does G. lucidum have cardioprotective impacts? Many studies have answered this question. Firstly, Sudheesh et al. [122] reported the presence of α-tocopherol in G. lucidum that protected the mitochondria, reducing cardiac toxicity and mitochondrial dysfunction. Additionally, Gao et al. [212] referred to the positive effects of ganopoly (G. lucidum polysaccharide extract) on coronary heart disease (CHD) patients. The same authors showed that a polysaccharide extract of G. lucidum induced decreased blood pressure and serum cholesterol levels.

3.7. Hepatoprotection

The GLPs and Ganoderma triterpenoids (GTs) can act on the immune system and effectively exhibit hepatoprotective effects and treat liver damage. The hepatoprotective effects of G. lucidum have been widely studied [213]. GLPs can protect hepatocyte injury by inhibiting lipid peroxidation, elevating antioxidant enzyme activity, and suppressing apoptosis and immune-inflammatory response [214]. GTs offered significant cytoprotection against the oxidative damage induced by tertbutyl hydrogen peroxide (t-BHP) in hepatocellular carcinoma cells by decreasing the level of malondialdehyde and increasing the contents of glutathione and superoxide dismutase (SOD) [215]. Analysis of histopathology and serum enzymes in mice revealed an important hepatoprotective function of an ethanol extract of G. lucidum (GLE). It was therefore assumed that GLE could improve alcohol-induced liver injury [216]. In addition, a G. lucidum mycelium-fermented liquid (GLFL) was reported to possess hepatoprotective properties in rats [217].

3.8. Anti-Inflammatory Effects

Inflammation is a normal physiological response to an infection or injury and is part of host defense and tissue healing [218]. GLPs can prevent inflammation, maintain intestinal homeostasis, and regulate the intestinal immunological barrier functions in mice [219]. The anti-inflammatory effect of GLPs plays an important role in the care of sensitive skin [213].

3.9. Prebiotic Potential

Prebiotics are defined as “a substrate that is selectively utilized by host microorganisms conferring a health benefit” [220,221]. Mushrooms are considered untapped sources of prebiotics such as fibers, oligosaccharides (major constituents of mushrooms), and polyphenols, which can boost the growth and metabolic activity of beneficial members of the gut microbiota. For example, nondigestible polysaccharides can prevent pathogen proliferation by improving the growth of probiotics in the gut [222]. During the last decade, the interplay between prebiotics and human gut microbiota and its implications in mitigating many diseases; e.g., cancer, diabetes, and obesity gained much focus and has emerged as one of the principal trending axes of food science and technology. Scientific evidence has accumulated on the critical role of gut microbiota dysbiosis in exacerbating inflammation in host tissues, from the intestinal environment to the brain. Likewise, critical data has established the gut microbiota’s regulatory role in energy metabolism, which may cause disturbances in the metabolism processes [223]. For instance, mushrooms are a rich source of prebiotics that may play a pivotal role in treating pneumonia and atherosclerosis, as well as in their antitumor activity [224]. In the same context, a study conducted on mice (C57BL/6) confirmed that the Mexican G. lucidum is a rich source of prebiotics that reduced blood cholesterol [225]. The same study attributed the ability of Mexican G. lucidum to lower the blood cholesterol level to the significant decrease in the lipid-generating gene expression (Hmgcr, Fasn, Srebp1c, Acaca), and Abcg5, Abcg8 as genes responsible for reverse cholesterol transport, simultaneous with an increase in Ldlr gene expression in the liver [225]. Another study showed the possibility that G. lucidum polysaccharide peptides (GLPP) may have a role in alleviating the disturbance in the metabolism of fats through the ability of these compounds to alter the composition of the gut microbiota, which in turn has a positive effect on controlling and reducing the disruption of fat metabolism, regulating genes involved in intestinal integrity, bile acid homeostasis, and extrauterine fat deposition (Figure 6). Thus, GLPP can be considered a potential functional food component for treating hyperlipidemia and gut microbiota dysbiosis [226].
Furthermore, to date, no extensive studies have examined the biological activities and functions of GLFL on the regulation of the gut microbiota and cardiovascular diseases (CVDs) [227].
Concerning this, it has been demonstrated that the gut microbiota could play an important role in host health through their influence on cardiovascular risk factors [228,229]. As was mentioned previously, the products associated with G. lucidum have positive effects on the gut microbiota, and thus these products can regulate the risk factors for cardiovascular disease in the intestine. Chang et al. [230] reported an altered gut microbiota composition in an obese mouse model treated with a water extract of G. lucidum mycelium. In addition, GLFL was shown to reduce plasma low-density lipoprotein (LDL-c) cholesterol, triglycerides, and total cholesterol, and increase high-density lipoprotein (HDL-c) cholesterol in mice [231]. Additionally, Wu et al. [227] reported that when GLFL was fed to humans, it profoundly altered the gut microbiota. In the post-feeding group, there was an evident difference in β diversity as compared to the case of the pre-feeding group; this suggested that GLFL had altered the composition of the gut microbiota. Furthermore, the same authors reported that GLFL could protect humans by stimulating the growth of probiotics (i.e., genus Lactobacillus (p < 0.05)) while inhibiting the growth of pathogens (i.e., genus Aggregatibacter and Campylobacter (p < 0.05)).

3.10. The Health Risks of Ganoderma lucidum and Its Products

Most research on G. lucidum and its products has reported positive clinical outcomes and potential therapeutic uses, while the innocuity and potential toxic effects in humans have been poorly investigated. For instance, G. lucidum extracts could cause toxicity in vitro [35]. Moreover, G. lucidum spore-powder treatment caused hepatotoxic effects, as reported by Wanmuang et al. [232] Although no adverse effects of consumption of G. lucidum on lactation were proven, it is not advised for pregnant or lactating women [233].

4. Future Trends

Thus far, many questions have arisen regarding why and how to expand and maximize the utilization of G. lucidum and its derivatives, and what to do about the new applications and innovative techniques used in this regard. These points will be discussed via three axes as follows.

4.1. Do the Beneficial Medical Properties of G. lucidum Need More Scientific Evidence?

Several publications have reported that G. lucidum may have diverse beneficial therapeutical characteristics via its myriad bioactive compounds, such as triterpenes, polysaccharides, and proteins; hence, G. lucidum and its products are still common as commercial products. Therefore, the efficacy and safety of the consumption of G. lucidum are still considered knowledge gaps that are poorly investigated. During the past three decades, in vitro/in vivo studies reported by Western researchers have shown biomedical benefits for G. lucidum, which helped promote this mushroom in the Western world. However, there still is an urgent need to fully understand the related biomechanisms, and thus unlock their biotherapeutic application. The isolation, purification, and identification of active compounds of G. lucidum should be carried out to decipher the bioactivity of these compounds within its nutraceutical and pharmaceutical products. This aspect is a big challenge when implementing the commercial standardization strategies of G. lucidum products [6,35].
More research is needed to re-examine and study the bioactive ingredients extracted from G. lucidum, and this will be beneficial for clinical applications due to the discrepancy in the research results for G. lucidum or its derivative products (e.g., GLFL, GLPP, and WEGL). For example, Wu et al. [231] reported that a GLFL was unsafe because it increased the number of opportunistic pathogens; e.g., Acinetobacter, and decreased probiotics; e.g., Lactococcus. These results were in contrast to what was obtained when they were applied to mice [226].
Continued genetic studies of G. lucidum will elucidate the biosynthesis of the therapeutically active compounds produced by this mushroom; e.g., the unique triterpenoid antitumor ganoderic acids (GAs). The clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR-CAS9) technology has positively identified active curative components in G. lucidum by constructing functional genes of GA biosynthesis in this mushroom, thus serving as a vital platform for metabolic engineering in G. lucidum. Therefore, the CRISPR-CAS9 technique can be a cornerstone in all biotechnological applications of G. lucidum, such as molecular breeding. Therefore, a complete understanding of the G. lucidum genome will pave the way for its future roles in medical and industrial applications [71,115,234].
Large-scale studies on G. lucidum mushrooms will be conducted with standard scientific methods in the near future.

4.2. Future of the G. lucidum Mushroom in the Food Industry

Nowadays, several Ganoderma lucidum-based products are available in nutraceutical form. Some of them are marketed as dietary supplements and are widely consumed in many countries such as the United States, where they are combined with many other ingredients; e.g., coffee and tea. Because there is no proper toolkit, the consistency of the quality of dietary supplements derived from G. lucidum is rarely evaluated. Additionally, G. lucidum could be considered a source of food preservatives [5,187,235].
To validate G. lucidum’s nutraceutical usage, more research on this mushroom is needed.

4.3. Is Tracing the Species and Geo-Origin of G. lucidum Essential?

Research conducted by Loyd and others [14] showed that manufactured G. lucidum-based products (e.g., dietary supplements), which are marketed as derived from G. lucidum, contain not only G. lucidum but also multiple Ganoderma species, are unfortunately sold for medicinal uses. Of course, not all Ganoderma species produce the same therapeutic compounds, the same quality, or the same quantities. This raises questions about the traceability and authenticity of mushroom species, and how important this is in the industry. Therefore, this question should be addressed in subsequent research focusing on G. lucidum and its products.
As mentioned by Qi et al. [236], the geographical-origin traceability of mushrooms and their products is critical to assure their quality and safety. Indeed, the nutritional and therapeutic properties of each mushroom species vary depending on the geo-origins [237,238,239,240]. Lu et al. [237] proved this fact through their research on samples of G. lucidum collected from different geographic regions, in which they found that the content of each G. lucidum sample of ganoderic acids A and B, polysaccharides, and triterpenoids varied according to their geographical origin (including the differences in cultivation and environmental conditions). Hence, the geo-origin traceability of G. lucidum will reinforce the value of this mushroom globally at all levels, whether industrial or economic.
Then, what is the best method that can be used for species and geo-traceability targets? El Sheikha and Hu [8] proposed the DNA barcoding approach as a new “cutting edge” technology to significantly enhance food traceability in general and in mushrooms, especially from the field to the table.

5. Infographic for Ganoderma lucidum: Current Scenario and Future Perspectives

Recently, research on G. lucidum and its products has achieved substantial progress and has become a focus of the attention of the scientific community in many fields. Many studies from different viewpoints elucidated the biological characteristics, chemical composition and active components, pharmacological effects and related mechanisms, and clinical applications based on G. lucidum. Furthermore, at the industrial level, G. lucidum has made some progress.
In the future, new chemical compositions and active components (as a promising functional food), cellular and molecular mechanisms of biological activities (e.g., prebiotic effects), rapid and confirmatory methods to identify effective ingredients, fermentation and cultivation techniques, double-blind large-scale clinical trials, and quality control monitoring of product will be the aims of G. lucidum research (see Figure 7).

6. Conclusions

Ganoderma lucidum (Lingzhi, Reishi, or Mannentake) is a promising source of prebiotics due to its abundance of several bioactive compounds that have nutritional and medicinal effects and are present in all parts of the fungus (fruit bodies, mycelium, and spores). Therefore, since ancient times, G. lucidum has been used traditionally in Chinese medicine to treat chronic diseases. In addition, Chinese tradition refers to G. lucidum as “the lucky fungus” for its power to alleviate conditions such as arthritis, insomnia, and chest tightness.
There has been an increased interest in G. lucidum as a dietary supplement containing Reishi, which is a widespread therapeutic agent worldwide. As for Western countries, the bioactive substances extracted from G. lucidum have been used in alternative medicine to support traditional medicine in treating severe diseases, including diabetes, hepatitis, and cancer. Nevertheless, continuing this trend requires more clinical trials, typically to confirm efficacy and safety. Soon, more studies will be conducted on this mushroom on a larger scale in terms of medicinal applications or the food industry. The geographical origin is considered one of the critical factors that greatly impact both the safety and quality of mushrooms. Therefore, the determination of geographical origin has become an essential requirement to provide consumers with safe and high-quality mushrooms, including G. lucidum. Although there are many challenges facing the production of nutraceuticals and functional foods from G. lucidum on a large scale, especially in light of the limited clinical trials in humans, there is a potential for innovation, development, and expansion of applications (e.g., in food and pharmaceutical applications) due to G. lucidum’s promising nutritional and health characteristics.

Funding

This research was supported by the Research Start-Up Fund of Jiangxi Agricultural University, China, with a grant to Aly El Sheikah (Fund No. 9232307245).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Chang, S.T. The world mushroom industry: Trends and technological development. Int. J. Med. Mushrooms 2006, 8, 297–314. [Google Scholar] [CrossRef]
  2. Wasser, S.P. Reishi (Ganoderma lucidum). In Encyclopedia of Dietary Supplements, 2nd ed.; Coates, P.M., Betz, J.M., Blackman, M.R., Cragg, G.M., Levine, M., Moss, J., White, J.D., Eds.; Informa Healthcare: London, UK, 2010; pp. 680–690. [Google Scholar]
  3. Zhao, X.-R.; Huo, X.-K.; Dong, P.-P.; Wang, C.; Huang, S.-S.; Zhang, B.-J.; Zhang, H.-L.; Deng, S.; Liu, K.-X.; Ma, X.-C. Inhibitory effects of highly oxygenated lanostane derivatives from the fungus Ganoderma lucidum on p-glycoprotein and α-glucosidase. J. Nat. Prod. 2015, 78, 1868–1876. [Google Scholar] [CrossRef]
  4. Money, N.P. Are mushrooms medicinal? Fungal Biol. 2016, 120, 449–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Nahata, A. Ganoderma lucidum: A potent medicinal mushroom with numerous health benefits. Pharm. Anal. Acta 2013, 4, e159. [Google Scholar] [CrossRef] [Green Version]
  6. Wachtel-Galor, S.; Yuen, J.; Buswell, J.A.; Benzie, I.F.F. Ganoderma lucidum (Lingzhi or Reishi). In Herbal Medicine: Biomolecular and Clinical Aspects, 2nd ed.; Benzie, I.F.F., Wachtel-Galor, S., Eds.; CRC Press, Taylor & Francis: Boca Raton, FL, USA, 2011. Available online: https://www.ncbi.nlm.nih.gov/books/NBK92757/ (accessed on 16 February 2022).
  7. Lindequist, U.; Niedermeyer, T.H.; Jülich, W.D. The pharmacological potential of mushrooms. Evid. Based Complement. Altern. Med. 2005, 2, 285–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. El Sheikha, A.F.; Hu, D.M. How to trace the geographic origin of mushrooms? Trends Food Sci. Technol. 2018, 78, 292–303. [Google Scholar] [CrossRef]
  9. Radwan, F.F.; Perez, J.M.; Haque, A. Apoptotic and immune restoration effects of ganoderic acids define a new prospective for complementary treatment of cancer. J. Clin. Cell Immunol. 2011, S3, 4. [Google Scholar] [CrossRef] [Green Version]
  10. 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]
  11. Lee, K.-H.; Morris-Natschke, S.L.; Yang, X.; Huang, R.; Zhou, T.; Wu, S.-F.; Shi, Q.; Itokawa, H. Recent progress of research on medicinal mushrooms, foods, and other herbal products used in traditional Chinese medicine. J. Tradit. Complement. Med. 2012, 2, 1–12. [Google Scholar] [CrossRef] [Green Version]
  12. Wu, G.-S.; Lu, J.-J.; Guo, J.-J.; Li, Y.-B.; Tan, W.; Dang, Y.-Y.; Zhong, Z.-F.; Xu, Z.-T.; Chen, X.-P.; Wang, Y.-T. Ganoderic acid DM, a natural triterpenoid, induces DNA damage, G1 cell cycle arrest and apoptosis in human breast cancer cells. Fitoterapia 2012, 83, 408–414. [Google Scholar] [CrossRef]
  13. Bishop, K.S.; Kao, C.H.; Xu, Y.; Glucina, M.P.; Paterson, R.R.; Ferguson, L.R. From 2000 years of Ganoderma lucidum to recent developments in nutraceuticals. Phytochemistry 2015, 114, 56–65. [Google Scholar] [CrossRef] [Green Version]
  14. Loyd, A.L.; Richter, B.S.; Jusino, M.A.; Truong, C.; Smith, M.E.; Blanchette, R.A.; Smith, J.A. Identifying the “mushroom of immortality”: Assessing the Ganoderma species composition in commercial Reishi products. Front. Microbiol. 2018, 9, 1557. [Google Scholar] [CrossRef] [PubMed]
  15. Anonymous. Shen Nong Materia Medica; People’s Hygiene Press: Beijing, China, 1955. (In Chinese) [Google Scholar]
  16. Jong, S.C.; Birmingham, J.M. Medicinal benefits of the mushroom Ganoderma. Adv. Appl. Microbiol. 1992, 37, 101–134, (Translated). [Google Scholar] [CrossRef]
  17. Zhou, S.; Gao, Y. The immunomodulating effects of Ganoderma lucidum (Curt.: Fr.) P. Karst. (Ling Zhi, reishi mushroom) (Aphyllophoromycetideae). Int. J. Med. Mushrooms 2002, 4, 1–11. [Google Scholar] [CrossRef]
  18. Sone, Y.; Okuda, R.; Wada, N.; Kishida, E.; Misaki, A. Structure and antitumor activities of the polysaccharide isolated from fruiting body and the growing culture of mycelium of Ganoderma lucidum. Agric. Biol. Chem. 1985, 49, 2641–2653. [Google Scholar] [CrossRef]
  19. Mizuno, T.; Wang, G.; Zhang, J.; Kawagishi, H.; Nishitoba, T.; Li, J. Reishi, Ganoderma lucidum and Ganoderma tsugae: Bioactive substances and medicinal effects. Food Rev. Int. 1995, 11, 151–166. [Google Scholar] [CrossRef]
  20. Arora, D. Mushroom Demystified: A Comprehensive Guide to the Fleshy Fungi, 2nd ed.; Ten Speed Press: Berekely, CA, USA, 1986. [Google Scholar]
  21. Chen, A.W. Cultivation of the medicinal mushroom Ganoderma lucidum (Curtis: Fr), P.karst. (Reishi) in North America. Int. J. Med. Mushrooms 1999, 1, 263–282. [Google Scholar] [CrossRef]
  22. Curtis, W. Flora Londinensis: Or Plates and Descriptions of Such Plants as Grow Wild in the Environs of London; Printed by the Author: London, UK, 1781. [Google Scholar]
  23. Fries, E.M. Systema Mycologicum, Sistens Fungorum Ordines, Genera et Species; Gryphiswaldiae, Sumtibus Ernesti Mauritti; The Horticultural Society of New York Inc.: New York, NY, USA, 1821; Volume 1. [Google Scholar]
  24. Teng, S.C. Notes on polyporaceae from China. Sinensia 1934, 5, 198–200. [Google Scholar]
  25. Cao, Y.; Wu, S.-H.; Dai, Y.-C. Species clarification of the prize medicinal Ganoderma mushroom “Lingzhi”. Fungal Divers. 2012, 56, 49–62. [Google Scholar] [CrossRef]
  26. Chang, S.-T.; Miles, P.G. Ganoderma lucidum—A leader of edible and medicinal mushrooms. In Mushrooms: Cultivation, Nutritional Value, Medicinal Effect, and Environmental Impact, 2nd ed.; Chang, S.-T., Miles, P.G., Eds.; CRC Press, Taylor & Francis: Boca Raton, FL, USA, 2004; pp. 357–372. [Google Scholar]
  27. Leung, P.C.; Xue, C.; Cheng, Y.C. A Comprehensive Guide to Chinese Medicine; Toh Tuck Link: World Scientific Publisher Co. Pte. Ltd.: Singapore, 2003. [Google Scholar]
  28. Zhao, S.; Ye, G.; Fu, G.; Cheng, J.-X.; Yang, B.B.; Peng, C. Ganoderma lucidum exerts anti-tumor effects on ovarian cancer cells and enhances their sensitivity to cisplatin. Int. J. Oncol. 2011, 38, 1319–1327. [Google Scholar] [CrossRef]
  29. Li, S.; Dong, C.; Wen, H.; Liu, X. Development of Ling-zhi industry in China—Emanated from the artificial cultivation in the Institute of Microbiology, Chinese Academy of Sciences (IMCAS). Mycology 2016, 7, 74–80. [Google Scholar] [CrossRef] [PubMed]
  30. Badalyan, S.M. The main groups of therapeutic compounds of medicinal mushrooms. Med. Mycol. 2001, 3, 16–23. [Google Scholar]
  31. Wu, Y.; Choi, M.-H.; Li, J.; Yang, H.; Shin, H.-J. Mushroom cosmetics: The present and future. Cosmetics 2016, 3, 22. [Google Scholar] [CrossRef]
  32. Lai, T.; Gao, Y.; Zhou, S.F. Global marketing of medicinal Ling Zhi mushroom Ganoderma lucidum (W.Curt.: Fr.)Lloyd (Aphyllophoromycetideae) products and safety concerns. Int. J. Med. Mushrooms 2004, 6, 189–194. [Google Scholar] [CrossRef]
  33. Perumal, K. Indigenous Technology on Organic Cultivation of Reishi; AMM Murugappa Chettiar Research Centre: Chennai, TN, India, 2009; pp. 1–12. [Google Scholar]
  34. Taofiq, O.; González-Paramás, A.M.; Martins, A.; Barreiro, M.F.; Ferreira, I.C.F.R. Mushrooms extracts and compounds in cosmetics, cosmeceuticals and nutricosmetics—A review. Ind. Crops Prod. 2016, 90, 38–48. [Google Scholar] [CrossRef] [Green Version]
  35. Hapuarachchi, K.K.; Elkhateeb, W.A.; Karunarathna, S.C.; Cheng, C.R.; Bandara, A.R.; Kakumyan, P.; Hyde, K.D.; Daba, G.M.; Wen, T.C. Current status of global Ganoderma cultivation, products, industry and market. Mycosphere 2018, 9, 1025–1052. [Google Scholar] [CrossRef]
  36. Shimizu, T. Health claims on functional foods: The Japanese regulations and an international comparison. Nutr. Res. Rev. 2003, 16, 241–252. [Google Scholar] [CrossRef] [Green Version]
  37. Food and Agriculture Organization (FAO). Authors Report on Functional Foods, Food Quality and Standards Service (AGNS). 2007. Available online: http://www.fao.org/ag/agn/agns/files/Functional_Foods_Report_Nov2007.pdf (accessed on 25 February 2010).
  38. Martirosyan, D.M.; Singharaj, B. Health Claims and Functional Food: The Future of Functional Foods under FDA and EFSA Regulation. In Functional Foods for Chronic Diseases; Food Science Publisher: Dallas, TX, USA, 2016; pp. 410–424. [Google Scholar]
  39. Martirosyan, D.; Pisarski, K. Bioactive Compounds: Their Role in Functional Food and Human Health, Classifications, and Definitions. In Bioactive Compounds and Cancer; Martirosyan, D., Zhou, J.-R., Eds.; Food Science Publisher: San Diego, CA, USA, 2017; pp. 238–277. [Google Scholar]
  40. MacAulay, J.; Petersen, B.; Shank, F. Functional Foods: Opportunities and Challenges; Institute of Food Technologists (IFT) Expert Report; Institute of Food Technologists: Chicago, IL, USA, 2005. [Google Scholar]
  41. Crowe, K.M.; Francis, C. Position of the academy of nutrition and dietetics: Functional foods. J. Acad. Nutr. Diet. 2013, 113, 1096–1103. [Google Scholar] [CrossRef]
  42. Link, R. What Are Functional Foods? All You Need to Know. 17 January 2020. Available online: https://www.healthline.com/nutrition/functional-foods?c=560339028350 (accessed on 16 February 2022).
  43. Arshad, M.S.; Khalid, W.; Ahmad, R.S.; Khan, M.K.; Ahmad, M.H.; Safdar, S.; Kousar, S.; Munir, H.; Shabbir, U.; Zafarullah, M.; et al. Functional Foods and Human Health: An Overview. In Functional Foods—Phytochemicals and Health Promoting Potential; Arshad, M.S., Ahmad, M.H., Eds.; IntechOpen Limited: London, UK, 2021; pp. 1–14. [Google Scholar] [CrossRef]
  44. Doyon, M. Functional foods: A conceptual definition. Br. Food J. 2008, 110, 1133–1149. [Google Scholar] [CrossRef]
  45. Cencic, A.; Chingwaru, W. The role of functional foods, nutraceuticals, and food supplements in intestinal health. Nutrients 2010, 2, 611–625. [Google Scholar] [CrossRef]
  46. Chang, S.-T. Overview of Mushroom Cultivation and Utilization as Functional Foods (Chapter 1). In Mushrooms as Functional Foods; Cheung, P.C.K., Ed.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2008; pp. 1–33. [Google Scholar]
  47. Cheung, P.C.K. Mushrooms as Functional Foods; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2008. [Google Scholar]
  48. Kumar, K. Role of edible mushrooms as functional foods—A review. South Asian J. Food Technol. Environ. 2015, 1, 211–218. [Google Scholar] [CrossRef]
  49. Cash, E.J. Mushrooms Need to Be Further Explored as Functional Foods, Say Researchers. Nutraingredients Newsletter, 20 October 2017. Available online: https://www.nutraingredients.com/Article/2017/10/20/Mushrooms-need-to-be-further-explored-as-functional-foods-say-researchers (accessed on 29 June 2021).
  50. Raghavendra, V.B.; Venkitasamy, C.; Pan, Z.; Nayak, C. Functional Foods from Mushroom. In Microbial Functional Foods and Nutraceuticals, 1st ed.; Gupta, V.K., Treichel, H., Shapaval, V., de Oliveira, L.A., Tuohy, M.G., Eds.; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2017; pp. 65–91. [Google Scholar]
  51. Reis, F.S.; Martins, A.; Vasconcelos, M.H.; Morales, P.; Ferreira, I.C.F.R. Functional foods based on extracts or compounds derived from mushrooms. Trends Food Sci. Technol. 2017, 66, 48–62. [Google Scholar] [CrossRef]
  52. Bryant, J.M.; Bouchard, M.; Haque, A. Anticancer activity of ganoderic acid DM: Current status and future perspective. J. Clin. Cell Immunol. 2017, 8, 535. [Google Scholar] [CrossRef] [PubMed]
  53. Kaul, T.N. Biology and Conservation of Mushrooms; Oxford and IBH Publishing Co. Pvt. Ltd.: New Delhi, India, 2001; pp. 117–145. [Google Scholar]
  54. Species Fungorum. Species FungorumInitiative. Coordinated by the Royal Botanic Gardens, Kew. 2020. Available online: http://www.speciesfungorum.org/Names/Names.asp (accessed on 16 February 2022).
  55. Hawksworth, D.L.; Lücking, R. Fungal Diversity Revisited: 2.2 to 3.8 Million Species. In The Fungal Kingdom; Heitman, J., Howlett, B.J., Crous, P.W., Stukenbrock, E.H., James, T.Y., Gow, N.A.R., Eds.; ASM Press: Washington, DC, USA, 2017; pp. 79–95. [Google Scholar] [CrossRef]
  56. Kamal, S.; Pandey, J.; Ghignone, S.; Varma, A. Mushroom biology and biotechnology an overview. In A Textbook of Molecular Biotechnology, 3rd ed.; Chauhan, A.K., Varma, A., Eds.; I. K. International Publishing House Pvt. Ltd.: New Delhi, India, 2009; pp. 573–628. [Google Scholar]
  57. Sadler, M. Nutritional properties of edible fungi. Nutr. Bull. 2003, 28, 305–308. [Google Scholar] [CrossRef]
  58. Zhou, X.; Lin, J.; Yin, Y.; Zhao, J.; Sun, X.; Tang, K. Ganodermataceae: Natural products and their related pharmacological functions. Am. J. Chin. Med. 2007, 35, 559–574. [Google Scholar] [CrossRef]
  59. Roy, D.N.; Azad, A.K.; Sultana, F.; Anisuzzaman, A.S.M.; Khondkar, P. Nutritional profile and mineral composition of two edible mushroom varieties consumed and cultivated in Bangladesh. J. Phytopharmacol. 2015, 4, 217–220. [Google Scholar] [CrossRef]
  60. Dietary Reference Intakes (DRIs). Dietary Reference Intakes of Nutrients-Based Reference Values. These Are Established by Nutrition Board of National Academy of Sciences; National Academy Press: Washington, DC, USA, 2004; Available online: http://www.nap.edu (accessed on 29 June 2021).
  61. Dietary Reference Intakes (DRIs). The Essential Guide to Nutrient Requirements; National Academy Press: Washington, DC, USA, 2006; Available online: https://www.nap.edu/read/11537/chapter/45 (accessed on 29 June 2021).
  62. Manzi, P.; Marconi, S.; Aguzzi, A.; Pizzoferrato, L. Commercial mushrooms: Nutritional quality and effect of cooking. Food Chem. 2004, 84, 201–206. [Google Scholar] [CrossRef]
  63. Casselbury, K. Recommended Daily Fat Intakes for Females. 2018. Available online: https://healthyeating.sfgate.com/recommended-daily-fat-intakes-females-6305.html (accessed on 29 June 2021).
  64. Duthie, G.G.; Susan, J.; Janet, A.; Kyle, M. Plant polyphenols in cancer and heart disease: Implications as nutritional antioxidants. Nutr. Res. Rev. 2000, 13, 79–106. [Google Scholar] [CrossRef] [Green Version]
  65. Rahman, M.A.; Al Masud, A.; Lira, N.Y.; Shakil, S. Proximate analysis, phtochemical screening and antioxidant activity of different strains of Ganoderma lucidum (Reishi Mushroom). Open J. Biol. Sci. 2020, 5, 24–27. [Google Scholar] [CrossRef]
  66. Zhang, H.; Jiang, H.; Zhang, X.; Yan, J. Amino acids from Ganoderma lucidum: Extraction optimization, composition analysis, hypoglycemic and antioxidant activities. Curr. Pharm. Anal. 2018, 14, 562–570. [Google Scholar] [CrossRef]
  67. Zhang, K.; Liu, Y.; Zhao, X.; Tang, Q.; Dernedde, J.; Zhang, J.; Fan, H. Anti-inflammatory properties of GLPss58, a sulfated polysaccharide from Ganoderma lucidum. Int. J. Biol. Macromol. 2018, 107, 486–493. [Google Scholar] [CrossRef] [PubMed]
  68. Ahmad, M.F. Ganoderma lucidum: A rational pharmacological approach to surmount cancer. J. Ethnopharmacol. 2020, 260, 113047. [Google Scholar] [CrossRef] [PubMed]
  69. Chang, S.T.; Wasser, S.P. The Cultivation and Environmental Impact of Mushrooms. Oxford Research Encyclopedia Environmental Science. 2017. Available online: https://oxfordre.com/environmentalscience/view/10.1093/acrefore/9780199389414.001.0001/acrefore-9780199389414-e-231 (accessed on 16 February 2022).
  70. Cör, D.; Knez, Z.; Hrnčič, M.K. Antitumour, antimicrobial, antioxidant and antiacetylcholinesterase effect of Ganoderma lucidum terpenoids and polysaccharides: A review. Molecules 2018, 23, 649. [Google Scholar] [CrossRef] [Green Version]
  71. Chen, S.; Xu, J.; Liu, C.; Zhu, Y.; Nelson, D.R.; Zhou, S.; Li, C.; Wang, L.; Guo, X.; Sun, Y.; et al. Genome sequence of the model medicinal mushroom Ganoderma lucidum. Nat. Commun. 2012, 3, 913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Martínez-Montemayor, M.M.; Ling, T.; Suárez-Arroyo, I.J.; Ortiz-Soto, G.; Santiago-Negrón, C.L.; Lacourt-Ventura, M.Y.; Valentín-Acevedo, A.; Lang, W.H.; Rivas, F. Identification of biologically active Ganoderma lucidum compounds and synthesis of improved derivatives that confer anti-cancer activities in vitro. Front. Pharmacol. 2019, 10, 115. [Google Scholar] [CrossRef] [Green Version]
  73. Sudheer, S.; Alzorqi, I.; Manickam, S.; Ali, A. Bioactive Compounds of the Wonder Medicinal Mushroom “Ganoderma lucidum”. In Bioactive Molecules in Food; Reference Series in Phytochemistry; Mérillon, J.M., Ramawat, K., Eds.; Springer: Cham, Switzerland, 2019; pp. 1863–1893. [Google Scholar] [CrossRef]
  74. Parepalli, Y.; Chavali, M.; Sami, R.; Khojah, E.; Elhakem, A.; El Askary, A.; Singh, M.; Sinha, S.; El-Chaghaby, G. Evaluation of some active nutrients, biological compounds and health benefits of reishi mushroom (Ganoderma lucidum) Int. J. Pharmacol. 2021, 17, 243–250. [Google Scholar] [CrossRef]
  75. El Mansy, S.M. Ganoderma: The mushroom of immortality. Microb. Biosyst. 2019, 4, 45–57. [Google Scholar]
  76. Zhang, Y.; Wang, D.; Chen, Y.; Liu, T.; Zhang, S.; Fan, H.; Liu, H.; Li, Y. Healthy function and high valued utilization of edible fungi. Food Sci. Hum. Wellness 2021, 10, 408–420. [Google Scholar] [CrossRef]
  77. Gong, P.; Wang, S.; Liu, M.; Chen, F.; Yang, W.; Chang, X.; Liu, N.; Zhao, Y.; Wang, J.; Chen, X. Extraction methods, chemical characterizations and biological activities of mushroom polysaccharides: A mini-review. Carbohydr. Res. 2020, 494, 108037. [Google Scholar] [CrossRef]
  78. Ma, H.T.; Hsieh, J.F.; Chen, S.T. Anti-diabetic effects of Ganoderma lucidum. Phytochemistry 2015, 114, 109–113. [Google Scholar] [CrossRef]
  79. Zhang, W.; Tao, J.; Yang, X.; Yang, Z.; Zhang, L.; Liu, H.; Wu, K.; Jianguo Wu, J. Antiviral effects of two Ganoderma lucidum triterpenoids against enterovirus 71 infection. Biochem. Biophys. Res. Commun. 2014, 449, 307–312. [Google Scholar] [CrossRef] [PubMed]
  80. Zhu, Q.; Bang, T.H.; Ohnuki, K.; Sawai, T.; Sawai, K.; Shimizu, K. Inhibition of neuraminidase by Ganoderma triterpenoids and implications for neuraminidase inhibitor design. Sci. Rep. 2015, 5, 13194. [Google Scholar] [CrossRef] [PubMed]
  81. Ferreira, I.C.F.R.; Heleno, S.A.; Reis, F.S.; Stojkovic, D.; Queiroz, M.J.R.P.; Vasconcelos, M.H.; Sokovic, M. Chemical features of Ganoderma polysaccharides with antioxidant, antitumor and antimicrobial activities. Phytochemistry 2015, 114, 38–55. [Google Scholar] [CrossRef] [Green Version]
  82. Chan, S.W.; Tomlinson, B.; Chan, P.; Lam, C.W.K. The beneficial effects of Ganoderma lucidum on cardiovascular and metabolic disease risk. Pharm. Biol. 2021, 59, 1161–1171. [Google Scholar] [CrossRef] [PubMed]
  83. Mehta, S. Studies on Genetic Variability and Bioactive Molecules Production by Ganoderma Species. Ph.D. Thesis, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, India, 2014. [Google Scholar]
  84. Lee, B.; Park, J.; Park, J.; Shin, H.-J.; Kwon, S.; Yeom, M.; Sur, B.; Kim, S.; Kim, M.; Lee, H.; et al. Cordyceps militaris improves neurite outgrowth in Neuro2A cells and reverses memory impairment in rats. Food Sci. Biotechnol. 2011, 20, 1599–1608. [Google Scholar] [CrossRef]
  85. Gao, P.; Hirano, T.; Chen, Z.; Yasuhara, T.; Nakata, Y.; Sugimoto, A. Isolation and identification of C-19 fatty acids with anti-tumor activity from the spores of Ganoderma lucidum (reishi mushroom). Fitoterapia 2012, 83, 490–499. [Google Scholar] [CrossRef] [PubMed]
  86. Nie, S.; Zhang, H.; Li, W.; Xie, M. Current development of polysaccharides from Ganoderma: Isolation, structure and bioactivities. Bioact. Carbohydr. Diet. Fibre 2013, 1, 10–20. [Google Scholar] [CrossRef]
  87. Jia, J.; Zhang, X.; Hu, Y.-S.; Wu, Y.; Wang, Q.-Z.; Li, N.-N.; Guo, Q.-C.; Dong, X.-C. Evaluation of in vivo antioxidant activities of Ganoderma lucidum polysaccharides in STZ-diabetic rats. Food Chem. 2009, 115, 32–36. [Google Scholar] [CrossRef]
  88. XiaoPing, C.; Yan, C.; Shuibing, L.; YouGou, C.; JianYun, L.; LanPing, L. Free radical scavenging of Ganoderma lucidum polysaccharides and its effect on antioxidant enzymes and immunity activities in cervical carcinoma rats. Carbohydr. Polym. 2009, 77, 389–393. [Google Scholar] [CrossRef]
  89. Shi, M.; Zhang, Z.; Yang, Y. Antioxidant and immunoregulatory activity of Ganoderma lucidum polysaccharide. Carbohydr. Polym. 2013, 95, 200–206. [Google Scholar] [CrossRef]
  90. Miyazaki, T.; Nishijima, M. Studies on fungal polysaccharides. XXVII. Structural examination of a water-soluble, antitumor polysaccharide of Ganoderma lucidum. Chem. Pharm. Bull. 1981, 29, 3611–3616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Hikino, H.; Konno, C.; Mirin, Y.; Hayashi, T. Isolation and hypoglycemic activity of ganoderans A and B, glycans of Ganoderma lucidum fruit bodies. Planta Med. 1985, 4, 339–340. [Google Scholar] [CrossRef] [PubMed]
  92. Tomoda, M.; Gonda, R.; Kasahara, Y.; Hikino, H. Glycan structures of ganoderans B and C, hypoglycemic glycans of Ganoderma lucidum fruit bodies. Phytochemistry 1986, 25, 2817–2820. [Google Scholar] [CrossRef]
  93. Bao, X.; Liu, C.; Fang, J.; Li, X. Structural and immunological studies of a major polysaccharide from spores of Ganoderma lucidum (Fr.) Karst. Carbohydr. Res. 2001, 332, 67–74. [Google Scholar] [CrossRef]
  94. Wachtel-Galor, S.; Buswell, J.A.; Tomlinson, B.; Benzie, I.F.F. Lingzhi polyphorous fungus. In Herbal and Traditional Medicine: Molecular Aspects of Health, 1st ed.; Wachtel-Galor, S., Ed.; Marcel Dekker Inc.: New York, NY, USA, 2004; pp. 179–228. [Google Scholar]
  95. Wang, P.-Y.; Zhu, X.-L.; Lin, Z.-B. Antitumor and immunomodulatory effects of polysaccharides from broken-spore of Ganoderma lucidum. Front. Pharmacol. 2012, 3, 135. [Google Scholar] [CrossRef] [Green Version]
  96. Liu, W.; Wang, H.; Pang, X.; Yao, W.; Gao, X. Characterization and antioxidant activity of two low-molecular-weight polysaccharides purified from the fruiting bodies of Ganoderma lucidum. Int. J. Biol. Macromol. 2010, 46, 451–457. [Google Scholar] [CrossRef]
  97. Kozarski, M.; Klaus, A.; Niksic, M.; Jakovljevic, D.; Helsper, J.P.F.G.; Van Griensven, L.J.L.D. Antioxidative and immunomodulating activities of polysaccharide extracts of the medicinal mushrooms Agaricus bisporus, Agaricus brasiliensis, Ganoderma lucidum and Phellinus linteus. Food Chem. 2011, 129, 1667–1675. [Google Scholar] [CrossRef]
  98. Ospina, N.M.; Alvarez, S.P.O.; Sierra, D.M.E.; Vahos, D.F.R.; Ocampo, P.A.Z.; Orozco, C.P.O. Isolation of chitosan from Ganoderma lucidum mushroom for biomedical applications. J. Mater. Sci. Mater. Med. 2015, 26, 135. [Google Scholar] [CrossRef]
  99. Ji, Z.; Tang, Q.; Zhang, J.; Yang, Y.; Jia, W.; Pan, Y. Immunomodulation of RAW264.7 macrophages by GLIS, a proteopolysaccharide from Ganoderma lucidum. J. Ethnopharmacol. 2007, 112, 445–450. [Google Scholar] [CrossRef]
  100. Nishitoba, T.; Sato, H.; Kasai, T.; Kawagishi, H.; Sakamura, S. New bitter C27 and C30 terpenoids from fungus Ganoderma lucidum (Reishi). Agric. Biol. Chem. 1984, 48, 2905–2907. [Google Scholar] [CrossRef]
  101. Sato, H.; Nishitoba, T.; Shirasu, S.; Oda, K.; Sakamura, S. Ganoderiol A and B, new triterpenoids from the fungus Ganoderma lucidum (Reishi). Agric. Biol. Chem. 1986, 50, 2887–2890. [Google Scholar] [CrossRef]
  102. Budavari, S. The Merck Index; Merck & Co., Inc.: Whitehouse Station, NJ, USA, 1989. [Google Scholar]
  103. Gonzalez, A.G.; Leon, F.; Rivera, A.; Munoz, C.M.; Bermejo, J. Lanostanoid triterpenes from Ganoderma lucidum. J. Nat. Prod. 1999, 62, 1700–1701. [Google Scholar] [CrossRef]
  104. Ma, J.; Ye, Q.; Hua, Y.; Zhang, D.; Cooper, R.; Chang, M.N.; Chang, J.Y.; Sun, H.H. New lanostanoids from the mushroom Ganoderma lucidum. J. Nat. Prod. 2002, 65, 72–75. [Google Scholar] [CrossRef] [PubMed]
  105. Akihisa, T.; Nakamura, Y.; Tagata, M.; Tokuda, H.; Yasukawa, K.; Uchiyama, E.; Suzuki, T.; Kimura, Y. Anti-inflammatory and anti-tumor-promoting effects of triterpene acids and sterols from the fungus Ganoderma lucidum. Chem. Biodivers. 2007, 4, 224–231. [Google Scholar] [CrossRef] [PubMed]
  106. Jiang, J.; Grieb, B.; Thyagarajan, A.; Sliva, D. Ganoderic acids suppress growth and invasive behavior of breast cancer cells by modulating AP-1 and NF-kappaB signaling. Int. J. Mol. Med. 2008, 21, 577–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Chen, Y.; Bicker, W.; Wu, J.; Xie, M.Y.; Lindner, W. Ganoderma species discrimination by dual-mode chromatographic fingerprinting: A study on stationary phase effects in hydrophilic interaction chromatography and reduction of sample misclassification rate by additional use of reversed-phase chromatography. J. Chromatogr. A 2010, 1217, 1255–1265. [Google Scholar] [CrossRef]
  108. Chiu, S.W.; Wang, Z.M.; Leung, T.M.; Moore, D. Nutritional value of Ganoderma extract and assessment of its genotoxicity and antigenotoxicity using comet assays of mouse lymphocytes. Food Chem. Toxicol. 2000, 38, 173–178. [Google Scholar] [CrossRef]
  109. Kolesnikova, O.P.; Tuzova, M.N.; Kozlov, V.A. Screening of immunoactive properties of alkanecarbonic acid derivatives and germanium-organic compounds in vivo. Immunologiya 1997, 10, 36–38. [Google Scholar]
  110. Van Der Hem, L.; Van Der Vliet, A.; Bocken, C.F.M.; Kino, K.; Hoitsma, A.J.; Tax, W.J.M. Lingzhi-8: Studies of a new immunomodulating agent. Transplantation 1995, 60, 438–443. [Google Scholar] [CrossRef]
  111. Sun, J.; He, H.; Xie, B.J. Novel antioxidant peptides from fermented mushroom Ganoderma lucidum. J. Agric. Food Chem. 2004, 52, 6646–6652. [Google Scholar] [CrossRef]
  112. Shi, Y.; Sun, J.; He, H.; Guo, H.; Zhang, S. Hepatoprotective effects of Ganoderma lucidum peptides against D-galactosamine-induced liver injury in mice. J. Ethnopharmacol. 2008, 117, 415–419. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, H.; Ng, T.B. Ganodermin, an antifungal protein from fruiting bodies of the medicinal mushroom Ganoderma lucidum. Peptides 2006, 27, 27–30. [Google Scholar] [CrossRef] [PubMed]
  114. Deepalakshmi, K.; Mirunalini, S. Therapeutic properties and current medical usage of medicinal mushroom: Ganoderma lucidum. Int. J. Pharm. Sci. Res. 2011, 2, 1922–1929. [Google Scholar] [CrossRef]
  115. Zhao, R.-L.; He, Y.-M. Network pharmacology analysis of the anti-cancer pharmacological mechanisms of Ganoderma lucidum extract with experimental support using Hepa1-6-bearing C57 BL/6 mice. J. Ethnopharmacol. 2018, 210, 287–295. [Google Scholar] [CrossRef]
  116. Liu, X.; Yuan, J.P.; Chung, C.K.; Chen, X.J. Antitumor activity of the sporoderm-broken germinating spores of Ganoderma lucidum. Cancer Lett. 2002, 182, 155–161. [Google Scholar] [CrossRef]
  117. Tang, W.; Gao, Y.; Chen, G.; Gao, H.; Dai, X.; Ye, J.; Chan, E.; Huang, M.; Zhou, S. A randomized, double-blind and placebo-controlled study of a Ganoderma lucidum polysaccharide extract in neurasthenia. J. Med. Food 2005, 8, 53–58. [Google Scholar] [CrossRef] [Green Version]
  118. Paterson, R.R. Ganoderma—A therapeutic fungal biofactory. Phytochemistry 2006, 67, 1985–2001. [Google Scholar] [CrossRef] [Green Version]
  119. Mir, M.A.; Sharma, T.; Sharma, K.; Anjum, S.; Mir, B.A. Anti-urolithiatic and anti-arthritis activity of various extracts of Ganoderma lucidum. Nat. Prod. Chem. Res. 2017, 5, 297. [Google Scholar] [CrossRef]
  120. Yen, G.C.; Wu, J.Y. Antioxidant and radical scavenging properties of extracts from Ganoderma tsugae. Food Chem. 1999, 65, 375–379. [Google Scholar] [CrossRef]
  121. Mau, J.L.; Lin, H.C.; Chen, C.C. Antioxidant properties of several medicinal mushrooms. J. Agric. Food Chem. 2002, 50, 6072–6077. [Google Scholar] [CrossRef]
  122. Sudheesh, N.A.; Ajith, T.A.; Janardhanan, K.K. Ganoderma lucidum ameliorate mitochondrial damage in isoproterenol-induced myocardial infarction in rats by enhancing the activities of TCA cycle enzymes and respiratory chain complexes. Int. J. Cardiol. 2013, 165, 117–125. [Google Scholar] [CrossRef] [PubMed]
  123. Gao, Y.; Lan, J.; Dai, X.; Ye, J.; Zhou, S. A phase I/II study of Lingzhi mushroom Ganoderma lucidum (W. Curt.: Fr.) Lloyd (Aphyllophoromycetideae) extract in patients with type II diabetes mellitus. Int. J. Med. Mushrooms 2004, 6, 33–40. [Google Scholar] [CrossRef]
  124. Teng, B.-S.; Wang, C.-D.; Yang, H.-J.; Wu, J.-S.; Zhang, D.; Zheng, M.; Fan, Z.-H.; Pan, D.; Zhou, P. A protein tyrosine phosphatase 1B activity inhibitor from the fruiting bodies of Ganoderma lucidum (Fr.) Karst and its hypoglycemic potency on streptozotocin-induced type 2 diabetic mice. J. Agric. Food Chem. 2011, 59, 6492–6500. [Google Scholar] [CrossRef] [PubMed]
  125. Basnet, B.B.; Liu, L.; Bao, L.; Liu, H. Current and future perspective on antimicrobial and anti-parasitic activities of Ganoderma sp.: An update. Mycology 2017, 8, 111–124. [Google Scholar] [CrossRef] [Green Version]
  126. Gao, Y.; Zhou, S.; Huang, M.; Xu, A. Antibacterial and antiviral value of the genus Ganoderma P. Karst. species (Aphyllophoromycetideae): A review. Int. J. Med. Mushrooms 2003, 5, 235–246. [Google Scholar] [CrossRef]
  127. Keypour, S.; Riahi, H.; Moradali, M.F.; Rafati, H. Investigation of the antibacterial activity of a chloroform extract of Lingzhi or Reishi medicinal mushroom, Ganoderma lucidum (W. Curt.: Fr.) P. Karst. (Aphyllophoromycetideae). Int. J. Med. Mushrooms 2008, 10, 345–349. [Google Scholar] [CrossRef]
  128. Jonathan, S.G.; Awotona, F.E. Studies on antimicrobial potentials of three Ganoderma species. Afr. J. Biomed. Res. 2010, 13, 133–139. [Google Scholar]
  129. Hernández-Márquez, E.; Lagunas-Martínez, A.; Bermudez-Morales, V.H.; Burgete-García, A.I.; León-Rivera, I.; Montiel-Arcos, E.; García-Villa, E.; Gariglio, P.; Madrid-Marina, V.V.; Ondarza-Vidaurreta, R.N. Inhibitory activity of Lingzhi or Reishi medicinal mushroom, Ganoderma lucidum (Higher Basidiomycetes) on transformed cells by Human Papillomavirus. Int. J. Med. Mushrooms 2014, 16, 179–187. [Google Scholar] [CrossRef]
  130. Shah, P.; Modi, H.A.; Shukla, M.D.; Lahiri, S.K. Preliminary phytochemical analysis and antibacterial activity of Ganoderma lucidum collected from Dang District of Gujarat, India. Int. J. Curr. Microbiol. Appl. Sci. 2014, 3, 246–255. [Google Scholar]
  131. Liu, D.Z.; Zhu, Y.Q.; Li, X.F.; Shan, W.G.; Gao, P.F. New triterpenoids from the fruiting bodies of Ganoderma lucidum and their bioactivities. Chem. Biodivers. 2014, 11, 982–986. [Google Scholar] [CrossRef]
  132. Shang, X.; Tan, Q.; Liu, R.; Yu, K.; Li, P.; Zhao, G.-P. In vitro anti-Helicobacter pylori effects of medicinal mushroom extracts, with special emphasis on the Lion’s Mane mushroom, Hericium erinaceus (higher Basidiomycetes). Int. J. Med. Mushrooms 2013, 15, 165–174. [Google Scholar] [CrossRef] [PubMed]
  133. Ćilerdžić, J.; Stajić, M.; Vukojević, J. Potential of submergedly cultivated mycelia of Ganoderma spp. as antioxidant and antimicrobial agents. Curr. Pharm. Biotechnol. 2016, 17, 275–282. [Google Scholar] [CrossRef] [PubMed]
  134. Karwa, A.; Rai, M. Naturally occurring medicinal mushroom-derived antimicrobials: A case-study using lingzhi or reishi Ganoderma lucidum (W. Curt.: Fr.) P. Karst. (Higher Basidiomycetes). Int. J. Med. Mushrooms 2012, 14, 481–490. [Google Scholar] [CrossRef] [PubMed]
  135. Ćilerdžić, J.; Vukojević, J.; Stajić, M.; Stanojković, T.; Glamočlija, J. Biological activity of Ganoderma lucidum basidiocarps cultivated on alternative and commercial substrate. J. Ethnopharmacol. 2014, 155, 312–319. [Google Scholar] [CrossRef] [PubMed]
  136. Yoon, S.Y.; Eo, S.K.; Kim, Y.S.; Lee, C.K.; Han, S.S. Antimicrobial activity of Ganoderma lucidum extract alone and in combination with some antibiotics. Arch. Pharm. Res. 1994, 17, 438–442. [Google Scholar] [CrossRef] [PubMed]
  137. Vazirian, M.; Faramarzi, M.A.; Ebrahimi, S.E.S.; Esfahani, H.R.M.; Samadi, N.; Hosseini, S.A.; Asghari, A.; Manayi, A.; Mousazadeh, A.; Asef, M.R.; et al. Antimicrobial effect of the Lingzhi or Reishi medicinal mushroom, Ganoderma lucidum (higher Basidiomycetes) and its main compounds. Int. J. Med. Mushrooms 2014, 16, 77–84. [Google Scholar] [CrossRef]
  138. Yang, F.; Zhang, T.; Hu, Y.; Wang, X.; Du, J.; Li, Y.; Sun, S.; Sun, X.; Li, Z.; Jin, Q. Survey of enterovirus infections from hand, foot and mouth disease outbreak in China, 2009. Virol. J. 2011, 8, 508. [Google Scholar] [CrossRef] [Green Version]
  139. Sa-Ard, P.; Sarnthima, R.; Khammuang, S.; Kanchanarach, W. Antioxidant, antibacterial and DNA protective activities of protein extracts from Ganoderma lucidum. J. Food Sci. Technol. 2015, 52, 2966–2973. [Google Scholar] [CrossRef] [Green Version]
  140. Heleno, S.A.; Ferreira, I.C.F.R.; Esteves, A.P.; Ćirić, A.; Glamočlija, J.; Martins, A.; Soković, M.; Queiroz, M.J.R.P. Antimicrobial and demelanizing activity of Ganoderma lucidum extract, p-hydroxybenzoic and cinnamic acids and their synthetic acetylated glucuronide methyl esters. Food Chem. Toxicol. 2013, 58, 95–100. [Google Scholar] [CrossRef]
  141. Sun, X.; Jin, X.; Pan, W.; Wang, J. Syntheses of new rare earth complexes with carboxymethylated polysaccharides and evaluation of their in vitro antifungal activities. Carbohydr. Polym. 2014, 113, 194–199. [Google Scholar] [CrossRef]
  142. El-Mekkawy, S.; Meselhy, M.R.; Nakamura, N.; Tezuka, Y.; Hattori, M.; Kakiuchi, N.; Shimotohno, K.; Kawahata, T.; Otake, T. Anti- HIV-1 and anti-HIV-1-protease substances from Ganoderma lucidum. Phytochemistry 1998, 49, 1651–1657. [Google Scholar] [CrossRef]
  143. Eo, S.K.; Kim, Y.S.; Lee, C.K.; Han, S.S. Possible mode of antiviral activity of acidic protein bound polysaccharide isolated from Ganoderma lucidum on herpes simplex viruses. J. Ethnopharmacol. 2000, 72, 475–481. [Google Scholar] [CrossRef]
  144. Donatini, B. Control of oral Human Papillomavirus (HPV) by medicinal mushrooms, trametes versicolor and Ganoderma lucidum: A preliminary clinical trial. Int. J. Med. Mushrooms 2014, 16, 497–498. [Google Scholar] [CrossRef] [PubMed]
  145. Shamaki, B.U.; Sandabe, U.K.; Ogbe, A.O.; Abdulrahman, F.I.; El-Yuguda, A.-D. Methanolic soluble fractions of lingzhi or reishi medicinal mushroom, Ganoderma lucidum (Higher basidiomycetes) Extract inhibit neuraminidase activity in newcastle disease virus (LaSota). Int. J. Med. Mushrooms 2014, 16, 579–583. [Google Scholar] [CrossRef] [PubMed]
  146. Iwatsuki, K.; Akihisa, T.; Tokuda, H.; Ukiya, M.; Oshikubo, M.; Kimura, Y.; Asano, T.; Nomura, A.; Nishino, H. Lucidenic acids P and Q, methyl lucidenate P, and other triterpenoids from the fungus Ganoderma lucidum and their inhibitory effects on epstein−barr virus activation. J. Nat. Prod. 2003, 66, 1582–1585. [Google Scholar] [CrossRef]
  147. Li, Y.; Yang, Y.; Fang, L.; Zhang, Z.; Jin, J.; Zhang, K. Antihepatitis activities in the broth of Ganoderma lucidum supplemented with a Chinese herbal medicine. Am. J. Chin. Med. 2006, 34, 341–349. [Google Scholar] [CrossRef] [Green Version]
  148. Li, Y.Q.; Wang, S.F. Anti-hepatitis B activities of ganoderic acid from Ganoderma lucidum. Biotechnol. Lett. 2006, 28, 837–841. [Google Scholar] [CrossRef]
  149. Zhu, Q.; Amen, Y.M.; Ohnuki, K.; Shimizu, K. Anti-influenza effects of Ganoderma lingzhi: An animal study. J. Funct. Foods 2017, 34, 224–228. [Google Scholar] [CrossRef]
  150. Blomberg, J.; Lycke, E.; Ahlfors, K.; Johnsson, T.; Wolontis, S.; von Zeipel, G. New enterovirus type associate with epidemic of aseptic meningitis and-or hand, foot, and mouth disease. Lancet 1974, 2, 112. [Google Scholar] [CrossRef]
  151. Zhang, D.; Lu, J.; Lu, J. Enterovirus 71 vaccine: Close but still far. Int. J. Infect. Dis. 2010, 14, e739–e743. [Google Scholar] [CrossRef] [Green Version]
  152. Shang, L.; Xu, M.; Yin, Z. Antiviral drug discovery for the treatment of enterovirus 71 infections. Antivir. Res. 2013, 97, 183–194. [Google Scholar] [CrossRef] [PubMed]
  153. Fowlkes, A.L.; Honarmand, S.; Glaser, C.; Yagi, S.; Schnurr, D.; Oberste, M.S.; Anderson, L.; Pallansch, M.A.; Khetsuriani, N. Enterovirus-associated encephalitis in the California encephalitis project, 1998–2005. J. Infect. Dis. 2008, 198, 1685–1691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Alexander, J.P., Jr.; Baden, L.; Pallansch, M.A.; Anderson, L.J. Enterovirus 71 infections and neurologic disease—United States, 1977–1991. J. Infect. Dis. 1994, 169, 905–908. [Google Scholar] [CrossRef] [PubMed]
  155. Chang, L.Y.; Huang, Y.C.; Lin, T.Y. Fulminant neurogenic pulmonary oedema with hand, foot, and mouth disease. Lancet 1998, 352, 367–368. [Google Scholar] [CrossRef] [PubMed]
  156. Huang, C.C.; Liu, C.C.; Chang, Y.C.; Chen, C.Y.; Wang, S.T.; Yeh, T.F. Neurologic complications in children with enterovirus 71 infection. N. Engl. J. Med. 1999, 341, 936–942. [Google Scholar] [CrossRef] [PubMed]
  157. Yang, F.; Ren, L.; Xiong, Z.; Li, J.; Xiao, Y.; Zhao, R.; He, Y.; Bu, G.; Zhou, S.; Wang, J.; et al. Enterovirus 71 outbreak in the People’s Republic of China in 2008. J. Clin. Microbiol. 2009, 47, 2351–2352. [Google Scholar] [CrossRef] [Green Version]
  158. Liu, M.Y.; Liu, W.; Luo, J.; Liu, Y.; Zhu, Y.; Berman, H.; Wu, J. Characterization of an outbreak of hand, foot, and mouth disease in Nanchang, China in 2010. PLoS ONE 2011, 6, e25287. [Google Scholar] [CrossRef] [Green Version]
  159. Kok, C.C. Therapeutic and prevention strategies against human enterovirus 71 infection. World J. Virol. 2015, 4, 78–95. [Google Scholar] [CrossRef]
  160. Zhu, Y.P.; Woerdenbag, H.J. Traditional Chinese herbal medicine. Pharm. World Sci. 1995, 17, 103–112. [Google Scholar] [CrossRef]
  161. Li, T.; Peng, T. Traditional Chinese herbal medicine as a source of molecules with antiviral activity. Antivir. Res. 2013, 97, 1–9. [Google Scholar] [CrossRef]
  162. Ma, B.; Ren, W.; Zhou, Y.; Ma, J.; Ruan, Y.; Wen, C.N. Triterpenoids from the spores of Ganoderma lucidum. N. Am. J. Med. Sci. 2011, 3, 495–498. [Google Scholar] [CrossRef] [PubMed]
  163. Martins, V.E.P.; Alencar, C.H.; Kamimura, M.T.; de Carvalho Araújo, F.M.; De Simone, S.G.; Dutra, R.F.; Guedes, M.I.F. Occurrence of natural vertical transmission of dengue-2 and dengue-3 viruses in Aedes aegypti and Aedes albopictus in Fortaleza, Ceará, Brazil. PLoS ONE 2012, 7, e41386. [Google Scholar] [CrossRef]
  164. Simmons, C.P.; Farrar, J.J.; van Vinh Chau, N.; Wills, B. Dengue. N. Engl. J. Med. 2012, 366, 1423–1432. [Google Scholar] [CrossRef] [PubMed]
  165. Akiner, M.M.; Demirci, B.; Babuadze, G.; Robert, V.; Schaffner, F. Spread of the invasive mosquitoes Aedes aegypti and Aedes albopictus in the Black Sea region increases risk of chikungunya, dengue, and Zika outbreaks in Europe. PLoS Negl. Trop. Dis. 2016, 10, e0004664. [Google Scholar] [CrossRef] [Green Version]
  166. Taguchi, Y. Principal components analysis based unsupervised feature extraction applied to gene expression analysis of blood from dengue haemorrhagic fever patients. Sci. Rep. 2017, 7, 44016. [Google Scholar] [CrossRef] [Green Version]
  167. Tang, T.H.-C.; Alonso, S.; Ng, L.F.-P.; Thein, T.-L.; Pang, V.J.-X.; Leo, Y.-S.; Lye, D.C.-B.; Yeob, T.-W. Increased serum hyaluronic acid and heparan sulfate in dengue fever: Association with plasma leakage and disease severity. Sci. Rep. 2017, 7, 46191. [Google Scholar] [CrossRef] [Green Version]
  168. Le Duyen, H.T.; Cerny, D.; Trung, D.T.; Pang, J.; Velumani, S.; Toh, Y.X.; Qui, P.T.; Hao, N.V.; Simmons, C.; Haniffa, M.; et al. Skin dendritic cell and T cell activation associated with dengue shock syndrome. Sci. Rep. 2017, 7, 14224. [Google Scholar] [CrossRef] [Green Version]
  169. Oliveira, M.; Lert-Itthiporn, W.; Cavadas, B.; Fernandes, V.; Chuansumrit, A.; Anunciação, O.; Casademont, I.; Koeth, F.; Penova, M.; Tangnararatchakit, K.; et al. Joint ancestry and association test indicate two distinct pathogenic pathways involved in classical dengue fever and dengue shock syndrome. PLoS Negl. Trop. Dis. 2018, 12, e0006202. [Google Scholar] [CrossRef]
  170. Mustafa, M.; Rasotgi, V.; Jain, S.; Gupta, V. Discovery of fifth serotype of dengue virus (DENV-5): A new public health dilemma in dengue control. Med. J. Armed Forces India 2015, 71, 67–70. [Google Scholar] [CrossRef] [Green Version]
  171. Dwivedi, V.D.; Tripathi, I.P.; Tripathi, R.C.; Bharadwaj, S.; Mishra, S.K. Genomics, proteomics and evolution of dengue virus. Brief Funct. Genom. 2017, 16, 217–227. [Google Scholar] [CrossRef]
  172. Mukhopadhyay, S.; Kuhn, R.J.; Rossmann, M.G. A structural perspective of the flavivirus life cycle. Nat. Rev. Microbiol. 2005, 3, 13–22. [Google Scholar] [CrossRef] [PubMed]
  173. Luo, D.; Vasudevan, S.G.; Lescar, J. The flavivirus NS2B–NS3 protease–helicase as a target for antiviral drug development. Antivir. Res. 2015, 118, 148–158. [Google Scholar] [CrossRef]
  174. Constant, D.A.; Mateo, R.; Nagamine, C.M.; Kirkegaard, K. Targeting intramolecular proteinase NS2B/3 cleavages for transdominant inhibition of dengue virus. Proc. Natl. Acad. Sci. USA 2018, 115, 10136–11014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Gibbs, A.C.; Steele, R.; Liu, G.; Tounge, B.A.; Montelione, G.T. Inhibitor bound dengue NS2B-NS3pro reveals multiple dynamic binding modes. Biochemistry 2018, 57, 1591–1602. [Google Scholar] [CrossRef] [PubMed]
  176. Bharadwaj, S.; Lee, K.E.; Dwivedi, V.D.; Yadava, U.; Panwar, A.; Lucas, S.J.; Pandey, A.; Kang, S.G. Discovery of Ganoderma lucidum triterpenoids as potential inhibitors against Dengue virus NS2B-NS3 protease. Sci. Rep. 2019, 9, 19059. [Google Scholar] [CrossRef] [Green Version]
  177. Gralinski, L.E.; Menachery, V.D. Return of the coronavirus: 2019-nCoV. Viruses 2020, 12, 135. [Google Scholar] [CrossRef] [Green Version]
  178. Burki, T.K. Coronavirus in China. Lancet Respir. Med. 2020, 8, P238. [Google Scholar] [CrossRef]
  179. World Health Organization (WHO). Director-General’s Remarks at the Media Briefing on 2019-nCoV on 11 February 2020. 2020. Available online: https://www.who.int/dg/speeches/detail/who-director-general-s-remarks-at-the-media-briefing-on-2019-ncov-on-11-february-2020 (accessed on 29 June 2021).
  180. Guan, W.-J.; Ni, Z.-Y.; Hu, Y.; Liang, W.-H.; Ou, C.-Q.; He, J.-X.; Liu, L.; Shan, H.; Lei, C.-L.; Hui, D.S.C.; et al. Clinical characteristics of 2019 novel coronavirus infection in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef] [PubMed]
  181. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  182. World Health Organization (WHO). Coronavirus Disease (COVID-19) Outbreak Situation. 2021. Available online: https://www.who.int/emergencies/diseases/novel-coronavirus-2019 (accessed on 29 June 2021).
  183. Cragg, G.M.; Newman, D.J. Natural products: A continuing source of novel drug leads. Biochim. Biophys. Acta 2013, 1830, 3670–3695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Yuan, H.; Ma, Q.; Ye, L.; Piao, G. The traditional medicine and modern medicine from natural products. Molecules 2016, 21, 559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Xu, J.; Xia, Z. Traditional Chinese Medicine (TCM)—Does its contemporary business booming and globalization really reconfirm its medical efficacy & safety? Med. Drug Discov. 2019, 1, 100003. [Google Scholar] [CrossRef]
  186. Gao, R.-R.; Hu, Y.-T.; Dan, Y.; Hao, L.-J.; Liu, X.; Song, J.-Y. Chinese herbal medicine resources: Where we stand. Chin. Herb. Med. 2020, 12, 3–13. [Google Scholar] [CrossRef]
  187. Yang, Y.; Islam, M.S.; Wang, J.; Li, Y.; Chen, X. Traditional Chinese medicine in the treatment of patients infected with 2019-new coronavirus (SARS-CoV-2): A review and perspective. Int. J. Biol. Sci. 2020, 16, 1708–1717. [Google Scholar] [CrossRef] [PubMed]
  188. Yu, M.S.; Lee, J.; Lee, J.M.; Kim, Y.; Chin, Y.-W.; Jee, J.-G.; Keum, Y.-S.; Jeong, Y.-J. Identification of myricetin and scutellarein as novel chemical inhibitors of the SARS coronavirus helicase, nsP13. Bioorg. Med. Chem. Lett. 2012, 22, 4049–4054. [Google Scholar] [CrossRef]
  189. Fung, K.P.; Leung, P.C.; Tsui, K.W.S.; Wan, C.C.D.; Wong, K.B.; Waye, M.Y.M.; Au, W.N.S.; Wong, C.K.; Lam, W.K.C.; Lau, B.S.C. Immunomodulatory activities of the herbal formula Kwan Du Bu Fei Dang in healthy subjects: A randomised, double-blind, placebo-controlled study. Hong Kong Med. J. 2011, 17, 41–43. [Google Scholar]
  190. Abdullah, N.; Ismail, S.M.; Aminudin, N.; Shuib, A.S.; Lau, B.F. Evaluation of selected culinary-medicinal mushrooms for antioxidant and ACE inhibitory activities. Evid. Based Complement. Altern. Med. 2012, 2012, 464238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Kana, Y.; Chen, T.; Wu, Y.; Wu, J. Antioxidant activity of polysaccharide extracted from Ganoderma lucidum using response surface methodology. Int. J. Biol. Macromol. 2015, 72, 151–157. [Google Scholar] [CrossRef] [PubMed]
  192. Zhang, N.; Chen, H.; Zhang, Y.; Xing, L.; Li, S.; Wang, X.; Sun, Z. Chemical composition and antioxidant properties of five edible Hymenomycetes mushrooms. Int. J. Food Sci. Technol. 2015, 50, 465–471. [Google Scholar] [CrossRef]
  193. Collins, A.R. Antioxidant intervention as a route to cancer prevention. Eur. J. Cancer 2005, 41, 1923–1930. [Google Scholar] [CrossRef]
  194. Benzie, I.F.F.; Wachtel-Galor, S. Biomarkers of long-term vegetarian diets. Adv. Clin. Chem. 2009, 47, 169–220. [Google Scholar]
  195. Mohan, K.; Padmanaban, M.; Uthayakumar, V. Isolation, structural characterization and antioxidant activities of polysaccharide from Ganoderma lucidum (Higher Basidiomycetes). Am. J. Biol. Life Sci. 2015, 3, 168–175. [Google Scholar]
  196. Zeng, Q.; Zhou, F.; Lei, L.; Chen, J.; Lu, J.; Zhou, J.; Cao, K.; Gao, L.; Xia, F.; Ding, S.; et al. Ganoderma lucidum polysaccharides protect fibroblasts against UVB-induced photoaging. Mol. Med. Rep. 2017, 15, 111–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. World Health Organization (WHO). Mortality Statistics. World Health Report. 2008. Available online: http://www.who.int/gho/publications/world_health_statistics/EN_WHS08_Full.pdf (accessed on 29 June 2021).
  198. El Sheikha, A.F. Medicinal plants: Ethno-uses to biotechnology era. In Biotechnology and Production of Anti-Cancer Compounds; Malik, S., Ed.; Part of Springer Nature; Springer International Publishing AG: Cham, Switzerland, 2017; pp. 1–38. [Google Scholar]
  199. Wasser, S.P.; Weis, A.L. Medicinal properties of substances occurring in higher basidiomycetes mushrooms: Current perspectives. Int. J. Med. Mushrooms 1999, 1, 31–62. [Google Scholar] [CrossRef] [Green Version]
  200. Borchers, A.T.; Krishnamurthy, A.; Keen, C.L.; Meyers, F.J.; Gershwin, M.E. The immunobiology of mushrooms. Exp. Biol. Med. 2008, 233, 259–276. [Google Scholar] [CrossRef] [PubMed]
  201. Tomasi, S.; Lohezic-Le, D.F.; Sauleau, P.; Bezivin, C.; Boustie, J. Cytotoxic activity of methanol extracts from Basidiomycete mushrooms on murine cancer cell lines. Pharmazie 2004, 59, 290–293. [Google Scholar] [PubMed]
  202. Zaidman, B.Z.; Yassin, M.; Mahajna, J.; Wasser, S.P. Medicinal mushroom modulators of molecular targets as cancer therapeutics. Appl. Microbiol. Biotechnol. 2005, 67, 453–468. [Google Scholar] [CrossRef] [PubMed]
  203. Yuen, J.W.; Gohel, M.D. The dual roles of Ganoderma antioxidants on urothelial cell DNA under carcinogenic attack. J. Ethnopharmacol. 2008, 118, 324–330. [Google Scholar] [CrossRef]
  204. Trajković, L.M.H.; Mijatović, S.A.; Maksimović-Ivanić, D.D.; Stojanović, I.D.; Momčilović, M.B.; Tufegdžić, S.J.; Maksimović, V.M.; Marjanovi, Z.S.; Stošić-Grujičić, S.D. Anticancer properties of Ganoderma lucidum methanol extracts in vitro and in vivo. Nutr. Cancer 2009, 61, 696–707. [Google Scholar] [CrossRef]
  205. Calviño, E.; Manjón, J.L.; Sancho, P.; Tejedor, M.C.; Herráez, A.; Diez, J.C. Ganoderma lucidum induced apoptosis in NB4 human leukemia cells: Involvement of Akt and Erk. J. Ethnopharmacol. 2010, 128, 71–78. [Google Scholar] [CrossRef]
  206. Thyagarajan, A.; Jedinak, A.; Nguyen, H.; Terry, C.; Baldridge, L.A.; Jiang, J.; Sliva, D. Triterpenes from Ganoderma lucidum induce autophagy in colon cancer through the inhibition of p38 mitogen-activated kinase (p38 MAPK). Nutr. Cancer 2010, 62, 630–640. [Google Scholar] [CrossRef] [PubMed]
  207. Hossain, A.; Radwan, F.F.Y.; Doonan, B.P.; God, J.M.; Zhang, L.; Bell, P.D.; Haque, A. A possible cross-talk between autophagy and apoptosis in generating an immune response in melanoma. Apoptosis 2012, 17, 1066–1078. [Google Scholar] [CrossRef] [PubMed]
  208. Liang, C.; Li, H.; Zhou, H.; Zhang, S.; Liu, Z.; Zhou, Q.; Sun, F. Recombinant Lz-8 from Ganoderma lucidum induces endoplasmic reticulum stress-mediated autophagic cell death in SGC-7901 human gastric cancer cells. Oncol. Rep. 2012, 27, 1079–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Oliveira, M.; Reis, F.S.; Sousa, D.; Tavares, C.; Lima, R.T.; Ferreira, I.C.F.R.; dos Santos, T.; Vasconcelos, M.H. A methanolic extract of Ganoderma lucidum fruiting body inhibits the growth of a gastric cancer cell line and affects cellular autophagy and cell cycle. Food Funct. 2014, 5, 1389–1394. [Google Scholar] [CrossRef]
  210. Reis, F.S.; Lima, R.T.; Morales, P.; Ferreira, I.C.F.R.; Vasconcelos, M.H. Methanolic extract of Ganoderma lucidum induces autophagy of AGS human gastric tumor cells. Molecules 2015, 20, 17872–17882. [Google Scholar] [CrossRef] [Green Version]
  211. Wang, F.; Zhou, Z.; Ren, X.; Wang, Y.; Yang, R.; Luo, J.; Strappe, P. Effect of Ganoderma lucidum spores intervention on glucose and lipid metabolism gene expression profiles in type 2 diabetic rats. Lipids Health Dis. 2015, 14, 49. [Google Scholar] [CrossRef] [Green Version]
  212. Gao, H.; Chan, E.; Zhou, F. Immunomodulating activities of Ganoderma, a mushroom with medicinal properties. Food Rev. Int. 2004, 20, 123–161. [Google Scholar] [CrossRef]
  213. Yang, Y.; Zhang, H.; Zuo, J.; Gong, X.; Yi, F.; Zhu, W.; Li, L. Advances in research on the active constituents and physiological effects of Ganoderma lucidum. Biomed. Dermatol. 2019, 3, 6. [Google Scholar] [CrossRef]
  214. Liu, Y.-J.; Du, J.-L.; Cao, L.-P.; Jia, R.; Shen, Y.-J.; Zhao, C.-Y.; Xu, P.; Yin, G.-J. Anti-inflammatory and hepatoprotective effects of Ganoderma lucidum polysaccharides on carbon tetrachloride-induced hepatocyte damage in common carp (Cyprinus carpio L.). Int. Immunopharmacol. 2015, 25, 112–120. [Google Scholar] [CrossRef]
  215. Wu, J.-G.; Kan, Y.-J.; Wu, Y.-B.; Yi, J.; Chen, T.-Q.; Wu, J.-Z. Hepatoprotective effect of ganoderma triterpenoids against oxidative damage induced by tert-butyl hydroperoxide in human hepatic HepG2 cells. Pharm. Biol. 2016, 54, 919–929. [Google Scholar] [CrossRef] [Green Version]
  216. Zhao, C.; Fan, J.; Liu, Y.; Guo, W.; Cao, H.; Xiao, J.; Wang, Y.; Liu, B. Hepatoprotective activity of Ganoderma lucidum triterpenoids in alcohol-induced liver injury in mice, an iTRAQ-based proteomic analysis. Food Chem. 2019, 271, 148–156. [Google Scholar] [CrossRef] [PubMed]
  217. Song, C.-H.; Yang, B.-K.; Ra, K.-S.; Shon, D.-H.; Park, E.-J.; Go, G.-I.; Kim, Y.-H. Hepatoprotective effect of extracellular polymer produced by submerged culture of Ganoderma lucidum WK-003. J. Microbiol. Biotechnol. 1998, 8, 277–279. [Google Scholar]
  218. Lee, C.-H.; Choi, E.Y. Macrophages and inflammation. J. Rheum. Dis. 2018, 25, 11–18. [Google Scholar] [CrossRef] [Green Version]
  219. Wei, B.; Zhang, R.; Zhai, J.; Zhu, J.; Yang, F.; Yue, D.; Liu, X.; Lu, C.; Sun, X. Suppression of Th17 cell response in the alleviation of dextran sulfate sodium-induced colitis by Ganoderma lucidum polysaccharides. J. Immunol. Res. 2018, 2018, 2906494. [Google Scholar] [CrossRef] [Green Version]
  220. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
  221. De Sousa, V.M.C.; Dos Santos, E.F.; Sgarbieri, V.C. The importance of prebiotics in functional foods and clinical practice. Food Nutr. Sci. 2011, 2, 133–144. [Google Scholar] [CrossRef] [Green Version]
  222. Bhakta, M.; Kumar, P. Mushroom polysaccharides as a potential prebiotics. Int. J. Health Sci. Res. 2013, 3, 77–84. [Google Scholar]
  223. Cani, P.D.; Delzenne, N.M. The role of the gut microbiota in energy metabolism and metabolic disease. Curr. Pharm. Des. 2009, 15, 1546–1558. [Google Scholar] [CrossRef] [Green Version]
  224. Jayachandran, M.; Xiao, J.; Xu, B. A critical review on health promoting benefits of edible mushrooms through gut microbiota. Int. J. Mol. Sci. 2017, 18, 1934. [Google Scholar] [CrossRef] [Green Version]
  225. Meneses, M.E.; Martínez-Carrera, D.; Torres, N.; Sánchez-Tapia, M.; Aguilar-López, M.; Morales, P.; Sobal, M.; Bernabé, T.; Escudero, H.; Granados-Portillo, O.; et al. Hypocholesterolemic properties and prebiotic effects of Mexican Ganoderma lucidum in C57BL/6 Mice. PLoS ONE 2016, 11, e0159631. [Google Scholar] [CrossRef] [Green Version]
  226. Lv, X.-C.; Guoa, W.-L.; Li, L.; Yu, X.-D.; Liu, B. Polysaccharide peptides from Ganoderma lucidum ameliorate lipid metabolic disorders and gut microbiota dysbiosis in high-fat diet-fed rats. J. Funct. Foods 2019, 57, 48–58. [Google Scholar] [CrossRef]
  227. Wu, Q.; Zhang, H.; Wang, P.G.; Chen, M. Evaluation of the efficacy and safety of Ganoderma lucidum mycelium-fermented liquid on gut microbiota and its impact on cardiovascular risk factors in human. RSC Adv. 2017, 7, 45093. [Google Scholar] [CrossRef] [Green Version]
  228. Clemente, J.C.; Ursell, L.K.; Parfrey, L.W.; Knight, R. The impact of the gut microbiota on human health: An integrative view. Cell 2012, 148, 1258–1270. [Google Scholar] [CrossRef] [Green Version]
  229. World Health Organization (WHO). Cardiovascular Diseases (CVDs). 2017. Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 29 June 2021).
  230. Chang, C.-J.; Lin, C.-S.; Lu, C.-C.; Martel, J.; Ko, Y.-F.; Ojcius, D.M.; Tseng, S.-F.; Wu, T.-R.; Chen, Y.-Y.M.; Young, J.D.; et al. Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota. Nat. Commun. 2015, 6, 7489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Yang, B.K.; Jeong, S.C.; Song, C.H. Hypolipidemic effect of exo- and endo-biopolymers produced from submerged mycelial culture of Ganoderma lucidum in rats. J. Microbiol. Biotechnol. 2002, 12, 872–877. [Google Scholar]
  232. Wanmuang, H.; Leopaircut, J.; Kositchaiwat, C. Fatal fulminant hepatitis associated with Ganoderma lucidum (Lingzhi) mushroom powder. J. Med. Assoc. Thai 2007, 90, 179–181. [Google Scholar]
  233. Ulbricht, C.; Isaac, R.; Milkin, T.; Poole, E.P.; Rusie, E.; Serrano, J.M.G.; Weissner, W.; Windsor, R.C.; Woods, J. An evidence-based systematic review of stevia by the Natural Standard Research Collaboration. Cardiovasc. Hematol. Agents Med. Chem. 2010, 8, 113–127. [Google Scholar] [CrossRef]
  234. 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]
  235. Wu, D.-T.; Deng, Y.; Chen, L.-X.; Zhao, J.; Bzhelyansky, A.; Li, S.-P. Evaluation on quality consistency of Ganoderma lucidum dietary supplements collected in the United States. Sci. Rep. 2017, 7, 7792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Qi, L.; Liu, H.; Li, J.; Li, T.; Wang, Y. Feature fusion of ICP-AES, UV-Vis and FTMIR for origin traceability of Boletus edulis mushrooms in combination with chemometrics. Sensors 2018, 18, 241. [Google Scholar] [CrossRef] [Green Version]
  237. Lu, J.; Qin, J.-Z.; Chen, P.; Chen, X.; Zhang, Y.-Z.; Zhao, S.-J. Quality difference study of six varieties of Ganoderma lucidum with different origins. Front. Pharmacol. 2012, 3, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Fernandes, Â.; Petrović, J.; Stojković, D.; Barros, L.; Glamočlija, J.; Soković, M.; Martins, A.; Ferreira, I.C.F.R. Polyporus squamosus (Huds.) Fr from different origins: Chemical characterization, screening of the bioactive properties and specific antimicrobial effects against Pseudomonas aeruginosa. LWT-Food Sci. Technol. 2016, 69, 91–97. [Google Scholar] [CrossRef] [Green Version]
  239. Tešanović, K.; Pejin, B.; Šibul, F.; Matavulj, M.; Rašeta, M.; Janjušević, L.; Karaman, M. A comparative overview of antioxidative properties and phenolic profiles of different fungal origins: Fruiting bodies and submerged cultures of Coprinus comatus and Coprinellus truncorum. J. Food Sci. Technol. 2017, 54, 430–438. [Google Scholar] [CrossRef] [Green Version]
  240. Yao, S.; Li, T.; Liu, H.; Li, J.; Wang, Y. Traceability of Boletaceae mushrooms using data fusion of UV–visible and FTIR combined with chemometrics methods. J. Sci. Food Agric. 2017, 98, 2215–2222. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Image of Ganoderma lucidum.
Figure 1. Image of Ganoderma lucidum.
Foods 11 01030 g001
Figure 2. Wide-scale applications of mushrooms including Ganoderma lucidum; i.e., pharmaceuticals, nutraceuticals, and cosmetics. Source: Reprinted from Wu et al. [31]. Licensed under CC BY 4.0.
Figure 2. Wide-scale applications of mushrooms including Ganoderma lucidum; i.e., pharmaceuticals, nutraceuticals, and cosmetics. Source: Reprinted from Wu et al. [31]. Licensed under CC BY 4.0.
Foods 11 01030 g002
Figure 3. A scientometric analysis of increasing interest in Ganoderma lucidum over the last 10 years. Reprinted with permission from Scopus. 2020, Elsevier.
Figure 3. A scientometric analysis of increasing interest in Ganoderma lucidum over the last 10 years. Reprinted with permission from Scopus. 2020, Elsevier.
Foods 11 01030 g003
Figure 4. Nutritional and health benefits conferred by Ganoderma lucidum.
Figure 4. Nutritional and health benefits conferred by Ganoderma lucidum.
Foods 11 01030 g004
Figure 5. Molecular docking for the interaction of antiviral compounds with EV71 capsid. (A) Stick conformer diagram. (B) Cartoon conformer diagram. Both GLTA and GLTB could bind stably in the viral capsid mainly through hydrophobic interactions at a hydrophobic pocket (F site) in the capsid of EV71 virion. Source: Reprinted with permission from Zhang et al. [79]. 2022, Elsevier.
Figure 5. Molecular docking for the interaction of antiviral compounds with EV71 capsid. (A) Stick conformer diagram. (B) Cartoon conformer diagram. Both GLTA and GLTB could bind stably in the viral capsid mainly through hydrophobic interactions at a hydrophobic pocket (F site) in the capsid of EV71 virion. Source: Reprinted with permission from Zhang et al. [79]. 2022, Elsevier.
Foods 11 01030 g005
Figure 6. Regulatory mechanism of GLPP on hyperlipidaemia, hypercholesterolemia, and gut microbiota dysbiosis in rats fed on HFD. GLPP: Ganoderma lucidum polysaccharide peptide; HFD: high-fat diet; TG: triglyceride; TC: total cholesterol; LDL-C: low-density lipoprotein cholesterol; FFA: free fatty acids; SCFA: short-chain fatty acids; OSTα: organic solute transporter alpha; CYP7A1: cholesterol 7α-hydroxylase; SREBP-1C: sterol regulatory element-binding protein-1C; PPARα: peroxisome proliferator-activated receptor alpha; HMG-Coa: 3-hydroxy-3-methylglutaryl coenzyme A; BSEP: bile salt export pump; MRP3: multidrug-resistance-associated protein 3; OATP2: organic-anion-transporting polypeptides 2. Source: Reprinted with permission from Lv et al. [226]. 2022, Elsevier.
Figure 6. Regulatory mechanism of GLPP on hyperlipidaemia, hypercholesterolemia, and gut microbiota dysbiosis in rats fed on HFD. GLPP: Ganoderma lucidum polysaccharide peptide; HFD: high-fat diet; TG: triglyceride; TC: total cholesterol; LDL-C: low-density lipoprotein cholesterol; FFA: free fatty acids; SCFA: short-chain fatty acids; OSTα: organic solute transporter alpha; CYP7A1: cholesterol 7α-hydroxylase; SREBP-1C: sterol regulatory element-binding protein-1C; PPARα: peroxisome proliferator-activated receptor alpha; HMG-Coa: 3-hydroxy-3-methylglutaryl coenzyme A; BSEP: bile salt export pump; MRP3: multidrug-resistance-associated protein 3; OATP2: organic-anion-transporting polypeptides 2. Source: Reprinted with permission from Lv et al. [226]. 2022, Elsevier.
Foods 11 01030 g006
Figure 7. Infographic for Ganoderma lucidum: current scenario and future perspectives.
Figure 7. Infographic for Ganoderma lucidum: current scenario and future perspectives.
Foods 11 01030 g007
Table 1. Some of the cosmetic products are produced commercially from the G. lucidum mushroom worldwide *.
Table 1. Some of the cosmetic products are produced commercially from the G. lucidum mushroom worldwide *.
Commercial Product Name/Producing CountryUses
CV Skinlabs Body Repair Lotion, USAWound healing and anti-inflammatory
Dr. Andrew Weil for Origins Mega-Mushroom Skin Relief Face Mask, USAAnti-inflammatory properties
Moon Juice Spirit Dust, USAImmune system
Estée Lauder Re-Nutriv Sun Supreme Rescue Serum sun care product, USATriple-action repair technology to enhance the skin’s own natural defenses against the visible effects of sun exposure and sun-stressed skin
Four Sigma Foods Instant Reishi Herbal Mushroom Tea, UKImmunity boost
Kat Burki Form Control Marine Collagen Gel, UKBoosting collagen, improving elasticity, and providing hydration
Tela Beauty Organics Encore Styling Cream, UKProviding hair with sun protection and preventing color fading
Menard Embellir Refresh Massage, FranceSkin antiaging
Yves Saint Laurent Temps Majeur Elixir De
Nuit, France
Antiaging
Pureology NanoWorks Shineluxe, FranceAntiaging and antifading
Hankook Sansim Firming Cream (Tan Ryuk
SANG), Korea
Making skin tight and vitalized
La Bella Figura Gentle Enzyme Cleanser, ItalyAntioxidants and vitamin D
DXNGanozhi Moisturizing Micro Emulsion, MalaysiaHydrating and nourishing the skin
Guangzhou Bocaly Bio-Tec. Ganoderma Cell-Repairing Antiaging Face Mask, ChinaAntiwrinkle, firming, lightening, moisturizer, and nourishing, pigmentation corrector; pore cleaning and whitening
Nanjing Zhongke Pharmaceuticals Ganoderma Face Cream Set (day/night cream and eye gel set), ChinaImmunity boost and antifatigue
Shenzhen Hai Li Xuan Technology HailiCare Skin Whitening Cream, ChinaRemoving freckles and whitening
Menard Embellir Night Cream, JapanEliminating toxins and helping repair skin damage associated with overexposure to UV radiation and free radicals
MAVEX Rejuvenating Treatment, Hong KongAntioxidant action and deep cellular renewal; fight degenerative processes and the negative action of free radicals
* Sources: Wu et al. [31], Taofiq et al. [34], Hapuarachchi et al. [35], www.vegamebeljepara.com, www.dazzlinggroup.com, www.dxnmalaysia.com, and www.vegamebeljepara.com (accessed on 16 February 2022) Adapted from Wu et al. [31]. Licensed under CC BY 4.0. Adapted with permission from Taofiq et al. [34]. 2022, Elsevier.
Table 2. Physicochemical properties and chemical composition of Ganoderma lucidum mushroom.
Table 2. Physicochemical properties and chemical composition of Ganoderma lucidum mushroom.
ConstituteContentDRIs * (g/day)Value in 100 g Mushroom/DRIs × 100
Valueg/100 g Mushroom (Wet-Weight Basis)g/100 g Mushroom (Dry-Weight Basis)
Moisture % 47
Total solids (TS) % 53
pH value5.6
Energy (kcal)238.98 ** Men: 2215 ***10.79
Women: 202511.80
Water-soluble proteins % 19.536.80Men (total proteins) ****: 5634.82
Women (total proteins): 4642.39
Total lipids % 3.005.6644–77 *****3.90–6.82
Total ash % 6.3
Reducing sugars % 4.398.28
Nonreducing sugars % 1.021.92
Total sugars % 5.4110.211304.16
Crude fibers % 3.5Men: 389.21
Women: 2514.00
Polyphenols “as gallic acid” 0.040.081 ******7.5
MineralMineral content (mg/100 g mushroom)DRIs (mg/day)Value in 100 g mushroom/DRIs × 100
Major minerals
Potassium43247009.19
Phosphorus22570032.14
Sulfur129200–15008.60–64.50
Magnesium7.95Men: 4002.00
Women: 3102.60
Sodium2.8215000.20
Calcium1.8810000.20
Trace minerals
Copper260.92889
Manganese22Men: 2.3956.52
Women: 1.81222.22
Iron2.22Men: 827.75
Women: 1812.33
Zinc0.7Men: 116.40
Women: 88.75
VitaminVitamin content (mg/100 g mushroom)DRIs (mg/day)Value in 100 g mushroom/DRIs × 100
Thiamine (B1)3.49Men: 1.2290.83
Women: 1.1317.27
Riboflavin (B2)17.10Men: 1.31315.38
Women: 1.11554.54
Niacin (B3)61.9Men: 16386.87
Women: 14442.14
Pyridoxine (B6)0.71Men: 1.450.71
Women: 1.259.16
Ascorbic acid32.2Men: 9035.77
Women: 7542.93
* DRIs: dietary recommended intakes for adults [60,61]; ** the total energy of 100 g of mushroom samples was calculated according to the equations of Manzi et al. [62]; *** based on 1.3 kcal/kg body weight/hour for the reference body weight; **** based on 0.8 g/kg body weight/day for the reference body weight; ***** Casselbury [63]; ****** Duthie et al. [64]. Sources: Roy et al. [59], Rahman et al. [65], and http://www.medicinabiomolecular.com.br/biblioteca/pdfs/Biomolecular/mb-0223.pdf. (accessed on 16 February 2022). Adapted from Rahman et al. [65]. Licensed under CC-BY.
Table 3. The major bioactive compounds of G. lucidum and their biological effects.
Table 3. The major bioactive compounds of G. lucidum and their biological effects.
Bioactive CompoundsBiological Effects References
Triterpenoids
Ganoderic acids, lucidumol, lucialdehyde, lucidenic acids, ganodermic, ganolucidic acids, ganoderals, ganoderiolsAnticancerWachtel-Galor et al. [6], El Mansy [75]
TriterpenoidsAntidiabeticAhmad [68], Ma et al. [78]
Ganoderic acids T-Q and lucideinic acids A, D2, E2, and PAnti-inflammatoryEl Mansy [75]
TriterpenesAntioxidantEl Mansy [75]
Ganoderic acids, ganodermin, ganoderic acid A, ganodermadiol, ganodermanondiol, lucidumol B, ganodermanontriol, ganoderic acid B, ganolucidic acid BAntimicrobialCör et al. [70], Sudheer et al. [73]
Triterpenoids, ganoderic acid, ganoderiol F, ganodermanontriolAntiviralBishop et al. [13], Zhang et al. [79], Zhu et al. [80]
Polysaccharides
1→3, 1→4, and 1→6-linked β and α-D (or L)-glucans, GLP-2BAnticancerWachtel-Galor et al. [6], Ferreira et al. [81]
PolysaccharidesAntidiabeticAhmad [68], Ma et al. [78]
PolysaccharidesAntioxidantEl Mansy [75]
PolysaccharidesAntimicrobialCör et al. [70]
Polysaccharides (ganopoly) Cardiovascular problemsChan et al. [82]
Proteins, Glycoproteins, and Peptidoglycans
Glycopeptides and peptidoglycansAnticancerWachtel-Galor et al. [6], Sudheer et al. [73], Ferreira et al. [81],
Protein Ling Zhi-8 (LZ-8), lectin, ribosome-inactivating proteins, antimicrobial proteins, glycopeptides/glycoproteins, peptidoglycans/proteoglycans, ganodermin A, ribonucleases, proteinases, metalloproteases, laccasesImmunomodulatory, anticancer, and antitumorWachtel-Galor et al. [6], El Mansy [75]
Proteoglycans, proteins (LZ-8)AntidiabeticAhmad [68], Ma et al. [78]
Polysaccharide–peptide complexAntioxidantMehta [83]
Phenolic compounds
Phenolic components, phenolic extractsAntioxidantMehta [83]
SaponinsAnticancer and antioxidantLee et al. [84]
Sterols; e.g., ergosterolProvitamin D2Wachtel-Galor et al. [6]
Long-chain fatty acidsAntitumorGao et al. [85]
Table 4. Antimicrobial activities of Ganoderma lucidum parts, products, and compounds.
Table 4. Antimicrobial activities of Ganoderma lucidum parts, products, and compounds.
Parts/Products/CompoundsTested Microorganism References
Antibacterial activity
Fruiting bodiesHelicobacter pylori ATCC 43504, Staphylococcus aureus ATCC 26003Liu et al. [131], Shang et al. [132]
Mycelia extractBacillus cereus (clinical isolate), Micrococcus flavus ATCC 10240, S. aureus ATCC 6538, Listeria monocytogenes NCTC 7973, Escherichia coli ATCC 35218, Enterobacter cloacae (human isolate), Pseudomonas aeruginosa ATCC 27853, Salmonella typhimurium ATCC 13311Ćilerdžić et al. [133]
Fruiting bodiesS. aureus (MTCC 96), B. cereus (MTCC 430), P. aeruginosa (MTCC 424)Karwa and Rai [134]
Fruiting bodiesS. aureus (ATCC 6538), Bacillus subtilis (ATCC 6633)Ćilerdžić et al. [135]
Ergosta-5,7,22-trien-3β-yl acetate, ergosta-7,22-dien-3β-yl acetate, ergosta-7,22-dien-3-one, ergosta-7,22-dien-3β-ol, ergosta-5,7,22-trien-3β-ol, ganodermadiolS. aureus (ATCC 6538), B. subtilis (ATCC 6633)Ćilerdžić et al. [135]
CarpophoresBacillus anthracis ATCC 6603, B. cereus ATCC 27348, B. subtilis ATCC 6633, Micrococcus luteus ATCC 9341, S. aureus ATCC 25923, E. coil ATCC 259 22, Klebsiella oxytoca ATCC 8724, Klebsiella pneumonia ATCC 10031, Proteus vulgaris ATCC 27853, S. typhi ATCC 6229Yoon et al. [136]
BasidiocarpsB. cereus (clinical isolate), M. flavus ATCC 10240, S. aureus ATCC 6538, L. monocytogenes NCTC 7973, E. coli ATCC 35218, E. cloacae (human isolate), P. aeruginosa ATCC 27853, S. typhimurium ATCC 13311Vazirian et al. [137]
12b-acetoxy-3β,7 β -dihydroxy-
11,15,23-trioxolanost-8-en-26-oic
acid butyl ester
S. aureus (ATCC 6538), B. subtilis (ATCC 6633)Yang et al. [138]
Mycelia (Protein extract)Staphylococcus epidermidis, B. subtilis, B. cereus, E. coli, P. aeruginosaSa-Ard et al. [139]
Fruiting bodies (Protein extract)S. epidermidis, S. aureus, B. subtilis, B. cereus, E. coli, P. aeruginosaSa-Ard et al. [139]
NG *S. aureus (ATCC 6538), B. cereus (clinical isolate), L. monocytogenes (NCTC 7973), M. flavus (ATCC 10240), P. aeruginosa (ATCC 27853), E. coli (ATCC 35210), S.
typhimurium (ATCC 13311), E. cloacae (human isolate)
Heleno et al. [140]
Antifungal activity
Fruiting bodiesAcremonium strictum BEOFB10m, Aspergillus glaucus BEOFB21m, Aspergillus flavus BEOFB22m, Aspergillus fumigatus BEOFB23m, Aspergillus nidulans BEOFB24m, Aspergillus niger BEOFB25m, Aspergillus terreus BEOFB26m, Trichoderma viride BEOFB61mVazirian et al. [137]
Fruiting bodiesA. fumigatus (human isolate), Aspergillus versicolor (ATCC 11730), Aspergillus ochraceus (ATCC 12066), A. niger (ATCC 6275), T. viride (IAMz5061), Penicillium funiculosum (ATCC 36839), Penicillium ochrochloron (ATCC 9112), Penicillium verrucosum var. cyclopium (food isolate)Heleno et al. [140]
Rare Earth-Carboxymethylated Ganoderma applanatum PolysaccharideValsa mali, Fusarium oxysporum, Gaeumannomyces graminis, Colletotrichum gloeosporioides, Alternaria brassicaeSun et al. [141]
GanoderminBotrytis cinerea, F. oxysporum, Physalo sporapiricolaWang and Ng [113]
MyceliaAcremonium strictum, A. glaucus, A. flavus, A. fumigatus, A. nidulans, A. niger, A. terreus, T. virideĆilerdžić et al. [133]
Antiviral activity
Ganoderiol F & GanodermanontriolHIV 1(HIV-1 protease)El-Mekkawy et al. [142]
CarpophoresHerpes simplex virus types 1 (HSV-1) and 2 (HSV-2),
influenza A virus (Flu A), and vesicular stomatitis virus
(VSV) Indiana and New Jersey strains
El-Mekkawy et al. [142]
Acidic protein-bound polysaccharideHSV-1 and HSV-2Eo et al. [143]
Fruiting bodiesOral human papillomavirus (HPV)Donatini [144]
NGNewcastle disease virus (anti-neuraminidase)Zhu et al. [80], Shamaki et al. [145]
Fruiting bodiesEpstein-Barr VirusIwatsuki et al. [146]
MyceliaHepatitis B virusLi et al. [147]
Mycelia (Ganoderic acid)Hepatitis BLi and Wang [148]
Lanosta-7,9(11),24-trien-3-one,15;26-dihydroxy (GLTA), Ganoderic acid YEnterovirus 71Zhang et al. [79]
* NG: data not given.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

El Sheikha, A.F. Nutritional Profile and Health Benefits of Ganoderma lucidum “Lingzhi, Reishi, or Mannentake” as Functional Foods: Current Scenario and Future Perspectives. Foods 2022, 11, 1030. https://doi.org/10.3390/foods11071030

AMA Style

El Sheikha AF. Nutritional Profile and Health Benefits of Ganoderma lucidum “Lingzhi, Reishi, or Mannentake” as Functional Foods: Current Scenario and Future Perspectives. Foods. 2022; 11(7):1030. https://doi.org/10.3390/foods11071030

Chicago/Turabian Style

El Sheikha, Aly Farag. 2022. "Nutritional Profile and Health Benefits of Ganoderma lucidum “Lingzhi, Reishi, or Mannentake” as Functional Foods: Current Scenario and Future Perspectives" Foods 11, no. 7: 1030. https://doi.org/10.3390/foods11071030

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