전체메뉴

Journal of Pharmacopuncture

Article Search

JoP

Review Article

Split Viewer

Related articles in JoP

More Related Articles

Article

Review Article

J Pharmacopuncture 2022; 25(4): 326-343

Published online December 31, 2022 https://doi.org/10.3831/KPI.2022.25.4.326

Copyright © The Korean Pharmacopuncture Institute.

Therapeutic Potential of Active Components from Acorus gramineus and Acorus tatarinowii in Neurological Disorders and Their Application in Korean Medicine

Cheol Ju Kim1 , Tae Young Kwak1 , Min Hyeok Bae1 , Hwa Kyoung Shin1,2* , Byung Tae Choi1,2*

1Department of Korean Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
2Graduate Training Program of Korean Medical Therapeutics for Healthy Aging, Pusan National University, Yangsan, Republic of Korea

Correspondence to:Hwa Kyoung Shin
Department of Korean Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Republic of Korea
Tel: +82-51-510-8476
E-mail: julie@pusan.ac.kr

Byung Tae Choi
Department of Korean Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Republic of Korea
Tel: +82-51-510-8475
E-mail: choibt@pusan.ac.kr

Received: August 26, 2022; Revised: August 26, 2022; Accepted: September 6, 2022

This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Neurological disorders represent a substantial healthcare burden worldwide due to population aging. Acorus gramineus Solander (AG) and Acorus tatarinowii Schott (AT), whose major component is asarone, have been shown to be effective in neurological disorders. This review summarized current information from preclinical and clinical studies regarding the effects of extracts and active components of AG and AT (e.g., α-asarone and β-asarone) on neurological disorders and biomedical targets, as well as the mechanisms involved. Databases, including PubMed, Embase, and RISS, were searched using the following keywords: asarone, AG, AT, and neurological disorders, including Alzheimer’s disease, Parkinson’s disease, depression and anxiety, epilepsy, and stroke. Meta-analyses and reviews were excluded. A total of 873 studies were collected. A total of 89 studies were selected after eliminating studies that did not meet the inclusion criteria. Research on neurological disorders widely reported that extracts or active components of AG and AT showed therapeutic efficacy in treating neurological disorders. These components also possessed a wide array of neuroprotective effects, including reduction of pathogenic protein aggregates, antiapoptotic activity, modulation of autophagy, anti-inflammatory and antioxidant activities, regulation of neurotransmitters, activation of neurogenesis, and stimulation of neurotrophic factors. Most of the included studies were preclinical studies that used in vitro and in vivo models, and only a few clinical studies have been performed. Therefore, this review summarizes the current knowledge on AG and AT therapeutic effects as a basis for further clinical studies, and clinical trials are required before these findings can be applied to human neurological disorders.

Keywords: alzheimer’s disease, asarone, depression, epilepsy, parkinson’s diseases, stroke

INTRODUCTION

Neurological disorders are the main cause of death and disability, and these diseases have risen rapidly due to the increasing elderly population worldwide. These disorders, particularly Alzheimer’s disease (AD), Parkinson’s disease (PD), epilepsy, and stroke, encompass diseases of the nervous system, cause irreversible damage, are difficult to treat, and show a wide range of sequelae [1]. These factors lead to poor prognosis, prolonged illness, and limited ability to perform personal and social roles during the disease period. Moreover, many of these disorders show only a minimal response to conventional therapies, thus necessitating the identification or development of innovative treatment modalities.

Recently, drugs of natural origin have attracted increasing interest in treating neurological disorders because of their potential efficacy and limited or nonexistent side effects. Traditional Korean medicines, which use natural raw materials, have a high potential for developing new constituents and conventional medicines. In Korean medicine, Acorus gramineus Solander (AG) and Acorus tatarinowii Schott (AT), which is widely known as “Shi Chang Pu,” have been commonly used alone or in combination with other herbs. These herbs have been used in Korean medicine to improve mental, cognitive, and learning capacities, thus suggesting that they may be effective against neurological disorders, such as meningitis and dementia [1]. These herbs have also attracted the attention of researchers because they have shown therapeutic efficacy against cognitive impairment, anxiety, gastritis, and gastric ulcers and have shown sedative activity in preclinical studies [2]. Among the bioactive phytochemicals of AG and AT, α-asarone and β-asarone are the most widely studied components in the treatment of various diseases (Fig. 1) [3].

Figure 1. Alpha (α)-asarone (1,2,4-trimethoxy-5-[(E)-prop-1-enyl]benzene; PubChem CID: 636822) and beta (β)-asarone (1,2,4-trimethoxy-5-[(Z)-prop-1-enyl]benzene; PubChem CID: 5281758).

Asarone plays a broad role in the nervous system and has shown antiepileptic, sedative, antidepressant, and neuroprotective effects in different neurological disease models [4]. Molecular investigations have shown that asarone may have antiapoptotic, anti-inflammatory, and antioxidant effects and may modulate neurotransmitter production and neurotrophic factor regulation in AD, PD, epilepsy, and stroke. In addition, asarone is able to cross the blood-brain barrier (BBB), thus increasing its potential as a therapeutic drug for neurological disorders.

In this study, we aimed to review the molecular mechanisms of extracts or active components of AG and AT (e.g., α-asarone and β-asarone) in neurological disorders, such as AD, PD, depression and anxiety, epilepsy, and stroke. This study also discusses the pharmacological potential and basis of the molecular mechanism of α-asarone and β-asarone in neurological disorders discussed in pre-clinical and clinical studies.

METHODS

1. Search strategy

Experimental studies focused on evaluating asarone in treating neurological disorders were identified in PubMed (https://pubmed.ncbi.nlm.nih.gov), Embase (https://www.embase.com), and RISS (http://www.riss.kr). The keywords used were as follows: asarone (“extract” OR “tang” OR “san” OR “wan” OR “decoction” OR “powder” OR “ball” OR “pill”) AND (“Acorus gramineus”[tiab] OR “Acorus tatarinowii”[tiab] OR “shi chang pu”[tiab]).

2. Inclusion and exclusion criteria

The inclusion criteria were as follows: (1) studies on asarone, AG or AT extracts, and herbal formulas containing AG or AT; (2) preclinical in vitro, in vivo, and clinical trials; and (3) studies that mainly focused on neurological disorders. The exclusion criteria were as follows: (1) non-English studies; (2) studies that showed no efficacy for neurological disorders; and (3) editorials, abstracts, comments, and reviews.

3. Data extraction

All data were drawn independently by two reviewers from the included studies (TK and MB). The following details were extracted from each study: (1) compounds used, (2) species and experimental model studied, and (3) results and outcome measures.

RESULTS

1. Study selection

Among the results obtained in the database search, 873 studies were collected after excluding duplicates. Among these studies, 715 were excluded on the basis of the exclusion criteria. Sixty-eight of the remaining 158 studies were excluded after reviewing their full texts. Thus, 89 studies were included for review, and the distribution of these studies by disease was as follows: AD and dementia (29); PD (12); depression and anxiety (10); epilepsy and seizure (12); stroke (10); herbal medicine (16) (Fig. 2).

Figure 2. Flow diagram.

2. Alzheimer’s disease and dementia (Table 1)

AD is a degenerative brain disease that shows clinical features of slowly progressing cognitive decline and behavioral disorders [5]. One specific pathogenesis of AD is an amyloid cascade characterized by the excessive formation of amyloid-beta (Aβ) plaques due to an abnormal degradation pathway of amyloid precursor protein (APP), which usually stabilizes microtubules, maintains neuronal shape, and reduces axonal transmission. A representative example is the Tau hypothesis, which explains the hyperphosphorylation of helper Tau proteins [5]. In addition, neuron damage due to the inflammatory response [5], oxidative stress [5], cholinergic neurotransmitter imbalance [6], and vascular-related factors, including hypercholesterolemia and hyperhomocysteinemia [5], have also been shown to mediate AD and dementia pathology.

SCP, Shi Chang Pu in Chinese; Aβ, amyloid-beta; polyQ, polyglutamine; APP, amyloid precursor protein; PS1, presenilin-1; P13K, phosphoinositide 3-kinases; Akt, protein kinase B; mTOR, mammalian target of rapamycin; CaMKII-α, calcium/calmodulin-dependent protein kinase II-alpha; p-CREB, phosphor-cAMP response element-binding protein; Bcl-2, B-cell lymphoma 2; Bax, BCL2-associated X; JNK, c-Jun N-terminal kinases; AD, Alzheimer’s disease; BACE, beta-secretase 1; PINK, PTEN-induced kinase 1; SAMP8, senescence accelerated mouse-prone 8; ROCK, Rho-associated protein kinase; IL-, interleukin-; TNF-α, tumor necrosis factor-α; AQP4, aquaporin4; LPS, lipopolysaccharide; MCP-1, monocyte chemoattractant protein-1; NO, nitric oxide; GPX, glutathione peroxidase; SOD, superoxide dismutase; ROS, reactive oxygen species; MDA, malondialdehyde; CAT, catalase; GSH-Px, glutathione peroxidase; Nrf2, nuclear factor erythroid-2-related factor 2; HO-1, heme oxygenase 1; AChE, acetylcholinesterase; SYP, synaptophysin; GluR1, glutamatergic receptor 1; GABA, γ-aminobutyric acid type; NMDA, N-methyl-d-aspartate; MMSE, Mini-mental State Examination; ADL, activities of daily living; rCBF, regional cerebral blood flow; NA-K-ATP, sodium-potassium adenosine triphosphatase, sodium–potassium pump; ET, endothelin; AT, Acorus tatarinowii; NPC, neural progenitor cell; ERK, extracellular signal-regulated kinase; NGF, nerve growth factor; BDNF, brain-derived neurotrophic factor; GDNF, glial-derived neurotrophic factor..

&md=tbl&idx=1' data-target="#file-modal"">Table 1

Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in Alzheimer’s disease.

EffectCompoundSpeciesExperimental modelExperiment resultMechanismsRef.
Anti-Aβ accumulationSCP-OilWormCaenorhabditis elegans modelSerotonin sensitivity and olfactory learning skill ↑Misfolded Aβ and polyQ proteins ↓[7]
β-asaroneMouseAPP/PSI double transgenic miceSenile plaques in the hippocampus & Aβ ↓↑Levels of Aβ40 and Aβ42 in hippocampus ↓[8]
β-asaroneMouseAPP/PSI transgenic miceLearning and memory function ↑Beclin-1-dependent autophagy (the PI3K/Akt/mTOR pathway) ↓[9]
Anti-apoptosisβ-asaroneMouseAPP/PSI double transgenic miceCognitive function ↑CaMKII-α/p-CREB/Bcl-2 pathway ↑[10]
β-asaroneCellAβ42 injury cellsNeuronal apoptosis ↓Bax ↓, Bcl-2 ↑[11]
β-asaroneCellΩAβ-induced PC12 cellsNeuronal apoptosis ↓JNK activation ↓
Bcl-w and Bcl-xL in a JNK-dependent manner ↑
Cytochrome c and activation of caspase-3 ↑
[12]
β-asaroneRatAD induced ratsSpatial memory ↑JNK activation ↓, caspase-3 activation & Bcl-w, and Bcl-2 ↑[13]
β-asaroneRatAD induced ratsNeuronal apoptosis ↓Bad expression & p-c-Jun activation, Bax expression ↓
Activation of caspase-9 ↑
[14]
Autophagy regulationβ-asaroneCellAβ1~42-induced PC12 cellsAβ ↓
Autophagy ↑
APP, PS1, Aβ, and BACE1 expression ↓
PINK1, Parkin & Autophagy ↑
[15]
β-asarone + IcariinCell, mouseAβ-induced PC12 cells, APP/PS1 miceMitochondrial damage ↓Clearance of toxic proteins & the formation of autophagosomes ↑
Beclin-1, PINK1, and p/Parkin ↑
[16]
β-asaroneMouseSAMP8 miceCognitive function ↑ROCK expression ↓, autophagy and synaptic loss ↓[17]
β-asaroneCellAβ-induced PC12 cellsNeuronal apoptosis ↓Beclin-1 expression ↓
p-Akt and p-mTOR ↑
[18]
Anti-inflammationβ-asaroneCellHuman neuroblastoma cells SH-SY5Y cellsAutophagy ↓
Inflammation ↓
Toxic effect of Aβ25-35 in SH-SY5Y cells ↓
Pro-inflammatory cytokines (IL-6, IL-1β and TNF-α) ↓
[19]
β-asaroneMouse, cellAβ1~42 injected rats
Aβ1-42 induced astrocytes
Spatial learning and memory ↑TNF-α, IL-1β & AQP4 expression ↓[20]
α-asaroneCellLPS-induced BV2 cellsMicroglial, morphological dynamics ↑Activated microglia ↓
MCP-1 ↓
[21]
Antioxidant-oxidantα-asaroneRatAβ-injected ratsSpatial memory ↑NO production & activation of astrocytes ↓[22]
β-asaroneMouseAβ-infused miceCell loss in the cerebral cortex and hippocampus ↓GPX and SOD ↑[23]
β-asaroneCellAβ-induced PC12 cellsAβ-induced damage ↓ROS, MDA ↓,
SOD, CAT, GSH-PX ↑, P13K/Akt/Nrf2 signaling pathway ↑, HO-1 ↑
[24]
β-asaroneRatAβ-infused ratsLearning and memory ability ↑Oxidative stress ↓
Pro-inflammatory cytokine ↓
Neurotransmitter and AChE activity ↑
[25]
Neurotransmitterβ-asaroneCellAPS/PSI double transgenic miceLearning and memory ability ↑Aβ neurotoxicity ↓
SYP and GluR1 ↑
[26]
α-asaroneRatAged ratsCognitive function ↑Aβ neurotoxicity ↓
GABA receptors ↑
[27]
α-asarone, β-asaroneCellNMDA or Glu-exposed cortical cells of ratNeuronal apoptosis ↓NMDA receptor function ↓[28]
β-asarone + tenuigeninHuman93 AD patientsTherapeutic effect ↑MMSE, ADL score ↑[29]
β-asarone + tenuigeninHuman152 AD patientsTherapeutic effect ↑MMSE, ADL score ↑[30]
β-asaroneRatAD induced ratsMemory impairment ↓rCBF of right parietal lober & the activity of NA-K-ATP ↑
ET-1 mRNA expression in hippocampus & pyruvic acid ↓
[31]
OthersVolatile oil fraction of ATMouseAβ-infused miceCognitive function ↑
Spatial memory ↑
Doublecortin and nestin ↓[32]
α-asarone, β-asaroneMouseAPS/PSI transgenic miceHippocampal neurogenesis ↑
NPCs ↑
ERK pathway & neurogenesis ↑[33]
α-asarone, β-asaroneCellPrimary astrocytes from ratsNGF, BDNF & GDNF ↑Neuronal action of AT ↑
Neurotrophic factors in astrocytes ↑
[34]
α-asarone, β-asaroneCellPC12 cellsNGF ↑Neurofilaments ↑[35]

SCP, Shi Chang Pu in Chinese; Aβ, amyloid-beta; polyQ, polyglutamine; APP, amyloid precursor protein; PS1, presenilin-1; P13K, phosphoinositide 3-kinases; Akt, protein kinase B; mTOR, mammalian target of rapamycin; CaMKII-α, calcium/calmodulin-dependent protein kinase II-alpha; p-CREB, phosphor-cAMP response element-binding protein; Bcl-2, B-cell lymphoma 2; Bax, BCL2-associated X; JNK, c-Jun N-terminal kinases; AD, Alzheimer’s disease; BACE, beta-secretase 1; PINK, PTEN-induced kinase 1; SAMP8, senescence accelerated mouse-prone 8; ROCK, Rho-associated protein kinase; IL-, interleukin-; TNF-α, tumor necrosis factor-α; AQP4, aquaporin4; LPS, lipopolysaccharide; MCP-1, monocyte chemoattractant protein-1; NO, nitric oxide; GPX, glutathione peroxidase; SOD, superoxide dismutase; ROS, reactive oxygen species; MDA, malondialdehyde; CAT, catalase; GSH-Px, glutathione peroxidase; Nrf2, nuclear factor erythroid-2-related factor 2; HO-1, heme oxygenase 1; AChE, acetylcholinesterase; SYP, synaptophysin; GluR1, glutamatergic receptor 1; GABA, γ-aminobutyric acid type; NMDA, N-methyl-d-aspartate; MMSE, Mini-mental State Examination; ADL, activities of daily living; rCBF, regional cerebral blood flow; NA-K-ATP, sodium-potassium adenosine triphosphatase, sodium–potassium pump; ET, endothelin; AT, Acorus tatarinowii; NPC, neural progenitor cell; ERK, extracellular signal-regulated kinase; NGF, nerve growth factor; BDNF, brain-derived neurotrophic factor; GDNF, glial-derived neurotrophic factor..



The essential oil of AT has been shown to ameliorate Aβ-induced toxicity via an autophagy pathway in a Caenorhabditis elegans model [7]. β-Asarone can ameliorate cognitive function by inhibiting Aβ accumulation, apoptotic neuronal death, and autophagy. In addition, β-asarone has been shown to decrease senile plaque formation and Aβ40 and Aβ42 levels in the hippocampus [8] and inhibit Beclin-1-dependent autophagy via PI3K/Akt/mTOR signaling in APP/PS1 mice [9]. Moreover, β-asarone has been shown to decrease apoptosis by activating calcium/calmodulin-dependent protein kinase II (CaMKII)/cAMP response element-binding protein (CREB)/B-cell lymphoma 2 (Bcl-2), thereby inducing an improvement in the cognitive function of APS/PSI mice [10]. In an Aβ-induced neurotoxicity in vitro study, β-asarone was shown to inhibit apoptotic neuronal death by inhibiting the proapoptotic Bax protein, activating the antiapoptotic Bcl protein [11] and c-Jun N-terminal kinase (JNK) pathway, and regulating Bcl-2 family proteins in PC12 cells [12]. After the in vivo Aβ injection in the rat hippocampus, β-asarone was shown to improve cognitive function by suppressing apoptotic neuronal death via the activation of Bcl-2 and Bcl-w and β-asarone reversed the downregulation of JNK phosphorylation [13]. In addition, β-asarone was shown to activate p-c-Jun and decrease the activation of Bcl-2-associated agonist of cell death (Bad), Bax, and caspase-9 [14]. The effects of β-asarone on autophagy are controversial. For instance, this component has been reported to promote autophagy, which is involved in the removal of harmful proteins, in vitro [15] and in an animal model of AD [16]. However, β-asarone has also been reported to decrease Rho-kinase expression, reduce autophagy, influence synaptic damage [17], and attenuate Aβ-induced autophagy via the induction of the Akt-mTOR pathway [18].

The antioxidant and anti-inflammatory effects of asarone are also well-studied in the context of AD. β-Asarone has been shown to delay inflammatory responses and autophagy by inhibiting the induction of tumor necrosis factor α (TNF-α), interleukin (IL)-6, and interleukin-1 beta (IL-1β) in Aβ-treated SH-SY5Y cells [19]. β-Asarone has also been shown to improve spatial learning and memory by reducing TNF-α and IL-1β production and aquaporin-4 (AQP4) expression and by protecting astrocytes [20]. α-Asarone has been shown to modulate the dynamics of microglial morphology and decrease the expression of monocyte chemoattractant protein in lipopolysaccharide (LPS)-induced BV2 cells [21]. Assessments of asarone’s antioxidant activity showed that nitrite levels in the temporal cortex and hippocampus of rats were significantly reduced with the improvement in spatial memory after α-asarone administration [22]. Moreover, the activities of glutathione peroxidase (GPX) and antioxidant enzymes superoxide dismutase (SOD) were induced after β-asarone administration in rats [23]. β-Asarone has also been shown to reduce reactive oxygen species and malondialdehyde expression, induce SOD, catalase (CAT), and GPX activities, and promote nuclear factor erythroid-2-related factor 2 (Nrf2) and heme oxygenase 1 expression by upregulating P13K/Akt/Nrf2 signaling in Aβ-induced PC12 cells [24].

Asarone improves cognitive function by modulating neurotransmitters. β-Asarone has been shown to improve cognitive function by restoring the levels of hippocampal neurotransmitters, such as dopamine (DA), serotonin, γ-aminobutyric acid (GABA), and norepinephrine, and acetylcholinesterase activity and reducing oxidative and neuroinflammatory damage in Aβ-infused rats [25]. β-Asarone was also shown to have neuroprotective efficacy for AD via the modulation of synaptic plasticity by inducing the expression of synaptophysin and glutamatergic receptor 1 [26]. In addition, α-asarone was shown to ameliorate cognitive dysfunction by reducing neuronal excitotoxicity via GABA A (GABAA) receptors in aged rats [27]. Both α-asarone and β-asarone showed neuroprotective activity against excitotoxicity due to N-methyl-d-aspartate (NMDA) or glutamate (Glu) blocking NMDA receptor function [28]. In a clinical study, a combined prescription of β-asarone and tenuigenin in AD patients was shown to increase clinical scores, which indicates the improvement in AD symptoms [29, 30].

Asarone has also been shown to have cerebrovascular protective and neurogenesis effects. Treatment with β-asarone was shown to improve cerebral metabolism and blood flow and to downregulate the mRNA expression of endothelin 1 in the hippocampus of AD rats [31]. Treatment with volatile oil fractions or water extracts of AG in Aβ1-42-injected mice was shown to ameliorate cognitive impairment and hippocampal neurogenesis via the upregulation of nestin and doublecortin in the hippocampus [32]. The extracts of AT and its major constituents, namely, α-asarone and β-asarone, have been shown to promote the proliferation of neural progenitor cells with activated extracellular signal-regulated kinase (ERK) in hippocampus-derived progenitor cells [33] and potentiate neuronal differentiation via nerve growth factor (NGF) in PC12 cells [34]. The volatile oil fractions from AG, which contain α-asarone and β-asarone, stimulate neurotrophic factor secretion, namely, brain-derived neurotrophic factor (BDNF), NGF, and glial-derived neurotrophic factor, via the PKA signaling pathway [35].

3. Parkinson’s disease (Table 2)

PD is a common chronic neurodegenerative disease and characteristically associated with dopaminergic neuronal loss in the substantia nigra and pars compacta [36]. Motor symptoms, such as bradykinesia, resting tremors, and stiffness, are the main dysfunctions exhibited by patients with PD. The treatment of motor symptoms in PD is primarily based on DA regulation; therefore, DA metabolites (levodopa and l-dopa), DA-degrading enzyme inhibitors (monoamine oxidase-B and MAO-B), and DA agonists are the drugs of choice for the initial treatment.

SD, Sprague Dawley; 6-OHDA, 6-hydroxydopamine; OFT, open-field test; RRT, rotarod test; LC3-II, light chain 3-II; HVA, homovanillic acid; Dopacl, 3,4-dihydroxyphenylacetic acid; 5-HIAA, 5-hydroxyindole acetic acid; JNK, c-Jun N-terminal kinase; Bcl-2, B-cell lymphoma; MPTP, 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine; TH, tyrosine hydroxylase; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; α-syn, α-synuclein; CHX, cycloheximide; MG132, a proteasome inhibitor; NO, nitric oxide; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2; TNF-α, tumor necrosis factor-alpha; IL-, interleukin-; NF-κB, nuclear factor kappa B; IκB, NF-κB inhibitor; Mac-1, macrophage Ag complex-1; CD-68, cluster of differentiation 68; Iba-1, ionized calcium-binding adapter molecule 1; DOPAC, 3, 4-dihydroxyphenylacetic acid; GRP78, glucose-regulated protein 78; p-PERK, phosphorylated protein kinase RNA-like endoplasmic reticulum kinase; CHOP, C/EBP homologous binding protein; ER, endoplasmic reticulum; IRE1, inositol-requiring enzyme 1; p-IRE1, phosphorylated IRE1; XBP1, X-box binding protein 1; CMA, chaperone-mediated autophagy; HSC70, heat-shock cognate protein 70; HSP70, heat-shock protein 70; MEF2D, myocyte enhancer factor 2D; LAMP-2A, lysosomal membrane protein receptor type 2A; BAX, B-cell lymphoma 2-associated X protein; TH, thyrosine hydroxylase; SOD, superoxide dismutase; CAT, catalase; GSH-Px, glutathione peroxidase; l-dopa, levodopa; DA, dopamine; COMT, catechol-O-methyltransferase; DAT, dopamine transporter; ST, stepping test; DDC, dopa decarboxylase; MAO-B, monoamine oxidase-B; BBB, blood–brain barrier; S100β, S100 calcium-binding protein β; NSE, neuron-specific enolase; P-gp, P-glycoprotein; ZO-1, zonula occludens-1; LC3B, microtubule-associated protein light chain 3B..

&md=tbl&idx=2' data-target="#file-modal"">Table 2

Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in Parkinson’s disease.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
Anti-cell deathβ-asaroneRat, cell6-OHDA-induced rats
SN4741 cells
Motor function ↑ (OFT, RRT, forelimb akinesia)In vitro: LC3-II ↓
In vivo: HVA, Dopacl, 5-HIAA, Bcl-2 ↑
Beclin-1, JNK, p-JNK ↓, Bcl-2 ↑
[37]
β-asaroneMouse, cellMPTP-induced mice
SH-SY5Y cells
Motor function ↑ (RRT)In vitro: TH+ cell ↑, MALAT1, α-syn, CHX, MG132 ↓
In vivo: MALAT1 ↓
[38]
Antioxidantα-asaroneMouse, cellMPTP-induced mice
BV-2 cells
Motor function ↑ (Y-maze test and pole test)In vitro: NO, iNOS, COX-2, TNF-α, IL-6, IL1β, NF-κB, IκB ↓
In vivo: Mac-1, CD-68, Iba-1, iNOS, COX-2, DOPAC ↓
[39]
β-asaroneRat6-OHDA-induced ratsER stress ↓GRP78, p-PERK, CHOP, Beclin-1 ↓
Bcl-2 ↑
[40]
β-asaroneRat6-OHDA-induced ratsER stress ↓IRE1, p-IRE1, XBP1 ↓[41]
β-asaroneRat6-OHDA-induced ratsCMA↑, Autophagy ↓HSC70, HSP70, MEF2D, LAMP-2A level ↑
α-Syn ↓
[42]
Anti-inflammationAG extractMouseMPTP-induced mice
BV-2 cells
Cell death ↓ Neuroinflammation ↓TH+ cell ↑
NO, iNOS, TNF-α, IL-6, IL1β, NF-κB, IκB ↓
[43]
β-asaroneRat6-OHDA-induced ratsMotor function ↑ (OFT, RRT, forelimb activity)α-Syn, Il-1β, TNF-α, NO, IL-6, BAX, Caspase ↓
TH, SOD, CAT, GSH-Px, Bcl-2 ↑
[44]
Coordination with levodopaβ-asarone + L-dopaRatSD ratL-Dopa, DA ↑DA ↑, COMT ↓[45]
β-asarone + L-dopaRat6-OHDA-induced ratsMotor function ↑ (OFT, ST, RRT)DDC level, DA level, MAO-B, COMT, DOPAC/DA, HVA/DA, TH, DAT ↑[46]
β-asarone + L-dopaRat6-OHDA-induced ratsL-Dopa BBB permeability ↑L-dopa, DA, DOPAC, HVA ↑
S100β ↑
NSE, P-gp, ZO-1, occludin, actin, claudin-5 ↓
[47]
β-asarone + L-dopaRat6-OHDA-induced ratsAutophagy activity ↓Beclin-1, LC3B ↓
p62 expression ↑
[48]

SD, Sprague Dawley; 6-OHDA, 6-hydroxydopamine; OFT, open-field test; RRT, rotarod test; LC3-II, light chain 3-II; HVA, homovanillic acid; Dopacl, 3,4-dihydroxyphenylacetic acid; 5-HIAA, 5-hydroxyindole acetic acid; JNK, c-Jun N-terminal kinase; Bcl-2, B-cell lymphoma; MPTP, 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine; TH, tyrosine hydroxylase; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; α-syn, α-synuclein; CHX, cycloheximide; MG132, a proteasome inhibitor; NO, nitric oxide; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2; TNF-α, tumor necrosis factor-alpha; IL-, interleukin-; NF-κB, nuclear factor kappa B; IκB, NF-κB inhibitor; Mac-1, macrophage Ag complex-1; CD-68, cluster of differentiation 68; Iba-1, ionized calcium-binding adapter molecule 1; DOPAC, 3, 4-dihydroxyphenylacetic acid; GRP78, glucose-regulated protein 78; p-PERK, phosphorylated protein kinase RNA-like endoplasmic reticulum kinase; CHOP, C/EBP homologous binding protein; ER, endoplasmic reticulum; IRE1, inositol-requiring enzyme 1; p-IRE1, phosphorylated IRE1; XBP1, X-box binding protein 1; CMA, chaperone-mediated autophagy; HSC70, heat-shock cognate protein 70; HSP70, heat-shock protein 70; MEF2D, myocyte enhancer factor 2D; LAMP-2A, lysosomal membrane protein receptor type 2A; BAX, B-cell lymphoma 2-associated X protein; TH, thyrosine hydroxylase; SOD, superoxide dismutase; CAT, catalase; GSH-Px, glutathione peroxidase; l-dopa, levodopa; DA, dopamine; COMT, catechol-O-methyltransferase; DAT, dopamine transporter; ST, stepping test; DDC, dopa decarboxylase; MAO-B, monoamine oxidase-B; BBB, blood–brain barrier; S100β, S100 calcium-binding protein β; NSE, neuron-specific enolase; P-gp, P-glycoprotein; ZO-1, zonula occludens-1; LC3B, microtubule-associated protein light chain 3B..



Dopaminergic neuron damage due to neurotoxicity, oxidative stress, and inflammation is considered the underlying pathogenesis of PD. β-asarone can improve motor function by inhibiting α-synuclein (α-syn) aggregation, dopaminergic cell death, and autophagy. The administration of β-asarone improves behavioral symptoms and elevates tyrosine hydroxylase (TH) levels via JNK/Bcl-2/Beclin-1 signaling in 6-hydrooxydopamine (6-OHDA)-treated rats and SN4741 cells [37]. β-Asarone has also shown a neuroprotective effect by modulating metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), which induces neuronal death and α-syn expression in a 1-methyl-4 phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mouse model and in 1-methyl-4-phenylpyridinium (MPP+)-treated SH-SY5Y cells, as the corresponding in vitro model [38].

Asarone has also been shown to attenuate behavioral deficits in PD animal models via its anti-inflammatory and antioxidant effects. α-Asarone attenuated microglia-mediated neuroinflammation by inhibiting the activation of NF-ĸB in LPS-treated BV2 cells and mitigated PD-like behavioral impairments in an MPTP-treated mouse model [39]. Moreover, β-asarone reduced autophagy and endoplasmic reticulum (ER) stress by inhibiting the protein kinase RNA-like ER kinase (PERK)/C/EBP homologous binding protein (CHOP)/Bcl-2/Beclin-1 pathway [40] and the inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (XBP1) pathway [41] and by modulating the heat-shock protein 70 (Hsp70)/mitogen-activated protein kinase (MAPK)/myocyte enhancer factor 2D (MEF2D)/Beclin-1 pathway [42] in a 6-OHDA-treated rat model. The aqueous extract of AG has been reported to inhibit neuroinflammation via the modulation of MAPKs, nuclear factor kappa B (NF-κB), and TIR domain-containing adapter-inducing interferon-β (TRIF) dependent signaling in LPS-stimulated BV2 cells and prevent neurotoxicity in an MPTP-treated mouse model [43]. In a recent study, β-asarone reduced neuron damage by decreasing α-syn and inhibiting oxidative stress, inflammatory reactions, and cell apoptosis in a 6-OHDA-treated parkinsonism rat model [44].

Asarone has been shown to further induce the efficacy of l-dopa via the co-treatment with l-dopa. This coadministration increased DA in the striatum of naive rats [45] and 6-OHDA rats [46-48]. Furthermore, β-asarone affected the transformation of l-dopa to DA by modulating catechol-O-methyltransferase (COMT) and DA metabolism [45], inhibiting autophagy activity via the downregulation of microtubule-associated protein light chain 3B (LC3B) and Beclin-1 expression and the upregulation of p62 expression [48], and promoting l-dopa into the brain via the modulation of tight junction proteins and P-glycoprotein in the BBB [47] and via the regulation of dopa decarboxylase, TH, COMT, MAO-B, and DA transporter levels [46].

4. Depression and anxiety (Table 3)

Depression is a representative mental illness that is accompanied by symptoms in the areas of mood, cognitive, and motor functions. Biological and psychosocial factors are involved in different ways during depression. However, the best-known theory for the biological etiology of depression is the monoamine hypothesis since monoamine neurotransmitters, such as serotonin, noradrenaline, and DA, are known to be involved in mood regulation. This theory is supported by the fact that selective serotonin-, norepinephrine-DA-, and serotonin-norepinephrine-reuptake inhibitors, which are drugs that act on these substances, are currently being used as antidepressants [49].

CUMS, chronic unpredictable mild stress; SPT, sucrose-preference test; FST, forced-swimming test; BDNF, brain-derived neurotrophic factor; ERK, extracellular signal-regulated kinases; CREB, cAMP response element-binding protein; OFT, open-field test; Trk-B, tropomyosin receptor kinase B; Bcl, B-cell lymphoma; Bad, Bcl-2-associated death promoter; MKP-1, mitogen-activated protein kinase phosphatase-1; p-ERK, phosphorylated extracellular signal-regulated kinases; p-CREB, phosphorylated cAMP response element-binding protein; EO, essential oil; AT, Acorus tatarinowii; TST, tail-suspension test; EPM, elevated plus maze; LDT, light/dark-transition test; NFC, novel–food-consumption test; MBT, marble-burying test; CFA: complete Freund’s adjuvant; MDA, malondialdehyde; CAT, catalase; GSH-R, glutathione reductase; GSH-Px, glutathione peroxide; AMPARs, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors; NMDARs, NR2A-containing N-methyl-d-aspartate recpetors; GABAAs, γ-aminobutyric acid type A receptors; HBT, hole-board test; TH, tyrosine hydroxylase..

&md=tbl&idx=3' data-target="#file-modal"">Table 3

Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in depression and anxiety.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
Anti-depressionβ-asaroneRatCUMS exposed ratsDepressant-like behavior ↓ (SPT, FST)BDNF ↑, ERK1/2 and CREB phosphorylation ↑[50]
β-asaroneRatCUMS exposed ratsDepressant-like behavior ↓ (SPT, OFT, FST)Apoptosis ↓, CREB ↑, BDNF ↑, Trk-B ↑, Bcl-2 ↑, Bad ↓, ERK ↑[51]
β-asaroneRatCUMS exposed ratsBody weight ↑
Depressant-like behavior ↓ (SPT, OFT)
MKP-1 ↓, p-ERK1/2 ↑, BDNF ↑[52]
α-asaroneMouseNicotine withdrawal induced miceDepressant-like behavior ↓ (FST)p-CREB ↓[53]
EO from AT, α-asarone, β-asaroneMouseNormal miceDepressant-like behavior ↓ (FST, TST)[54]
α-asaroneMouseNormal miceDepressant-like behavior ↓ (TST)[55]
Anti-anxietyα-asaroneMouseNormal miceAnxiolytic-like behavior ↑ (EPM, LDT, NFC, MBT)[56]
α-asaroneRatSleep deprived ratsAnxiolytic-like behavior ↑ (EPM, OFT)MDA ↓, CAT ↑, GSH-R ↑, GSH-Px ↑[57]
α-asaroneMouseCFA-induced chronic
Inflammatory pain mice
Anxiolytic-like behavior ↑ (EPM, OFT)AMPARs ↓, NMDARs ↓, GABAARs ↑, hyper-excitability of pyramidal neurons ↓[58]
α-asaroneRatCorticosterone-induced anxiety ratsAnxiolytic-like behavior ↑ (EPM, HBT)TH ↓, BDNF ↓, TrkB ↓[59]

CUMS, chronic unpredictable mild stress; SPT, sucrose-preference test; FST, forced-swimming test; BDNF, brain-derived neurotrophic factor; ERK, extracellular signal-regulated kinases; CREB, cAMP response element-binding protein; OFT, open-field test; Trk-B, tropomyosin receptor kinase B; Bcl, B-cell lymphoma; Bad, Bcl-2-associated death promoter; MKP-1, mitogen-activated protein kinase phosphatase-1; p-ERK, phosphorylated extracellular signal-regulated kinases; p-CREB, phosphorylated cAMP response element-binding protein; EO, essential oil; AT, Acorus tatarinowii; TST, tail-suspension test; EPM, elevated plus maze; LDT, light/dark-transition test; NFC, novel–food-consumption test; MBT, marble-burying test; CFA: complete Freund’s adjuvant; MDA, malondialdehyde; CAT, catalase; GSH-R, glutathione reductase; GSH-Px, glutathione peroxide; AMPARs, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors; NMDARs, NR2A-containing N-methyl-d-aspartate recpetors; GABAAs, γ-aminobutyric acid type A receptors; HBT, hole-board test; TH, tyrosine hydroxylase..



However, many studies describing the effects of asarone in improving depression are related to the improvement of brain functions, mainly by increasing the production of BDNF and other neurotrophic factors rather than the production of neurotransmitters. The administration of β-asarone reversed depression-like behaviors in an unpredictable chronic mild stress (UCMS)-treated model, which is related to enhanced neurogenesis and decreased neuronal cell death in the hippocampus [50-52]. In addition to these therapeutic effects, β-asarone increased BDNF by enhancing ERK1/2, CREB phosphorylation, tropomyosin receptor kinase B (Trk-B), and Bcl-2 and by reducing Bad and mitogen-activated protein kinase phosphatase-1 (MKP-1) [50-52]. In addition, α-asarone also attenuated depression-like behavior via the modulation of hippocampal pCREB levels in a nicotine-withdrawn mouse model of depression [53].

The antidepressant results of asarone are also demonstrated in normal mouse models. The major essential oil components from AT (essential oils and asarone) have shown antidepressant-like efficacy in normal mice [54], and the effects of α-asarone promoted mediation via the noradrenergic and serotonergic systems [55].

The effects of asarone on anxiety symptoms have also been studied. In particular, α-asarone alleviated anxiety in various experimental animal models. First, the anxiolytic potential of α-asarone has been observed in normal mice using various behavioral tests, and the effects of α-asarone were similar to that of diazepam [56]. The administration of α-asarone improved insomnia-associated anxiety and cognitive functions by inhibiting lipid peroxidation and enhancing the activities of CAT and glutathione reductase (GSH-R) in a sleep-deprived rat model [57]. α-Asarone has also shown an anxiolytic-like activity in a chronic pain-related anxiety model because of the maintenance of the stability between excitatory and inhibitory communications and the attenuation of the hyperexcitability of excitatory neurons in the basolateral amygdala [58]. Moreover, the administration of α-asarone prior to corticosterone treatment improved anxiety. This effect was related to the regulation of the noradrenergic system and BDNF via the modulation of the Trk-B signaling process in rats [59].

5. Epilepsy (Table 4)

Epilepsy is a chronic neurological disorder that is characterized by persistent seizures despite the absence of physical abnormalities [60]. The antiepileptic and anticonvulsive effects of α-asarone have been investigated in various rodent seizure models [61, 62]. Decoctions and volatile oils extracted from AT prevented convulsions associated with convulsion-induced GABAergic neuron injury in a pentylenetetrazole (PTZ)-treated model [63]. The water extracts and essential oils of AG acted on the central nervous system via the GABAergic system [64, 65] and exhibited neuroprotective effects by blockading NMDA receptor activity [66]. Moreover, α-asarone modulated neurotransmitters, thereby suppressing the seizures caused by intense abnormal excitability. However, the antiepileptic action of α-asarone was mediated by GABAergic regulation and not by the antagonism of acetylcholine receptors [67-70].

MES, maximal electroshock; scPTZ, subcutaneous pentylenetetrazol seizure; LI-PILO, lithium-pilocarpine; PTZ, pentylenetetrazol; SE, status epilepticus; SRS, spontaneous recurrent seizures; AT, Acorus tatarinowii; GABA-IR, GABA-like immunoreactivity; AG, Acorus gramineus; GABA, γ-aminobutyric acid; EO, essential oil; NMDAR, N-methyl-d-aspartate receptor; LCA, locomotor activity; BT, body temperature; nAChRs, nicotinic acetylcholine receptor; TLE, temporal lobe epilepsy; GAD67, glutamic acid decarboxylase 67; GABAAR, γ-aminobutyric acid type A receptor; GABA-T, GABA transaminase; CNaIIA, type IIA Na+ channel; Nav1.2 channel, a dominant rat brain Na+ channel subtype; NMDA, N-methyl-d-aspartate; WMT, water maze test; LPS, lipopolysaccharide; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells..

&md=tbl&idx=4' data-target="#file-modal"">Table 4

Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in epilepsy.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
α-asaroneMouse, ratMES-induced seizure mice
scPTZ-induced seizure mice
LI-PILO-induced epilepsy rats
Seizure incidence, severity, frequency ↓, latency ↑[61]
α-asaroneMouse, ratMES-induced seizure mice
PTZ-induced seizure mice
LI-PILO-induced SE rats
SRS induced rats
Seizure onset, incidence, latency, severity, mortality, frequency ↓[62]
NeurotransmitterDecoction and volatile oil of ATMouse, ratMES-induced seizure mice
PTZ-induced seizure mice
Prolonged PTZ-induced seizure rats
Convulsant ↓, mortality ↓, seizure latency ↑, seizure intensity ↓GABA-IR neurons ↑, GABA-IR neuron damage ↓[63]
Water extract of AGMousePTZ-induced seizure miceOnset of seizure and death ↓GABA agonist[64]
EO of AGMousePTZ-induced seizure miceConvulsion ↓GABA transaminase ↓, GABA ↑, glutamate content ↓[65]
EO of AGCellGlutamate-induced excitotoxicity in primary rat cortical cellsExcitotoxicity ↓, neuroprotection ↑NMDAR antagonist[66]
α-asaroneRatPTZ-induced epilepsy rats
Kainate-induced epilepsy rats
Latency of seizures ↑, Susceptibility to seizure ↓Firing rate of spontaneous spiking ↓, tonic GABAergic inhibition ↑, inducing inward currents when picotoxin and bicuculline together ↓[67]
α-asaroneMouseNicotine-induced seizure miceLCA and BT ↓, onset time of seizures ↑[68]
α-asaroneRatLI-PILO-induced TLE ratsGABAergic modulationGABA ↑, GAD67 ↑, GABAAR-mRNA ↑, GABA-T ↓[69]
α-asaroneCellCNaIIA cell lineSpontaneous firing of mitral cells and Na+ channel ↓Spontaneous firing of output neurons, mitral cells ↓, Nav1.2 currents ↓[70]
Antioxidantα-asaroneMousePTZ-induced seizure mice
Picrotoxin-induced seizure mice
NMDA-induced seizure mice
PILO-induced seizure mice
MES-induced seizure mice
Treadmill performance and LCA ↓, hypothermia ↑, sleep ↑, onset of seizures ↓Antioxidant enzymes ↑[71]
Anti-inflammationα-asaroneRatPILO-induced TLE ratsCognitive function ↑ (WMT), behavioral score of SRSs ↓, frequency of seizures ↓Microglial activation ↓, proinflammatory cytokine ↓, LPS-stimulated neuroinflammatory responses ↓, NF-κB ↓[72]

MES, maximal electroshock; scPTZ, subcutaneous pentylenetetrazol seizure; LI-PILO, lithium-pilocarpine; PTZ, pentylenetetrazol; SE, status epilepticus; SRS, spontaneous recurrent seizures; AT, Acorus tatarinowii; GABA-IR, GABA-like immunoreactivity; AG, Acorus gramineus; GABA, γ-aminobutyric acid; EO, essential oil; NMDAR, N-methyl-d-aspartate receptor; LCA, locomotor activity; BT, body temperature; nAChRs, nicotinic acetylcholine receptor; TLE, temporal lobe epilepsy; GAD67, glutamic acid decarboxylase 67; GABAAR, γ-aminobutyric acid type A receptor; GABA-T, GABA transaminase; CNaIIA, type IIA Na+ channel; Nav1.2 channel, a dominant rat brain Na+ channel subtype; NMDA, N-methyl-d-aspartate; WMT, water maze test; LPS, lipopolysaccharide; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells..



In addition, α-asarone modulated neuronal excitability in rat hippocampal cell cultures and suppressed the epileptic symptoms of mice in a PTZ or kainate seizure animal model, presumably based on the activation of GABAA receptors [67]. Furthermore, α-asarone produced antiepileptic effects in a lithium-pilocarpine-treated rat model via modulating GABAergic homeostasis, decreasing GABA degradation by lowering the activity of GABA-T, increasing GABA levels by increasing the expression of glutamic acid decarboxylase 67 (GAD67), and increasing GABA-mediated inhibition by increasing the expression of GABAA receptor [69]. α-Asarone has also been shown to block the Na+ channel and activate GABAA receptors in the mitral cells of the olfactory bulb in mice brain slices [70]. According to a recent study, α-asarone pretreatment prolonged the onset time of nicotine-treated mice seizures but not the relationship to nicotinic acetylcholine receptors [68].

In addition to these effects, α-asarone has been shown to have antioxidant and anti-inflammatory effects; therefore, it could provide protection from brain damage. Treatment with α-asarone delayed the onset time of clonic and tonic seizures in various animal seizure models and induced antioxidant enzymes, such as SOD, GPX, and GSH-R, in the brain, particularly in the cortex, striatum, and hippocampus [71]. α-Asarone has also been shown to arrest the inflammatory process via the transcriptional level regulation of NF-κB by inhibiting the degradation pathways of NF-κB inhibitor (IκB) alpha (IκBα) and IκB beta (IκBβ) in pilocarpine-treated status epilepticus rats and LPS-treated microglial cells [72].

6. Stroke (Table 5)

Stroke is a neurological disorder wherein the blood vessels supplying blood to the brain become blocked or rupture, thus causing damage to the brain. Pathophysiological events arise in stroke, including energy deprivation, glutamate-induced excitotoxicity, oxidative stress, inflammation, and BBB breakdown [73].

OGD/R, 2 hours of oxygen-glucose deprivation followed by 24 hours of reperfusion; LPS, lipopolysaccharide; MMP, mitochondrial membrane potential; MCAO, middle cerebral artery occlusion; JNK, c-Jun N-terminal kinase; p-JNK, phosphorylated c-Jun N-terminal kinase; Bcl-2, B-cell lymphoma 2; GFAP, glial fibrillary acidic protein; Iba-1, ionized calcium-binding adaptor molecule-1; LC3, microtubule-associated protein light chain 3; SOD, superoxide dismutase; LDH, lactate dehydrogenase; MDA, malondialdehyde; ROS, reactive oxidative species; RPPH1, ribonuclease P RNA component H1; PAPs, pro-apoptotic proteins; AAPs, antiapoptotic proteins; Nrf2, nuclear factor erythroid 2-related factor 2; ARE, antioxidant response elements; NPC, neural progenitor cell; CIR, cerebral ischemia-reperfusion; GSH, glutathione; LPO, lipid peroxidation; GPx, glutathione peroxidase; GR, glutathione reductase; CAT, catalase; GST, glutathione S transferase; LPS, lipopolysaccharides; NO, nitric oxide; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2; NF-κB, nuclear factor-κB; BBB, blood–brain barrier; AT, Acorus tatarinowii; NKCC1, Na-K-Cl cotransporter; AQP4, aquaporin 4; ICAM, Na-K-Cl cotransporter-1; MMP-9, matrix metallopeptidase 9..

&md=tbl&idx=5' data-target="#file-modal"">Table 5

Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in stroke.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
Neuroprotectionβ-asaroneCellPC12 cells (OGD/R)Cell viability ↑, autophagy ↓MMP ↑, Beclin-1 ↓, [Ca2+]I ↓[74]
β-asaroneRatMCAOAutophagy ↓Beclin1 ↓, JNK ↓, p-JNK ↓, Bcl-2 ↑[75]
α-asaroneRatMCAOInfarct volume ↓
Epilepsy ↓
Neurological function ↑
Apoptosis ↓, GFAP ↓, Iba-1 ↓, LC3II/LC3I ↓, p62 ↑[76]
β-asaroneCellHypoxia induced PC12 cellsCell viability ↑SOD ↑, MMP ↑, apoptosis ↓, LDH ↓, ROS ↓, RPPH1 ↓[77]
α-asaroneRat4-Vessel occlusionNeuroprotection ↑Cell death ↓, damaged pyramidal neurons ↓[78]
β-asaroneRatMCAOInfarction volume ↓Apoptosis ↓, PAPs ↓, AAPs ↑, Nrf2-ARE pathway-related proteins ↑[79]
Antioxidantβ-asaroneRatMCAOMotor function ↑LDH ↓, GSH ↑, LPO ↓, GPx ↑, GR ↑, CAT ↑, GST ↑[80]
Anti-inflammationβ-asaroneCellLPS-stimulated BV-2 microglial cellsAnti-inflammatory effects ↑NO ↓, iNOS ↓, COX-2 ↓, NF-κB ↓[81]
BBB protectionAT extractRatMCAOInfarct size, edema ↓
BBB permeability ↓
Neurological function ↑
Astrocytic NKCC1/AQP4 ↓
JNK/iNOS-mediated ICAM-1/MMP-9 signaling ↓
[82]
Neurogenesisα-asaroneMouseMCAOMotor function ↑Differentiation of transplanted NPCs ↑[83]

OGD/R, 2 hours of oxygen-glucose deprivation followed by 24 hours of reperfusion; LPS, lipopolysaccharide; MMP, mitochondrial membrane potential; MCAO, middle cerebral artery occlusion; JNK, c-Jun N-terminal kinase; p-JNK, phosphorylated c-Jun N-terminal kinase; Bcl-2, B-cell lymphoma 2; GFAP, glial fibrillary acidic protein; Iba-1, ionized calcium-binding adaptor molecule-1; LC3, microtubule-associated protein light chain 3; SOD, superoxide dismutase; LDH, lactate dehydrogenase; MDA, malondialdehyde; ROS, reactive oxidative species; RPPH1, ribonuclease P RNA component H1; PAPs, pro-apoptotic proteins; AAPs, antiapoptotic proteins; Nrf2, nuclear factor erythroid 2-related factor 2; ARE, antioxidant response elements; NPC, neural progenitor cell; CIR, cerebral ischemia-reperfusion; GSH, glutathione; LPO, lipid peroxidation; GPx, glutathione peroxidase; GR, glutathione reductase; CAT, catalase; GST, glutathione S transferase; LPS, lipopolysaccharides; NO, nitric oxide; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2; NF-κB, nuclear factor-κB; BBB, blood–brain barrier; AT, Acorus tatarinowii; NKCC1, Na-K-Cl cotransporter; AQP4, aquaporin 4; ICAM, Na-K-Cl cotransporter-1; MMP-9, matrix metallopeptidase 9..



Asarone has been shown to provide neuroprotective effects against stroke-induced damage by inhibiting autophagy and neuronal cell death. Both α-asarone and β-asarone attenuate ischemia-induced injury by inhibiting autophagy [74-76]. β-Asarone has previously been shown to attenuate Beclin-1-dependent autophagy in PC12 cells after oxygen-glucose deprivation followed by reperfusion [74] and ischemia-reperfusion-induced autophagy by regulating Bcl-2, Beclin 1, JNK, and p-JNK in a middle cerebral artery occlusion (MCAO) rat model [75]. α-Asarone treatment has also been shown to reduce the infarct volume, improve neurological functions, decrease the expression of ionized calcium-binding adaptor molecule-1 (IBA1) and LC3, and increase the expression of p62 in MCAO rats, thus suggesting that α-asarone attenuates ischemic brain injury by modulating the activation of glia and autophagy [76]. Asarone has also been shown to play a role in inhibiting neuronal cell death [77, 78]. Treatment with β-asarone mitigated neuronal death by negatively regulating the ribonuclease P RNA component H1 (H1RNA)/MiR-542-3p/death effector domain (DED)-containing 2 signaling pathway in hypoxia-treated PC12 cells [77]. β-Asarone also decreased the infarction volume and apoptotic cell death via the activation of Nrf2-antioxidant response element signaling in MCAO rats [79]. Additionally, α-asarone reduced neuronal death in the hippocampus in a study using a four-vessel occlusion model of rats [78].

Asarone has also been shown to improve brain function via antioxidant, anti-inflammatory, and BBB-protecting activities in ischemic models. β-Asarone treatment increased glutathione (GSH) levels by decreasing lipid peroxidation and restoring the activity of endogenous antioxidant enzymes involving GPX, GSH-R, CAT, and GSH S transferases, thus indicating that β-asarone might have antioxidant activity against MCAO ischemic rats [80]. In addition, AG ethanolic extract and its active component, β-asarone, exhibited anti-inflammatory outcomes by suppressing proinflammatory mediators via NF-κB and JNK signaling in LPS-treated BV2 microglia cells [81]. Furthermore, AT extract reduced brain edema by alleviating astrocytic swelling and BBB breakdown, which are associated with the downregulation of astrocytic Na-K-Cl cotransporter 1 (NKCC1)/AQP4 and JNK/inducible nitric oxide synthase (iNOS)-mediated NKCC1/mitochondrial membrane potential 9 signaling [82].

A recent report suggested that α-asarone influences primary cultured NPCs and an ischemic stroke mouse model [83]. α-Asarone promoted the proliferation of NPCs and the differentiation of neuron-lineage cells via the activation of ERK, β-catenin, and cyclin D1, thereby facilitating neurofunctional recovery after NPC transplantation and ischemic brain injury.

7. Effects of formulas and decoctions, including A. gramineus and A. tatarinowii on neurological disorders (Table 6)

The therapeutic potentials of nine AG- and AT-containing herbal formulas and decoctions against neurological disorders were reviewed. The composition of each formula is listed in Table 7. The most studied prescription is Kai Xin San, an herbal formula composed of Radix ginseng, poria, R. polygalae, and A. tatarinowii rhizome. This formula has been used to improve memory, cognition, depression, and other neurological symptoms for thousands of years in China [84-90]. Kai Xin San ameliorated cognitive dysfunction in Aβ-treated mice [85] and rats [90], scopolamine-induced mice [88], and multi-infarct dementia rats [87]. In addition, Kai Xin San has also been shown to have an antidepressant effect in rat models with chronic mild stress [84] and UCMS [86, 89].

CUMS, chronic unpredictable mild stress; SPT, sucrose-preference test; NIH, novelty-induced hypophagia; AD, Alzheimer’s disease; Aβ, amyloid-beta; SDT, step-down test; OFT, open-field test; FST, forced-swim test; ICA, internal carotid artery; MWM, Morris water maze; SCOP, scopolamine; NOR, novel-object recognition; IDE, insulin-degrading enzyme; CCA, common carotid artery; GPCRAC, the combination of Gastrodia elata, Polygala tenuifolia, Cistanche deserticola, Rehmannia lutinosa, Acorus gramineus, Curcuma longa; BA, broaden area; PD, Parkinson’s disease; MPP+, 1-methyl-4-phenylpyridinium; MPTP, 1-methyl-4 phenyl-1, 2, 3, 6-tetrahydropyridine; TH-IR, tyrosine hydroxylase-immunoreacting..

&md=tbl&idx=6' data-target="#file-modal"">Table 6

Therapeutic potential of formulas including Acorus gramineus Solander and Acorus tatarinowii Schott in neurological disorders.

ComponentDisordersSpeciesResultsRef.
Kai Xin SanDepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, NIH, Two-Way Active Avoidance Test)[84]
ADAβ-injected ratsMemory ↑ (SDT)[85]
DepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, OFT, FST)[86]
StrokeICA embolic ratsCognitive function ↑ (MWM)[87]
DementiaSCOP induced miceCognitive function ↑ (MWM, Y-maze)[88]
DepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, OFT), weight ↑[89]
ADAβ-injected ratsCognitive function ↑ (NOR), injured neurons ↓, Aβ level ↓, IDE expression ↑[90]
Kaixin JieyuDepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, OFT)[91]
StrokeCCA ligation ratsAntidepressant-like behavior ↑ (SPT, OFT)[92]
Qisheng Wan formulaADAβ-injected ratsCognitive function ↑ (MWM)[93]
Bushen Tisansui DecoctionADAβ-injected ratsCognitive function ↑ (MWM)[94]
GPCRACDementiaSCOP induced miceMemory ↑ (SDT)Cognitive function ↑ (MWM)[95]
Bazhu DecoctionAD5×FAD transgenic miceCognitive function ↑ (OFT, Y-maze, MWM)[96]
Chong Myung TangDementiaSCOP induced miceCognitive & memory function ↑ (passive avoidance test, MWM)[97]
Yishen Huazhuo DecoctionADAD patient (human)Cerebral activity ↑ [supramarginal gyrus (BA 40), superior temporal gyrus (BA 22)][98]
Yeolda Hanso TangPDMPP+ -induced cells
MPTP-induced mice
Cell viability ↑ (survival ratio of TH-IR cell ↑)[99]

CUMS, chronic unpredictable mild stress; SPT, sucrose-preference test; NIH, novelty-induced hypophagia; AD, Alzheimer’s disease; Aβ, amyloid-beta; SDT, step-down test; OFT, open-field test; FST, forced-swim test; ICA, internal carotid artery; MWM, Morris water maze; SCOP, scopolamine; NOR, novel-object recognition; IDE, insulin-degrading enzyme; CCA, common carotid artery; GPCRAC, the combination of Gastrodia elata, Polygala tenuifolia, Cistanche deserticola, Rehmannia lutinosa, Acorus gramineus, Curcuma longa; BA, broaden area; PD, Parkinson’s disease; MPP+, 1-methyl-4-phenylpyridinium; MPTP, 1-methyl-4 phenyl-1, 2, 3, 6-tetrahydropyridine; TH-IR, tyrosine hydroxylase-immunoreacting..



Table 7

Compositions of the formulas including Acorus gramineus Solander and Acorus tatarinowii Schott.

FormulaComposition
Kai-Xin-SanPanax ginseng, Poria cocos, Polygala tenuifolia, and Acori Tatarinowii
Kaixin-jieyuRadix Ginseng, Radix Paeoniae rubra, Acorus Gramineus soland, Fructus Aurantii immaturus, Radix Polygalae, Poria, Morinda Officinalis and Glycyrrhiza
Qisheng Wan FormulaPoria cocos, Cinnamomum cassia, Polygala tenuifolia, Panax ginseng, Asparagus cochinchinensis, Acorus tatarinowii, Lycium chinense
Bushen TiansuiEpimedium brevicornum, Polygonum multiflorum, Chinemys reevesii, Fossilia Ossis Mastodi, Polygala, Acorus tatarinowii
GPCRACGastrodia elata, Polygala tenuifolia, Cistanche deserticola, Rehmannia lutinosa, Acorus gramineus, Curcuma longa
Yishen Huazhuo DecoctionEpimedium, Fructus ligustri, Psoralea fruit, Radix polygoni multiflori, Radix astragali, Ligusticum wallichi franchat, Acorus gramineus
Bazhu DecoctionRadix Morindae Officinalis, Asiatic Cornelian Cherry Fruit, Grassleaf Sweetflag Rhizome, Earth Worm, Arisaema Cum Bile
Yeolda-Hanso TangPueraria lobata, Angelica tenuissima, Scutellaria baicalensis, Platycodon grandiflorum, Angelicae Dahurica, Cimicifuga heracleifolia, Raphanus sativa, Polygala tenuifolia, Acorus gramineus, Dimocarpus longan
Chong-Myung TangAcorus gramineus, Polygala tenuifolia, Poria cocos


Kaixin Jieyu decoction is an herbal medicine preparation from Sini powder and Kai Xin San. This preparation has been shown to reduce depression-like behavior via the production of monoamines in a UCMS rat model [91] and the expression of glial fibrillary acidic protein (GFAP) and BDNF in the hippocampus [92]. In addition, other studies have reported that the Qisheng Wan formula [93]; Bushen Tiansui decoction [94]; GPCRAC with extracts from Polygala tenuifolia, Rehmannia lutinosa, Gastrodia elata, Cistanche deserticola, AG, and Curcuma longa [95]; Bazhu decoction [96]; and Chong Myung Tang [97] ameliorated the cognitive impairment of AD animals. Moreover, a study reported that the Yishen Huazhuo decoction improved brain activity in AD patients [98]. The curative potential of Yeolda Hanso Tang in the prevention and treatment of PD has been studied using a PC12 cell model and an MPTP-treated PD mouse model [99].

CONCLUSION

AG and AT, which is commonly referred to as “Shi Chang Pu,” have been widely used for improving mental, cognitive, and learning capacities in Korean medicine, and α- and β-asarone, which are the bioactive phytochemicals of AG and AT, are the most studied agents in the treatment of various diseases. In this review, 73 studies showed the potential neuroprotective function of the extracts and the compounds of AG or AT (α-asarone and β-asarone) in neurological disorders. They improved behavioral functions and neuronal cell survival, and their effects were associated with several potential mechanisms of action, including reduction of pathogenic protein aggregates, antiapoptotic activity, regulation of autophagy, anti-inflammatory and antioxidant activities, modulation of neurotransmitters, and activation of neurotrophic factors and neurogenesis (Fig. 3).

Figure 3. Mechanism of action of extracts or active components of Acorus gramineus Solander and Acorus tatarinowii Schott in neurological disorders.

These neuroprotective features make asarones from AG or AT a potential therapeutic for treating neurological disorders, such as AD, PD, depression, anxiety, epilepsy, and stroke. These results can also explain the therapeutic effects of traditional Korean medicines, including Shi Chang Pu, on neurological diseases. However, the most studied AG- and AT-containing formulas and decoctions against neurological disorders were of Chinese traditional medicine origin. Therefore, more research is needed on the therapeutic potentials of the Korean medical formulas and decoctions, including Shi Chang Pu, against neurological disorders. In this review, several limitations of this study also require consideration. First, we searched only databases written in English, and the exclusion of studies published in non-English languages may lead to some selection bias. Second, many studies evaluated the neuroprotective effects on neurological diseases by employing preclinical in vitro and in vivo studies, which differ greatly from human patients. Therefore, we hope this review will encourage clinical trials on AG and AT, as well as their active components, in patients with neurological disorders.

ACKNOWLEDGMENT

This work was supported by a 2-Year Research Grant of Pusan National University.

CONFLICT OF INTEREST

The authors have no conflicts of interest to declare.

Fig 1.

Figure 1.Alpha (α)-asarone (1,2,4-trimethoxy-5-[(E)-prop-1-enyl]benzene; PubChem CID: 636822) and beta (β)-asarone (1,2,4-trimethoxy-5-[(Z)-prop-1-enyl]benzene; PubChem CID: 5281758).
Journal of Pharmacopuncture 2022; 25: 326-343https://doi.org/10.3831/KPI.2022.25.4.326

Fig 2.

Figure 2.Flow diagram.
Journal of Pharmacopuncture 2022; 25: 326-343https://doi.org/10.3831/KPI.2022.25.4.326

Fig 3.

Figure 3.Mechanism of action of extracts or active components of Acorus gramineus Solander and Acorus tatarinowii Schott in neurological disorders.
Journal of Pharmacopuncture 2022; 25: 326-343https://doi.org/10.3831/KPI.2022.25.4.326

Table 1 . Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in Alzheimer’s disease.

EffectCompoundSpeciesExperimental modelExperiment resultMechanismsRef.
Anti-Aβ accumulationSCP-OilWormCaenorhabditis elegans modelSerotonin sensitivity and olfactory learning skill ↑Misfolded Aβ and polyQ proteins ↓[7]
β-asaroneMouseAPP/PSI double transgenic miceSenile plaques in the hippocampus & Aβ ↓↑Levels of Aβ40 and Aβ42 in hippocampus ↓[8]
β-asaroneMouseAPP/PSI transgenic miceLearning and memory function ↑Beclin-1-dependent autophagy (the PI3K/Akt/mTOR pathway) ↓[9]
Anti-apoptosisβ-asaroneMouseAPP/PSI double transgenic miceCognitive function ↑CaMKII-α/p-CREB/Bcl-2 pathway ↑[10]
β-asaroneCellAβ42 injury cellsNeuronal apoptosis ↓Bax ↓, Bcl-2 ↑[11]
β-asaroneCellΩAβ-induced PC12 cellsNeuronal apoptosis ↓JNK activation ↓
Bcl-w and Bcl-xL in a JNK-dependent manner ↑
Cytochrome c and activation of caspase-3 ↑
[12]
β-asaroneRatAD induced ratsSpatial memory ↑JNK activation ↓, caspase-3 activation & Bcl-w, and Bcl-2 ↑[13]
β-asaroneRatAD induced ratsNeuronal apoptosis ↓Bad expression & p-c-Jun activation, Bax expression ↓
Activation of caspase-9 ↑
[14]
Autophagy regulationβ-asaroneCellAβ1~42-induced PC12 cellsAβ ↓
Autophagy ↑
APP, PS1, Aβ, and BACE1 expression ↓
PINK1, Parkin & Autophagy ↑
[15]
β-asarone + IcariinCell, mouseAβ-induced PC12 cells, APP/PS1 miceMitochondrial damage ↓Clearance of toxic proteins & the formation of autophagosomes ↑
Beclin-1, PINK1, and p/Parkin ↑
[16]
β-asaroneMouseSAMP8 miceCognitive function ↑ROCK expression ↓, autophagy and synaptic loss ↓[17]
β-asaroneCellAβ-induced PC12 cellsNeuronal apoptosis ↓Beclin-1 expression ↓
p-Akt and p-mTOR ↑
[18]
Anti-inflammationβ-asaroneCellHuman neuroblastoma cells SH-SY5Y cellsAutophagy ↓
Inflammation ↓
Toxic effect of Aβ25-35 in SH-SY5Y cells ↓
Pro-inflammatory cytokines (IL-6, IL-1β and TNF-α) ↓
[19]
β-asaroneMouse, cellAβ1~42 injected rats
Aβ1-42 induced astrocytes
Spatial learning and memory ↑TNF-α, IL-1β & AQP4 expression ↓[20]
α-asaroneCellLPS-induced BV2 cellsMicroglial, morphological dynamics ↑Activated microglia ↓
MCP-1 ↓
[21]
Antioxidant-oxidantα-asaroneRatAβ-injected ratsSpatial memory ↑NO production & activation of astrocytes ↓[22]
β-asaroneMouseAβ-infused miceCell loss in the cerebral cortex and hippocampus ↓GPX and SOD ↑[23]
β-asaroneCellAβ-induced PC12 cellsAβ-induced damage ↓ROS, MDA ↓,
SOD, CAT, GSH-PX ↑, P13K/Akt/Nrf2 signaling pathway ↑, HO-1 ↑
[24]
β-asaroneRatAβ-infused ratsLearning and memory ability ↑Oxidative stress ↓
Pro-inflammatory cytokine ↓
Neurotransmitter and AChE activity ↑
[25]
Neurotransmitterβ-asaroneCellAPS/PSI double transgenic miceLearning and memory ability ↑Aβ neurotoxicity ↓
SYP and GluR1 ↑
[26]
α-asaroneRatAged ratsCognitive function ↑Aβ neurotoxicity ↓
GABA receptors ↑
[27]
α-asarone, β-asaroneCellNMDA or Glu-exposed cortical cells of ratNeuronal apoptosis ↓NMDA receptor function ↓[28]
β-asarone + tenuigeninHuman93 AD patientsTherapeutic effect ↑MMSE, ADL score ↑[29]
β-asarone + tenuigeninHuman152 AD patientsTherapeutic effect ↑MMSE, ADL score ↑[30]
β-asaroneRatAD induced ratsMemory impairment ↓rCBF of right parietal lober & the activity of NA-K-ATP ↑
ET-1 mRNA expression in hippocampus & pyruvic acid ↓
[31]
OthersVolatile oil fraction of ATMouseAβ-infused miceCognitive function ↑
Spatial memory ↑
Doublecortin and nestin ↓[32]
α-asarone, β-asaroneMouseAPS/PSI transgenic miceHippocampal neurogenesis ↑
NPCs ↑
ERK pathway & neurogenesis ↑[33]
α-asarone, β-asaroneCellPrimary astrocytes from ratsNGF, BDNF & GDNF ↑Neuronal action of AT ↑
Neurotrophic factors in astrocytes ↑
[34]
α-asarone, β-asaroneCellPC12 cellsNGF ↑Neurofilaments ↑[35]

SCP, Shi Chang Pu in Chinese; Aβ, amyloid-beta; polyQ, polyglutamine; APP, amyloid precursor protein; PS1, presenilin-1; P13K, phosphoinositide 3-kinases; Akt, protein kinase B; mTOR, mammalian target of rapamycin; CaMKII-α, calcium/calmodulin-dependent protein kinase II-alpha; p-CREB, phosphor-cAMP response element-binding protein; Bcl-2, B-cell lymphoma 2; Bax, BCL2-associated X; JNK, c-Jun N-terminal kinases; AD, Alzheimer’s disease; BACE, beta-secretase 1; PINK, PTEN-induced kinase 1; SAMP8, senescence accelerated mouse-prone 8; ROCK, Rho-associated protein kinase; IL-, interleukin-; TNF-α, tumor necrosis factor-α; AQP4, aquaporin4; LPS, lipopolysaccharide; MCP-1, monocyte chemoattractant protein-1; NO, nitric oxide; GPX, glutathione peroxidase; SOD, superoxide dismutase; ROS, reactive oxygen species; MDA, malondialdehyde; CAT, catalase; GSH-Px, glutathione peroxidase; Nrf2, nuclear factor erythroid-2-related factor 2; HO-1, heme oxygenase 1; AChE, acetylcholinesterase; SYP, synaptophysin; GluR1, glutamatergic receptor 1; GABA, γ-aminobutyric acid type; NMDA, N-methyl-d-aspartate; MMSE, Mini-mental State Examination; ADL, activities of daily living; rCBF, regional cerebral blood flow; NA-K-ATP, sodium-potassium adenosine triphosphatase, sodium–potassium pump; ET, endothelin; AT, Acorus tatarinowii; NPC, neural progenitor cell; ERK, extracellular signal-regulated kinase; NGF, nerve growth factor; BDNF, brain-derived neurotrophic factor; GDNF, glial-derived neurotrophic factor..


Table 2 . Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in Parkinson’s disease.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
Anti-cell deathβ-asaroneRat, cell6-OHDA-induced rats
SN4741 cells
Motor function ↑ (OFT, RRT, forelimb akinesia)In vitro: LC3-II ↓
In vivo: HVA, Dopacl, 5-HIAA, Bcl-2 ↑
Beclin-1, JNK, p-JNK ↓, Bcl-2 ↑
[37]
β-asaroneMouse, cellMPTP-induced mice
SH-SY5Y cells
Motor function ↑ (RRT)In vitro: TH+ cell ↑, MALAT1, α-syn, CHX, MG132 ↓
In vivo: MALAT1 ↓
[38]
Antioxidantα-asaroneMouse, cellMPTP-induced mice
BV-2 cells
Motor function ↑ (Y-maze test and pole test)In vitro: NO, iNOS, COX-2, TNF-α, IL-6, IL1β, NF-κB, IκB ↓
In vivo: Mac-1, CD-68, Iba-1, iNOS, COX-2, DOPAC ↓
[39]
β-asaroneRat6-OHDA-induced ratsER stress ↓GRP78, p-PERK, CHOP, Beclin-1 ↓
Bcl-2 ↑
[40]
β-asaroneRat6-OHDA-induced ratsER stress ↓IRE1, p-IRE1, XBP1 ↓[41]
β-asaroneRat6-OHDA-induced ratsCMA↑, Autophagy ↓HSC70, HSP70, MEF2D, LAMP-2A level ↑
α-Syn ↓
[42]
Anti-inflammationAG extractMouseMPTP-induced mice
BV-2 cells
Cell death ↓ Neuroinflammation ↓TH+ cell ↑
NO, iNOS, TNF-α, IL-6, IL1β, NF-κB, IκB ↓
[43]
β-asaroneRat6-OHDA-induced ratsMotor function ↑ (OFT, RRT, forelimb activity)α-Syn, Il-1β, TNF-α, NO, IL-6, BAX, Caspase ↓
TH, SOD, CAT, GSH-Px, Bcl-2 ↑
[44]
Coordination with levodopaβ-asarone + L-dopaRatSD ratL-Dopa, DA ↑DA ↑, COMT ↓[45]
β-asarone + L-dopaRat6-OHDA-induced ratsMotor function ↑ (OFT, ST, RRT)DDC level, DA level, MAO-B, COMT, DOPAC/DA, HVA/DA, TH, DAT ↑[46]
β-asarone + L-dopaRat6-OHDA-induced ratsL-Dopa BBB permeability ↑L-dopa, DA, DOPAC, HVA ↑
S100β ↑
NSE, P-gp, ZO-1, occludin, actin, claudin-5 ↓
[47]
β-asarone + L-dopaRat6-OHDA-induced ratsAutophagy activity ↓Beclin-1, LC3B ↓
p62 expression ↑
[48]

SD, Sprague Dawley; 6-OHDA, 6-hydroxydopamine; OFT, open-field test; RRT, rotarod test; LC3-II, light chain 3-II; HVA, homovanillic acid; Dopacl, 3,4-dihydroxyphenylacetic acid; 5-HIAA, 5-hydroxyindole acetic acid; JNK, c-Jun N-terminal kinase; Bcl-2, B-cell lymphoma; MPTP, 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine; TH, tyrosine hydroxylase; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; α-syn, α-synuclein; CHX, cycloheximide; MG132, a proteasome inhibitor; NO, nitric oxide; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2; TNF-α, tumor necrosis factor-alpha; IL-, interleukin-; NF-κB, nuclear factor kappa B; IκB, NF-κB inhibitor; Mac-1, macrophage Ag complex-1; CD-68, cluster of differentiation 68; Iba-1, ionized calcium-binding adapter molecule 1; DOPAC, 3, 4-dihydroxyphenylacetic acid; GRP78, glucose-regulated protein 78; p-PERK, phosphorylated protein kinase RNA-like endoplasmic reticulum kinase; CHOP, C/EBP homologous binding protein; ER, endoplasmic reticulum; IRE1, inositol-requiring enzyme 1; p-IRE1, phosphorylated IRE1; XBP1, X-box binding protein 1; CMA, chaperone-mediated autophagy; HSC70, heat-shock cognate protein 70; HSP70, heat-shock protein 70; MEF2D, myocyte enhancer factor 2D; LAMP-2A, lysosomal membrane protein receptor type 2A; BAX, B-cell lymphoma 2-associated X protein; TH, thyrosine hydroxylase; SOD, superoxide dismutase; CAT, catalase; GSH-Px, glutathione peroxidase; l-dopa, levodopa; DA, dopamine; COMT, catechol-O-methyltransferase; DAT, dopamine transporter; ST, stepping test; DDC, dopa decarboxylase; MAO-B, monoamine oxidase-B; BBB, blood–brain barrier; S100β, S100 calcium-binding protein β; NSE, neuron-specific enolase; P-gp, P-glycoprotein; ZO-1, zonula occludens-1; LC3B, microtubule-associated protein light chain 3B..


Table 3 . Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in depression and anxiety.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
Anti-depressionβ-asaroneRatCUMS exposed ratsDepressant-like behavior ↓ (SPT, FST)BDNF ↑, ERK1/2 and CREB phosphorylation ↑[50]
β-asaroneRatCUMS exposed ratsDepressant-like behavior ↓ (SPT, OFT, FST)Apoptosis ↓, CREB ↑, BDNF ↑, Trk-B ↑, Bcl-2 ↑, Bad ↓, ERK ↑[51]
β-asaroneRatCUMS exposed ratsBody weight ↑
Depressant-like behavior ↓ (SPT, OFT)
MKP-1 ↓, p-ERK1/2 ↑, BDNF ↑[52]
α-asaroneMouseNicotine withdrawal induced miceDepressant-like behavior ↓ (FST)p-CREB ↓[53]
EO from AT, α-asarone, β-asaroneMouseNormal miceDepressant-like behavior ↓ (FST, TST)[54]
α-asaroneMouseNormal miceDepressant-like behavior ↓ (TST)[55]
Anti-anxietyα-asaroneMouseNormal miceAnxiolytic-like behavior ↑ (EPM, LDT, NFC, MBT)[56]
α-asaroneRatSleep deprived ratsAnxiolytic-like behavior ↑ (EPM, OFT)MDA ↓, CAT ↑, GSH-R ↑, GSH-Px ↑[57]
α-asaroneMouseCFA-induced chronic
Inflammatory pain mice
Anxiolytic-like behavior ↑ (EPM, OFT)AMPARs ↓, NMDARs ↓, GABAARs ↑, hyper-excitability of pyramidal neurons ↓[58]
α-asaroneRatCorticosterone-induced anxiety ratsAnxiolytic-like behavior ↑ (EPM, HBT)TH ↓, BDNF ↓, TrkB ↓[59]

CUMS, chronic unpredictable mild stress; SPT, sucrose-preference test; FST, forced-swimming test; BDNF, brain-derived neurotrophic factor; ERK, extracellular signal-regulated kinases; CREB, cAMP response element-binding protein; OFT, open-field test; Trk-B, tropomyosin receptor kinase B; Bcl, B-cell lymphoma; Bad, Bcl-2-associated death promoter; MKP-1, mitogen-activated protein kinase phosphatase-1; p-ERK, phosphorylated extracellular signal-regulated kinases; p-CREB, phosphorylated cAMP response element-binding protein; EO, essential oil; AT, Acorus tatarinowii; TST, tail-suspension test; EPM, elevated plus maze; LDT, light/dark-transition test; NFC, novel–food-consumption test; MBT, marble-burying test; CFA: complete Freund’s adjuvant; MDA, malondialdehyde; CAT, catalase; GSH-R, glutathione reductase; GSH-Px, glutathione peroxide; AMPARs, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors; NMDARs, NR2A-containing N-methyl-d-aspartate recpetors; GABAAs, γ-aminobutyric acid type A receptors; HBT, hole-board test; TH, tyrosine hydroxylase..


Table 4 . Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in epilepsy.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
α-asaroneMouse, ratMES-induced seizure mice
scPTZ-induced seizure mice
LI-PILO-induced epilepsy rats
Seizure incidence, severity, frequency ↓, latency ↑[61]
α-asaroneMouse, ratMES-induced seizure mice
PTZ-induced seizure mice
LI-PILO-induced SE rats
SRS induced rats
Seizure onset, incidence, latency, severity, mortality, frequency ↓[62]
NeurotransmitterDecoction and volatile oil of ATMouse, ratMES-induced seizure mice
PTZ-induced seizure mice
Prolonged PTZ-induced seizure rats
Convulsant ↓, mortality ↓, seizure latency ↑, seizure intensity ↓GABA-IR neurons ↑, GABA-IR neuron damage ↓[63]
Water extract of AGMousePTZ-induced seizure miceOnset of seizure and death ↓GABA agonist[64]
EO of AGMousePTZ-induced seizure miceConvulsion ↓GABA transaminase ↓, GABA ↑, glutamate content ↓[65]
EO of AGCellGlutamate-induced excitotoxicity in primary rat cortical cellsExcitotoxicity ↓, neuroprotection ↑NMDAR antagonist[66]
α-asaroneRatPTZ-induced epilepsy rats
Kainate-induced epilepsy rats
Latency of seizures ↑, Susceptibility to seizure ↓Firing rate of spontaneous spiking ↓, tonic GABAergic inhibition ↑, inducing inward currents when picotoxin and bicuculline together ↓[67]
α-asaroneMouseNicotine-induced seizure miceLCA and BT ↓, onset time of seizures ↑[68]
α-asaroneRatLI-PILO-induced TLE ratsGABAergic modulationGABA ↑, GAD67 ↑, GABAAR-mRNA ↑, GABA-T ↓[69]
α-asaroneCellCNaIIA cell lineSpontaneous firing of mitral cells and Na+ channel ↓Spontaneous firing of output neurons, mitral cells ↓, Nav1.2 currents ↓[70]
Antioxidantα-asaroneMousePTZ-induced seizure mice
Picrotoxin-induced seizure mice
NMDA-induced seizure mice
PILO-induced seizure mice
MES-induced seizure mice
Treadmill performance and LCA ↓, hypothermia ↑, sleep ↑, onset of seizures ↓Antioxidant enzymes ↑[71]
Anti-inflammationα-asaroneRatPILO-induced TLE ratsCognitive function ↑ (WMT), behavioral score of SRSs ↓, frequency of seizures ↓Microglial activation ↓, proinflammatory cytokine ↓, LPS-stimulated neuroinflammatory responses ↓, NF-κB ↓[72]

MES, maximal electroshock; scPTZ, subcutaneous pentylenetetrazol seizure; LI-PILO, lithium-pilocarpine; PTZ, pentylenetetrazol; SE, status epilepticus; SRS, spontaneous recurrent seizures; AT, Acorus tatarinowii; GABA-IR, GABA-like immunoreactivity; AG, Acorus gramineus; GABA, γ-aminobutyric acid; EO, essential oil; NMDAR, N-methyl-d-aspartate receptor; LCA, locomotor activity; BT, body temperature; nAChRs, nicotinic acetylcholine receptor; TLE, temporal lobe epilepsy; GAD67, glutamic acid decarboxylase 67; GABAAR, γ-aminobutyric acid type A receptor; GABA-T, GABA transaminase; CNaIIA, type IIA Na+ channel; Nav1.2 channel, a dominant rat brain Na+ channel subtype; NMDA, N-methyl-d-aspartate; WMT, water maze test; LPS, lipopolysaccharide; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells..


Table 5 . Therapeutic potential of Acorus gramineus Solander and Acorus tatarinowii Schott in stroke.

EffectCompoundSpeciesExperimental modelResultsMechanismsRef.
Neuroprotectionβ-asaroneCellPC12 cells (OGD/R)Cell viability ↑, autophagy ↓MMP ↑, Beclin-1 ↓, [Ca2+]I ↓[74]
β-asaroneRatMCAOAutophagy ↓Beclin1 ↓, JNK ↓, p-JNK ↓, Bcl-2 ↑[75]
α-asaroneRatMCAOInfarct volume ↓
Epilepsy ↓
Neurological function ↑
Apoptosis ↓, GFAP ↓, Iba-1 ↓, LC3II/LC3I ↓, p62 ↑[76]
β-asaroneCellHypoxia induced PC12 cellsCell viability ↑SOD ↑, MMP ↑, apoptosis ↓, LDH ↓, ROS ↓, RPPH1 ↓[77]
α-asaroneRat4-Vessel occlusionNeuroprotection ↑Cell death ↓, damaged pyramidal neurons ↓[78]
β-asaroneRatMCAOInfarction volume ↓Apoptosis ↓, PAPs ↓, AAPs ↑, Nrf2-ARE pathway-related proteins ↑[79]
Antioxidantβ-asaroneRatMCAOMotor function ↑LDH ↓, GSH ↑, LPO ↓, GPx ↑, GR ↑, CAT ↑, GST ↑[80]
Anti-inflammationβ-asaroneCellLPS-stimulated BV-2 microglial cellsAnti-inflammatory effects ↑NO ↓, iNOS ↓, COX-2 ↓, NF-κB ↓[81]
BBB protectionAT extractRatMCAOInfarct size, edema ↓
BBB permeability ↓
Neurological function ↑
Astrocytic NKCC1/AQP4 ↓
JNK/iNOS-mediated ICAM-1/MMP-9 signaling ↓
[82]
Neurogenesisα-asaroneMouseMCAOMotor function ↑Differentiation of transplanted NPCs ↑[83]

OGD/R, 2 hours of oxygen-glucose deprivation followed by 24 hours of reperfusion; LPS, lipopolysaccharide; MMP, mitochondrial membrane potential; MCAO, middle cerebral artery occlusion; JNK, c-Jun N-terminal kinase; p-JNK, phosphorylated c-Jun N-terminal kinase; Bcl-2, B-cell lymphoma 2; GFAP, glial fibrillary acidic protein; Iba-1, ionized calcium-binding adaptor molecule-1; LC3, microtubule-associated protein light chain 3; SOD, superoxide dismutase; LDH, lactate dehydrogenase; MDA, malondialdehyde; ROS, reactive oxidative species; RPPH1, ribonuclease P RNA component H1; PAPs, pro-apoptotic proteins; AAPs, antiapoptotic proteins; Nrf2, nuclear factor erythroid 2-related factor 2; ARE, antioxidant response elements; NPC, neural progenitor cell; CIR, cerebral ischemia-reperfusion; GSH, glutathione; LPO, lipid peroxidation; GPx, glutathione peroxidase; GR, glutathione reductase; CAT, catalase; GST, glutathione S transferase; LPS, lipopolysaccharides; NO, nitric oxide; iNOS, inducible nitric oxide synthase; COX-2, cyclooxygenase-2; NF-κB, nuclear factor-κB; BBB, blood–brain barrier; AT, Acorus tatarinowii; NKCC1, Na-K-Cl cotransporter; AQP4, aquaporin 4; ICAM, Na-K-Cl cotransporter-1; MMP-9, matrix metallopeptidase 9..


Table 6 . Therapeutic potential of formulas including Acorus gramineus Solander and Acorus tatarinowii Schott in neurological disorders.

ComponentDisordersSpeciesResultsRef.
Kai Xin SanDepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, NIH, Two-Way Active Avoidance Test)[84]
ADAβ-injected ratsMemory ↑ (SDT)[85]
DepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, OFT, FST)[86]
StrokeICA embolic ratsCognitive function ↑ (MWM)[87]
DementiaSCOP induced miceCognitive function ↑ (MWM, Y-maze)[88]
DepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, OFT), weight ↑[89]
ADAβ-injected ratsCognitive function ↑ (NOR), injured neurons ↓, Aβ level ↓, IDE expression ↑[90]
Kaixin JieyuDepressionCUMS exposed ratsAntidepressant-like behavior ↑ (SPT, OFT)[91]
StrokeCCA ligation ratsAntidepressant-like behavior ↑ (SPT, OFT)[92]
Qisheng Wan formulaADAβ-injected ratsCognitive function ↑ (MWM)[93]
Bushen Tisansui DecoctionADAβ-injected ratsCognitive function ↑ (MWM)[94]
GPCRACDementiaSCOP induced miceMemory ↑ (SDT)Cognitive function ↑ (MWM)[95]
Bazhu DecoctionAD5×FAD transgenic miceCognitive function ↑ (OFT, Y-maze, MWM)[96]
Chong Myung TangDementiaSCOP induced miceCognitive & memory function ↑ (passive avoidance test, MWM)[97]
Yishen Huazhuo DecoctionADAD patient (human)Cerebral activity ↑ [supramarginal gyrus (BA 40), superior temporal gyrus (BA 22)][98]
Yeolda Hanso TangPDMPP+ -induced cells
MPTP-induced mice
Cell viability ↑ (survival ratio of TH-IR cell ↑)[99]

CUMS, chronic unpredictable mild stress; SPT, sucrose-preference test; NIH, novelty-induced hypophagia; AD, Alzheimer’s disease; Aβ, amyloid-beta; SDT, step-down test; OFT, open-field test; FST, forced-swim test; ICA, internal carotid artery; MWM, Morris water maze; SCOP, scopolamine; NOR, novel-object recognition; IDE, insulin-degrading enzyme; CCA, common carotid artery; GPCRAC, the combination of Gastrodia elata, Polygala tenuifolia, Cistanche deserticola, Rehmannia lutinosa, Acorus gramineus, Curcuma longa; BA, broaden area; PD, Parkinson’s disease; MPP+, 1-methyl-4-phenylpyridinium; MPTP, 1-methyl-4 phenyl-1, 2, 3, 6-tetrahydropyridine; TH-IR, tyrosine hydroxylase-immunoreacting..


Table 7 . Compositions of the formulas including Acorus gramineus Solander and Acorus tatarinowii Schott.

FormulaComposition
Kai-Xin-SanPanax ginseng, Poria cocos, Polygala tenuifolia, and Acori Tatarinowii
Kaixin-jieyuRadix Ginseng, Radix Paeoniae rubra, Acorus Gramineus soland, Fructus Aurantii immaturus, Radix Polygalae, Poria, Morinda Officinalis and Glycyrrhiza
Qisheng Wan FormulaPoria cocos, Cinnamomum cassia, Polygala tenuifolia, Panax ginseng, Asparagus cochinchinensis, Acorus tatarinowii, Lycium chinense
Bushen TiansuiEpimedium brevicornum, Polygonum multiflorum, Chinemys reevesii, Fossilia Ossis Mastodi, Polygala, Acorus tatarinowii
GPCRACGastrodia elata, Polygala tenuifolia, Cistanche deserticola, Rehmannia lutinosa, Acorus gramineus, Curcuma longa
Yishen Huazhuo DecoctionEpimedium, Fructus ligustri, Psoralea fruit, Radix polygoni multiflori, Radix astragali, Ligusticum wallichi franchat, Acorus gramineus
Bazhu DecoctionRadix Morindae Officinalis, Asiatic Cornelian Cherry Fruit, Grassleaf Sweetflag Rhizome, Earth Worm, Arisaema Cum Bile
Yeolda-Hanso TangPueraria lobata, Angelica tenuissima, Scutellaria baicalensis, Platycodon grandiflorum, Angelicae Dahurica, Cimicifuga heracleifolia, Raphanus sativa, Polygala tenuifolia, Acorus gramineus, Dimocarpus longan
Chong-Myung TangAcorus gramineus, Polygala tenuifolia, Poria cocos

References

  1. GBD 2016 Neurology Collaborators. Global, regional, and national burden of neurological disorders, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(5):459-80.
    Pubmed KoreaMed CrossRef
  2. Parés-Badell O, Barbaglia G, Jerinic P, Gustavsson A, Salvador-Carulla L, Alonso J. Cost of disorders of the brain in Spain. PLoS One. 2014;9(8):e105471.
    Pubmed KoreaMed CrossRef
  3. Park CH, Kim KH, Lee IK, Lee SY, Choi SU, Lee JH, et al. Phenolic constituents of Acorus gramineus. Arch Pharm Res. 2011;34(8):1289-96.
    Pubmed CrossRef
  4. Balakrishnan R, Cho DY, Kim IS, Seol SH, Choi DK. Molecular mechanisms and therapeutic potential of α- and β-asarone in the treatment of neurological disorders. Antioxidants (Basel). 2022;11(2):281.
    Pubmed KoreaMed CrossRef
  5. Jakob-Roetne R, Jacobsen H. Alzheimer's disease: from pathology to therapeutic approaches. Angew Chem Int Ed Engl. 2009;48(17):3030-59.
    Pubmed CrossRef
  6. Kim SY. Past and future of drug treatments for Alzheimer's disease. J Korean Neuropsychiatr Assoc. 2018;57(1):30-42.
    CrossRef
  7. Chen XY, Liao DC, Sun ML, Cui XH, Wang HB. Essential oil of Acorus tatarinowii schott ameliorates -induced toxicity in Caenorhabditis elegans through an autophagy pathway. Oxid Med Cell Longev. 2020;2020:3515609.
    Pubmed KoreaMed CrossRef
  8. Deng M, Huang L, Zhong X. β-asarone modulates Beclin-1, LC3 and p62 expression to attenuate Aβ40 and Aβ42 levels in APP/PS1 transgenic mice with Alzheimer's disease. Mol Med Rep. 2020;21(5):2095-102.
    Pubmed KoreaMed CrossRef
  9. Deng M, Huang L, Ning B, Wang N, Zhang Q, Zhu C, et al. β-asarone improves learning and memory and reduces Acetyl Cholinesterase and Beta-amyloid 42 levels in APP/PS1 transgenic mice by regulating Beclin-1-dependent autophagy. Brain Res. 2016;1652:188-94.
    Pubmed CrossRef
  10. Wei G, Chen YB, Chen DF, Lai XP, Liu DH, Deng RD, et al. β-Asarone inhibits neuronal apoptosis via the CaMKII/CREB/Bcl-2 signaling pathway in an in vitro model and AβPP/PS1 mice. J Alzheimers Dis. 2013;33(3):863-80.
    Pubmed CrossRef
  11. Liang ZH, Cheng XH, Ruan ZG, Wang H, Li SS, Liu J, et al. Protective effects of components of the Chinese herb grassleaf sweetflag rhizome on PC12 cells incubated with amyloid-beta42. Neural Regen Res. 2015;10(8):1292-7.
    Pubmed KoreaMed CrossRef
  12. Li C, Xing G, Dong M, Zhou L, Li J, Wang G, et al. Beta-asarone protection against beta-amyloid-induced neurotoxicity in PC12 cells via JNK signaling and modulation of Bcl-2 family proteins. Eur J Pharmacol. 2010;635(1-3):96-102.
    Pubmed CrossRef
  13. Geng Y, Li C, Liu J, Xing G, Zhou L, Dong M, et al. Beta-asarone improves cognitive function by suppressing neuronal apoptosis in the beta-amyloid hippocampus injection rats. Biol Pharm Bull. 2010;33(5):836-43.
    Pubmed CrossRef
  14. Liu J, Li C, Xing G, Zhou L, Dong M, Geng Y, et al. Beta-asarone attenuates neuronal apoptosis induced by Beta amyloid in rat hippocampus. Yakugaku Zasshi. 2010;130(5):737-46.
    Pubmed CrossRef
  15. Wang N, Wang H, Li L, Li Y, Zhang R. β-asarone inhibits amyloid-β by promoting autophagy in a cell model of Alzheimer's disease. Front Pharmacol. 2020;10:1529.
    Pubmed KoreaMed CrossRef
  16. Wang N, Wang H, Pan Q, Kang J, Liang Z, Zhang R. The combination of β-asarone and icariin inhibits amyloid-β and reverses cognitive deficits by promoting mitophagy in models of Alzheimer's disease. Oxid Med Cell Longev. 2021;2021:7158444.
    Pubmed KoreaMed CrossRef
  17. Chen Y, Wei G, Nie H, Lin Y, Tian H, Liu Y, et al. β-Asarone prevents autophagy and synaptic loss by reducing ROCK expression in asenescence-accelerated prone 8 mice. Brain Res. 2014;1552:41-54.
    Pubmed CrossRef
  18. Xue Z, Guo Y, Zhang S, Huang L, He Y, Fang R, et al. Beta-asarone attenuates amyloid beta-induced autophagy via Akt/mTOR pathway in PC12 cells. Eur J Pharmacol. 2014;741:195-204.
    Pubmed CrossRef
  19. Chang W, Teng J. β-asarone prevents Aβ25-35-induced inflammatory responses and autophagy in SH-SY5Y cells: down expression Beclin-1, LC3B and up expression Bcl-2. Int J Clin Exp Med. 2015;8(11):20658-63.
    Pubmed KoreaMed
  20. Yang Y, Xuan L, Chen H, Dai S, Ji L, Bao Y, et al. Neuroprotective effects and mechanism of β-asarone against Aβ1-42-induced injury in astrocytes. Evid Based Complement Alternat Med. 2017;2017:8516518.
    Pubmed KoreaMed CrossRef
  21. Cai Q, Li Y, Mao J, Pei G. Neurogenesis-promoting natural product α-asarone modulates morphological dynamics of activated microglia. Front Cell Neurosci. 2016;10:280.
    Pubmed KoreaMed CrossRef
  22. Limón ID, Mendieta L, Díaz A, Chamorro G, Espinosa B, Zenteno E, et al. Neuroprotective effect of alpha-asarone on spatial memory and nitric oxide levels in rats injected with amyloid-beta((25-35)). Neurosci Lett. 2009;453(2):98-103.
    Pubmed CrossRef
  23. Saki G, Eidi A, Mortazavi P, Panahi N, Vahdati A. Effect of β-asarone in normal and β-amyloid-induced Alzheimeric rats. Arch Med Sci. 2020;16(3):699-706.
    Pubmed KoreaMed CrossRef
  24. Meng M, Zhang L, Ai D, Wu H, Peng W. β-asarone ameliorates β-amyloid-induced neurotoxicity in PC12 cells by activating P13K/Akt/Nrf2 signaling pathway. Front Pharmacol. 2021;12:659955.
    Pubmed KoreaMed CrossRef
  25. Kumar P, Deshmukh R, Bariwal J, Shree P. β-asarone restores hippocampal neurochemistry and improve cognitive functions in Aβ (1-42) infused rats. Alzheimer's Dement. 2019;15(7):P1292.
    CrossRef
  26. Liu SJ, Yang C, Zhang Y, Su RY, Chen JL, Jiao MM, et al. Neuroprotective effect of β-asarone against Alzheimer's disease: regulation of synaptic plasticity by increased expression of SYP and GluR1. Drug Des Devel Ther. 2016;10:1461-9.
    Pubmed KoreaMed CrossRef
  27. Chen Y, Gao X, Liu Q, Zeng L, Zhang K, Mu K, et al. Alpha-asarone improves cognitive function of aged rats by alleviating neuronal excitotoxicity via GABAA receptors. Neuropharmacology. 2020;162:107843.
    Pubmed CrossRef
  28. Cho J, Kim YH, Kong JY, Yang CH, Park CG. Protection of cultured rat cortical neurons from excitotoxicity by asarone, a major essential oil component in the rhizomes of Acorus gramineus. Life Sci. 2002;71(5):591-9.
    Pubmed CrossRef
  29. Dong H, Wu S, Hu N, Xing G. Efficacy of tenuigenin and β-asarone as augmentations for memantine in the treatment of Alzheimer's disease. Neuroreport. 2018;29(3):203-7.
    Pubmed CrossRef
  30. Chang W, Teng J. Combined application of tenuigenin and β-asarone improved the efficacy of memantine in treating moderate-to-severe Alzheimer's disease. Drug Des Devel Ther. 2018;12:455-62.
    Pubmed KoreaMed CrossRef
  31. Li Z, Zhao G, Qian S, Yang Z, Chen X, Chen J, et al. Cerebrovascular protection of β-asarone in Alzheimer's disease rats: a behavioral, cerebral blood flow, biochemical and genic study. J Ethnopharmacol. 2012;144(2):305-12.
    Pubmed CrossRef
  32. Ma Y, Tian S, Sun L, Yao S, Liang Z, Li S, et al. The effect of acori graminei rhizoma and extract fractions on spatial memory and hippocampal neurogenesis in amyloid beta 1-42 injected mice. CNS Neurol Disord Drug Targets. 2015;14(3):411-20.
    Pubmed CrossRef
  33. Mao J, Huang S, Liu S, Feng XL, Yu M, Liu J, et al. A herbal medicine for Alzheimer's disease and its active constituents promote neural progenitor proliferation. Aging Cell. 2015;14(5):784-96.
    Pubmed KoreaMed CrossRef
  34. Lam KYC, Wu QY, Hu WH, Yao P, Wang HY, Dong TTX, et al. Asarones from Acori Tatarinowii Rhizoma stimulate expression and secretion of neurotrophic factors in cultured astrocytes. Neurosci Lett. 2019;707:134308.
    Pubmed CrossRef
  35. Gao N, Liu H, Li S, Tu X, Tian S, Liu J, et al. Volatile oil from acorus gramineus ameliorates the injury neurons in the hippocampus of amyloid beta 1-42 injected mice. Anat Rec (Hoboken). 2019;302(12):2261-70.
    Pubmed CrossRef
  36. Bloem BR, Okun MS, Klein C. Parkinson's disease. Lancet. 2021;397(10291):2284-303.
    Pubmed KoreaMed CrossRef
  37. Zhang S, Gui XH, Huang LP, Deng MZ, Fang RM, Ke XH, et al. Neuroprotective effects of β-asarone against 6-hydroxy dopamine-induced Parkinsonism via JNK/Bcl-2/Beclin-1 pathway. Mol Neurobiol. 2016;53(1):83-94.
    Pubmed CrossRef
  38. Zhang QS, Wang ZH, Zhang JL, Duan YL, Li GF, Zheng DL. Beta-asarone protects against MPTP-induced Parkinson's disease via regulating long non-coding RNA MALAT1 and inhibiting α-synuclein protein expression. Biomed Pharmacother. 2016;83:153-9.
    Pubmed CrossRef
  39. Kim BW, Koppula S, Kumar H, Park JY, Kim IW, More SV, et al. α-Asarone attenuates microglia-mediated neuroinflammation by inhibiting NF kappa B activation and mitigates MPTP-induced behavioral deficits in a mouse model of Parkinson's disease. Neuropharmacology. 2015;97:46-57.
    Pubmed CrossRef
  40. Ning B, Zhang Q, Wang N, Deng M, Fang Y. β-asarone regulates ER stress and autophagy via inhibition of the PERK/CHOP/Bcl-2/Beclin-1 pathway in 6-OHDA-induced parkinsonian rats. Neurochem Res. 2019;44(5):1159-66.
    Pubmed CrossRef
  41. Ning B, Deng M, Zhang Q, Wang N, Fang Y. β-asarone inhibits IRE1/XBP1 endoplasmic reticulum stress pathway in 6-OHDA-induced parkinsonian rats. Neurochem Res. 2016;41(8):2097-101.
    Pubmed CrossRef
  42. Huang L, Deng M, He Y, Lu S, Liu S, Fang Y. β-asarone increases MEF2D and TH levels and reduces α-synuclein level in 6-OHDA-induced rats via regulating the HSP70/MAPK/MEF2D/Beclin-1 pathway: chaperone-mediated autophagy activation, macroautophagy inhibition and HSP70 up-expression. Behav Brain Res. 2016;313:370-9.
    Pubmed CrossRef
  43. Jiang J, Kim JJ, Kim DY, Kim MK, Oh NH, Koppula S, et al. Acorus gramineus inhibits microglia mediated neuroinflammation and prevents neurotoxicity in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of Parkinson's disease. J Ethnopharmacol. 2012;144(3):506-13.
    Pubmed CrossRef
  44. Liu H, Ren M, Wang H, Zhang M, Xiong J. Effect and mechanism of beta asarone from Acorus tatarinowii Schott on neuron injury in rats with Parkinson's disease. Indian J Pharm Sci. 2021;84(2):400-6.
    CrossRef
  45. Huang L, Deng M, Zhang S, Fang Y, Li L. Coadministration of β-asarone and levodopa increases dopamine in rat brain by accelerating transformation of levodopa: a different mechanism from Madopar. Clin Exp Pharmacol Physiol. 2014;41(9):685-90.
    Pubmed CrossRef
  46. Huang L, Deng M, Zhang S, Lu S, Gui X, Fang Y. β-asarone and levodopa coadministration increases striatal levels of dopamine and levodopa and improves behavioral competence in Parkinson's rat by enhancing dopa decarboxylase activity. Biomed Pharmacother. 2017;94:666-78.
    Pubmed CrossRef
  47. Huang L, Deng M, He Y, Lu S, Ma R, Fang Y. β-asarone and levodopa co-administration increase striatal dopamine level in 6-hydroxydopamine induced rats by modulating P-glycoprotein and tight junction proteins at the blood-brain barrier and promoting levodopa into the brain. Clin Exp Pharmacol Physiol. 2016;43(6):634-43.
    Pubmed CrossRef
  48. Huang LP, Deng MZ, He YP, Fang YQ. β-asarone and levodopa co-administration protects against 6-hydroxydopamine-induced damage in parkinsonian rat mesencephalon by regulating autophagy: down-expression Beclin-1 and light chain 3B and up-expression P62. Clin Exp Pharmacol Physiol. 2015;42(3):269-77.
    Pubmed CrossRef
  49. Willner P, Scheel-Krüger J, Belzung C. The neurobiology of depression and antidepressant action. Neurosci Biobehav Rev. 2013;37(10 Pt 1):2331-71.
    Pubmed CrossRef
  50. Dong H, Gao Z, Rong H, Jin M, Zhang X. β-asarone reverses chronic unpredictable mild stress-induced depression-like behavior and promotes hippocampal neurogenesis in rats. Molecules. 2014;19(5):5634-49.
    Pubmed KoreaMed CrossRef
  51. Dong H, Cong W, Guo X, Wang Y, Tong S, Li Q, et al. β-asarone relieves chronic unpredictable mild stress induced depression by regulating the extracellular signal-regulated kinase signaling pathway. Exp Ther Med. 2019;18(5):3767-74.
    Pubmed KoreaMed CrossRef
  52. Sun YR, Wang XY, Li SS, Dong HY, Zhang XJ. β-asarone from Acorus gramineus alleviates depression by modulating MKP-1. Genet Mol Res. 2015;14(2):4495-504.
    Pubmed CrossRef
  53. Chellian R, Pandy V, Mohamed Z. Alpha-asarone attenuates depression-like behavior in nicotine-withdrawn mice: evidence for the modulation of hippocampal pCREB levels during nicotine-withdrawal. Eur J Pharmacol. 2018;818:10-6.
    Pubmed CrossRef
  54. Han P, Han T, Peng W, Wang XR. Antidepressant-like effects of essential oil and asarone, a major essential oil component from the rhizome of Acorus tatarinowii. Pharm Biol. 2013;51(5):589-94.
    Pubmed CrossRef
  55. Chellian R, Pandy V, Mohamed Z. Biphasic effects of α-asarone on immobility in the tail suspension test: evidence for the involvement of the noradrenergic and serotonergic systems in its antidepressant-like activity. Front Pharmacol. 2016;7:72.
    Pubmed KoreaMed CrossRef
  56. Liu S, Chen SW, Xu N, Liu XH, Zhang H, Wang YZ, et al. Anxiolytic-like effect of α-asarone in mice. Phytother Res. 2012;26(10):1476-81.
    Pubmed CrossRef
  57. Radhakrishnan A, Jayakumari N, Kumar VM, Gulia KK. α-Asarone in management of sleep deprivation induced memory deficits and anxiety in rat model. Sleep Biol Rhythms. 2019;17(1):37-47.
    CrossRef
  58. Tian J, Tian Z, Qin SL, Zhao PY, Jiang X, Tian Z. Anxiolytic-like effects of α-asarone in a mouse model of chronic pain. Metab Brain Dis. 2017;32(6):2119-29.
    Pubmed CrossRef
  59. Lee B, Sur B, Yeom M, Shim I, Lee H, Hahm DH. Alpha-asarone, a major component of acorus gramineus, attenuates corticosterone-induced anxiety-like behaviours via modulating TrkB signaling process. Korean J Physiol Pharmacol. 2014;18(3):191-200.
    Pubmed KoreaMed CrossRef
  60. Chang BS, Lowenstein DH. Epilepsy. N Engl J Med. 2003;349(13):1257-66.
    Pubmed CrossRef
  61. Miao JK, Chen QX, Wu XM, Li C, Zhang XP. Antiepileptic properties of alpha-asarone from acori graminei rhizoma in mice and rats seizure models. Int J Pharmacol. 2012;8(6):567-71.
    CrossRef
  62. Chen QX, Miao JK, Li C, Li XW, Wu XM, Zhang XP. Anticonvulsant activity of acute and chronic treatment with a-asarone from Acorus gramineus in seizure models. Biol Pharm Bull. 2013;36(1):23-30.
    Pubmed CrossRef
  63. Liao WP, Chen L, Yi YH, Sun WW, Gao MM, Su T, et al. Study of antiepileptic effect of extracts from Acorus tatarinowii Schott. Epilepsia. 2005;46 Suppl 1:21-4.
    Pubmed CrossRef
  64. Liao JF, Huang SY, Jan YM, Yu LL, Chen CF. Central inhibitory effects of water extract of Acori graminei rhizoma in mice. J Ethnopharmacol. 1998;61(3):185-93.
    Pubmed CrossRef
  65. Koo BS, Park KS, Ha JH, Park JH, Lim JC, Lee DU. Inhibitory effects of the fragrance inhalation of essential oil from Acorus gramineus on central nervous system. Biol Pharm Bull. 2003;26(7):978-82.
    Pubmed CrossRef
  66. Cho J, Kong JY, Jeong DY, Lee KD, Lee DU, Kang BS. NMDA recepter-mediated neuroprotection by essential oils from the rhizomes of Acorus gramineus. Life Sci. 2001;68(13):1567-73.
    Pubmed CrossRef
  67. Huang C, Li WG, Zhang XB, Wang L, Xu TL, Wu D, et al. α-asarone from Acorus gramineus alleviates epilepsy by modulating A-type GABA receptors. Neuropharmacology. 2013;65:1-11.
    Pubmed CrossRef
  68. Chellian R, Pandy V. Protective effect of α-asarone against nicotine-induced seizures in mice, but not by its interaction with nicotinic acetylcholine receptors. Biomed Pharmacother. 2018;108:1591-5.
    Pubmed CrossRef
  69. Miao JK, Chen QX, Li C, Li XW, Wu XM, Zhang X. Modulation effects of α-asarone on the GABA homeostasis in the lithium-pilocarpine model of temporal lobe epilepsy. Int J Pharmacol. 2013;9(1):24-32.
    CrossRef
  70. Wang ZJ, Levinson SR, Sun L, Heinbockel T. Identification of both GABAA receptors and voltage-activated Na(+) channels as molecular targets of anticonvulsant α-asarone. Front Pharmacol. 2014;5:40.
    Pubmed KoreaMed CrossRef
  71. Pages N, Maurois P, Delplanque B, Bac P, Stables JP, Tamariz J, et al. Activities of α-asarone in various animal seizure models and in biochemical assays might be essentially accounted for by antioxidant properties. Neurosci Res. 2010;68(4):337-44.
    Pubmed CrossRef
  72. Liu HJ, Lai X, Xu Y, Miao JK, Li C, Liu JY, et al. α-Asarone attenuates cognitive deficit in a pilocarpine-induced status epilepticus rat model via a decrease in the nuclear factor-κB activation and reduction in microglia neuroinflammation. Front Neurol. 2017;8:661.
    Pubmed KoreaMed CrossRef
  73. Donnan GA, Fisher M, Macleod M, Davis SM. Stroke. Lancet. 2008;371(9624):1612-23.
    Pubmed KoreaMed CrossRef
  74. Mo ZT, Fang YQ, He YP, Zhang S. β-Asarone protects PC12 cells against OGD/R-induced injury via attenuating Beclin-1-dependent autophagy. Acta Pharmacol Sin. 2012;33(6):737-42.
    Pubmed KoreaMed CrossRef
  75. Liu L, Fang YQ, Xue ZF, He YP, Fang RM, Li L. Beta-asarone attenuates ischemia-reperfusion-induced autophagy in rat brains via modulating JNK, p-JNK, Bcl-2 and Beclin 1. Eur J Pharmacol. 2012;680(1-3):34-40.
    Pubmed CrossRef
  76. Zhang K, Liu Q, Luo L, Feng X, Hu Q, Fan X, et al. Neuroprotective effect of alpha-asarone on the rats model of cerebral ischemia-reperfusion stroke via ameliorating glial activation and autophagy. Neuroscience. 2021;473:130-41.
    Pubmed CrossRef
  77. Yang F, Duan H, Ye N, Zeng Y, Yang P, Shao B, et al. β-Asarone protects PC12 cells against hypoxia-induced injury via negatively regulating RPPH1/MiR-542-3p/DEDD2 axis. Cell Transplant. 2022;31:9636897221079336.
    Pubmed KoreaMed CrossRef
  78. Kim YO, Kwon YS, Lee SE, Lee SW, Lee HJ. Effects of α-asarone against global cerebral ischemia in rats. Nat Prod Sci. 2009;15(4):198-202.
  79. Pan H, Xu Y, Cai Q, Wu M, Ding M. Effects of β-asarone on ischemic stroke in middle cerebral artery occlusion rats by an Nrf2-antioxidant response elements (ARE) pathway-dependent mechanism. Med Sci Monit. 2021;27:e931884.
    Pubmed KoreaMed CrossRef
  80. Yang YX, Chen YT, Zhou XJ, Hong CL, Li CY, Guo JY. Beta-asarone, a major component of Acorus tatarinowii Schott, attenuates focal cerebral ischemia induced by middle cerebral artery occlusion in rats. BMC Complement Altern Med. 2013;13:236.
    Pubmed KoreaMed CrossRef
  81. Lim HW, Kumar H, Kim BW, More SV, Kim IW, Park JI, et al. β-Asarone (cis-2,4,5-trimethoxy-1-allyl phenyl), attenuates pro-inflammatory mediators by inhibiting NF-κB signaling and the JNK pathway in LPS activated BV-2 microglia cells. Food Chem Toxicol. 2014;72:265-72.
    Pubmed CrossRef
  82. Lee YC, Kao ST, Cheng CY. Acorus tatarinowii Schott extract reduces cerebral edema caused by ischemia-reperfusion injury in rats: involvement in regulation of astrocytic NKCC1/AQP4 and JNK/iNOS-mediated signaling. BMC Complement Med Ther. 2020;20(1):374.
    Pubmed KoreaMed CrossRef
  83. Lee HJ, Ahn SM, Pak ME, Jung DH, Lee SY, Shin HK, et al. Positive effects of α-asarone on transplanted neural progenitor cells in a murine model of ischemic stroke. Phytomedicine. 2018;51:151-61.
    Pubmed CrossRef
  84. Dang H, Sun L, Liu X, Peng B, Wang Q, Jia W, et al. Preventive action of Kai Xin San aqueous extract on depressive-like symptoms and cognition deficit induced by chronic mild stress. Exp Biol Med (Maywood). 2009;234(7):785-93.
    Pubmed CrossRef
  85. Zhang B, Li Y, Liu JW, Liu XW, Wen W, Cui Y, et al. Postsynaptic GluR2 involved in amelioration of Aβ-induced memory dysfunction by KAIXIN-San through rescuing hippocampal LTP in mice. Rejuvenation Res. 2019;22(2):131-7.
    Pubmed CrossRef
  86. Yu S, Liu S, Wang N, Yu D, Qin M, Wu J, et al. Novel insights into antidepressant mechanism of Kai Xin San formula: inhibiting NLRP3 inflammasome activation by promoting autophagy. Phytomedicine. 2021;93:153792.
    Pubmed CrossRef
  87. Li X, Wen W, Li P, Fu Y, Chen H, Wang F, et al. Mitochondrial protection and against glutamate neurotoxicity via Shh/Ptch1 signaling pathway to ameliorate cognitive dysfunction by Kaixin San in multi-infarct dementia rats. Oxid Med Cell Longev. 2021;2021:5590745.
    Pubmed KoreaMed CrossRef
  88. Xu YM, Wang XC, Xu TT, Li HY, Hei SY, Luo NC, et al. Kai Xin San ameliorates scopolamine-induced cognitive dysfunction. Neural Regen Res. 2019;14(5):794-804.
    Pubmed KoreaMed CrossRef
  89. Huang YL, Liang XB, Qian LQ, Cai C, Guo J, Gao C, et al. Effects of Kaixin Powder on melatonin receptor expression and (125)I-Mel binding affinity in a rat model of depression. Chin J Integr Med. 2015;21(7):507-15.
    Pubmed CrossRef
  90. Wang N, Jia YM, Zhang B, Xue D, Reeju M, Li Y, et al. Neuroprotective mechanism of Kai Xin San: upregulation of hippocampal insulin-degrading enzyme protein expression and acceleration of amyloid-beta degradation. Neural Regen Res. 2017;12(4):654-9.
    Pubmed KoreaMed CrossRef
  91. Huang SJ, Zhang XH, Wang YY, Pan JH, Cui HM, Fang SP, et al. Effects of Kaixin Jieyu Decoction (开心解郁汤) on behavior, monoamine neurotransmitter levels, and serotonin receptor subtype expression in the brain of a rat depression model. Chin J Integr Med. 2014;20(4):280-5.
    Pubmed CrossRef
  92. Zhang XH, Huang SJ, Wang YY, Zhang Y, Pan JH, Zheng J, et al. Effects of kaixin jieyu decoction on behavior and glial fibrillary acidic protein expression in cerebral hippocampus of a rat vascular depression model. Chin J Integr Med. 2015;21(3):223-8.
    Pubmed CrossRef
  93. Xiong W, Zhao X, Xu Q, Wei G, Zhang L, Fan Y, et al. Qisheng Wan formula ameliorates cognitive impairment of Alzheimer's disease rat via inflammation inhibition and intestinal microbiota regulation. J Ethnopharmacol. 2022;282:114598.
    Pubmed CrossRef
  94. Hui S, Yang Y, Peng WJ, Sheng CX, Gong W, Chen S, et al. Protective effects of Bushen Tiansui decoction on hippocampal synapses in a rat model of Alzheimer's disease. Neural Regen Res. 2017;12(10):1680-6.
    Pubmed KoreaMed CrossRef
  95. Huang Q, Zhang C, Qu S, Dong S, Ma Q, Hao Y, et al. Chinese herbal extracts exert neuroprotective effect in Alzheimer's disease mouse through the dopaminergic synapse/apoptosis signaling pathway. Front Pharmacol. 2022;13:817213.
    Pubmed KoreaMed CrossRef
  96. Peng A, Gao Y, Zhuang X, Lin Y, He W, Wang Y, et al. Bazhu decoction, a traditional Chinese medical formula, ameliorates cognitive deficits in the 5xFAD mouse model of Alzheimer's disease. Front Pharmacol. 2019;10:1391.
    Pubmed KoreaMed CrossRef
  97. Lee MR, Yun BS, Park SY, Ly SY, Kim SN, Han BH, et al. Anti-amnesic effect of Chong-Myung-Tang on scopolamine-induced memory impairments in mice. J Ethnopharmacol. 2010;132(1):70-4.
    Pubmed CrossRef
  98. Lin C, Zhou Z, Xu J, Li Q, Guo J, Long M, et al. Changes of brain activity during a functional magnetic resonance imaging stroop task study: effect of Chinese herbal formula in Alzheimer's disease. Eur J Integr Med. 2017;16:46-53.
    CrossRef
  99. Bae N, Ahn T, Chung S, Oh MS, Ko H, Oh H, et al. The neuroprotective effect of modified Yeoldahanso-tang via autophagy enhancement in models of Parkinson's disease. J Ethnopharmacol. 2011;134(2):313-22.
    Pubmed CrossRef