Mushrooms: Immune-Boosting Insights
Mushrooms: Immune-Boosting Insights
Journal of Autoimmunity
journal homepage: www.elsevier.com/locate/jautimm
A R T I C L E I N F O A B S T R A C T
Keywords: In the wide field of nutraceuticals, the effects of mushrooms on immunity, cancer and including autoimmunity
Immunity have been proposed for centuries but in recent years a growing interest has led scientists to elucidate which
Leukocytes specific compounds have bioactive properties and through which mechanisms. Glucans and specific proteins are
Cytokines
responsible for most of the biological effects of mushrooms, particularly in terms of immunomodulatory and anti-
Mushrooms
Glucans
tumor results. Proteins with bioactive effects include lectins, fungal immunomodulatory proteins (FIPs), ribo
Nutraceutical some inactivating proteins (RIPs), ribonucleases, laccases, among others. At the present status of knowledge,
numerous studies have been performed on cell lines and murine models while only a few clinical trials have been
conducted. As in most cases of dietary components, the multitude of variables implicated in the final effect and
an inadequate standardization are expected to affect the observed differences, thus making the available evi
dence insufficient to justify the treatment of human diseases with mushrooms extracts. We will herein provide a
comprehensive review and critically discussion the biochemical changes induced by different mushroom com
pounds as observed in in vitro studies, particularly on macrophages, dendritic cells, T cells, and NK cells,
compared to in vivo and human studies. Additional effects are represented by lipids which constitute a minor part
of mushrooms but may have a role in reducing serum cholesterol levels or phenols acting as antioxidant and
reducing agents. Human studies provide a minority of available data, as well illustrated by a placebo-controlled
study of athletes treated with β-glucan from Pleurotus ostreatus. Variables influencing study outcomes include
different mushrooms strains, growing conditions, developmental stage, part of mushroom used, extraction
method, and storage conditions. We foresee that future rigorous research will be needed to determine the po
tential of mushroom compounds for human health to reproduce the effects of some compounds such as lentinan
which a metaanalysis demonstrated to increase the efficacy of chemotherapy in the treatment of lung cancer and
in the improvement of the patients quality of life.
* Corresponding author. Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center IRCCS, Via A. Manzoni 56, 20089, Rozzano,
Milan, Italy.
E-mail address: [email protected] (C. Selmi).
https://doi.org/10.1016/j.jaut.2020.102576
Received 20 October 2020; Received in revised form 12 November 2020; Accepted 13 November 2020
Available online 1 December 2020
0896-8411/© 2020 Elsevier Ltd. All rights reserved.
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
still relies on the use of mushroom for remedies preparation [7]. Sci on hundreds of different species. Glucans are the main polysaccharides
entific studies on the mechanisms underlying their medical properties found in mushrooms, as they constitute the fungal cell wall, and are
were lacking until the last decades, when progresses have been made excreted into the cell growth medium, therefore their recovery, purifi
and molecular studies, clinical trials, papers, journals and congresses cation and characterization is generally feasible [7,10]. As glucans are
have been dedicated to mushrooms [2]. recognized by different immune cells, their effects are pleiotropic and
several experiments have tried to elucidate them. Different methods are
2. Mushroom type and composition available to extract and purify glucans, potentially affecting their ac
tivities [11]. The most common type of glucan and the most important
The gross composition of mushrooms is water (90%), and the for biological activities consists of a backbone of D-glucose-linked β-(1
remaining 10% consists of protein and amino acids (10%–40%) → 3) frequently branched at O-6 by β-D-glucose residues as side chains
including primarily leucine, valine, glutamine, glutamic and aspartic (hereafter referred to as β-glucan) [10]. A large variability can be
acids, fats (2%–8%), mainly linoleic, oleic and palmitic fatty acid, car observed in mushroom species and its concentrations range from 0.21 to
bohydrates (3%–28%) such as chitin, glycogen, trehalose and mannitol, 0.53 g/100 g dry weight [12]. In addition, α- and mixed D-glucans can
fibers (3%–32%), and ash (8%–10%), the fraction of dry matter that be found [11], even though the majority are β-glucans, including len
remains after incineration, mainly composed of salts and metals, such as tinan from Lentinus edodes, schizophyllan from Schizophyllum commune,
potassium, calcium, phosphorus, magnesium, iron, zinc and copper. krestin from Coriolus versicolor, grifolan from Grifola frondosa and scle
Mushrooms also contain vitamins such as thiamine, riboflavin, niacin, roglucan from Sclerotinia sclerotiorum [13].
tocopherol and vitamin D and antioxydants. Growth features, stage, Glucans cannot be synthesized and their structure includes
harvest and storage condition may influence the chemical composition pathogen-associated molecular patterns (PAMPs), a highly conserved
and the nutritional value of edible mushrooms [7–9]. While in the last pattern able to induce the immune response. Different membrane re
centuries whole mushrooms were used for treatment and analysed in ceptors, the pattern-recognition receptors (PRRs), can recognize PAMPs,
early studies, it is now ascertained that medical properties are attrib such as Dectin-1, complement receptor 3 (CR3), NOD-like (NLR), RIG-I-
utable to polysaccharide fractions, some specific proteins or other like (RLR), and Toll-like receptors (TLR) [14,15]. These are found on
bioactive compounds. They have been identified and purified from fruit macrophages, monocytes, neutrophils, dendritic cells (DCs) and natural
bodies, cultured mycelium and cultured broth and studies now focus on killer (NK) cells [14] and bind with the highest affinity the polymers
their activities [7]. Medicinal mushrooms appear to have several func with the greatest molecular weight, such as schizophyllan and scle
tions, including immunomodulating, antitumor, antioxidant, car roglucan [13]. After binding, the signal is transduced through direct
dioprotective, hypocholesterolemic, antiviral, antibiotic, anti-parasitic, receptor activation and/or cellular pathway activation. Studies in ani
antifungal, hepatoprotective and hypoglycemic effects [2]. In this re mal models demonstrate that a possible mechanism could be the
view we will focus on the effects on immunity. Bioactive beneficial glucan-induced clustering of Dectin-1, with subsequent recruitment of
properties have been found in both edible and non-edible mushrooms. downstream proteins activating the nuclear factor
For example, Ganoderma and Coriolus are considered as medicinal but kappa-light-chain-enhancer of activated B cells (NF-κB) and mediating
not edible. The most common medicinal mushrooms are listed in Table 1 the transcription of genes [16]. Dectin-1 is one of the most important
[7]. innate immune receptors for glucans and notably knock out mice for this
receptor show an increased susceptibility to chemically induced colitis,
3. Mushroom bioactive compounds as a consequence of altered responses to fungi. Moreover, in humans, a
polymorphism in the gene for Dectin-1 is linked to a severe form of ul
3.1. Polysaccharides cerative colitis [17]. Other studies in vitro showed how the macrophage
response to β-glucans could be influenced by the inflammatory milieu. In
Polysaccharides are the mushroom-derived molecules with the most fact, immune cells can be activated by membrane PRRs, by direct
biologically potent effects and numerous studies have been performed membrane binding or by intracellular PRRs, depending on factors as
2
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
Table 1
Classification of the most common medicinal mushrooms.
Genus Latin name Common name Family Class
priming with lipopolysaccharide (LPS) and various cytokines and immunomodulatory action involves the activation of NF-κB [23] and
different inflammatory pathways are initiated [18]. stimulates the generation of reactive oxygen species (ROS) and nitric
The route of administration of glucans has been an issue, and the oxide (NO), which contribute to the death of microorganisms. The effect
bioavailability after oral intake remains debated, with only intraperi on cytokines has been demonstrated in several studies on different
toneal or intravenous applications being considered adequate. However, mushrooms mostly for β-glucan, in both murine and human macro
further experiments showed that orally given glucan has similar effects phages, showing increased expression of interleukin (IL)-1, IL-6, tumor
to an injected dose [19] and that the gut-associated lymphoid tissue is necrosis factor (TNF)-α and other pro-inflammatory cytokines and
the principal site of interaction between mushroom extracts and im decreased expression or no induction of anti-inflammatory factors, as
mune cells, which have membrane PRRs [20]. IL-10 [24–32]. Table 2 depicts the detailed effects of glucans on mac
rophages in different studies and Fig. 2 illustrates them.
3.2. Effects of polysaccharides on macrophages Macrophage stimulation by glucans appears to be important in mu
rine models to mitigate chemotherapy-induced myelosuppression [33,
Macrophages play a role in tissue homeostasis, immune surveillance, 34], to induce wound healing [35], as a defence against viral [36] and
response against pathogens and in the resolution of inflammation. They bacterial infections, i.e. against Salmonella [37] and Cryptococcus [38],
are able to secrete cytokines and mediators and to polarize to M1, as well as against cancer [39,40].
eliciting a cytotoxic immune response, or to M2, with homeostatic and In a few studies α-glucan effect was examined. An α-glucan from
anti-inflammatory effects [21,22]. In macrophages, the β-glucans Grifola frondosa was administered to diabetic mice, leading to reduction
Table 2
Effects of mushroom glucans on macrophages.
Mushroom Type of glucan Type of Pro- or anti-inflammatory Other effect Ref
macrophages effects
3
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
Fig. 2. Effects of mushroom polysaccharides on monocytes and macrophages. Mushroom polysaccharides seem to have a pro-inflammatory activity, inducing M1
macrophages. On the other hand, if monocytes are stimulated with glucans as PCPS at early stages, the differentiation in M2 anti-inflammatory macrophages
is promoted.
of NO and IL-1 macrophages production, improvement of glucose, tri militaris have different effects depending on the type of extraction. The
glycerides, cholesterol and free fatty acid levels and amelioration of aqueous extract, which is more rich of mannose and galactose and poor
ultrastructural changes of pancreatic β-cells, suggesting a protective role of glucose, stimulates a pro-inflammatory response, while the alkaline
on pancreatic islets [41]. extract, with a higher amount of glucose and a lower content of the two
Of note, in a recent study Li and Colleague obtained a β-glucan (LeP- other monosaccharides, show the inhibition of inflammatory genes, as
N2) from Lentinus edodes and showed its ability to activate macrophages observed for the purified β-(1 → 3)-D-glucan [46]. Moreover, when
with ROS and NO production in a dose-dependent manner, with no analysed in detail, the effect of glucans might be dual with
cytotoxicity until the dose of 100 μg/mL [42]. pro-inflammatory properties in some pathways and inhibitory in others.
In a study on human macrophages, nine commercially available In this regards, Ahn and Colleagues described that lentinan had an effect
preparations from three mushroom species, Ganoderma lucidum, Lenti on inflammasome activation and cytokine maturation in murine bone
nula edodes, and Grifola frondosa, were analysed for β- and α-glucan marrow-derived macrophages. Lentinan up-regulated pro-inflammatory
contents and three extracts were selected based on the highest β-glucan cytokines and the expression of the inflammasome components NLRP3
contents. These extracts led to a dose-dependent increase in cytokine and pro-IL-1β. However, lentinan also selectively inhibited the IL-1β
expression in both non-LPS and LPS stimulated macrophages, with a maturation in response to AIM2 inflammasome activation, ameliorated
synergistic effect on the expression of IL-1α, IL-6 and TNF-α and an LPS-induced lethality and reduced IL-1β secretion induced by Listeria
antagonistic effect on the expression of IL-10. A combined mushroom monocytogenes-mediated AIM2 inflammasome activation. Therefore,
formula had EC50 values lower than 100 μg/mL and even lower in TNF-α while stimulating the expression of pro-inflammatory cytokines, it
expression from LPS treated macrophages compared to the individual selectively attenuates the cytokine maturation in response to AIM2
extracts, suggesting a potential synergistic effect of the mushroom for inflammasome activation. The Authors suggest lentinan as an anti-in
mulas [43]. flammasome agent, especially for AIM2 inflammasome [47]. On the
As already mentioned, the capability of mushrooms to stimulate the contrary, in previous studies, other glucans as paramylon and zymosan
production of pro-inflammatory cytokines is likely dependent on com failed to modulate IL-1β secretion [48,49]. It could be possible that
plex factors, among which an important variable is the stage of matu polysaccharides have different effect depending on the type of glucans,
ration of immune cell precursors. In fact, a polysaccharide derived from their chemical structures, the method for extraction, the cells and the
Pleurotus citrinopileatus (PCPS) showed anti-inflammatory effect on setting used to study their effects and a multitude of variables not yet
macrophages, modulating the monocyte-to-macrophage differentiation completely understood. For example, a high molecular weight fraction
early, at the monocyte stage, promoting the development of alternative from Grifola frondosa inhibits TNF-α, IL-6 and NF-κB activation in
activated macrophages with anti-inflammatory properties. The response LPS-induced macrophages. On the base of chemical and enzymatic an
of PCPS-treated monocytes to interferon (IFN)-γ + LPS led to reduced alyses, a glucan with a β-(1 → 4)-linked backbone and β-(1 → 6)-linked
levels of TNF, IL-6 and CCR2 and a tendency to increase IL-10, CCL2 and branches has been identified and may contribute to the
CCL8, with anti-inflammatory effect. Therefore, an early stimulation of anti-inflammatory activity, interacting with TLR2 rather than Dectin-1
macrophage precursor with mushroom glucans might induce a long- or CR3 receptors [50]. Murphy and Colleagues compared an ‘in house’
lasting anti-inflammatory effect [44]. Zymosan and mushroom poly lentinan and a commercial extract and reported that the latter has higher
saccharide extracts led to a different response of THP-1 monocytes amounts of α-glucans and less β-glucans while both reduce
compared to macrophages, the latter producing higher levels of cyto cytokine-induced NF-κB activation in human alveolar epithelial A549
kines upon stimulation [45]. cells, with the ‘in house’ extract being more potent. However, in acti
Other factors implicated in the bioactivity of mushroom poly vated THP-1 derived macrophages, the commercial extract is more
saccharides are their composition and structure. It has been demon effective in attenuating pro-inflammatory cytokine production (TNF-α,
strated that polysaccharides derived from the Ascomycete Cordyceps IL-8, IL-2, IL-6, IL-22), transforming growth factor (TGF)-β and IL-10.
4
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
Moreover, it reduces the oxidative stress-induced early apoptosis, while administered orally to tumor-bearing mice is captured by macro
the ‘in house’ extract attenuated late apoptosis [51]. Other studies phages and DCs in the Peyer’s patches, transported to the spleen
support the observation of anti-inflammatory properties of glucans on significantly changing the cellular composition, increasing the number
macrophages, thus reducing the production of NO and cytokines even of CD4, CD8, B, NK cells and DCs, while decreasing the number of
after stimulation [52–56]. Of note, an Agaricus blazei Murill extract, splenic myeloid-derived suppressor cells. It also induces the immune
known to be pro-inflammatory in vitro, showed reduction of cytokines response against cancer by increasing the IFN-γ expression in Peyer’s
levels in human whole blood of healthy volunteers after oral intake of patches, spleen tumor and mesenteric lymph nodes. Moreover, the
the extract for several days [57]. proliferation of T lymphocytes induced by MD/tumor lysate DCs was
Taken altogether, the unconclusive and in some cases conflicting higher than that induced by tumor lysate alone-stimulated DCs [63].
results account for the great heterogeneity of mushrooms, derived A similar effect can be obtained with schizophyllan [64] and with
polysaccharides, extraction techniques, biological pathways involved sparan derived from Sparassis crispa, which increased maturation and
and underline the importance of rigorous studies and trials to settle the cytokine production in murine DCs and promote T lymphocyte prolif
issue. eration and activation with release of IFN-γ [60]. A Ganoderma lucidum
extract activates DCs, induces secretion of IL-23 but not IL-12 and pro
3.3. Effects of polysaccharides on dendritic (DCs) and T cells motes the differentiation of Th17 cells. T cells produce large amounts of
IL-17 but not of IFN-γ, different from cells cultured with LPS-stimulated
DCs have an important role in immune defence, contributing to an DCs [61]. An extract from Agaricus brasiliensis induces the production
tigen presentation to cluster of differentiation (CD)4+ and CD8+ T cells, IL-6 and TNF-α from bone marrow derived DCs in a dose dependent
thus stimulating acquired immune response against pathogens and manner, in the presence of exogenous granulocyte macrophage-colony
tumor cells [58]. The immunomodulatory effects of glucans on DCs are stimulating factor (GM-CSF). In addition, DCs have a crucial role in
mediated by Dectin-1 for some compounds [59], by TLR4 for others [60] stimulating IFN-γ production by splenocytes [65].
and the intracellular signaling appears to be transduced through the The DCs and T cells-mediated immunomodulating potential of
MEK-ERK cascade [61]. The effects of glucans on DCs and T cells in vitro mushrooms is effective in murine models of colitis, where Grifola fron
are reported in Table 3 and illustrated in Fig. 3. dosa and Agaricus bisporus extracts stimulate IL-23 [66], malaria parasite
The α-glucan YM-2A, isolated from Grifola frondosa, was shown to infection, with lentinan-induced DCs maturation and Th1 immune
have an impact on maturation and function of DCs, increasing the response [67], allergy, with Th1 polarization and reduced IgE produc
expression of maturation markers, as major histocompatibility complex tion elicited by Agaricus blazei Murill extract [68]. In addition, para
(MHC) class II molecules, CD80 and CD86, in dose-dependent manner, mylon, a β-1,3-D-glucan isolated from Euglena gracilis Z, has been shown
increasing IL-12p40, TNF-α, and IL-6 production by DCs, but not IL-10 to inhibit the development of atopic dermatitis in mice models [69].
and promoting CD4 and CD8 lymphocytes proliferation and produc Through activation of DCs in the Peyer’s patches and induction of T-cell
tion of IFN-γ. In addition, it had an adjuvant effect on a vaccine against response, the oral administration of glucan fractions inhibits tumor
murine tumors, increasing IFN-γ producing T cells thus reducing tumor growth and improves survival rate in murine models of colon carcinoma
growth and improving survival in murine models [59]. and melanoma [70].
A soluble β-glucan D-fraction extracted from Grifola frondosa, in In human DCs a mushroom extract leads to significant dose-
combination with a TLR9 agonist, synergistically increases the surface dependent elevation of IL-8, CCL4, granulocyte-colony stimulating fac
expression of CD80, CD86 and MHC class II and cytokine production in tor (G-CSF), TNF-α, IL-1β and IL-6. The increase of G-CSF, TNF-α and IL-
DCs, but not IL-10, generating a polarized type 1 T cell response. As for 1β is greater than those induced by LPS, whereas IL-2, IL-8 and IFN-γ
α-glucan, it acts as an anti-tumor, increasing DCs in the tumor site and levels are similar after extract and LPS stimulation. IL-5, IL-10, IL-12 and
inducing activation of CD4 and CD8 T lymphocytes [62]. A different IL-13 are not increased by the same mushroom extracts [71]. In a
study on Grifola frondosa demonstrated that labelled MD-extract different study, human DCs analysed in vitro show no cytokine
Table 3
Effects of mushroom glucans on dendritic cells and lymphocytes.
Mushroom Type of glucan Type of Effects on DC and Other effects Ref
DCs lymphocytes
Grifola frondosa α-glucan YM-2A Murine DC maturation, CD4 and Enhanced DCs vaccine [59]
CD8 lymphocytes as an adjuvant
stimulation
Grifola frondosa Soluble β-glucan D-fraction Murine DC maturation, CD4 and Enhanced DCs anti- [62]
CD8 lymphocytes tumor effect
stimulation
Grifola frondosa MD-fraction Murine DC maturation, CD4 and Reduction of tumor [63]
CD8 lymphocytes volume
stimulation
Schizophyllum commune Schizophyllan K3-SPG Murine CD8 lymphocyte [64]
stimulation
Sparassis crispa Sparan (1,3-β-D-glucan) Murine DC maturation, T [60]
lymphocyte stimulation
Ganoderma lucidum Water soluble extract Murine DC maturation, CD4 [61]
lymphocyte stimulation
Agaricus brasiliensis 1,6-β-glucan with a small amount of 1,3- Murine Splenocyte stimulation [65]
β-glucan extract
AndoSan™ extract (82.4% from Agaricus blazei Moisture 5.8 g, protein 2.6 g, fat 0.3 g, Human Pro-inflammatory [71]
Murill, 14.7% from Hericium erinaceum and carbohydrates 89.4 g (of which β-glucan cytokine release
2.9% from G. frondosa) constitutes 2.8 g) and ash 1.9 g
Pleurotus sajor-caju β-glucan extract Human CD4 and CD8 lymphocytes IgG neutralizing [72]
stimulation antibodies to HPV (in
mice)
Legend. DCs: dendritic cells. CD: cluster of differentiation. IgG: immunoglobulin G. HPV: human papilloma virus.
5
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
production and no increase of maturation markers, albeit T cells stim [76]. The glucan-induced maturation of DCs into more immunostimu
ulated by glucans and immunoglobulin against viral antigens are latory cells is a promising tool for a more efficient immunotherapy
significantly elevated [72]. against cancer.
The molecular structure of glucans and their ability to stimulate DCs
to activate acquired immune response led to test their potential role in
enhancing vaccine delivery and efficacy. Glucans are good antigen 3.4. Effects of polysaccharides on NK cells
carriers and can be used for nasal delivery of vaccines, as they are up-
taken in the nasal-associated lymphoid tissue [73,74]. Moreover, it NK cells are part of the innate immune system and play a role in
has been shown that carbon nanotubes binding lentinan are able to in response against pathogens, in viral infected and cancer cells through
crease antigen accumulation in cells and potentiate cellular and humoral recognition and lysis of target cells and induction of cytokines able to
immunity [75]. It has also been demonstrated that the intravenous in stimulate innate and adaptive immune response [77]. Table 4 summa
jection of K3-fraction from schizophyllan, but not a TLR9 agonist alone, rizes the effect of mushroom polysaccharides on NK cells.
is accumulated in the tumor microenvironment and stimulates immu In vitro studies on murine cells show that the oral intake of an extract
nogenic cell death of malignant cells through IFN and IL-12 production combination of Grifola frondosa and Lentinula edodes is the most active in
stimulating both the cellular and humoral branch of immune reactions,
Table 4
Effects of mushroom glucans on NK cells.
Mushroom Type of glucan Type of Effects on NK cells Other effects Ref
NK
Grifola frondosa - MaitakeGold 404 (MTG404) extract Murine MTG404-Shiitake combination and MTG Increased phagocytosis (highest for [78]
and Lentinula - an α-glucan-rich whole Shiitake 404 alone activated NK. AHCC activated combination of MTG404 and Shiitake)
edodes mushroom powder from Gourmet NK only in the highest NK cells to target
Mushrooms, Inc. (Sebastopol, CA, cells ratio.
USA)
- AHCC (Amino-Up Chemical
Company, Sapporo, Japan)
- an α-glucan-rich nutraceutical
ingredient prepared by culturing
Shiitake and other Basidiomycetes
Pleurotus ostreatus Polysaccharide or glucans from Murine Increased NK cytotoxicity Decrease in malignanant cell volume and [79]
mycelia and fruit body number
Astraeus AE2 heteroglucan Murine NK activation Macrophages activation Tumor regression. [80]
hygrometricus Increased survival.
Grifola frondosa MD-fraction Murine NK activation Enhanced cisplatin antitumor and [81]
antimetastatic activity compared to cisplatin
alone. Reduced cisplatin-induced
nephrotoxicity and myelotoxicity
Trametes versicolor Mycelium and fermented substrate Human NK activation Activation of monocytes and lymphocytes [82]
Pleurotus ostreatus Extract rich in carbohydrate Human NK activation Anti-tumor activity against breast and lung cell [83]
lines
Pleurotus ostreatus Pleuran Human Increased NK cell number Reduction of upper respiratory infection [84]
symptoms. No reduction of phagocytosis in
glucan-treated group
6
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
compared to the compounds alone. The combination and Grifola extract production of antibodies [92]. The immunization with a Reishi poly
alone are able to activate NK cells [78]. Glucans isolated from the saccharide fraction induces the production of antibodies against murine
mycelia and fruit body of Pleurotus ostreatus can induce lymphocyte Lewis lung carcinoma cells, with increased antibody-mediated cyto
proliferation, macrophage activation, macrophage and NK cell mediated toxicity and reduced production of tumor-associated inflammatory
cytotoxicity. A study on Dalton’s lymphoma mice tumor model showed mediators, in particular CCL2 [93].
the ability of mycelia and fruit body glucan supplementation to inhibit
tumor growth, with increased survival, and this effect is associated with 3.6. Proteins
an enhanced immune response and apoptosis of malignant cells [79].
Similar results were obtained with a heteroglucan isolated from Astraeus As previously mentioned, mushrooms consist of polysaccharides and
hygrometricus [80]. of a proportion of proteins that are receiving a growing focus due to the
A MD-fraction from Grifola frondosa also exhibits antitumor and bioactive properties, including lectins, fungal immunomodulatory pro
antimetastatic activities. In tumor-bearing mice it effectively enhances teins (FIPs), ribosome inactivating proteins (RIPs), ribonucleases, lac
cisplatin antitumor activity compared to cisplatin alone. The fraction cases, and other proteins. These are usually isolated through a protocol
also induces IL-12 production, leading to increased NK cells activity in including different step as extraction, ammonium sulfate precipitation,
cisplatin-treated mice and mRNA expression of GM-CSF, G-CSF, chromatography and filtration [94].
macrophage-colony stimulating factor (M-CSF) and IFN-γ in spleno Lectins are non-immunoglobulin proteins or glycoproteins able to
cytes, thus reducing myelotoxicity. Of clinical relevance, it is able to specifically bind to cell surface carbohydrates, inducing cell agglutina
reduce cisplatin-induced nephrotoxicity [81]. tion. Their molecular masses, subunit number and carbohydrate speci
A study on human peripheral blood cells in vitro showed that the ficity vary among species and from different parts of the mushroom and
mycelium of Trametes versicolor and its fermented substrate activate NK the expression levels may change depending on fruit-body age, season,
cells as well as lymphocytes and monocytes [82]. An extract from location and year [95]. Lectins have antiproliferative, antitumor, anti
Pleurotus ostreatus is effective in activating human NK cells in vitro and to viral and immune stimulating properties; proteins from different
stimulate their cytotoxic activity with induction of IFN-γ against lung mushrooms have shown antitumor activity, probably by cross-linking
and breast cancer cells, with the largest effect against breast cancer cells, the altered cancer cell glycoproteins. A ricin B-like lectin from Clito
enhanced in the presence of IL-2. It also causes the up-regulation of cybe nebularis is able to induce hemoagglutination by binding to blood
KIR2DL gene expression, which stimulates cytokine and chemokine group A carbohydrates and has antiproliferative activity specific to
secretion and of NKG2D receptors on NK cells, which increased the human leukemic T cells [96]. Lectins from Pholiota adiposa have mito
production of IFN-γ [83]. genic activity toward mouse splenocytes, inhibitory effect on human
Pleuran, an insoluble β-(1,3/1,6) glucan from Pleurotus ostreatus, is immunodeficiency virus (HIV)-1 reverse transcriptase and anti
effective in reducing upper respiratory tract infections risk in athletes, as proliferative activity toward hepatoma Hep G2 cells and breast cancer
illustrated in two human studies. In a randomized trial, a group of MCF7 cells [97]. Analogous antiviral and antitumor effects have been
athletes supplemented with pleuran for 3 months have a significantly demonstrated for lectins from Russula mushroom [98,99], Lactarius
lower number of respiratory infection symptoms and increased number flavidulus [100] and Hericium erinaceum [97]. A lectin from Pleurotus
of circulating NK cells compared with a placebo-treated control group ostreatus activated TLR6 signaling pathway in DCs and increased hepa
[84]. Similarly, in a double-blind trial, twenty athletes were randomized titis B virus (HBV) specific antibody levels and T helper response,
to dietary supplementation with 100 mg of a β-glucan from Pleurotus overcoming HBV tolerance in transgenic mice [101]. A
ostreatus or placebo, once a day for 2 months. A significant reduction in mannose-binding lectin from Narcissus tazetta showed in vitro antiviral
NK cell activity is observed in the placebo group after intensive exercise, activity against several viruses in a dose-dependent manner, including
whereas no significant reduction in number and activity ensues in the influenza A (H1N1, H3N2, H5N1), influenza B viruses and human res
β-glucan group, suggesting a protective role of this mushroom extract in piratory syncytial virus [102]. Lectins can also stimulate mitosis in
modulating exercise-induced changes in NK cells in athletes [85]. lymphocytes and splenocytes, by binding to T-cell receptors, triggering
the signaling cascade and IL-2 gene expression [95].
3.5. Effects of polysaccharides on B cells While lectins do no possess enzymatic activity, FIPs are a family of
enzymes (named LZ-8, gts, jap, fve, vvo, gsi, among others), purified
Limited data are available on the effects of mushroom poly from different edible mushrooms, with a molecular weight around 13
saccharides on B lymphocytes. While no significant effect has been re kDa and including 110–114 amino acids. These share common
ported in some studies [86], some lines of evidence showed conserved motifs, essential for their functions and are studied predom
immunomodulatory properties on B cells but the net effects remain inantly for their antitumor activities, based on in vitro or murine models
unclear (Fig. 3). Zhuling polysaccharide, from the fruit bodies of Poly effects showing capability to elicit the immune response toward cancer
porus umbellatus, is constituted by a (1 → 6, 1 → 4)-linked β-D-gluco cells. Possible mechanisms of action involve T cell receptor binding,
pyranosyl backbone, substituted at O-3 position of (1 → 6)-linked activation and signal transduction with production of cytokines as IFN-γ
β-D-glucopyranosyl by (1 → 3)-linked β-D-glucopyranosyl branches and or IL-2 with consequent cytotoxic response, inhibition of telomerase
is a potent activator of B cells, macrophages and DCs in a murine model. activity and cell apoptosis [103]. FIPs are in fact able to suppress telo
The B cell stimulating properties appear to be determined by its merase, inducing premature senescence, for example in human bladder
branches, as their depletion causes a reduction of the activating effect. cancer cells [104], even in those resistant to cisplatin [105] and human
The polysaccharide also elicits a specific IgM production in mice. lung adenocarcinoma cells [106]. Another type of programmed cell
Human subjects have circulating antibodies against Zhuling backbone, death is autophagy, induced by FIPs in non-small cell lung cancer [107,
suggesting that its epitope is shared by environmental antigens, as 108]. Furthermore, the potential application of FIPs as adjuvants for
β-glucans constitute the cell wall of saprophytic bacteria and fungi, and cancer immunotherapy has been analysed, with results revealing
is able to induce an adaptive humoral responses [87]. significantly stronger adjuvant effect with enhancement of type 1 im
Other β-glucans are known to induce B cell activation, as Ganoderma mune response when administered in association with tumor antigen,
lucidum’s [88]. Moreover, crude polysaccharides from Pleurotus nebro compared to the antigen alone. Tumor-bearing mice co-immunized with
densis extracted in hot water enhances the activity of B lymphocytes an oncoprotein and a FIP had a significantly longer tumor-free survival.
[89], polysaccharides extracted from Flammulina velutipes increases FIPs can induce DCs maturation and Th1 polarization and the IFN-γ
the production of IgM and IgG [90], from Cantharellus cibarius Fr have a response elicited by CD4+ and CD8+ T lymphocytes had a crucial role in
proliferative effect on B cells [91], from Coriolus versicolor induces the the antitumor effect. In addition, the production of specific antibodies
7
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
8
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
9
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
effects in vitro, through T cell-mediated immune response [103], adju [16] S.E. Elcombe, S. Naqvi, M.W.M. Van Den Bosch, K.F. MacKenzie, F. Cianfanelli, G.
D. Brown, et al., Dectin-1 regulates IL-10 production via a MSK1/2 and CREB
vant properties on vaccine against oncoproteins [109], or induction of
dependent pathway and promotes the induction of regulatory macrophage
autophagy [107]. The potential beneficial properties in cancer include markers, PloS One 8 (2013) e60086-e.
also the reduced chemotherapy-induced myelosuppression, through [17] I.D. Iliev, V.A. Funari, K.D. Taylor, Q. Nguyen, C.N. Reyes, S.P. Strom, et al.,
glucans-mediated macrophage stimulation [34]. In the case of allergies, Interactions between commensal fungi and the C-type lectin receptor Dectin-1
influence colitis, Science 336 (2012) 1314–1317.
the type 1 T cell response polarization may account for the protection [18] C. Municio, Y. Alvarez, O. Montero, E. Hugo, M. Rodríguez, E. Domingo, et al.,
against atopy by glucans [136,138], while the pro-inflammatory activity The response of human macrophages to β-glucans depends on the inflammatory
on macrophages can have application for wound healing [35] or as milieu, PloS One 8 (2013) e62016-e.
[19] V. Vetvicka, B. Dvorak, J. Vetvickova, J. Richter, J. Krizan, P. Sima, et al., Orally
enhancer of immune response against viral or bacterial infections administered marine (1→3)-β-d-glucan Phycarine stimulates both humoral and
[36–38]. On the other hand, some evidence has shown an cellular immunity, Int. J. Biol. Macromol. 40 (2007) 291–298.
anti-inflammatory effect of mushroom extracts, as on monocytes, in [20] R. Wismar, S. Brix, H. Frøkiær, H.N. Lærke, Dietary fibers as immunoregulatory
compounds in health and disease, Ann. N. Y. Acad. Sci. 1190 (2010) 70–85.
particular when administered before M1/M2 polarization [44]. [21] C.D. Mills, Anatomy of a discovery: m1 and m2 macrophages, Front. Immunol. 6
Accordingly, there seems to be a role of mushrooms in the prevention of (2015) 212.
autoimmunity, as a protective contribution against the development of [22] L.C. Davies, S.J. Jenkins, J.E. Allen, P.R. Taylor, Tissue-resident macrophages,
Nat. Immunol. 14 (2013) 986–995.
rheumatoid arthritis has been shown [140]. This findings deserve to be [23] S. Batbayar, M.J. Kim, H.W. Kim, Medicinal mushroom lingzhi or Reishi,
study in depth, as, if confirmed, could provide an option for the pro Ganoderma lucidum (W.curt.:Fr.) P. Karst., β-glucan induces toll-like receptors
tection against autoimmunity, or for the reduction of flares in estab and fails to induce inflammatory cytokines, in: NF-κB Inhibitor-Treated
Macrophages, vol. 13, 2011, 213-25.
lished disease.
[24] K.-I. Minato, A. Ohara, M. Mizuno, A proinflammatory effect of the β-glucan from
Future studies need an improvement in the methodological quality, pleurotus cornucopiae mushroom on macrophage action 2017, Mediators
in standardization and in the size of the population analysed. This could Inflamm, 2017, p. 8402405.
allow to elucidate the real beneficial effects of mushroom compounds [25] R.H. de Melo, A.E. do Amaral, R.A. Menolli, T.S. Ayala, R. de Cassia Garcia Simao,
A.P. de Santana-Filho, et al., β-(1→3)-Glucan of the southern bracket mushroom,
and whether they could have an impact on human health. Ganoderma australe (Agaricomycetes), stimulates phagocytosis and interleukin-6
production in mouse peritoneal macrophages 18 (2016) 313–320.
[26] C. Liu, P.C.K. Cheung, Structure and immunomodulatory activity of
Funding microparticulate mushroom sclerotial β-glucan prepared from Polyporus
rhinocerus, J. Agric. Food Chem. 67 (2019) 9070–9078.
This research did not receive any specific grant from funding [27] E.R. Carbonero, A.C. Ruthes, C.S. Freitas, P. Utrilla, J. Gálvez, E.V.d. Silva, et al.,
Chemical and biological properties of a highly branched β-glucan from edible
agencies in the public, commercial, or not-for-profit sectors. mushroom Pleurotus sajor-caju, Carbohydr. Polym. 90 (2012) 814–819.
Legend. PCPS: Pleurotus citrinopileatus derived polysaccharide. ROS: [28] J. Yan, Z. Han, Y. Qu, C. Yao, D. Shen, G. Tai, et al., Structure elucidation and
reactive oxygen species. NO: nitric oxyde. IL: interleukin. TNF: tumor immunomodulatory activity of a β-glucan derived from the fruiting bodies of
Amillariella mellea, Food Chem. 240 (2018) 534–543.
necrosis factor. IFN: interferon. LPS: lipopolysaccharide. CCL: C–C motif
[29] W. Chanput, M. Reitsma, L. Kleinjans, J.J. Mes, H.F.J. Savelkoul, H.J. Wichers,
chemokine ligand. Red arrows indicate pro-inflammatory stimuli. Blu β-Glucans are involved in immune-modulation of THP-1 macrophages, Mol. Nutr.
arrows indicate anti-inflammatory stimuli. Food Res. 56 (2012) 822–833.
[30] F. Cui, L. Jiang, L. Qian, W. Sun, T. Tao, X. Zan, et al., A macromolecular α-glucan
from fruiting bodies of Volvariella volvacea activating RAW264. 7 macrophages
through MAPKs pathway, Carbohydr. Polym. 230 (2020) 115674.
Declaration of competing interest [31] S. Ghosh, S. Khatua, K. Acharya, Crude polysaccharide from a wild mushroom
enhances immune response in murine macrophage cells by TLR/NF-κB pathway,
None. J. Pharm. Pharmacol. 71 (2019) 1311–1323.
[32] S. Khatua, K. Acharya, Alkaline extractive crude polysaccharide from Russula
senecis possesses antioxidant potential and stimulates innate immunity response,
References J. Pharm. Pharmacol. 69 (2017) 1817–1828.
[33] Q. Liu, L. Dong, H. Li, J. Yuan, Y. Peng, S. Dai, Lentinan mitigates therarubicin-
induced myelosuppression by activating bone marrow-derived macrophages in an
[1] D.L. Hawksworth, Mushrooms: the extent of the unexplored potential 3 (2001) 5.
MAPK/NF-κB-dependent manner, Oncol. Rep. 36 (2016) 315–323.
[2] S. Wasser, Medicinal mushroom science: current perspectives, advances,
[34] H. Lin, E. de Stanchina, X.K. Zhou, F. Hong, A. Seidman, M. Fornier, et al.,
evidences, and challenges, Biomed. J. 37 (2014).
Maitake beta-glucan promotes recovery of leukocytes and myeloid cell function in
[3] S. Wasser, Medicinal mushroom science: history, current status, future trends,
peripheral blood from paclitaxel hematotoxicity, Cancer Immunol. Immunother.
and unsolved problems, Int. J. Med. Mushrooms 12 (2010) 1–16.
59 (2010) 885–897.
[4] A.T. Borchers, J.S. Stern, R.M. Hackman, C.L. Keen, M.E. Gershwin, Mushrooms,
[35] A.H. Kwon, Z. Qiu, M. Hashimoto, K. Yamamoto, T. Kimura, Effects of medicinal
tumors, and immunity, PSEBM (Proc. Soc. Exp. Biol. Med.) 221 (1999) 281–293.
mushroom (Sparassis crispa) on wound healing in streptozotocin-induced
[5] K.K. Pennerman, G. Yin, J.W. Bennett, Health effects of small volatile compounds
diabetic rats, Am. J. Surg. 197 (2009) 503–509.
from East asian medicinal mushrooms, Mycobiology 43 (2015) 9–13.
[36] D. Muramatsu, A. Iwai, S. Aoki, H. Uchiyama, K. Kawata, Y. Nakayama, et al.,
[6] L. Capasso, 5300 years ago, the Ice Man used natural laxatives and antibiotics,
β-Glucan derived from Aureobasidium pullulans is effective for the prevention of
Lancet 352 (1998) 1864.
influenza in mice, PloS One 7 (2012) e41399–e.
[7] M.E. Valverde, T. Hernández-Pérez, O. Paredes-López, Edible mushrooms:
[37] S.P. Kim, E. Moon, S.H. Nam, M. Friedman, Hericium erinaceus mushroom
improving human health and promoting quality life, Int. J. Microbiol. 2015
extracts protect infected mice against Salmonella typhimurium-induced liver
(2015) 376387.
damage and mortality by stimulation of innate immune cells, J. Agric. Food
[8] W.M. Breene, Nutritional and medicinal value of specialty mushrooms, J. Food
Chem. 60 (2012) 5590–5596.
Protect. 53 (1990) 883–894.
[38] A.M.M. Basso, R.J.A. De Castro, T.B. de Castro, H.I. Guimarães, V.L.P. Polez, E.
[9] X.-M. Wang, J. Zhang, L.-H. Wu, Y.-L. Zhao, T. Li, J.-Q. Li, et al., A mini-review of
R. Carbonero, et al., Immunomodulatory activity of β-glucan-containing
chemical composition and nutritional value of edible wild-grown mushroom from
exopolysaccharides from Auricularia auricular in phagocytes and mice infected
China, Food Chem. 151 (2014) 279–285.
with Cryptococcus neoformans, Med. Mycol. 58 (2019) 227–239.
[10] N. Dalonso, G.H. Goldman, R.M.M. Gern, β-(1→3),(1→6)-Glucans: medicinal
[39] J. Fang, Y. Wang, X. Lv, X. Shen, X. Ni, K. Ding, Structure of a β-glucan from
activities, characterization, biosynthesis and new horizons, Appl. Microbiol.
Grifola frondosa and its antitumor effect by activating Dectin-1/Syk/NF-κB
Biotechnol. 99 (2015) 7893–7906.
signaling, Glycoconj. J. 29 (2012) 365–377.
[11] A.C. Ruthes, F.R. Smiderle, M. Iacomini, d-Glucans from edible mushrooms: a
[40] S.P. Kim, M.Y. Kang, J.H. Kim, S.H. Nam, M. Friedman, Composition and
review on the extraction, purification and chemical characterization approaches,
mechanism of antitumor effects of Hericium erinaceus mushroom extracts in
Carbohydr. Polym. 117 (2015) 753–761.
tumor-bearing mice, J. Agric. Food Chem. 59 (2011) 9861–9869.
[12] P. Manzi, L. Pizzoferrato, Beta-glucans in edible mushrooms, Food Chem. 68
[41] H. Lei, S. Guo, J. Han, Q. Wang, X. Zhang, W. Wu, Hypoglycemic and
(2000) 315–318.
hypolipidemic activities of MT-α-glucan and its effect on immune function of
[13] I.A. Schepetkin, M.T. Quinn, Botanical polysaccharides: macrophage
diabetic mice, Carbohydr. Polym. 89 (2012) 245–250.
immunomodulation and therapeutic potential, Int. Immunopharm. 6 (2006)
[42] J. Li, C. Cai, M. Zheng, J. Hao, Y. Wang, M. Hu, et al., Alkaline extraction,
317–333.
structural characterization, and bioactivities of (1→6)-β-d-glucan from Lentinus
[14] V. Vetvicka, L. Vannucci, P. Sima, J. Richter, Beta glucan: supplement or drug?
edodes, Molecules 24 (2019) 1610.
From laboratory to clinical trials, Molecules 24 (2019) 1251.
[43] B. Mallard, D.N. Leach, H. Wohlmuth, J. Tiralongo, Synergistic immuno-
[15] A. Plato, S.E. Hardison, G.D. Brown, Pattern recognition receptors in antifungal
modulatory activity in human macrophages of a medicinal mushroom
immunity, Semin. Immunopathol. 37 (2015) 97–106.
10
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
formulation consisting of Reishi, Shiitake and Maitake, PloS One 14 (2019) infection by evoking adaptive immune-responses, Int. Immunopharm. 9 (2009)
e0224740-e. 455–462.
[44] K.-i. Minato, L.C. Laan, I. van Die, M. Mizuno, Pleurotus citrinopileatus [68] L.K. Ellertsen, G. Hetland, An extract of the medicinal mushroom Agaricus blazei
polysaccharide stimulates anti-inflammatory properties during monocyte-to- Murill can protect against allergy, Clin. Mol. Allergy 7 (2009) 6.
macrophage differentiation, Int. J. Biol. Macromol. 122 (2019) 705–712. [69] A. Sugiyama, S. Hata, K. Suzuki, E. Yoshida, R. Nakano, S. Mitra, et al., Oral
[45] J. van de Velde, R.H.P. Wilbers, L.B. Westerhof, D.R. van Raaij, I. Stavrakaki, A.S. administration of paramylon, a β-1,3-D-glucan isolated from Euglena gracilis Z
M. Sonnenberg, et al., Assessing the immunomodulatory potential of high- inhibits development of atopic dermatitis-like skin lesions in NC/Nga mice,
molecular-weight extracts from mushrooms; an assay based on THP-1 J. Vet. Med. Sci. 72 (2010), 755-63.
macrophages, J. Sci. Food Agric. 95 (2015) 344–350. [70] Y. Masuda, Y. Nakayama, A. Tanaka, K. Naito, M. Konishi, Antitumor activity of
[46] F.R. Smiderle, C.H. Baggio, D.G. Borato, A.P. Santana-Filho, G.L. Sassaki, orally administered maitake α-glucan by stimulating antitumor immune response
M. Iacomini, et al., Anti-inflammatory properties of the medicinal mushroom in murine tumor, PloS One 12 (2017) e0173621-e.
Cordyceps militaris might be related to its linear (1→3)-β-D-glucan, PloS One 9 [71] D.T. Førland, E. Johnson, A.M.A. Tryggestad, T. Lyberg, G. Hetland, An extract
(2014) e110266–e. based on the medicinal mushroom Agaricus blazei Murill stimulates monocyte-
[47] H. Ahn, E. Jeon, J.-C. Kim, S.G. Kang, S.-I. Yoon, H.-J. Ko, et al., Lentinan from derived dendritic cells to cytokine and chemokine production in vitro, Cytokine
shiitake selectively attenuates AIM2 and non-canonical inflammasome activation 49 (2010) 245–250.
while inducing pro-inflammatory cytokine production, Sci. Rep. 7 (2017) 1314. [72] P.E. Roopngam, Increased response of human T-lymphocytes by dendritic cells
[48] P. Kankkunen, L. Teirilä, J. Rintahaka, H. Alenius, H. Wolff, S. Matikainen, (1,3)- pulsed with HPV16E7 and pleurotus sajor-caju-β-glucan (PBG), Iranian J.
β-Glucans activate both dectin-1 and NLRP3 inflammasome in human Immunol. 15 (2018) 246–255.
macrophages, J. Immunol. 184 (2010) 6335. [73] K. Baert, B.G. De Geest, H. De Greve, E. Cox, B. Devriendt, Duality of β-glucan
[49] W. Liu, J. Gu, J. Qi, X.-N. Zeng, J. Ji, Z.-Z. Chen, et al., Lentinan exerts synergistic microparticles: antigen carrier and immunostimulants, Int. J. Nanomed. 11
apoptotic effects with paclitaxel in A549 cells via activating ROS-TXNIP-NLRP3 (2016) 2463–2469.
inflammasome, J. Cell Mol. Med. 19 (2015) 1949–1955. [74] E. Cevher, S.K. Salomon, A. Makrakis, X.W. Li, S. Brocchini, H.O. Alpar,
[50] C.-H. Su, M.-K. Lu, T.-J. Lu, M.-N. Lai, L.-T. Ng, A (1→6)-branched (1→4)-β-d- Development of chitosan–pullulan composite nanoparticles for nasal delivery of
glucan from Grifola frondosa inhibits lipopolysaccharide-induced cytokine vaccines: optimisation and cellular studies, J. Microencapsul. 32 (2015) 755–768.
production in RAW264.7 macrophages by binding to TLR2 rather than dectin-1 or [75] J. Xing, Z. Liu, Y. Huang, T. Qin, R. Bo, S. Zheng, et al., Lentinan-modified carbon
CR3 receptors, J. Nat. Prod. 83 (2020) 231–242. nanotubes as an antigen delivery system modulate immune response in vitro and
[51] E.J. Murphy, C. Masterson, E. Rezoagli, D. O’Toole, I. Major, G.D. Stack, et al., in vivo, ACS Appl. Mater. Interfaces 8 (2016) 19276–19283.
β-Glucan extracts from the same edible shiitake mushroom Lentinus edodes [76] Y. Kitahata, T. Kanuma, M. Hayashi, N. Kobayashi, K. Ozasa, T. Kusakabe, et al.,
produce differential in-vitro immunomodulatory and pulmonary cytoprotective Circulating nano-particulate TLR9 agonist scouts out tumor microenvironment to
effects - implications for coronavirus disease (COVID-19) immunotherapies, Sci. release immunogenic dead tumor cells, Oncotarget 7 (2016) 48860–48869.
Total Environ. 732 (2020) 139330. [77] J. Pahl, A. Cerwenka, Tricking the balance: NK cells in anti-cancer immunity,
[52] A. Jedinak, S. Dudhgaonkar, Q.-L. Wu, J. Simon, D. Sliva, Anti-inflammatory Immunobiology 222 (2017) 11–20.
activity of edible oyster mushroom is mediated through the inhibition of NF-κB [78] V. Vetvicka, J. Vetvickova, Immune-enhancing effects of maitake (Grifola
and AP-1 signaling, Nutr. J. 10 (2011) 52. frondosa) and shiitake (Lentinula edodes) extracts, Ann. Transl. Med. 2 (2014)
[53] X. Xu, M. Yasuda, S. Nakamura-Tsuruta, M. Mizuno, H. Ashida, β-Glucan from 14.
Lentinus edodes inhibits nitric oxide and tumor necrosis factor-α production and [79] K.S.P. Devi, B. Behera, D. Mishra, T.K. Maiti, Immune augmentation and Dalton’s
phosphorylation of mitogen-activated protein kinases in lipopolysaccharide- Lymphoma tumor inhibition by glucans/glycans isolated from the mycelia and
stimulated murine RAW 264.7 macrophages, J. Biol. Chem. 287 (2012) 871–878. fruit body of Pleurotus ostreatus, Int. Immunopharm. 25 (2015) 207–217.
[54] C.-F. Chen, C.-H. Su, M.-N. Lai, L.-T. Ng, Differences in water soluble non- [80] S.K. Mallick, S. Maiti, S.K. Bhutia, T.K. Maiti, Antitumor properties of a
digestible polysaccharides and anti-inflammatory activities of fruiting bodies heteroglucan isolated from Astraeus hygrometricus on Dalton’s lymphoma
from two cultivated Xylaria nigripes strains, Int. J. Biol. Macromol. 116 (2018) bearing mouse, Food Chem. Toxicol. 48 (2010) 2115–2121.
728–734. [81] Y. Masuda, M. Inoue, A. Miyata, S. Mizuno, H. Nanba, Maitake β-glucan enhances
[55] L. Byung Ryong, lt, gt sup, K. So Young, K. Dae Won, et al., Agrocybe chaxingu therapeutic effect and reduces myelosupression and nephrotoxicity of cisplatin in
polysaccharide prevent inflammation through the inhibition of COX-2 and NO mice, Int. Immunopharm. 9 (2009) 620–626.
production, BMB Rep 42 (2009) 794–799. [82] K.F. Benson, P. Stamets, R. Davis, R. Nally, A. Taylor, S. Slater, et al., The
[56] H. Cui, X. Zhu, Z. Huo, B. Liao, J. Huang, Z. Wang, et al., A β-glucan from Grifola mycelium of the Trametes versicolor (Turkey tail) mushroom and its fermented
frondosa effectively delivers therapeutic oligonucleotide into cells via dectin-1 substrate each show potent and complementary immune activating properties in
receptor and attenuates TNFα gene expression, Int. J. Biol. Macromol. 149 (2020) vitro, BMC Compl. Alternative Med. 19 (2019) 342.
801–808. [83] N.M. El-Deeb, H.I. El-Adawi, A.E.A. El-Wahab, A.M. Haddad, H.A. El Enshasy, Y.-
[57] E. Johnson, D.T. Førland, L. Sætre, S.V. Bernardshaw, T. Lyberg, G. Hetland, W. He, et al., Modulation of NKG2D, KIR2DL and cytokine production by
Effect of an extract based on the medicinal mushroom Agaricus blazei Murill on pleurotus ostreatus glucan enhances natural killer cell cytotoxicity toward cancer
release of cytokines, chemokines and leukocyte growth factors in human blood ex cells, Front Cell Dev Biol 7 (2019) 165.
vivo and in vivo, Scand. J. Immunol. 69 (2009) 242–250. [84] K. Bergendiova, E. Tibenska, J. Majtan. Pleuran (β-glucan from Pleurotus
[58] A. Waisman, D. Lukas, B.E. Clausen, N. Yogev, Dendritic cells as gatekeepers of ostreatus) supplementation, cellular immune response and respiratory tract
tolerance, Semin. Immunopathol. 39 (2017) 153–163. infections in athletes, Eur. J. Appl. Physiol. 111 (2011) 2033–2040.
[59] Y. Masuda, Y. Nakayama, T. Mukae, A. Tanaka, K. Naito, M. Konishi, Maturation [85] M. Bobovčák, R. Kuniaková, J. Gabriž, J. Majtán, Effect of Pleuran (β-glucan from
of dendritic cells by maitake α-glucan enhances anti-cancer effect of dendritic cell Pleurotus ostreatus) supplementation on cellular immune response after intensive
vaccination, Int. Immunopharm. 67 (2019) 408–416. exercise in elite athletes, Appl. Physiol. Nutr. Metabol. 35 (2010) 755–762.
[60] H.S. Kim, J.Y. Kim, H.S. Ryu, H.-G. Park, Y.O. Kim, J.S. Kang, et al., Induction of [86] B. Kaleta, A. Górski, R. Zagożdżon, M. Cieślak, J. Kaźmierczak-Barańska,
dendritic cell maturation by β-glucan isolated from Sparassis crispa, Int. B. Nawrot, et al., Selenium-containing polysaccharides from Lentinula
Immunopharm. 10 (2010) 1284–1294. edodes—biological activity, Carbohydr. Polym. 223 (2019) 115078.
[61] H. Yoshida, M. Suzuki, R. Sakaguchi, I. Tani, H. Kotani, N. Shudo, et al., [87] H. Dai, X.-Q. Han, F.-Y. Gong, H. Dong, P.-F. Tu, X.-M. Gao, Structure elucidation
Preferential induction of Th17 cells in vitro and in vivo by Fucogalactan from and immunological function analysis of a novel β-glucan from the fruit bodies of
Ganoderma lucidum (Reishi), Biochem. Biophys. Res. Commun. 422 (2012) Polyporus umbellatus (Pers.) Fries, Glycobiology 22 (2012) 1673–1683.
174–180. [88] B.-M. Shao, H. Dai, W. Xu, Z.-B. Lin, X.-M. Gao, Immune receptors for
[62] Y. Masuda, D. Nawa, Y. Nakayama, M. Konishi, H. Nanba, Soluble β-glucan from polysaccharides from Ganoderma lucidum, Biochem. Biophys. Res. Commun. 323
Grifola frondosa induces tumor regression in synergy with TLR9 agonist via (2004) 133–141.
dendritic cell-mediated immunity, J. Leukoc. Biol. 98 (2015) 1015–1025. [89] R.M. Collard, H. Boter, R.A. Schoevers, R.C. Oude Voshaar, Prevalence of frailty
[63] Y. Masuda, H. Inoue, H. Ohta, A. Miyake, M. Konishi, H. Nanba, Oral in community-dwelling older persons: a systematic review, J. Am. Geriatr. Soc. 60
administration of soluble β-glucans extracted from Grifola frondosa induces (2012) 1487–1492.
systemic antitumor immune response and decreases immunosuppression in [90] W.-H. Wang, J.-S. Zhang, T. Feng, J. Deng, C.-C. Lin, H. Fan, et al., Structural
tumor-bearing mice, Int. J. Canc. 133 (2013) 108–119. elucidation of a polysaccharide from Flammulina velutipes and its
[64] K. Kobiyama, B. Temizoz, T. Kanuma, K. Ozasa, M. Momota, T. Yamamoto, et al., immunomodulation activities on mouse B lymphocytes, Sci. Rep. 8 (2018) 3120.
Species-dependent role of type I IFNs and IL-12 in the CTL response induced by [91] D. Zhao, X. Ding, Y. Hou, W. Hou, L. Liu, T. Xu, et al., Structural characterization,
humanized CpG complexed with β-glucan, Eur. J. Immunol. 46 (2016) immune regulation and antioxidant activity of a new heteropolysaccharide from
1142–1151. Cantharellus cibarius Fr, Int. J. Mol. Med. 41 (2018) 2744–2754.
[65] D. Yamanaka, R. Tada, Y. Adachi, K.-i. Ishibashi, M. Motoi, Y. Iwakura, et al., [92] S.-f. Yang, T.-f. Zhuang, Y.-m. Si, K.-y. Qi, J. Zhao, Coriolus versicolor mushroom
Agaricus brasiliensis-derived β-glucans exert immunoenhancing effects via a polysaccharides exert immunoregulatory effects on mouse B cells via membrane
dectin-1-dependent pathway, Int. Immunopharm. 14 (2012) 311–319. Ig and TLR-4 to activate the MAPK and NF-κB signaling pathways, Mol. Immunol.
[66] L.C. Chandra, D. Traoré, C. French, D. Marlow, J. D’Offay, S.L. Clarke, et al., 64 (2015) 144–151.
White button, portabella, and shiitake mushroom supplementation up-regulates [93] S.-F. Liao, C.-H. Liang, M.-Y. Ho, T.-L. Hsu, T.-I. Tsai, Y.S.Y. Hsieh, et al.,
interleukin-23 secretion in acute dextran sodium sulfate colitis C57BL/6 mice and Immunization of fucose-containing polysaccharides from Reishi mushroom
murine macrophage J.744.1 cell line, Nutr. Res. 33 (2013) 388–396. induces antibodies to tumor-associated Globo H-series epitopes, Proc. Natl. Acad.
[67] L.-d. Zhou, Q.-h. Zhang, Y. Zhang, J. Liu, Y.-m. Cao, The shiitake mushroom- Sci. U. S. A. 110 (2013) 13809–13814.
derived immuno-stimulant lentinan protects against murine malaria blood-stage [94] X. Xu, H. Yan, J. Chen, X. Zhang, Bioactive proteins from mushrooms, Biotechnol.
Adv. 29 (2011) 667–674.
11
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
[95] M.A.A. Hassan, R. Rouf, E. Tiralongo, T.W. May, J. Tiralongo, Mushroom lectins: [121] J.E. Libbey, I. Tsunoda, R.S. Fujinami, Studies in the modulation of experimental
specificity, structure and bioactivity relevant to human disease, Int. J. Mol. Sci. autoimmune encephalomyelitis, J. Neuroimmune Pharmacol. 5 (2010) 168–175.
16 (2015) 7802–7838. [122] H. Yoshitomi, N. Sakaguchi, K. Kobayashi, G.D. Brown, T. Tagami, T. Sakihama,
[96] J. Pohleven, N. Obermajer, J. Sabotic, S. Anzlovar, K. Sepcić, J. Kos, et al., et al., A role for fungal {beta}-glucans and their receptor Dectin-1 in the induction
Purification, characterization and cloning of a ricin B-like lectin from mushroom of autoimmune arthritis in genetically susceptible mice, J. Exp. Med. 201 (2005)
Clitocybe nebularis with antiproliferative activity against human leukemic T 949–960.
cells, Biochim. Biophys. Acta 1790 (2009) 173–181. [123] Y. Maeda, T. Kurakawa, E. Umemoto, D. Motooka, Y. Ito, K. Gotoh, et al.,
[97] Y. Li, G. Zhang, T.B. Ng, H. Wang, A novel lectin with antiproliferative and HIV-1 Dysbiosis contributes to arthritis development via activation of Autoreactive T
reverse transcriptase inhibitory activities from dried fruiting bodies of the cells in the intestine, Arthritis & Rheumatology 68 (2016) 2646–2661.
monkey head mushroom Hericium erinaceum, J. Biomed. Biotechnol. 2010 [124] M. Ruutu, G. Thomas, R. Steck, M.A. Degli-Esposti, M.S. Zinkernagel,
(2010) 716515. K. Alexander, et al., β-glucan triggers spondylarthritis and Crohn’s disease–like
[98] G. Zhang, J. Sun, H. Wang, T.B. Ng, First isolation and characterization of a novel ileitis in SKG mice, Arthritis Rheum. 64 (2012) 2211–2222.
lectin with potent antitumor activity from a Russula mushroom, Phytomedicine [125] Y. Zhang, M. Zhang, Y. Jiang, X. Li, Y. He, P. Zeng, et al., Lentinan as an
17 (2010) 775–781. immunotherapeutic for treating lung cancer: a review of 12 years clinical studies
[99] S. Zhao, Y. Zhao, S. Li, J. Zhao, G. Zhang, H. Wang, et al., A novel lectin with in China, J. Canc. Res. Clin. Oncol. 144 (2018) 2177–2186.
highly potent antiproliferative and HIV-1 reverse transcriptase inhibitory [126] X. Jin, J. Ruiz Beguerie, D.M.-Y. Sze, G.C.F. Chan, Ganoderma lucidum (Reishi
activities from the edible wild mushroom Russula delica, Glycoconj. J. 27 (2010) mushroom) for cancer treatment 4, Cochrane Database Syst Rev, 2016.
259–265. CD007731-CD.
[100] Y. Wu, H. Wang, T.B. Ng, Purification and characterization of a lectin with [127] P. Twardowski, N. Kanaya, P. Frankel, T. Synold, C. Ruel, S.K. Pal, et al., A phase I
antiproliferative activity toward cancer cells from the dried fruit bodies of trial of mushroom powder in patients with biochemically recurrent prostate
Lactarius flavidulus, Carbohydr. Res. 346 (2011) 2576–2581. cancer: roles of cytokines and myeloid-derived suppressor cells for Agaricus
[101] M. He, D. Su, Q. Liu, W. Gao, Y. Kang, Mushroom lectin overcomes hepatitis B bisporus-induced prostate-specific antigen responses, Cancer 121 (2015)
virus tolerance via TLR6 signaling, Sci. Rep. 7 (2017) 5814. 2942–2950.
[102] L.S.M. Ooi, W.-S. Ho, K.L.K. Ngai, L. Tian, P.K.S. Chan, S.S.M. Sun, et al., [128] W.Y. Chay, C.K. Tham, H.C. Toh, H.Y. Lim, C.K. Tan, C. Lim, et al., Coriolus
Narcissus tazetta lectin shows strong inhibitory effects against respiratory versicolor (Yunzhi) use as therapy in advanced hepatocellular carcinoma patients
syncytial virus, influenza A (H1N1, H3N2, H5N1) and B viruses, J. Biosci. 35 with poor liver function or who are unfit for standard therapy, J. Alternative
(2010) 95–103. Compl. Med. 23 (2017) 648–652.
[103] Q.-Z. Li, Y.-Z. Zheng, X.-W. Zhou, Fungal immunomodulatory proteins: [129] M.-Y. Tsai, Y.-C. Hung, Y.-H. Chen, Y.-H. Chen, Y.-C. Huang, C.-W. Kao, et al.,
characteristic, potential antitumor activities and their molecular mechanisms, A preliminary randomised controlled study of short-term Antrodia cinnamomea
Drug Discov. Today 24 (2019) 307–314. treatment combined with chemotherapy for patients with advanced cancer, BMC
[104] M.H. Han, D.-S. Lee, J.-W. Jeong, S.-H. Hong, I.-W. Choi, H.-J. Cha, et al., Compl. Alternative Med. 16 (2016) 322.
Fucoidan induces ROS-dependent apoptosis in 5637 human bladder cancer cells [130] J.-M. Tangen, A. Tierens, J. Caers, M. Binsfeld, O.K. Olstad, A.-M.S. Trøseid, et al.,
by downregulating telomerase activity via inactivation of the PI3K/Akt signaling Immunomodulatory effects of the Agaricus blazei Murrill-based mushroom
pathway, Drug Dev. Res. 78 (2017) 37–48. extract AndoSan in patients with multiple myeloma undergoing high dose
[106] C.-H. Liao, Y.-M. Hsiao, C.-H. Lin, C.-S. Yeh, J.C.-H. Wang, C.-H. Ni, et al., chemotherapy and autologous stem cell transplantation: a randomized, double
Induction of premature senescence in human lung cancer by fungal blinded clinical study, BioMed Res. Int. 2015 (2015) 718539.
immunomodulatory protein from Ganoderma tsugae, Food Chem. Toxicol. 46 [131] S.P. Therkelsen, G. Hetland, T. Lyberg, I. Lygren, E. Johnson, Cytokine levels after
(2008) 1851–1859. consumption of a medicinal Agaricus blazei murill-based mushroom extract,
[107] I.L. Hsin, C.-C. Ou, T.-C. Wu, M.-S. Jan, M.-F. Wu, L.-Y. Chiu, et al., GMI, an AndoSan™, in patients with Crohn’s disease and ulcerative colitis in a
immunomodulatory protein from Ganoderma microsporum, induces autophagy randomized single-blinded placebo-controlled study, Scand. J. Immunol. 84
in non-small cell lung cancer cells, Autophagy 7 (2011) 873–882. (2016) 323–331.
[108] I.L. Hsin, G.-T. Sheu, M.-S. Jan, H.-L. Sun, T.-C. Wu, L.-Y. Chiu, et al., Inhibition of [132] D.T. Førland, E. Johnson, L. Sætre, T. Lyberg, I. Lygren, G. Hetland, Effect of an
lysosome degradation on autophagosome formation and responses to GMI, an extract based on the medicinal mushroom Agaricus blazei Murill on expression of
immunomodulatory protein from Ganoderma microsporum, Br. J. Pharmacol. cytokines and calprotectin in patients with ulcerative colitis and crohn’s disease,
167 (2012) 1287–1300. Scand. J. Immunol. 73 (2011) 66–75.
[109] Y. Ding, S.V. Seow, C.H. Huang, L.M. Liew, Y.C. Lim, I.C. Kuo, et al., [133] S.P. Therkelsen, G. Hetland, T. Lyberg, I. Lygren, E. Johnson, Effect of the
Coadministration of the fungal immunomodulatory protein FIP-Fve and a tumour- medicinal Agaricus blazei murill-based mushroom extract, AndoSanTM, on
associated antigen enhanced antitumour immunity, Immunology 128 (2009) symptoms, fatigue and quality of life in patients with Crohn’s disease in a
e881–e894. randomized single-blinded placebo controlled study, PloS One 11 (2016)
[110] F. Sheu, P.-J. Chien, K.-Y. Hsieh, K.-L. Chin, W.-T. Huang, C.-Y. Tsao, et al., e0159288-e.
Purification, cloning, and functional characterization of a novel [134] S.P. Therkelsen, G. Hetland, T. Lyberg, I. Lygren, E. Johnson, Effect of a medicinal
immunomodulatory protein from Antrodia camphorata (bitter mushroom) that Agaricus blazei murill-based mushroom extract, AndoSan™, on symptoms,
exhibits TLR2-dependent NF-κB activation and M1 polarization within murine fatigue and quality of life in patients with ulcerative colitis in a randomized
macrophages, J. Agric. Food Chem. 57 (2009) 4130–4141. single-blinded placebo controlled study, PloS One 11 (2016) e0150191-e.
[111] J.H. Wong, H. Bao, T.B. Ng, H.H.L. Chan, C.C.W. Ng, G.C.W. Man, et al., New [135] F. Mahmood, G. Hetland, I. Nentwich, M.R. Mirlashari, R. Ghiasvand, L.S.
ribosome-inactivating proteins and other proteins with protein H. Nissen-Meyer, Agaricus blazei-based mushroom extract supplementation to
synthesis–inhibiting activities, Appl. Microbiol. Biotechnol. 104 (2020) birch allergic blood donors: a randomized clinical trial, Nutrients 11 (2019) 2339.
4211–4226. [136] M. Jesenak, S. Urbancek, J. Majtan, P. Banovcin, J. Hercogova, β-Glucan-based
[112] E.F. Fang, T.B. Ng, Ribonucleases of different origins with a wide spectrum of cream (containing pleuran isolated from pleurotus ostreatus) in supportive
medicinal applications, Biochim. Biophys. Acta Rev. Canc 1815 (2011) 65–74. treatment of mild-to-moderate atopic dermatitis, J. Dermatol. Treat. 27 (2016)
[113] T.B. Ng, R.C.F. Cheung, J.H. Wong, Y.S. Chan, X. Dan, W. Pan, et al., Fungal 351–354.
proteinaceous compounds with multiple biological activities, Appl. Microbiol. [137] M. Jesenak, I. Urbancikova, P. Banovcin, Respiratory tract infections and the role
Biotechnol. 100 (2016) 6601–6617. of biologically active polysaccharides in their management and prevention,
[114] E. Guillamón, A. García-Lafuente, M. Lozano, M. DÁrrigo, M.A. Rostagno, Nutrients 9 (2017) 779.
A. Villares, et al., Edible mushrooms: role in the prevention of cardiovascular [138] M. Jesenak, M. Hrubisko, J. Majtan, Z. Rennerova, P. Banovcin, Anti-allergic
diseases, Fitoterapia 81 (2010) 715–723. effect of pleuran (β-glucan from pleurotus ostreatus) in children with recurrent
[115] P. Saiki, Y. Kawano, L.J.L.D. Van Griensven, K. Miyazaki, The anti-inflammatory respiratory tract infections, Phytother Res. 28 (2014) 471–474.
effect of Agaricus brasiliensis is partly due to its linoleic acid content, Food & [139] E.K. Li, L.-S. Tam, C.K. Wong, W.C. Li, C.W.K. Lam, S. Wachtel-Galor, et al., Safety
Function 8 (2017) 4150–4158. and efficacy of Ganoderma lucidum (lingzhi) and San Miao San supplementation
[116] A. Grzywacz, J.G. Argasinska, K. Kala, W. Opoka, B. Muszynska, Anti- in patients with rheumatoid arthritis: a double-blind, randomized, placebo-
inflammatory activity of biomass extracts of the bay mushroom, Imleria badia controlled pilot trial, Arthritis Care Res. 57 (2007) 1143–1150.
(Agaricomycetes), in RAW 264, 7 Cells 18 (2016) 769–779. [140] J. He, Y. Wang, M. Feng, X. Zhang, Y.-B. Jin, X. Li, et al., Dietary intake and risk of
[117] B. Muszyńska, A. Grzywacz-Kisielewska, K. Kała, J. Gdula-Argasińska, Anti- rheumatoid arthritis-a cross section multicenter study, Clin. Rheumatol. 35
inflammatory properties of edible mushrooms: a review, Food Chem. 243 (2018) (2016) 2901–2908.
373–381. [141] D. Collado Mateo, F. Pazzi, F.J. Domínguez Muñoz, J.P. Martín Martínez, P.
[118] O.T. Burton, P. Zaccone, J.M. Phillips, H. De La Peña, Z. Fehérvári, M. Azuma, et R. Olivares, N. Gusi, et al., Ganoderma lucidum improves physical fitness in
al., Roles for TGF-β and programmed cell death 1 ligand 1 in regulatory T cell women with fibromyalgia, Nutr. Hosp. 32 (2015) 2126–2135.
expansion and diabetes suppression by zymosan in nonobese diabetic mice, [142] F. Corana, V. Cesaroni, B. Mannucci, R.M. Baiguera, A.M. Picco, E. Savino, et al.,
J. Immunol. 185 (2010) 2754–2762. Array of metabolites in Italian Hericium erinaceus mycelium, primordium, and
[119] H. Li, P. Gonnella, F. Safavi, G. Vessal, B. Nourbakhsh, F. Zhou, et al., Low dose sporophore, Molecules 24 (2019) 3511.
zymosan ameliorates both chronic and relapsing experimental autoimmune [143] G. Koutrotsios, N. Kalogeropoulos, P. Stathopoulos, A.C. Kaliora, G.I. Zervakis,
encephalomyelitis, J. Neuroimmunol. 254 (2013) 28–38. Bioactive compounds and antioxidant activity exhibit high intraspecific
[120] L. Li, G. Wu, B.Y. Choi, B.G. Jang, J.H. Kim, G.H. Sung, et al., A mushroom extract variability in Pleurotus ostreatus mushrooms and correlate well with cultivation
Piwep from Phellinus igniarius ameliorates experimental autoimmune performance parameters, World J. Microbiol. Biotechnol. 33 (2017) 98.
encephalomyelitis by inhibiting immune cell infiltration in the spinal cord,
BioMed Res. Int. 2014 (2014) 218274.
12
F. Motta et al. Journal of Autoimmunity 117 (2021) 102576
[144] M. Sari, A. Prange, J.I. Lelley, R. Hambitzer, Screening of beta-glucan contents in [148] C. Yin, X. Fan, Z. Fan, D.-F. Shi, H. Gao, Effects of various treatments on
commercially cultivated and wild growing mushrooms, Food Chem. 216 (2017) extraction of the main bioactive components and determination of biological
45–51. activity of extracts from the caterpillar medicinal mushroom Cordyceps militaris
[145] D. Satria, S. Tamrakar, H. Suhara, S. Kaneko, K. Shimizu, Mass spectrometry- (Ascomycetes) 20 (2018) 873–885.
based untargeted metabolomics and α-glucosidase inhibitory activity of lingzhi [149] T. Bakir, M. Karadeniz, S. Unal, Investigation of antioxidant activities of Pleurotus
(Ganoderma lingzhi) during the developmental stages, Molecules 24 (2019) 2044. ostreatus stored at different temperatures, Food Sci. Nutr. 6 (2018) 1040–1044.
[146] X. Ren, J. Wang, L. Huang, K. Cheng, M. Zhang, H. Yang, Comparative studies on [150] M. Jayachandran, J. Chen, S.S.M. Chung, B. Xu, A critical review on the impacts
bioactive compounds, ganoderic acid biosynthesis, and antioxidant activity of of β-glucans on gut microbiota and human health, J. Nutr. Biochem. 61 (2018)
pileus and stipes of lingzhi or Reishi medicinal mushroom, Ganoderma lucidum 101–110.
(Agaricomycetes), Fruiting Body at Different Growth Stages 22 (2020) 133–144. [151] M. Jayachandran, J. Xiao, B. Xu, A critical review on health promoting benefits of
[147] X. Li, L. Wang, Effect of extraction method on structure and antioxidant activity of edible mushrooms through gut microbiota, Int. J. Mol. Sci. 18 (2017) 1934.
Hohenbuehelia serotina polysaccharides, Int. J. Biol. Macromol. 83 (2016)
270–276.
13