Next Article in Journal
Human Palatine Tonsils Are Linked to Alzheimer’s Disease through Function of Reservoir of Amyloid Beta Protein Associated with Bacterial Infection
Next Article in Special Issue
Febrile Seizures Cause Depression and Anxiogenic Behaviors in Rats
Previous Article in Journal
Reprogramming of Amino Acid Metabolism Differs between Community-Acquired Pneumonia and Infection-Associated Exacerbation of Chronic Obstructive Pulmonary Disease
Previous Article in Special Issue
Applying a Fast-Scan Cyclic Voltammetry to Explore Dopamine Dynamics in Animal Models of Neuropsychiatric Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Monkey Head Mushroom and Memory Enhancement in Alzheimer’s Disease

1
School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
2
Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(15), 2284; https://doi.org/10.3390/cells11152284
Submission received: 18 June 2022 / Revised: 15 July 2022 / Accepted: 19 July 2022 / Published: 24 July 2022

Abstract

:
Alzheimer’s disease (AD) is a neurodegenerative disorder, and no effective treatments are available to treat this disorder. Therefore, researchers have been investigating Hericium erinaceus, or the monkey head mushroom, an edible medicinal mushroom, as a possible treatment for AD. In this narrative review, we evaluated six preclinical and three clinical studies of the therapeutic effects of Hericium erinaceus on AD. Preclinical trials have successfully demonstrated that extracts and bioactive compounds of Hericium erinaceus have potential beneficial effects in ameliorating cognitive functioning and behavioral deficits in animal models of AD. A limited number of clinical studies have been conducted and several clinical trials are ongoing, which have thus far shown analogous outcomes to the preclinical studies. Nonetheless, future research on Hericium erinaceus needs to focus on elucidating the specific neuroprotective mechanisms and the target sites in AD. Additionally, standardized treatment parameters and universal regulatory systems need to be established to further ensure treatment safety and efficacy. In conclusion, Hericium erinaceus has therapeutic potential and may facilitate memory enhancement in patients with AD.

1. Introduction

Neurodegenerative disorders are an umbrella term for a wide range of conditions. Alzheimer’s disease (AD) constitutes nearly 50–70% of all neurodegenerative disorder-related illnesses for dementia [1]. With the increasing older population worldwide, it is predicted that over 130 million individuals over the age of 65 will develop AD by 2050 [2]. Alzheimer’s disease causes considerable social and economic burdens, especially for patients, their families, and caregivers. Accumulation of amyloid-beta (Aβ) plaques and hyperphosphorylation of tau leading to neurofibrillary tangles (NFTs) are characteristic hallmarks of AD, resulting in key symptoms such as memory loss, difficulty in recalling recent incidents, and overall behavior changes [3,4,5]. In AD, the symptoms can range from mild to severe. In mild condition, AD patients have a greater tendency to misplace items and show poor judgment. As AD progresses, individuals can have difficulty identifying their family members, cannot learn new information, and lose the ability to categorize thoughts. Individuals with severe AD can lose the ability to communicate and are prone to infections and chronic inflammation, eventually leading to death [1]. Early diagnosis and treatment are important, as progression of the disease leads to irreversible cognitive decline.
Although there are some treatments currently available for AD, the multifactorial and complex nature of this disease have led to questions about their effectiveness [6]. Current AD therapeutics primarily focus on alleviating the symptoms and slowing down the disease progression [7,8]. Acetylcholinesterase inhibitors (AChEl) including donepezil and galantamine and memantine (N-methyl-D-aspartic acid (NMDA) receptor antagonists) can potentially treat the cognitive decline in AD [9]. It has been shown that AChEls can increase transmission of acetylcholine in the brain including the cerebral cortex via preventing acetylcholine breakdown by cholinesterase in synapses [10], whereas NMDA receptor antagonists can protect neuronal cells against excitotoxicity due to excessive activation of NMDA receptors [11]. Monoclonal antibodies such as Solanezumab and Aducanumab target aggregated Aβ peptides to remove excess Aβ plaques [10,12]. Several potential approaches for blocking tau hyperphosphorylation have also been developed [13]. Despite these current treatments, new and safer drugs are needed, as patients on AD medications often experience adverse side effects [14,15]. In addition, these medications only inhibit the symptoms of AD for a limited period and have low efficacy in decelerating the progressive deterioration. Further research should focus on the therapeutic management of AD with consideration of ethical concerns and international collaboration of interdisciplinary research to tackle the mechanisms of AD [14,15].
The limitations of current treatments can be countered by the use of complementary and alternative medicines. Highly nutritious culinary and medicinal mushrooms are rich sources of antioxidants that have been shown to delay the progression of neurodegenerative disorders [16,17]. Mushrooms contain a large number of bioactive compounds including alkaloids, flavonoids, polyketides, steroids, terpenes, polysaccharides, proteins, micronutrients, and unsaturated fatty acids. The phenolic compounds have the highest antioxidant activities and are mainly responsible for attenuating oxidative damage. Hericium erinaceus (HE), also known as the monkey’s head mushroom, lion’s mane mushroom, or Yamabushitake, is commonly found in East Asia [18] (Figure 1). It is well-known for its diverse therapeutic activities, including neuroprotection [16,17,19,20] and neuroregeneration [21,22,23,24], which are attributed to its neurogenesis, antioxidative, and anti-neuroinflammatory functions [17,19,20,23,25].
The structural characterization and isolation of compounds in the polysaccharide fraction (mainly beta-glucans) and secondary metabolites (e.g., hericenones, erinacines, hericerins, resorcinols, monoterpenes, diterpenes, steroids, and volatile aromatic compounds) of HE fruiting bodies and mycelia have been shown to have immune-modulatory activities, neurogenesis, antioxidative, anti-inflammatory, and anti-cancer properties [17,19,20,23]. These neurotrophic compounds have potential as treatments for AD, due to their potential to penetrate through the blood–brain barrier [26]. Additionally, recent studies have reported that HE can modulate neural activity, such as increasing synaptic plasticity, reducing apoptosis, decreasing Aβ plaques, and inhibiting acetylcholinesterase (AChE) and BACE1 [27], supporting its therapeutic potential in the management of AD. This narrative review discusses recent clinical and animal studies on the therapeutic efficacy of HE for AD.

2. Etiology of Alzheimer’s Disease

Alzheimer’s disease is associated with complex multifaceted pathologies, leading to neuronal dysfunction and degeneration. In AD, the deposition of senile plaques and intraneuronal NFTs tend to form in stereotypical neuroanatomical patterns throughout the brain [3,4,5]. The accumulation of Aβ peptides arises from the sequential cleavage of amyloid precursor protein (APP) by β-Site APP-cleaving enzyme 1 (BACE1) to generate soluble C99 and APPβ fragments, followed by further cleavage by γ-secretase [3,4,5]. It has also been shown that mutations in Presenilin-1 and -2 in AD can modify the activity of γ-secretase, which increases the accumulation of Aβ peptides [28]. The Aβ peptides form soluble oligomers that aggregate and deposit as senile plaques, suppressing the growth and differentiation of neuronal cells. The downregulation of β-catenin by oligomeric Aβ senile plaques consequently impairs Wnt/β-catenin signaling, which is an essential signaling pathway of neurogenesis [29]. Inhibition of this pathway increases the production of Aβ senile plaques and tau hyperphosphorylation through activation of GSK3β signaling [30]. Abnormally hyperphosphorylated tau form NFTs that also contribute to the suppression of neurogenesis [31]. The NFTs disrupt microtubule-associated proteins that regulate microtubule dynamics, which impairs axonal transport and leads to the progressive loss of neuronal cells [32,33].
Oxidative stress, which is the overproduction of reactive oxygen species (ROS), has been implicated in the progression of AD. ROS participates in various cellular functions, including gene transcription and signal transduction; however, overproduction of ROS and free radicals lead to the deterioration of cellular antioxidant defense mechanisms [34]. Intracellular ROS is observed to be generated by various endogenous and exogenous sources. For instance, mitochondria generate approximately 90% of cellular ROS. The mitochondrial electron transport chain (ETC) is considered to be the primary endogenous source of ROS including superoxide (O2−•), hydrogen peroxide (H2O2), singlet oxygen (1O2), nitric oxide (NO), and hydroxyl radical (OH•), which are involved in cellular functioning and signaling [35,36].
The univalent metabolic reduction status of oxygen can lead to the production of potentially harmful ROS [35]. Mutations and metabolite deficits can result in a major shift in the redox balance and mitochondrial dysfunction, which inhibit enzyme and protein synthesis, leading to excessive ROS production. In the brains of AD patients, increased oxygen consumption, elevated polyunsaturated lipid levels, and impaired antioxidant defense systems lead to further generation of ROS. Neuron and glial cells are vulnerable to oxidative stress and the accumulation of excess ROS modifies the function of major biomolecules, including nucleic acids (DNA, RNA), proteins, and lipids [35]. In addition, alterations in the activity of antioxidant enzymes (e.g., glutathione peroxidase, superoxide dismutase) have been shown to decrease the proliferation rate of neuronal cells in AD [37].
The accumulation of NFTs and Aβ deposition have demonstrated to activate neuroinflammation, which plays a harmful role in AD by causing the increased inflammatory mediators expression in the vicinity of Aβ peptide deposits and NFTs [27]. The neuroinflammation defense system is supported by glial cells such as microglia that act as the primary defense line in the CNS. Glial cells phagocytize exogenous and harmful substances and also produce inflammatory cytokines including interferon-gamma (IFN-γ), interleukin one beta (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α) [27,38,39]. Chronic and sustained activation of microglia causes overproduction of neurotoxins and cytokines, resulting in neuroinflammation, triggering a series of intrinsic apoptotic pathways, and gradually leading to neuronal cell death [27,39]. Insult to the central nervous system also results in molecular, structural, and functional modifications to astrocytes, a process known as reactive gliosis [4]. Reactive gliosis has beneficial effects during the initial phases of AD development by eliminating neurotoxic peptides and inducing inflammatory signals [4,40]. However, sustained and excessive activation of pro-inflammatory mediators and ROS levels cause adverse neurotoxic effects on neighboring neurons [27]. Research has shown inflammatory mediators have effects in the pathogenesis of AD through the generation of Aβ peptides from amyloid precursor proteins via β- and γ-secretase enzymes [27,41].
Lastly, altered neurotransmitter expression and loss of neuronal synapses are closely associated with AD pathogenesis [5]. Acetylcholine (ACh), an essential neurotransmitter that contributes to learning and memory, is found to be deficient in AD [5,42,43]. A study by Slotkin et al. found that reduced choline acetyltransferase levels resulted in decreased synthesis of ACh. This further reduced its uptake by acetylcholine receptors (AChRs), leading to the loss of memory function [42,43]. The presence of Aβ peptides and NFTs in AD was also found to influence the expression of acetylcholinesterase, which further affects the synthesis of ACh [44]. Reduced levels of AChRs have also been observed in the mild condition of AD, leading to a decreased uptake of ACh [45]. Glutamate, an essential excitatory neurotransmitter associated with learning and memory, is also impaired in AD. Glutamate uptake or recycling can be impaired in AD due to alterations in the expression of glutamate transporters. It is well-established that increased presynaptic release of glutamate and failure of glutamate uptake lead to the activation of NMDA ionotropic receptors [46]. Excessive activation of NMDA receptors leads to excitotoxicity and decreases neuronal cell proliferation and survival [47]. Mitochondrial dysfunction is also known to be a contributing factor to glutamate excitotoxicity in AD [46].

3. Preclinical Studies of Hericium erinaceus in AD

Hericium erinaceus has recently attracted considerable research attention due to its therapeutic potential in treating the debilitating conditions in AD [48,49,50]. This review analyzes six essential animal studies on the therapeutic potential of HE in AD models (Table 1). The commonly used HE products include erinacine A-enriched HE-mycelia (HE-A), ethanol extracts of erinacine A-enriched HE-mycelia (HE-Et), and erinacine S-enriched HE mycelia (HE-S). Culinary and medicinal mushrooms are consumed by humans as a functional food and as a nutraceutical source. Hence, the use of mushrooms with beneficial health effects in humans requires in vivo experimentation to ensure their safety and efficacy. Aqueous extracts of HE rich in polysaccharides were shown to promote neuronal growth and differentiation [17,23,51,52,53] via the reduction in endoplasmic reticulum stress-induced cell atrophy, expression of neurotrophic factors in astrocytes, and decrease in neurodegenerative-induced cell atrophy [51,54,55,56]. The mechanism of action of HE has been further investigated in several preclinical studies. The effects of HE were investigated in Aβ (25–35)-treated mice that had learning and memory deficits, as seen by reduced discrimination in the novel object recognition (NOR) test and poor working memory in the Y-maze test [48]. Oral administration of HE rescued the learning and memory impairment in Aβ (25–35)-injected mice through elevation of nerve growth factor (NGF) mRNA abundance in the hippocampus, suggesting that it can enhance hippocampal neurogenesis. However, this study only investigated NGF expression and did not report the physiological effects of HE treatment on learning and memory. Moreover, the short duration of the HE treatment might not provide insights on its long-term beneficial effects.
Tsai-Teng et al. (2016) further investigated the effects of HE-A and HE-Et on APPswe/PS1dE9 mice, which resulted in a reduction in Aβ buildup in the cerebral cortex in a short-term and long-term administration treatment paradigm [49]. After 30 days of oral administration, there were decreased astrocytes and plaque-activated microglia in the cerebral and hippocampal regions. Additionally, there was an increase in the ratio of NGF to neural growth factor precursor, indicating hippocampal neurogenesis, which correlated with the ameliorated nesting behavior of the AD mice. Long-term administration demonstrated improved nesting behavior, which has been previously associated with progressive cognitive impairment in APPswe/PS1dE9 mice, suggesting the potential role of HE as a treatment for AD [57].
Tzeng et al. (2018) also conducted a short-term and long-term administration treatment paradigm that reported the effects of ethanol extracts for HE-A and HE-S in APPswe/PS1dE9 mice [50]. After 1 month of oral administration, both HE-A and HE-S showed beneficial effects, including inhibiting cerebral plaque growth and reducing glial cell activation. Moreover, HE-A reduced insoluble Aβ fragments and the C-terminal fragment of APP, leading to hippocampal neurogenesis and attenuating Aβ production. After 100 days, the increase in neurogenesis was found to be correlated with recovering behavioral and memory deficits in the nesting, burrowing, and Morris water maze (MWM) tests.
Zhang et al. (2016) investigated the mechanisms of polysaccharide-enriched aqueous extract of HE mycelia in an AD model of mice which was generated by subcutaneously injecting D-galactose and intragastrically administrating aluminum chloride (AlCl3) [47]. The brains of AD mice demonstrated reduced AChE and acetyltransferase (ChAT) levels, resulting in learning and memory deficits [5]. Mice treated with the HE extracts depicted dose-dependent increased concentrations of AChE and ChAT in the hypothalamus and blood serum, which correlated with enhanced learning and memory in the behavioral tests, suggesting that the neuroprotective effects of polysaccharide-enriched HE is mediated through the cholinergic signaling pathway.
A recent study by Cordaro et al. (2021) demonstrated the effects of HE in a Wistar rat model of AD induced by AlCl3, which exhibited AlCl3 accumulation in the hippocampus [58]. Treatment with HE ameliorated the AlCl3 accumulation, improved behavioral deficits, and enhanced hippocampal neurogenesis, leading to reduced levels of tau phosphorylation, APP overexpression, and Aβ accumulation. Furthermore, the HE treatment was found to mitigate oxidative stress through suppressing NLRP3 inflammasome activation. These results indicate that HE also has anti-oxidative and anti-inflammatory activity.
Lee et al. (2021) expanded the previous research on the effects of HE-A on brain aging, learning, and memory [50,59]. They found that a preparation of HE-A significantly improved cognitive function in active and passive avoidance behavioral tests and delayed cognitive degeneration due to aging. In addition, HE-A significantly decreased the levels of induced nitric oxidase synthase (iNOS), thiobarbituric acid-reactive substances (TBARS), and 8-hydroxy-2′-deoxyguanosine (8-OHdG) in a dose-dependent manner, leading to the attenuation of oxidative stress. The findings support that HE has beneficial anti-oxidative and anti-inflammatory properties.
Table 1. Preclinical studies of Hericium erinaceus in AD animal models.
Table 1. Preclinical studies of Hericium erinaceus in AD animal models.
AuthorsAnimal ModelsTreatment MethodBehavioral TestBehavioral OutcomeMechanism and Physiological Effect
Mori et al., 2011 [48]5-week-old male ICR mice with Aβ (25–35)
and Aβ (35–25)
10 μg of amyloid β (25–35) peptide administered intracerebroventricularly on days 7 and 14 and fed with HE diet (powdered mixture of normal diet and HE), containing 5.5% of (w/w) for 23 daysY-Maze test
NOR
No significant difference observed in alternation behavior between Aβ (25–35) and Aβ (35–25) group
HE increased exploration time for novel object than for familiar object in Aβ (25–35) mice, but not Aβ (35–25) mice
Increased hippocampal NGF mRNA expression
Tsai-Teng et al., 2016 [49]5-month-old female APPswe/PS1dE9 double transgenic miceShort-term: Oral administration of HE-A and HE-Et (300 mg/kg/day) for 30 days
Long-term: Oral administration of HE-My (300 mg/kg/day) for 70–90 days
NestingHE-My for 81 days improved nesting behaviorsHE-A or HE-Et for 30 days:
Eliminated Aβ plaque burden
Prevented recruitment and activation of plaque-associated microglia and astrocytes
Promoted proliferation of neuron progenitors
Increased neuronal proliferation in the dentate gyrus
Tzeng et al., 2018 [50]5-month-old female APPswe/PS1dE9 double transgenic miceShort-term: HE-A or HE-S (30 mg/kg/day) administered through gavage with vehicle for 30 days
Long-term: Oral administration of HE-A (10 and 30 mg/kg/day) for 100 days
Burrowing
Nesting
MWM
HE-A ameliorated learning and spatial memory during the probe trial
Deficits in spontaneous burrowing behavior significantly recovered at both 10 and 30 mg/kg of HE-A
Impaired nesting behavior significantly recovered at 30 mg/kg of HE-A
HE-A and HE-S decreased Aβ plaque burden and increased cerebral Aβ degradation
HE-A decreased Aβ accumulation by inhibiting Aβ production in the cerebrum
HE-A and HE-S reduced activation of glial cells in the cerebrum
HE-A and HE-S promoted neurogenesis and dendritic complexity in the hippocampus
Zhang et al., 2016 [47]10-week-old Balb/c female mice with 120 mg/kg of D-gal 20 mg/kg of kg of AlCl3
Subcutaneous injection of 120 mg/kg of D-gal and intragastric administration of 20 mg/kg of AlCl3 once per day for 10 weeks
Intragastric administration of polysaccharide-enriched aqueous extract of HE mycelia at dose of 0.3, 1.0, and 3.0 g/kg for 4 weeks
Autonomic activities test
MWM
Fatigue rotarod test
HE enhanced vertical and horizontal movements in the autonomic activity test
HE ameliorated rotarod test endurance time
HE reduced MWM escape latency time
Increasing the dose of HE-enhanced AChE and ChAT concentrations in the serum and hypothalamus
Cordaro et al., 2021 [58]6–8-week-old male Wistar rats with AlCl3Intraperitoneally administered 70 mg/kg of AlCl3 daily for 6 weeks
Control + HE:
Oral administration of HE (200 mg/kg) daily by gavage
AD + HE:
Oral administration of HE (200 mg/kg) daily by gavage
MWM
EPM
NOR
HE increased animal permanence in target quadrant
HE increased time of novel object recognition with high discrimination ratio
HE reduced AlCl3-induced CA1 neuronal degeneration
HE increased Nrf2 expression in the hippocampus
HE increased antioxidant defense including SOD, CAT, and GSH levels
HE reduced NLRP3 inflammasome activation
HE decreased phosphorylated Tau, APP overexpression, and Aβ aggregation
Lee et al., 2021 [59]3-month-old male and female (SAMP8) miceLow-dose group (108 mg/kg/bw/day), intermediate-dose group (215 mg/kg/bw/day), and high-dose group (431 mg/kg/bw/day) of oral HE-A administration for 13 weeksPassive Avoidance Task
Active Shuttle Avoidance Task
HE-A significantly increased number of avoidance responses
Latency time after training increased for passive avoidance test in HE-A groups
HE-A lowered iNOS expression, lowering oxidative stress/inflammation
HE-A decreased TBARS levels, decreasing lipid peroxidation
HE-A resulted in a downward trend in Aβ plaque (%)
Abbreviations: HE, Hericium erinaceus; APPswe, Amyloid precursor protein; PS1dE9, Presenilin-1; BrdU, Bromodeoxyuridine; HE-A, erinacine A-enriched Hericium erinaceus mycelia; ADL, Activities of daily living; HE-Et, ethanol extract of erinacine A-enriched Hericium erinaceus mycelium; HE-S, ethanol extract of erinacine S-enriched Hericium erinaceus mycelium; MWM, Morris water maze; Aβ, Amyloid-beta; Balb/c, Bagg and albino; AlCl3, Aluminum; AChE, Acetylcholinesterase; ChAT, Choline acetyltransferase; EPM, Elevated plus maze; NOR, Novel objection recognition; CA1, Carbonic anhydrase 1; Nrf2, Nuclear factor-erythroid 2-related factor 2; SOD, Superoxide dismutase; CAT, Catalase; GSH, Glutathione; NLRP3, NLR family pyrin domain containing 3; NGF, Nerve growth factor; mRNA, Messenger RNA; SAMP8, Senescence accelerated mouse prone 8; iNOS, Induced nitric oxidase synthase; TBARS, Thiobarbituric acid-reactive substances.

4. Clinical Studies of Hericium erinaceus in AD

Only three clinical trials have thus far investigated the potential therapeutic effects of HE on AD in humans (Table 2). In a completed clinical trial conducted by Li et al. (2020), the safety and efficacy of HE-A were investigated in patients with a mild form of AD [60]. Participants were administered daily for 49 weeks with 350 mg mycelia-based capsules of erinacine A. The treatment group depicted a notable improvement in the activity of daily living (ADL), cognitive ability, and mini-mental state scores. Moreover, HE-A treatment improved contrast sensitivity in the ophthalmologic examination compared to the placebo group. Although HE-A was largely found to be safe and effective, four participants dropped out of the study due to adverse reactions, including nausea, abdominal pain, and skin rash. Clinical studies with a much larger sample size are required to further verify the neurocognitive benefits of HE-A in AD patients.
A double-blind, parallel group study by Mori et al. (2009) investigated the effects of oral administration of four 250 mg capsules containing 96% HE three times daily for 16 weeks in Japanese women and men with mild cognitive impairment. The treatment group demonstrated significant improvement in cognitive functioning scores compared to the placebo group with no apparent side effects [61]. Although there was a positive correlation between the treatment and improved cognitive function, this only lasted during the drug administration period and cognitive function scores decreased thereafter, which suggests the need for long-term HE use for the management of AD.
The most recent study was a double-blind, parallel group clinical trial by Saitsu et al. examining the effects of the consumption of four HE supplements containing 0.8 g of powdered fruiting bodies daily for 12 weeks. Cognitive abilities were assessed by the Mini-Mental State Examination (MMSE), standard verbal paired-associate learning, and Benton visual retention tests [62]. The HE treatment was found to prevent short-term memory decline and improve cognitive function in the MMSE, indicating the beneficial effects on neural network regeneration and its overall safety.

5. Mechanisms of Hericium erinaceus in AD

Several erinacines and hericenones have been isolated from the fruiting bodies and mycelia of HE, respectively [63]. Among them, 15 erinacines and cyathane diterpenoids were reported to possess various biological activities. Erinacines A–I were demonstrated to have neuroprotective properties through enhancing the release of neurotrophic factors, increasing the expression of insulin-degrading enzymes (erinacines A and S), reducing Aβ aggregation, and managing neuropathic pain (erinacine E) (Figure 2) [63]. The majority of hericenones were demonstrated to have been correlated with improved cognitive function through the activation of NGF synthesis in astrocytes, whereas erinacine B was found to prevent thrombosis, increase cerebral blood flow, and confer protection against cerebrovascular risk and vascular dementia [62,64].

5.1. Anti-Amyloidogenic Functions

Hericium erinaceus was reported to have anti-amyloid properties in reducing Aβ synthesis and accumulation and protecting neuronal cells against Aβ cytotoxicity [28]. Multiple mechanisms have been implicated in the clearance of Aβ plaque, including a reduction in CTF-β, SDS-soluble Aβ1-40, and SDS-insoluble Aβ levels [50]. Treatment with EAHEM reduced levels of Aβ1-42, which is the variant most prone to aggregation. Moreover, HE was found to prevent the deposition of Aβ peptides through the proteolytic degradation of Aβ and APP intracellular domain (AICD) by insulin-degrading enzyme (IDE) [65]. Farris et al. (2004) reported that IDE is a key proteolytic enzyme in Aβ reduction. Rats with partial loss-of-function mutation of IDE and an IDE-knockout mouse model demonstrated enhanced Aβ accumulation in the cerebral region [66]. In addition, a mouse model with AD-associated ApoE4 allele showed reduced levels of IDE associated with increased Aβ [67]. Remarkably, Tzeng et al. (2018) found that both HE-A and HE-S were able to increase the expression of IDE in AD animal models accompanied by a reduction in Aβ, as seen in the immunohistochemical analysis [50].

5.2. Anti-Oxidative Function

Several studies have suggested that the neuroprotective effects of HE result from upregulated antioxidant enzymes (e.g., glutathione peroxidase, catalase, and SOD) and reduced MDA levels that are implicated in the cellular defense mechanisms against ROS [68]. Furthermore, HE was shown to exert its antioxidant effects through the regulation of the transcriptional activity of nuclear factor-erythroid 2-related factor 2 (Nrf2) [69]. The Nrf2 signaling pathway regulates genes encoding various proteins that function as endogenous stress–response proteins, antioxidant enzymes, and redox-maintaining factors [58].
The antioxidant capacity of HE has been demonstrated in several preclinical animal studies. In a study by Lee et al. (2021), administration of increasing concentrations of EAHEM in SAMP8 mice over 13 weeks restored the level of TBARS, which is an index of lipid peroxidation [59]. This restoration is important considering that the long-term accumulation of lipid peroxidation is a key contributor to the aging brain and cognitive deterioration [70]. Besides, an ethanol extract of HE was also found to reduce apoptotic activity by inhibiting Bax/Bcl-2 and caspase-3 signaling pathways in a cellular model of glutamate-induced oxidative stress [71].

5.3. Anti-Neuroinflammation

Recently, Cordaro et al. (2021) demonstrated the anti-neuroinflammatory effects of HE by ameliorating NLRP3 inflammasome activation, which was found to involve the antioxidant properties of HE [58]. The inflammasome complex consists of various proteins, including DAMPS or PAMPS receptor (damage- or pathogen-derived molecular patterns), NLRP3 (NLR family pyrin domain containing 3), and pro-caspase-1 activated through ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) [58,72]. The NLRP3 inflammasome can sense a wide range of stimuli to trigger inflammation, mediating the activation of DAMPs or PAMPs, recruitment of ASC, and cleavage of pro-caspase-1 (pro-IL1β and pro-IL18) to generate pro-inflammatory cytokines [58,73]. The findings by Cordaro et al. (2021) revealed the anti-inflammatory mechanisms through the downregulation of the inflammasome network by decreasing the expression levels of ASC, NLRP3, and pro-caspase-1. Additionally, HE was also shown to inhibit the activation of NF-kB, a pro-inflammatory transcription factor [58].
In addition, HE was found to reduce the inflammatory responses by regulating iNOS expression. Three nitric oxide synthase (NOS) isoforms (i.e., neuronal NOS (nNOS), endothelial NOS (eNOS), and inducible NOS (iNOS)) [59], and among these isoforms, increased iNOS expression has been correlated with oxidative stress and inflammatory processes [74]. Lee et al. (2021) observed a reduction in iNOS expression in mice administered EAHEM, which suggests that its neuroprotective effects were mediated through the attenuation of inflammation and oxidative stress.

5.4. Neurotrophic Mechanisms

Hericium erinaceus has been shown to stimulate the release of neurotrophic factors, including NGF and brain-derived neurotrophic factors, which are known to regulate the development, maintenance, function, and survival of neuronal cells [54]. Apart from being the major players in neuroplasticity, these neurotrophic factors can also activate neurogenesis and protect neuronal cells against apoptosis. HE extracts stimulate NGF release by promoting NGF mRNA expression in astrocytes via the c-jun N-terminal kinase signaling [75]. The increased levels of NGF released from astrocytes transmit into the nerve cells and have been associated with neurogenesis and neuroplasticity in the hippocampus, pituitary glands, and cerebral cortex [16,76]. The binding of NGF to tropomyosin receptor kinase A (TrkA) receptors results in the activation of extracellular signal-regulated protein kinase (Erk)-cyclic adenosine monophosphate (cAMP)-response element-binding protein (CREB) signaling cascade, which modulates proliferation, maintenance, and memory development in neural precursor cells [76]. Furthermore, NGF-mediated neuronal differentiation also promoted an extensive mitochondrial remodeling [77] and increased fusion proteins (Mfn2 and Opa1), Drp1-dependent mitochondrial fission, activation of Sirt3 and PPARγ, and mtTFA transcription factors, ultimately controlling bioenergetic capacity. Martorana et al. (2018) reported that NGF was important for mitochondrial remodeling and contributed to neurogenesis and nerve regeneration [77].
Various studies have shown that HE treatments can have long-lasting effects on increasing Ki67-positive, PCNA-positive, and BrdU immunoreactive cells in the dentate gyrus of the hippocampus, leading to the development of neural progenitor cells in the hippocampus [16,78]. Current evidence suggests that the regulation of hippocampal neurogenesis by HE involves NGF by increasing its mRNA and protein expression levels, which also demonstrates the ability of HE bioactive compounds to pass through the blood–brain barrier [16,78].

5.5. Neurotransmission

The mechanism by which HE modulates the expression of neurotransmitters has been investigated in preclinical studies. Treatment with HE was found to improve cholinergic function by enhancing ACh and choline acetyltransferase levels in AD mouse models [47]. Brandalise et al. (2017) found that dietary HE supplementation enhanced the release of glutamate neurotransmitter from the hippocampal mossy fiber terminals, as evident by the increased spontaneous excitatory activities in the mossy fiber-CA3 synapses that were found to be dependent on glutamate release [79]. Further studies are required to examine the effects of HE on other memory-related neurotransmitters to better understand its modulating pathways.
Overall, the results of these studies indicate that HE treatments improved memory, which was accompanied with enhanced hippocampal neurogenesis and modulation of the anti-amyloidogenic, anti-oxidative, anti-neuroinflammatory, and neurotransmitter pathways (Figure 3).

6. Limitations and Future Perspective

Hericium erinaceus and its bioactive compounds have been shown to target neuropathological hallmarks associated with AD and has potential for treating the debilitating symptoms. Nevertheless, the precise mechanisms and therapeutic outcomes of HE in AD remain unclear and will require further research. For example, whether these compounds can cross the blood–brain barrier will require the investigation of their physicochemical and pharmacokinetic properties [16].
Considering only erinacines A and S have been revealed to possess beneficial effects in AD, there needs to be more research on other erinacines and cyathane diterpenoids to facilitate the development of new therapeutics. For this reason, bioreactor designs and scale-up principles could be ideal technologies to produce pure mycelial materials without any feedstock residue. Such processes would ensure that high-quality HE mycelia can be consistently produced by controlling the substrate, HE strain, growth conditions, and post-processing.
On the contrary, as a complementary and alternative medicine, HE may elicit its neuroprotective effects synergistically through multiple compounds and via multiple targets. Many herbal formulations and alternative medicines act synergistically through the various constituents to elicit their therapeutic effects [80,81]. However, the safety and effectiveness of some HE formulations have been questioned, mainly due to concerns about the proprietary formulas from different manufacturers [82,83]. Therefore, guidelines on good pharmacovigilance practices and monitoring the safety of HE formulations, as well as ethical concerns on memory modulation, are required to achieve quality compliance in clinical trials [14,15].
Recent progress in nanotechnology [84] and neuromodulation techniques [85,86,87,88,89] could pave the way for revolutionizing the development of compounds to tackle dementia. In particular, one could combine HE treatment with an invasive/non-invasive brain stimulation approach to enhance the therapeutic potential of memory function in AD patients [85,86,87,88,89]. In this respect, the development of nanotherapeutics with multi-functionalities has considerable potential to bridge the gap between the challenges associated with current therapeutics and their clinical application as treatments for AD. Biosynthesis of HE nanoparticles can enhance the delivery or transportation of bioactive compounds by preventing drug resistance, through increasing their bioavailability at the target sites and bioactivity that results in prolonged action of sustained drug release.
The therapeutic effects of HE have mostly been tested in preclinical animal models with only a few clinical studies. To overcome such limitations, different preparations and formulations of HE should be incorporated into phase 1 clinical trials to evaluate their safety in healthy volunteers. Presently, although medicinal mushrooms are readily available, information regarding their dosage, preparation, and manufacturing processes may differ significantly among manufacturers. The lack of standardized parameters in terms of dosage, adverse effects, and active ingredients may hamper conducting clinical studies and affect the reliability and validity of the outcomes [90]. Furthermore, a universal regulatory system needs to be established to ensure the safety and efficacy of phytopharmaceuticals, as evidence-based verification of the activity of alternative medicines including medicinal mushrooms is often not available.

7. Conclusions

Several preclinical and clinical investigations have established the therapeutic potential of HE in ameliorating behavior and cognitive function deficits, and overall beneficial effect in delaying AD pathogenesis. However, present research on the effects of HE, particularly their mechanisms of action, is still in the early phase. Further studies are required to establish the efficacy and safety of these compounds and to understand their specific mechanisms with interdisciplinary research collaboration of international societies [14,15].

Author Contributions

Conceptualization, original draft preparation, and revision: Y., W.S.Y. and L.W.L. Data collection and preparation of tables/figures: Y. and K.H.W. Supervision and manuscript editing: C.W.L., L.W.L. and K.H.W. Intellectual inputs and manuscript review: Y., W.S.Y., M.L.F., C.W.L., L.W.L. and K.H.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by University of Malaya Faculty of Medicine Research Grant GPF003C-2019.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The figures of this review paper were constructed using BioRender.com (accessed on 18 July 2022). The research activities were conducted at the Neuromodulation Laboratory (www.drlimlab.com) (accessed on 18 July 2022) of The University of Hong Kong. This work was funded by University of Malaya Faculty of Medicine Research Grant GPF003C-2019.

Conflicts of Interest

The authors declare no conflict of interest or financial conflict that may bias the data interpretation and presentation of this work.

References

  1. Kumar, A.; Sidhu, J.; Goyal, A.; Tsao, J.W. Alzheimer Disease. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2021. [Google Scholar]
  2. Yuan, J.; Zhang, Z.; Wen, H.; Hong, X.; Hong, Z.; Qu, Q.; Tang, M.; Wu, J.; Xu, Q.; Li, H.; et al. Incidence of dementia and subtypes: A cohort study in four regions in China. Alzheimer’s Dement. 2016, 12, 262–271. [Google Scholar] [CrossRef] [PubMed]
  3. Poon, C.H.; Wang, Y.; Fung, M.L.; Zhang, C.; Lim, L.W. Rodent Models of Amyloid-Beta Feature of Alzheimer’s Disease: Development and Potential Treatment Implications. Aging Dis. 2020, 11, 1235–1259. [Google Scholar] [CrossRef]
  4. Uddin, M.S.; Lim, L.W. Glial cells in Alzheimer’s disease: From neuropathological changes to therapeutic implications. Ageing Res. Rev. 2022, 78, 101622. [Google Scholar] [CrossRef]
  5. Wong, K.Y.; Roy, J.; Fung, M.L.; Heng, B.C.; Zhang, C.; Lim, L.W. Relationships between Mitochondrial Dysfunction and Neurotransmission Failure in Alzheimer’s Disease. Aging Dis. 2020, 11, 1291–1316. [Google Scholar] [CrossRef]
  6. Lanctôt, K.L.; Rajaram, R.D.; Herrmann, N. Therapy for Alzheimer’s Disease: How Effective are Current Treatments? Adv. Neurol. Disord. 2009, 2, 163–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Tanaka, M.; Török, N.; Vécsei, L. Novel Pharmaceutical Approaches in Dementia. NeuroPsychopharmacotherapy 2020, 1–18. [Google Scholar] [CrossRef]
  8. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef]
  9. Shega, J.W.; Ellner, L.; Lau, D.T.; Maxwell, T.L. Cholinesterase inhibitor and N-methyl-D-aspartic acid receptor antagonist use in older adults with end-stage dementia: A survey of hospice medical directors. J. Palliat. Med. 2009, 12, 779–783. [Google Scholar] [CrossRef]
  10. Chan, H.J.; Yanshree; Roy, J.; Tipoe, G.L.; Fung, M.-L.; Lim, L.W. Therapeutic Potential of Human Stem Cell Implantation in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 10151. [Google Scholar] [CrossRef]
  11. Liu, J.; Chang, L.; Song, Y.; Li, H.; Wu, Y. The Role of NMDA Receptors in Alzheimer’s Disease. Front. Neurosci. 2019, 13, 43. [Google Scholar] [CrossRef] [Green Version]
  12. Uddin, M.S.; Kabir, M.T.; Rahman, M.S.; Behl, T.; Jeandet, P.; Ashraf, G.M.; Najda, A.; Bin-Jumah, M.N.; El-Seedi, H.R.; Abdel-Daim, M.M. Revisiting the Amyloid Cascade Hypothesis: From Anti-Aβ Therapeutics to Auspicious New Ways for Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 5858. [Google Scholar] [CrossRef]
  13. Congdon, E.E.; Sigurdsson, E.M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399–415. [Google Scholar] [CrossRef] [PubMed]
  14. Tan, S.Z.K.; Lim, L.W. A practical approach to the ethical use of memory modulating technologies. BMC Med. Ethics 2020, 21, 89. [Google Scholar] [CrossRef] [PubMed]
  15. Tan, S.Z.K.; Zhao, R.C.; Chakrabarti, S.; Stambler, I.; Jin, K.; Lim, L.W. Interdisciplinary Research in Alzheimer’s Disease and the Roles International Societies Can Play. Aging Dis. 2021, 12, 36–41. [Google Scholar] [CrossRef] [PubMed]
  16. Chong, P.S.; Fung, M.-L.; Wong, K.H.; Lim, L.W. Therapeutic Potential of Hericium erinaceus for Depressive Disorder. Int. J. Mol. Sci. 2020, 21, 163. [Google Scholar] [CrossRef] [Green Version]
  17. Chong, P.S.; Khairuddin, S.; Tse, A.C.K.; Hiew, L.F.; Lau, C.L.; Tipoe, G.L.; Fung, M.-L.; Wong, K.H.; Lim, L.W. Hericium erinaceus potentially rescues behavioural motor deficits through ERK-CREB-PSD95 neuroprotective mechanisms in rat model of 3-acetylpyridine-induced cerebellar ataxia. Sci. Rep. 2020, 10, 14945. [Google Scholar] [CrossRef]
  18. Thongbai, B.; Rapior, S.; Hyde, K.D.; Wittstein, K.; Stadler, M. Hericium erinaceus, an amazing medicinal mushroom. Mycol. Prog. 2015, 14, 91. [Google Scholar] [CrossRef]
  19. Lew, S.Y.; Lim, S.H.; Lim, L.W.; Wong, K.H. Neuroprotective effects of Hericium erinaceus (Bull.: Fr.) Pers. against high-dose corticosterone-induced oxidative stress in PC-12 cells. BMC Complement. Med. Ther. 2020, 20, 340. [Google Scholar] [CrossRef]
  20. Lew, S.-Y.; Yow, Y.-Y.; Lim, L.-W.; Wong, K.-H. Antioxidant-mediated protective role of Hericium erinaceus (Bull.: Fr.) Pers. against oxidative damage in fibroblasts from Friedreich’s ataxia patient. Food Sci. Technol. 2020, 40, 264–272. [Google Scholar] [CrossRef] [Green Version]
  21. Wong, K.H.; Naidu, M.; David, P.; Abdulla, M.A.; Abdullah, N.; Kuppusamy, U.R.; Sabaratnam, V. Peripheral Nerve Regeneration Following Crush Injury to Rat Peroneal Nerve by Aqueous Extract of Medicinal Mushroom Hericium erinaceus (Bull.: Fr) Pers. (Aphyllophoromycetideae). Evid.-Based Complement. Altern. Med. 2011, 2011, 580752. [Google Scholar] [CrossRef] [Green Version]
  22. Samberkar, S.; Gandhi, S.; Naidu, M.; Wong, K.H.; Raman, J.; Sabaratnam, V. Lion’s Mane, Hericium erinaceus and Tiger Milk, Lignosus rhinocerotis (Higher Basidiomycetes) Medicinal Mushrooms Stimulate Neurite Outgrowth in Dissociated Cells of Brain, Spinal Cord, and Retina: An In Vitro Study. Int. J. Med. Mushrooms 2015, 17, 1047–1054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chong, P.S.; Poon, C.H.; Roy, J.; Tsui, K.C.; Lew, S.Y.; Phang, M.W.L.; Tan, R.J.Y.; Cheng, P.G.; Fung, M.L.; Wong, K.H.; et al. Neurogenesis-dependent antidepressant-like activity of Hericium erinaceus in an animal model of depression. Chin. Med. 2021, 16, 132. [Google Scholar] [CrossRef] [PubMed]
  24. Yow, Y.Y.; Goh, T.K.; Nyiew, K.Y.; Lim, L.W.; Phang, S.M.; Lim, S.H.; Ratnayeke, S.; Wong, K.H. Therapeutic Potential of Complementary and Alternative Medicines in Peripheral Nerve Regeneration: A Systematic Review. Cells 2021, 10, 2194. [Google Scholar] [CrossRef]
  25. Kah Hui, W.; Sabaratnam, V.; Abdullah, N.; Kuppusamy, U.R.; Naidu, M. Effects of Cultivation Techniques and Processing on Antimicrobial and Antioxidant Activities of Hericium erinaceus (Bull.:Fr.) Pers. Extracts. Food Technol. Biotechnol. 2008, 47, 47–55. [Google Scholar]
  26. Zhang, C.-C.; Yin, X.; Cao, C.-Y.; Wei, J.; Zhang, Q.; Gao, J.-M. Chemical constituents from Hericium erinaceus and their ability to stimulate NGF-mediated neurite outgrowth on PC12 cells. Bioorg. Med. Chem. Lett. 2015, 25, 5078–5082. [Google Scholar] [CrossRef]
  27. Kushairi, N.; Tarmizi, N.A.K.A.; Phan, C.W.; Macreadie, I.; Sabaratnam, V.; Naidu, M.; David, P. Modulation of neuroinflammatory pathways by medicinal mushrooms, with particular relevance to Alzheimer’s disease. Trends Food Sci. Technol. 2020, 104, 153–162. [Google Scholar] [CrossRef]
  28. Sadigh-Eteghad, S.; Sabermarouf, B.; Majdi, A.; Talebi, M.; Farhoudi, M.; Mahmoudi, J. Amyloid-Beta: A Crucial Factor in Alzheimer’s Disease. Med. Princ. Pract. 2015, 24, 1–10. [Google Scholar] [CrossRef]
  29. He, P.; Shen, Y. Interruption of β-Catenin Signaling Reduces Neurogenesis in Alzheimer’s Disease. J. Neurosci. 2009, 29, 6545. [Google Scholar] [CrossRef] [Green Version]
  30. Phiel, C.J.; Wilson, C.A.; Lee, V.M.; Klein, P.S. GSK-3alpha regulates production of Alzheimer’s disease amyloid-beta peptides. Nature 2003, 423, 435–439. [Google Scholar] [CrossRef]
  31. Serrano-Pozo, A.; Frosch, M.P.; Masliah, E.; Hyman, B.T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2011, 1, a006189. [Google Scholar] [CrossRef]
  32. Komuro, Y.; Xu, G.; Bhaskar, K.; Lamb, B.T. Human tau expression reduces adult neurogenesis in a mouse model of tauopathy. Neurobiol. Aging 2015, 36, 2034–2042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rajmohan, R.; Reddy, P.H. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer’s disease Neurons. J. Alzheimer’s Dis. 2017, 57, 975–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Poddar, M.K.; Apala, C.; Soumyabrata, B. Neurodegeneration: Diagnosis, Prevention, and Therapy In Oxidoreductase; Mahmoud Ahmed Mansour, M.A., Blumenberg, M., Eds.; IntechOpen: London, UK, 2021; pp. 131–167. [Google Scholar]
  35. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kim, G.H.; Kim, J.E.; Rhie, S.J.; Yoon, S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef] [PubMed]
  37. Uttara, B.; Singh, A.V.; Zamboni, P.; Mahajan, R.T. Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Curr. Neuropharmacol. 2009, 7, 65–74. [Google Scholar] [CrossRef] [Green Version]
  38. Guzman-Martinez, L.; Maccioni, R.B.; Andrade, V.; Navarrete, L.P.; Pastor, M.G.; Ramos-Escobar, N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front. Pharmacol. 2019, 10, 1008. [Google Scholar] [CrossRef] [Green Version]
  39. Harry, G.J.; Kraft, A.D. Neuroinflammation and microglia: Considerations and approaches for neurotoxicity assessment. Expert Opin. Drug Metab. Toxicol. 2008, 4, 1265–1277. [Google Scholar] [CrossRef]
  40. Pekny, M.; Pekna, M. Reactive gliosis in the pathogenesis of CNS diseases. Biochim. Biophys. Acta-Mol. Basis Dis. 2016, 1862, 483–491. [Google Scholar] [CrossRef]
  41. Sun, X.; Chen, W.-D.; Wang, Y.-D. β-Amyloid: The Key Peptide in the Pathogenesis of Alzheimer’s Disease. Front. Pharmacol. 2015, 6, 221. [Google Scholar] [CrossRef] [Green Version]
  42. Slotkin, T.A.; Seidler, F.J.; Crain, B.J.; Bell, J.M.; Bissette, G.; Nemeroff, C.B. Regulatory changes in presynaptic cholinergic function assessed in rapid autopsy material from patients with Alzheimer disease: Implications for etiology and therapy. Proc. Natl. Acad. Sci. USA 1990, 87, 2452–2455. [Google Scholar] [CrossRef] [Green Version]
  43. Schliebs, R.; Arendt, T. The significance of the cholinergic system in the brain during aging and in Alzheimer’s disease. J. Neural Transm. 2006, 113, 1625–1644. [Google Scholar] [CrossRef] [PubMed]
  44. García-Ayllón, M.-S.; Small, D.H.; Avila, J.; Saez-Valero, J. Revisiting the Role of Acetylcholinesterase in Alzheimer’s Disease: Cross-Talk with P-tau and β-Amyloid. Front. Mol. Neurosci. 2011, 4, 22. [Google Scholar] [CrossRef] [Green Version]
  45. Strac, D.S.; Muck-Seler, D.; Pivac, N. Neurotransmitter measures in the cerebrospinal fluid of patients with Alzheimer’s disease: A review. Psychiatr. Danub. 2015, 27, 14–24. [Google Scholar] [PubMed]
  46. Kandimalla, R.; Reddy, P.H. Therapeutics of Neurotransmitters in Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 57, 1049–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zhang, J.; An, S.; Hu, W.; Teng, M.; Wang, X.; Qu, Y.; Liu, Y.; Yuan, Y.; Wang, D. The Neuroprotective Properties of Hericium erinaceus in Glutamate-Damaged Differentiated PC12 Cells and an Alzheimer’s Disease Mouse Model. Int. J. Mol. Sci. 2016, 17, 1810. [Google Scholar] [CrossRef] [PubMed]
  48. Mori, K.; Obara, Y.; Moriya, T.; Inatomi, S.; Nakahata, N. Effects of Hericium erinaceus on amyloid β(25-35) peptide-induced learning and memory deficits in mice. Biomed. Res. 2011, 32, 67–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Tsai-Teng, T.; Chin-Chu, C.; Li-Ya, L.; Wan-Ping, C.; Chung-Kuang, L.; Chien-Chang, S.; Chi-Ying, H.F.; Chien-Chih, C.; Shiao, Y.-J. Erinacine A-enriched Hericium erinaceus mycelium ameliorates Alzheimer’s disease-related pathologies in APPswe/PS1dE9 transgenic mice. J. Biomed. Sci. 2016, 23, 49. [Google Scholar] [CrossRef] [Green Version]
  50. Tzeng, T.-T.; Chen, C.-C.; Chen, C.-C.; Tsay, H.-J.; Lee, L.-Y.; Chen, W.-P.; Shen, C.-C.; Shiao, Y.-J. The Cyanthin Diterpenoid and Sesterterpene Constituents of Hericium erinaceus Mycelium Ameliorate Alzheimer’s Disease-Related Pathologies in APP/PS1 Transgenic Mice. Int. J. Mol. Sci. 2018, 19, 598. [Google Scholar] [CrossRef] [Green Version]
  51. Sabaratnam, V.; Kah-Hui, W.; Naidu, M.; Rosie David, P. Neuronal health—Can culinary and medicinal mushrooms help? J. Tradit. Complement. Med. 2013, 3, 62–68. [Google Scholar] [CrossRef] [Green Version]
  52. Park, Y.S.; Lee, H.S.; Won, M.H.; Lee, J.H.; Lee, S.Y.; Lee, H.Y. Effect of an exo-polysaccharide from the culture broth of Hericium erinaceus on enhancement of growth and differentiation of rat adrenal nerve cells. Cytotechnology 2002, 39, 155–162. [Google Scholar] [CrossRef]
  53. Kah Hui, W.; Sabaratnam, V.; Abdullah, N.; Naidu, M.; Keynes, R. Activity of Aqueous Extracts of Lion’s Mane Mushroom Hericium erinaceus (Bull.: Fr.) Pers. (Aphyllophoromycetideae) on the Neural Cell Line NG108-15. Int. J. Med. Mushrooms 2007, 9, 57–65. [Google Scholar] [CrossRef]
  54. Kawagishi, H.; Shimada, A.; Shirai, R.; Okamoto, K.; Ojima, F.; Sakamoto, H.; Ishiguro, Y.; Furukawa, S. Erinacines A, B and C, strong stimulators of nerve growth factor (NGF)-synthesis, from the mycelia of Hericium erinaceum. Tetrahedron Lett. 1994, 35, 1569–1572. [Google Scholar] [CrossRef]
  55. Nagai, K.; Chiba, A.; Nishino, T.; Kubota, T.; Kawagishi, H. Dilinoleoyl-phosphatidylethanolamine from Hericium erinaceum protects against ER stress-dependent Neuro2a cell death via protein kinase C pathway. J. Nutr. Biochem. 2006, 17, 525–530. [Google Scholar] [CrossRef] [PubMed]
  56. Uddin, M.S.; Yu, W.S.; Lim, L.W. Exploring ER stress response in cellular aging and neuroinflammation in Alzheimer’s disease. Ageing Res. Rev. 2021, 70, 101417. [Google Scholar] [CrossRef]
  57. Filali, M.; Lalonde, R. Age-related cognitive decline and nesting behavior in an APPswe/PS1 bigenic model of Alzheimer’s disease. Brain Res. 2009, 1292, 93–99. [Google Scholar] [CrossRef]
  58. Cordaro, M.; Salinaro, A.T.; Siracusa, R.; D’Amico, R.; Impellizzeri, D.; Scuto, M.; Ontario, M.L.; Cuzzocrea, S.; Di Paola, R.; Fusco, R.; et al. Key Mechanisms and Potential Implications of Hericium erinaceus in NLRP3 Inflammasome Activation by Reactive Oxygen Species during Alzheimer’s Disease. Antioxidants 2021, 10, 1664. [Google Scholar] [CrossRef]
  59. Lee, L.-Y.; Chou, W.; Chen, W.-P.; Wang, M.-F.; Chen, Y.-J.; Chen, C.-C.; Tung, K.-C. Erinacine A-Enriched Hericium erinaceus Mycelium Delays Progression of Age-Related Cognitive Decline in Senescence Accelerated Mouse Prone 8 (SAMP8) Mice. Nutrients 2021, 13, 3659. [Google Scholar] [CrossRef]
  60. Li, I.C.; Chang, H.H.; Lin, C.H.; Chen, W.P.; Lu, T.H.; Lee, L.Y.; Chen, Y.W.; Chen, Y.P.; Chen, C.C.; Lin, D.P. Prevention of Early Alzheimer’s Disease by Erinacine A-Enriched Hericium erinaceus Mycelia Pilot Double-Blind Placebo-Controlled Study. Front. Aging Neurosci. 2020, 12, 155. [Google Scholar] [CrossRef]
  61. Mori, K.; Inatomi, S.; Ouchi, K.; Azumi, Y.; Tuchida, T. Improving effects of the mushroom Yamabushitake (Hericium erinaceus) on mild cognitive impairment: A double-blind placebo-controlled clinical trial. Phytother. Res. 2009, 23, 367–372. [Google Scholar] [CrossRef]
  62. Saitsu, Y.; Nishide, A.; Kikushima, K.; Shimizu, K.; Ohnuki, K. Improvement of cognitive functions by oral intake of Hericium erinaceus. Biomed. Res. 2019, 40, 125–131. [Google Scholar] [CrossRef] [Green Version]
  63. Li, I.C.; Lee, L.-Y.; Tzeng, T.-T.; Chen, W.-P.; Chen, Y.-P.; Shiao, Y.-J.; Chen, C.-C. Neurohealth Properties of Hericium erinaceus Mycelia Enriched with Erinacines. Behav. Neurol. 2018, 2018, 5802634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lee, A.Y. Vascular dementia. Chonnam Med. J. 2011, 47, 66–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Qiu, W.Q.; Folstein, M.F. Insulin, insulin-degrading enzyme and amyloid-beta peptide in Alzheimer’s disease: Review and hypothesis. Neurobiol. Aging 2006, 27, 190–198. [Google Scholar] [CrossRef] [PubMed]
  66. Farris, W.; Mansourian, S.; Leissring, M.A.; Eckman, E.A.; Bertram, L.; Eckman, C.B.; Tanzi, R.E.; Selkoe, D.J. Partial Loss-of-Function Mutations in Insulin-Degrading Enzyme that Induce Diabetes also Impair Degradation of Amyloid β-Protein. Am. J. Pathol. 2004, 164, 1425–1434. [Google Scholar] [CrossRef]
  67. Cook, D.G.; Leverenz, J.B.; McMillan, P.J.; Kulstad, J.J.; Ericksen, S.; Roth, R.A.; Schellenberg, G.D.; Jin, L.-W.; Kovacina, K.S.; Craft, S. Reduced Hippocampal Insulin-Degrading Enzyme in Late-Onset Alzheimer’s Disease Is Associated with the Apolipoprotein E-ε4 Allele. Am. J. Pathol. 2003, 162, 313–319. [Google Scholar] [CrossRef]
  68. Liang, B.; Guo, Z.; Xie, F.; Zhao, A. Antihyperglycemic and antihyperlipidemic activities of aqueous extract of Hericium erinaceus in experimental diabetic rats. BMC Complement. Altern. Med. 2013, 13, 253. [Google Scholar] [CrossRef] [Green Version]
  69. Bellezza, I.; Giambanco, I.; Minelli, A.; Donato, R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2018, 1865, 721–733. [Google Scholar] [CrossRef]
  70. Montine, T.J.; Neely, M.D.; Quinn, J.F.; Beal, M.F.; Markesbery, W.R.; Roberts, L.J.; Morrow, J.D. Lipid peroxidation in aging brain and Alzheimer’s disease. Free Radic. Biol. Med. 2002, 33, 620–626. [Google Scholar] [CrossRef]
  71. Chang, C.-H.; Chen, Y.; Yew, X.-X.; Chen, H.-X.; Kim, J.-X.; Chang, C.-C.; Peng, C.-C.; Peng, R.Y. Improvement of erinacine A productivity in Hericium erinaceus mycelia and its neuroprotective bioactivity against the glutamate-insulted apoptosis. LWT-Food Sci. Technol. 2016, 65, 1100–1108. [Google Scholar] [CrossRef]
  72. Kim, Y.K.; Shin, J.S.; Nahm, M.H. NOD-Like Receptors in Infection, Immunity, and Diseases. Yonsei Med. J. 2016, 57, 5–14. [Google Scholar] [CrossRef] [Green Version]
  73. Davis, B.K.; Wen, H.; Ting, J.P.-Y. The Inflammasome NLRs in Immunity, Inflammation, and Associated Diseases. Annu. Rev. Immunol. 2011, 29, 707–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Malinski, T. Nitric oxide and nitroxidative stress in Alzheimer’s disease. J. Alzheimer’s Dis. 2007, 11, 207–218. [Google Scholar] [CrossRef] [PubMed]
  75. Mori, K.; Obara, Y.; Hirota, M.; Azumi, Y.; Kinugasa, S.; Inatomi, S.; Nakahata, N. Nerve Growth Factor-Inducing Activity of Hericium erinaceus in 1321N1 Human Astrocytoma Cells. Biol. Pharm. Bull. 2008, 31, 1727–1732. [Google Scholar] [CrossRef] [Green Version]
  76. Ji, S.; Wu, H.; Ding, X.; Chen, Q.; Jin, X.; Yu, J.; Yang, M. Increased hippocampal TrkA expression ameliorates cranial radiation-induced neurogenesis impairment and cognitive deficit via PI3K/AKT signaling. Oncol. Rep. 2020, 44, 2527–2536. [Google Scholar] [CrossRef] [PubMed]
  77. Martorana, F.; Gaglio, D.; Bianco, M.R.; Aprea, F.; Virtuoso, A.; Bonanomi, M.; Alberghina, L.; Papa, M.; Colangelo, A.M. Differentiation by nerve growth factor (NGF) involves mechanisms of crosstalk between energy homeostasis and mitochondrial remodeling. Cell Death Dis. 2018, 9, 391. [Google Scholar] [CrossRef] [PubMed]
  78. Hericium erinaceus Extract Reduces Anxiety and Depressive Behaviors by Promoting Hippocampal Neurogenesis in the Adult Mouse Brain. J. Med. Food 2018, 21, 174–180. [CrossRef]
  79. Brandalise, F.; Cesaroni, V.; Gregori, A.; Repetti, M.; Romano, C.; Orrù, G.; Botta, L.; Girometta, C.; Guglielminetti, M.L.; Savino, E.; et al. Dietary Supplementation of Hericium erinaceus Increases Mossy Fiber-CA3 Hippocampal Neurotransmission and Recognition Memory in Wild-Type Mice. Evid.-Based Complement. Altern. Med. 2017, 2017, 3864340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. John, P.A.; Wong, K.H.; Naidu, M.; Sabaratnam, V.; David, P. Combination Effects of Curcumin and Aqueous Extract of Lignosus rhinocerotis Mycelium on Neurite Outgrowth Stimulation Activity in PC-12 Cells. Nat. Prod. Commun. 2013, 8, 1934578X1300800. [Google Scholar] [CrossRef] [Green Version]
  81. Phang, M.W.L.; Lew, S.Y.; Chung, I.; Lim, W.K.; Lim, L.W.; Wong, K.H. Therapeutic roles of natural remedies in combating hereditary ataxia: A systematic review. Chin. Med. 2021, 16, 15. [Google Scholar] [CrossRef]
  82. Liu, S.H.; Chuang, W.C.; Lam, W.; Jiang, Z.; Cheng, Y.C. Safety surveillance of traditional Chinese medicine: Current and future. Drug Saf. 2015, 38, 117–128. [Google Scholar] [CrossRef] [Green Version]
  83. Wang, C.; Su, W.; Su, X.; Ni, G.; Liu, T.; Kong, Y. Synergy Effects of Three Plant Extracts on Protection of Gastric Mucosa. Nat. Prod. Commun. 2015, 10, 1989–1991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Vaiserman, A.; Koliada, A.; Zayachkivska, A.; Lushchak, O. Nanodelivery of Natural Antioxidants: An Anti-aging Perspective. Front. Bioeng. Biotechnol. 2019, 7, 447. [Google Scholar] [CrossRef] [Green Version]
  85. Liu, A.; Jain, N.; Vyas, A.; Lim, L.W. Ventromedial prefrontal cortex stimulation enhances memory and hippocampal neurogenesis in the middle-aged rats. eLife 2015, 4, e04803. [Google Scholar] [CrossRef] [PubMed]
  86. Tan, S.Z.K.; Neoh, J.; Lawrence, A.J.; Wu, E.X.; Lim, L.W. Prelimbic Cortical Stimulation Improves Spatial Memory Through Distinct Patterns of Hippocampal Gene Expression in Aged Rats. Neurotherapeutics 2020, 17, 2054–2068. [Google Scholar] [CrossRef]
  87. Tan, S.Z.K.; Poon, C.H.; Chan, Y.S.; Lim, L.W. Prelimbic cortical stimulation disrupts fear memory consolidation through ventral hippocampal dopamine D2 receptors. Br. J. Pharm. 2021, 178, 3587–3601. [Google Scholar] [CrossRef] [PubMed]
  88. Yu, W.S.; Aquili, L.; Wong, K.H.; Lo, A.C.Y.; Chan, L.L.H.; Chan, Y.S.; Lim, L.W. Transcorneal electrical stimulation enhances cognitive functions in aged and 5XFAD mouse models. Ann. N. Y. Acad. Sci. 2022. [Google Scholar] [CrossRef] [PubMed]
  89. Yu, W.S.; Tse, A.C.; Guan, L.; Chiu, J.L.Y.; Tan, S.Z.K.; Khairuddin, S.; Agadagba, S.K.; Lo, A.C.Y.; Fung, M.L.; Chan, Y.S.; et al. Antidepressant-like effects of transcorneal electrical stimulation in rat models. Brain Stimul. 2022, 15, 843–856. [Google Scholar] [CrossRef]
  90. Venturella, G.; Ferraro, V.; Cirlincione, F.; Gargano, M.L. Medicinal Mushrooms: Bioactive Compounds, Use, and Clinical Trials. Int. J. Mol. Sci. 2021, 22, 634. [Google Scholar] [CrossRef]
Figure 1. Hericium erinaceus fruiting body (A) and mycelia (B).
Figure 1. Hericium erinaceus fruiting body (A) and mycelia (B).
Cells 11 02284 g001
Figure 2. Bioactive compounds isolated from Hericium erinaceus with therapeutic effects on Alzheimer’s disease.
Figure 2. Bioactive compounds isolated from Hericium erinaceus with therapeutic effects on Alzheimer’s disease.
Cells 11 02284 g002
Figure 3. A schematic diagram summarizing the functions of Hericium erinaceus in AD. Abbreviations: ROS, Reactive oxygen species; BAX, Bcl-2- associated X protein; TBARS, Thiobarbituric acid reactive substances; MDA, Malondialdehyde; Nrf2, Nuclear factor-erythroid factor 2-related factor 2; SOD, Superoxide dismutase; CAT, Catalase; GSH, Glutathione; Bcl-2, B-cell lymphoma 2; SDS, Sodium dodecyl sulfate; CTF-β, Beta-carboxyl-terminal fragment; Aβ, Amyloid-beta; IDE, Insulin-degrading enzyme; iNOS, Nitric oxide synthase; ASC, Apoptosis-associated speck-like protein containing a caspase recruitment domain; NLRP3, NLR family pyrin domain containing 3; NF-kB, Nuclear factor-kappa B; ACh, Acetylcholine; ChAT, Choline acetyltransferase; TrkA, Tropomyosin receptor kinase A; RAS-GTP, Ras protein guanine triphosphatase; Raf, Rapidly accelerated fibrosarcoma; MEK, Mitogen-activated protein kinase; Erk, Extracellular signal-regulated kinase; CREB, cAMP-response element binding protein.
Figure 3. A schematic diagram summarizing the functions of Hericium erinaceus in AD. Abbreviations: ROS, Reactive oxygen species; BAX, Bcl-2- associated X protein; TBARS, Thiobarbituric acid reactive substances; MDA, Malondialdehyde; Nrf2, Nuclear factor-erythroid factor 2-related factor 2; SOD, Superoxide dismutase; CAT, Catalase; GSH, Glutathione; Bcl-2, B-cell lymphoma 2; SDS, Sodium dodecyl sulfate; CTF-β, Beta-carboxyl-terminal fragment; Aβ, Amyloid-beta; IDE, Insulin-degrading enzyme; iNOS, Nitric oxide synthase; ASC, Apoptosis-associated speck-like protein containing a caspase recruitment domain; NLRP3, NLR family pyrin domain containing 3; NF-kB, Nuclear factor-kappa B; ACh, Acetylcholine; ChAT, Choline acetyltransferase; TrkA, Tropomyosin receptor kinase A; RAS-GTP, Ras protein guanine triphosphatase; Raf, Rapidly accelerated fibrosarcoma; MEK, Mitogen-activated protein kinase; Erk, Extracellular signal-regulated kinase; CREB, cAMP-response element binding protein.
Cells 11 02284 g003
Table 2. Clinical studies of Hericium erinaceus in AD individuals.
Table 2. Clinical studies of Hericium erinaceus in AD individuals.
AuthorsDisease, Clinical Phase, and DurationResearch DesignAdministration MethodDosage Outcome MeasuresClinical EvaluationAdverse Effects
Li et al., 2020 [60]Mild Alzheimer’s disease
Completed
52 weeks
Double-blind, two parallel groups, randomized, placebo-controlled
49 participants,
50 to 90 years old
Oral route350 mg mycelia-based capsule containing 5 mg/g of erinacine A per day or identically appearing placebo capsulesSignificant improvement in MMSE, IADL, CASI scores, and better contrast sensitivity than placebo
HE-A was well-tolerated, safe, and improved cognitive function
Adverse events evaluation
NPI
MMSE
CASI
IADL
Ophthalmic examination
Biomarker collection
Neuroimaging
1 subject lost to follow-up
3 subjects lost to unsatisfactory efficacy
4 subjects lost to side effects: nausea, abdominal discomfort, nausea, and skin rash
Mori et al., 2009 [61]Mild cognitive impairment
Completed
22 weeks
Double-blind, parallel group, placebo-controlled trial, randomized
30 participants,
50 to 80 years old
Oral routeFour 250 mg tablets of dry powder of HE (96% of HE) or three placebo tablets per day for 16 weeksImproved cognitive function scale scores
HDS-R
Adverse effect evaluation
Blood chemistry
N.A.
Saitsu et al., 2019 [62]Normal cognitive functions
Completed
12 weeks
Double-blind, placebo-controlled trial, parallel group, randomized
34 participants,
50 years old and above
Oral routeFour HE supplements containing 0.8 g of powdered fruiting body or four placebo supplements per day for 12 weeksSignificantly improved cognitive functions in MMSEMMSE
Benton visual retention test
S-PA
N.A.
Abbreviations: MMSE, Mini-mental state examination; IADL, Instrumental activities of daily living; CASI, Cognitive abilities screening instrument; HE-A, erinacine A-enriched Hericium erinaceus mycelia; NPI, Neuropsychiatric inventory; HDS-R, Revised Hasegawa Dementia scale; S-PA, Standard verbal paired-associate learning test.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yanshree; Yu, W.S.; Fung, M.L.; Lee, C.W.; Lim, L.W.; Wong, K.H. The Monkey Head Mushroom and Memory Enhancement in Alzheimer’s Disease. Cells 2022, 11, 2284. https://doi.org/10.3390/cells11152284

AMA Style

Yanshree, Yu WS, Fung ML, Lee CW, Lim LW, Wong KH. The Monkey Head Mushroom and Memory Enhancement in Alzheimer’s Disease. Cells. 2022; 11(15):2284. https://doi.org/10.3390/cells11152284

Chicago/Turabian Style

Yanshree, Wing Shan Yu, Man Lung Fung, Chi Wai Lee, Lee Wei Lim, and Kah Hui Wong. 2022. "The Monkey Head Mushroom and Memory Enhancement in Alzheimer’s Disease" Cells 11, no. 15: 2284. https://doi.org/10.3390/cells11152284

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