Next Article in Journal
Could Inflammatory Indices and Metabolic Syndrome Predict the Risk of Cancer Development? Analysis from the Bagnacavallo Population Study
Next Article in Special Issue
Kurarinone Inhibits HCoV-OC43 Infection by Impairing the Virus-Induced Autophagic Flux in MRC-5 Human Lung Cells
Previous Article in Journal
Both Human Hematoma Punctured from Pelvic Fractures and Serum Increase Muscle Resident Stem Cells Response to BMP9: A Multivariate Statistical Approach
Previous Article in Special Issue
Pharmacological Therapeutics Targeting RNA-Dependent RNA Polymerase, Proteinase and Spike Protein: From Mechanistic Studies to Clinical Trials for COVID-19
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Vacha (Acorus calamus Linn.) in Neurological and Metabolic Disorders: Evidence from Ethnopharmacology, Phytochemistry, Pharmacology and Clinical Study

1
Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, BHU, Varanasi, Uttar Pradesh 221005, India
2
Department of Chemistry, Faculty of Science, University of Hradec Králové, Rokitanskeho 62, 50003 Hradec Králové, Czech Republic
3
Faculty of Medicine, University of Porto, Alameda Prof. Hernani Monteiro, 4200-319 Porto, Portugal
4
Institute for research and Innovation in Heath (i3S), University of Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal
*
Authors to whom correspondence should be addressed.
Submission received: 23 March 2020 / Revised: 12 April 2020 / Accepted: 14 April 2020 / Published: 19 April 2020
(This article belongs to the Special Issue Bioactive Phytochemicals in Health and Disease)

Abstract

:
Vacha (Acorus calamus Linn. (Acoraceae)) is a traditional Indian medicinal herb, which is practiced to treat a wide range of health ailments, including neurological, gastrointestinal, respiratory, metabolic, kidney, and liver disorders. The purpose of this paper is to provide a comprehensive up-to-date report on its ethnomedicinal use, phytochemistry, and pharmacotherapeutic potential, while identifying potential areas for further research. To date, 145 constituents have been isolated from this herb and identified, including phenylpropanoids, sesquiterpenoids, and monoterpenes. Compelling evidence is suggestive of the biopotential of its various extracts and active constituents in several metabolic and neurological disorders, such as anticonvulsant, antidepressant, antihypertensive, anti-inflammatory, immunomodulatory, neuroprotective, cardioprotective, and anti-obesity effects. The present extensive literature survey is expected to provide insights into the involvement of several signaling pathways and oxidative mechanisms that can mitigate oxidative stress, and other indirect mechanisms modulated by active biomolecules of A. calamus to improve neurological and metabolic disorders.

1. Introduction

Globally, an estimated 450 million people are suffering from mental disorders and about 425 million are known diabetics [1,2]. In 2016, 650 million adults were obese and about 23.6 million people were estimated to die of cardiovascular diseases (CVDs) by the year 2030 [3]. Metabolic disorders are characterized by hypertension, hyperglycemia, abdominal obesity, and hyperlipidemia, which may worsen the neurological disease risk. Improper diet (high calorie intake), lifestyle (e.g., smoking, chronic alcohol consumption, sedentary habits), and/or low level of nitrosamines (through processed food, tobacco smoke, and nitrate-containing fertilizers) affect the liver and can further lead to fatty liver disease [4,5]. In this condition, fatty changes may be due to increased production or decreased use of fatty acids, which may lead to inflammatory injury of hepatocytes, where inflammatory mediators, such as cytokines and interleukins, are released, which, along with lower adipokines, may eventually develop hepatic insulin resistance [6]. The same pathology also mediates diabetes, obesity, and peripheral insulin resistance. Insulin resistance also promotes the release of ceramides and other toxic lipids which enter the circulation and cross the blood–brain barrier leading to brain insulin resistance, inflammatory changes, and further progression to neurodegeneration and neurological disorders (Figure 1) [7].
Acorus calamus Linn. (Acoraceae), also known as Vacha in Sanskrit, is a mid-term, perennial, fragrant herb which is practiced in the Ayurvedic (Indian traditional) and the Chinese system of medicine. The plant’s rhizomes are brown in color, twisted, cylindrical, curved, and shortly nodded. The leaves are radiant green, with a sword-like structure, which is thicker in the middle and has curvy margins (Figure 2) [8]. Several reports ascertained a wide range of biological activities involving its myriad of active phytoconstituents. In this sense, the intent of this review is to assemble and summarize the geographical distribution, ethnopharmacology, phytochemistry, mechanism of action of A. calamus along with preclinical and clinical claims that are relevant to manage neurological and metabolic disorders. To the best of our knowledge, so far, none of the published reviews has described all the characteristics of this medicinal plant [9,10,11]. The present report is expected to produce a better understanding of the characteristics, bioactivities, and mechanistic aspects of this plant and to provide new leads for future research.

2. Methodology

The literature available in the Ayurvedic classical texts, technical reports, online scientific records such as SciFinder, Google Scholar, MEDLINE, EMBASE, Scopus directory were explored for ethnomedicinal uses, geographical distribution, phytochemistry, pharmacology, and biomedicine by applying the following keywords: “Acorus calamus”, “Vacha”, “Medhya”, “neuroprotective”, “phytochemistry”, “obesity”, “oxidative stress”, “anticonvulsant”, “antidepressant”, “antihypertensive”, “anti-inflammatory”, “immunomodulator”, “antioxidant”, “diabetes”, “mechanism of action” with their corresponding medical subject headings (MeSH) terms using conjunctions OR/AND. The search was focused on identifying Ayurvedic claims in the available ethnomedicinal, phytochemical, preclinical, clinical, and toxicity reports to understand the role of A. calamus in neurological and metabolic disorders. This search was undertaken between January 2018 and January 2020. Searches were restricted to the English language. The search methodology as per the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) is stipulated in the flowchart in Figure 3.

3. Geographical Distribution

A. calamus grows in high (1800 m) and low (900 m) altitudes and it is found to be geographically available in 42 countries [8]. Furthermore, as per the Global Biodiversity Information Facility records [12], the distribution of this plant in several parts of the world, as well as in India, is highlighted in Figure 4.

4. Ethnomedicinal Use

This plant is being practiced traditionally in the Indian Ayurvedic tradition, as well as in the Chinese system of medicine for analgesic, antipyretic, tonic, anti-obesity, and healing purposes; it is highly effective for skin diseases, along with neurological, gastrointestinal, respiratory, and several other health disorders. Rhizomes and leaves are found to be profusely practiced in the form of infusion, powder, paste, or decoction [13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72]. The ethnomedicinal uses of the A. calamus are detailed in Table 1.
A. calamus rhizomes and leaves are also used as an active pharmaceutical ingredient in various Ayurvedic formulations (Table 2).

5. Phytochemistry

The phytochemical investigation of this plant has been ongoing since the year 1957 [73,74]. To date, about 145 compounds were isolated from A. calamus rhizomes and leaves, viz. phenylpropanoids, sterols, triterpene glycosides, triterpenoid saponins, sesquiterpenoids, monoterpenes, and alkaloids (Table 3). Amongst those, phenylpropanoids (chiefly, asarone and eugenol) and sesquiterpenoids have been considered the principal effective compounds of A. calamus. Chemical structures of isolated compounds from A. calamus are illustrated in Figure 5.

5.1. Phenylpropanoids

Phenylpropanoids have an aromatic ring with a structurally diverse group of phenylalanine-derived secondary plant metabolites (C6–C3), like α-asarone, β-asarone, eugenol, isoeugenol, etc. [75]. A number of phenylpropanoids have been identified from A. calamus rhizome and leaves (1-45). α and β-asarone isolated from the rhizome are the predominant compounds present in this plant. A series of aromatic oils from the rhizome with diverse structures are also reported [74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98].

5.2. Sesquiterpenoids

About 44 sesquiterpenes, including lactones, were characterized and identified in A. calamus rhizomes. Sesquiterpene lactones are produced of 3 isoprene units and composed of lactone rings. αβ unsaturated γ-lactonic ring in sesquiterpene lactones is believed to be responsible for pharmacological activity (46-99) [74,78,89,91,93,98,99,100,101,102,103,104].

5.3. Monoterpenes

Monoterpenes (C-10) are the simplest class of the terpene series that belongs to two isoprene units (tricyclic, bicyclic, monocyclic, etc.). Monoterpenes can have different functional groups, like aldehydes, ketones, esters, ethers, phenols, and alcohols [80]. These organic compounds emit the characteristic flavor and fragrance of A. calamus leaves and rhizomes (100-122) [74,78,89,91,93,97,98].

5.4. Triterpenoid Saponins

Triterpenoid saponins are made up of a pentacyclic C-30 terpene skeleton as a pillar. Limited reports studying triterpenoid saponins in A. calamus are available, and only two triterpenoid saponins (124, 125) have been isolated from A. calamus rhizomes (Table 3) [85].

5.5. Other Compounds

To date, one xanthone glycoside (123) [82,83], two alkaloids (126-127) [84], one triterpene glycoside (128), one steroid (129) [85], 12 amino acids (130-141) [86,87], and 4 fatty acids (142-145) [88] have been identified in A. calamus rhizomes [83,84,85,86,87,88].

6. Pharmacological Properties

Diverse bioactivities of A. calamus extracts are evident from preclinical (in vitro and in vivo) and clinical reports, such as antidiabetic, anti-obesity, antihypertensive, antioxidant, anti-inflammatory, immunomodulatory, anticonvulsant, and neuroprotective [105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173]. The summarized information on A. calamus botanical parts, extract type, and their bioactivities in neurological and metabolic disorders is stipulated in Table 4.

6.1. Antidiabetic Effect

The antidiabetic effect of A. calamus ethyl acetate fraction was evaluated in streptozotocin (STZ)-induced and diabetic (db/db) mice. Glucagon-like peptide-1 (GLP-1) levels, plasma insulin, “and related gene expression were evaluated. The fraction (100 mg/kg, intragastric (i.g.)) indicated a significant reduction in blood glucose levels. For in vitro, at the concentration of 12.5 μg/mL, a significant increment in GLP-1 levels was found in the insulin-secreting L-cell culture medium [108]. The ethyl acetate radix fraction exhibited a significant effect on the HIT-T15 cell line and α-glucosidase enzyme. The ethyl acetate fraction also enhanced insulin secretion in HIT-T15 cells and blocked the α-glucosidase in vitro activity with 0.41 μg/mL of inhibitory concentration (IC50) [109].”

6.2. Anti-Obesity Effect

The β-asarone compound isolated from the rhizome was investigated against high-fat diet (HFD)-induced obesity in animals. β-Asarone-treated adipose rats showed weight loss, but also inhibited metabolic transformations, as well as glucose intolerance, elevated cholesterol, and adipokine variance [143]. The in vitro investigation on the A. calamus aqueous extract showed lipid-lowering activity through inhibition of the pancreatic lipase percentage (28.73%) [144].

6.3. Antihypertensive Effect

The antihypertensive effects of A. calamus were studied on their own, in isolation, and in combination with Gymnema sylvestre in the HFD-induced hypertension in rats. The HFD was given for 4 weeks, which significantly increased the average systolic blood pressure (SBP). At a 200 mg/kg dose, A. calamus in combination with G. sylvestre reduced the SBP and heart rate significantly. A. calamus with G. sylvestre exhibited synergistic effect as compared with individual herbs [145].

6.4. Anti-Inflammatory and Immunomodulatory Effect

The methanolic A. calamus rhizome extract (12.5 µg/mL) prevented the VCAP-1 and intercellular expression on the surface of mouse myeloid leukemia cells and murine endothelial cells, respectively [146]. In an in vitro anti-inflammatory study (Red blood cell membrane stabilization method), the A. calamus aqueous rhizome extract at the highest concentration of 10 mg/mL showed insignificant activity against hemolysis inhibition and the RBC membrane stabilization percentage [144]. Aqueous A. calamus leave extract was studied on HaCaT cells and restricted the characteristics of interleukin (IL)-8, IL-6 RNA protein levels alongside interferon regulatory factor 3 (IRF3) and nuclear factor kB (NF-κB) activation [147]. N-hexane, butanolic, and aqueous fractions of A. calamus were evaluated against cyclooxygenase (COX) and lipoxygenase (LOX)-mediated eicosanoid production by arachidonic acid. The butanolic fraction inhibited the COX-mediated production of thromboxane B2 (TXB2) and lipoxygenase product 1 (LP1). Investigation of the underlying signaling pathways revealed that the butanolic fraction inhibited phospholipase C (PLC) pathway in platelets, presumably acting on protein kinase C (PKC) [148]. The essential oil isolated from A. calamus was evaluated by protein denaturation assay, where at the concentration level of 300 μg/mL, 69.56% of the inhibition level was observed [149].

6.5. Antioxidant Effect

The in vitro antioxidant activity of acetone, acetonitrile, alcoholic, and aqueous extracts of A. calamus rhizomes exhibited free radical scavenging activity on the [2,2′-azinobis (3-ethylbenzothiazoline-6-sulphonic acid)] free radical scavenging activity assay (ABTS), the (1, 1-diphenyl-2-picrylhydrazyl) free radical scavenging activity assay (DPPH), and the ferric ion reducing antioxidant power assay (FRAP). Strong antioxidant effect was noticed in the acetone extract, followed by acetonitrile and methanol, while in the aqueous extract, poor antioxidant activity was found [150]. The aqueous extract exhibited superior antioxidant effects in metal ion chelation, lipid peroxidation (LPO), and DPPH assays [144,151]. The in vitro antioxidant activity of ethanol, hydro-ethanol, and aqueous whole plant extracts of A. calamus was investigated using FRAP, DPPH, nitric oxide, hydroxyl radical, reductive ability, and superoxide radical scavenging activity. The existence of phenolics and flavonoids in A. calamus are believed to contribute to the promising antioxidant effect. IC50 values of the ethanol extract were found to be 54.82, 109.85, 38.3, 118.802 µg/mL for the scavenging activities of DPPH, hydroxyl radical, superoxide radical, and nitric oxide, respectively. The irreversible potential of the above results and the FRAP values of the extracts were found to augment in a concentration-dependent manner [152]. “Ethanol and hydro-alcoholic extracts of A. calamus roots and rhizomes were studied for antioxidant potential against DPPH compared with butylated hydroxyanisole (BHA) and silymarin. Ethanol and hydro-alcoholic extracts showed free radical scavenging activity of 59.13 ± 18.95 and 56.71 ± 19.54, respectively [153,154,155]. The essential oil isolated from A. calamus showed strong antioxidant efficacy against the β-carotene/linoleic acid bleaching test and DPPH free radicals [156]. The methanol extract of the A. calamus rhizome was evaluated against the free radical scavenging activity, and the reported IC50 value was 704 µg/mL [157]. The IC50 of the essential oil was 1.68 μg/mL, which showed virtuous free radical scavenging activity in the DPPH test [149].”

6.6. Anticonvulsant Effect

The methanol extract shows anticonvulsant effects feasibly through potentiating the action of gamma-aminobutyric acid (GABA) pathway in the central nervous system [124]. When it comes to the purification of A. calamus rhizome in cow urine, it is advocated in the Ayurvedic pharmacopoeia of India (API) before its therapeutic use. The purified rhizome was investigated in a maximal electroshock (MES) seizure model, and phenytoin was used as the standard drug. The raw and processed rhizome (11 mg/kg, p.o.) exhibited notable anticonvulsant activity by minimizing the span of the tonic extensor period in rats, whereas the processed rhizome showed better therapeutic activity than when it was raw [158]. The calamus oil isolated from the A. calamus rhizome was evaluated at varying dose levels of 30, 100, and 300 mg/kg, p.o., body weight (b.w.), against MES, pentylenetetrazol (PTZ), and minimal clonic seizure (MCS) models. The calamus oil was found to be neurotoxic at 300 mg/kg, though it was effective in the MCS test at 6 Hz. The protective index value of calamus oil was found to be 4.65 [125].

6.7. Antidepressant Effect

Interaction of the methanolic A. calamus rhizome extract with the adrenergic, dopaminergic, serotonergic, and GABAergic system was found responsible for the expression of antidepressant activity [128]. In another study, the methanolic A. calamus leave extract showed significant activity through a reduction in the immobility period in the TST and FST [129]. Through interaction with the adrenergic and dopaminergic system, the hydro-alcoholic extract was normalized to the over-activity of the hypothalamic pituitary adrenal (HPA) axis [131]. Sobers capsules (a herbo-mineral formulation containing A. calamus) were evaluated by tail suspension and forced swimming tests in mice. At the oral dose of 50 mg/kg for 14 days, capsules exhibited insignificant impact on locomotor activity, and caused antidepressant effects in experimental animals [159]. Tensarin (the traditional medicine of Nepal containing A. calamus) was evaluated for the anxiolytic effect in mice using the open field test (OFT), activity monitoring along with the passive avoidance test. At all three dose levels (50, 100, 200 mg/kg), Tensarin produced an anxiolytic effect in a dose-dependent way by an improvement in rearing, number of passages, and duration of the period employed by mice [160].

6.8. Neuroprotective Effect

The ethanolic extract was studied (25, 50, and 100 mg/kg doses, oral and intraperitoneal routes) for learning and memory-enhancing activity. The subjects used consisted of male rates, through Y maze and shuttle box tests models. The findings showed an increase in acquisition–recalling and spatial recognition data [161]. The ethanolic A. calamus rhizome extract (0.5 mL/kg, i.p.) potentiated pentobarbitone-created sleep periods, which caused significant inhibition of conditioned avoidance response in rats and marked (40–60%) protection against PTZ-induced convulsions, although it did not show any spontaneous motor activity and impact the aggressive or fighting behavior response in male rat pairs [162].

6.9. Cardioprotective Effect

The alcoholic A. calamus rhizome extract (100 and 200 mg/kg) considerably attenuated isoproterenol-led cardiomyopathy in rats and showed a significant reduction in the heart/body weight ratio, level of serum calcineurin, serum nitric oxide, serum lactate dehydrogenase (LDH), and thiobarbituric acid reactive substances (TBARS) level. However, the level of the antioxidant enzyme was found increased at the 100 mg/kg extract dose level [163]. The crude extract and its fractions (0.01–10 mg/mL) were investigated in an isolated rabbit heart, which showed mild reduction in the force of forced vital capacity (FVC), hazard ratio (HR), and cystic fibrosis (CF), while the ethyl acetate extract exhibited complete suppression, and the n-hexane fraction showed the same effect on FVC and HR, but enhanced CF. The extract and its fractions exhibited controlled coronary vasodilator effect, interceded maybe by an endothelial-derived hyperpolarizing factor [164]. The cardioprotective potential of the whole plant’s ethanolic extract (100 and 200 mg/kg) reduced serum enzyme levels and shielded the myocardium from the lethal effect of DOX [141].

6.10. Cytochrome Inhibitory Activities

Cytochromes P450 (CYPs) are the prime enzymes that catalyze the oxidative metabolism of a wide variety of xenobiotics. It is known that 2,4,5-trimethoxycinnamic acid is the main metabolite of α- or β- asarone [165]. The metabolism rate of α- and β-asarone was shown to be directly proportional to the CYPs concentration in rat hepatocytes and liver microsomes [166,167]. CYP3A4 (CYP isoforms) has been reported for bioactivation of α-asarone [168]. The hydro-alcoholic A. calamus extract and α-asarone were evaluated by the CYPs-carbon monoxide complex method. The extract exhibited moderate potential interaction in CYP3A4 (IC50 = 46.84 μg/mL) and CYP2D6 (IC50 = 36.81 μg/mL), while α-asarone showed higher interaction in CYP3A4 (IC50 = 65.16 μg/mL) and CYP2D6 (IC50 = 55.17 μg/mL) [169]. These outcomes indicated that both extracts and α-asarone interacted quite well in drug metabolism and also had an inhibitory effect on CYP3A4 and CYP2D6. The drug-drug interaction effect of the A. calamus extract and its main chemical constituent (α and β-asarone) needs to be studied in more CYPs isomers, like CYP2C9 and CYP2E1.

6.11. Toxicity and Safety Concerns

In acute and sub-acute toxicity of the hydro-alcoholic extract of A. calamus in rats, at the highest dose level of 10 gm/kg, no severe changes were observed, and the lethal dose (LD50) was found to be 5 g/kg [170]. The petroleum ether extracts (obtained by cold rolling, water distillation, and Soxhlet extraction methods) of the A. calamus rhizome showed mild toxicity in two-day-old oriental fruit flies [171]. The ethanolic extract of the A. calamus rhizome at oral dosage of 175, 550, 1750, and 5000 mg/kg b.w. was given for 14 days within an acute toxicity study, while at the dose level of 0, 200, 400, and 600 mg/kg, p.o., the extract was given for 90 days within a chronic toxicity study. At the doses of 1750 and 5000 mg/kg, piloerection, tremors, and abdominal breathing were found for 30 min [172]. In that study, A. calamus was purified for 3 h in cow urine, decoction of Sphaeranthus indicus, and decoction of leaves of Mangifera indica, Eugenia jambolana, Feronia limonia, Citrus medica, and Aegle marmelos, followed by fomentation with Gandhodaka (decoction of six aromatic herbs) for 1 h. The acute oral toxicity test of raw and purified A. calamus was performed in albino rats at 2000 mg/kg for 2 weeks. At the 2000 mg/kg dose, A. calamus did not produce any toxic symptoms within 14 days [173].
The β-asarone compound isolated from A. calamus was found to be carcinogenic and toxic [174]. The LD50 value of β-asarone by oral and intraperitoneal route was found to be 1010 and 184 mg/kg, respectively, in mice and rats [175]. The LD50 of calamus oil was found to be 8.88 gm/kg b.w. [176], while in the calamus oil obtained from Jammu, India, the LD50 was 777 mg/kg b.w. [177]. Overall, several investigations have been carried out on A. calamus regarding its toxicity; however, no noticeable data on toxicity have been found so far.

7. Clinical Reports

A. calamus has also been clinically investigated as a monotherapy as well as in combination with other medicinal herbs in healthy subjects and sufferers of various metabolic and neurological ailments. Most clinical research has looked at the A. calamus effect on obesity, depression, neuroprotection, and cardiovascular disease [178,179,180,181,182,183,184,185,186,187,188,189,190,191]. The data obtained so far can be found in Table 5. Furthermore, a systematic review reveals that A. calamus (alone or in combination therapy) exhibits anti-obesity, antidepressant, and cardioprotective effects, as well as helps physical and mental performance.

8. Mechanistic Role

The proposed mechanism of action of A. calamus in neurological and metabolic disorders includes a synergic integration of antioxidant defense, GABAergic transmission, brain stress hormones modulation, pro-inflammatory cytokines, leptin and resistin levels, adipocytes inhibition, calcium channel blocker effect, protein synthesis, oxidative stress, acetylcholinesterase (AChE) inhibition, and anti-dopaminergic properties. A compendium of mechanisms of action of A. calamus in neurological and metabolic protection is illustrated in Figure 6 and Table 6. A. calamus significantly affects fasting blood sugar, insulin resistance, HbA1c, and the adipogenic transcription expression factor through various mechanisms, viz. antioxidant, anti-inflammatory, β-cells regeneration, improving insulin sensitivity, gluconeogenesis, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and glucose transporter type 4 (GLUT-4)-mediated transport inhibition.
The antihypertensive effect of A. calamus may be explained by Ca2+ antagonists that affect the nitric oxide pathway. The chemical constituents of A. calamus upregulate the antioxidant effect, suppress pro-inflammatory cytokines, and act as detoxifying enzymes through the NF-κB and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways. The Nrf2 pathway may be activated by phenylpropanoids, sesquiterpenoids, and monoterpenes by interaction of active phytoconstituents with nitric oxide derivatives react with thiol groups between KEAP1 and Nrf2, along with Nrf2 phosphorylation. “When Nrf2 is released from the Kelch-like erythroid-derived CNC (cap’n’collar) homology protein (ECH)-associated protein 1 (KEAP1), it transfers into the nucleus, where it induces the genes encoding protein expression impenetrable in glutathione (GSH) synthesis, antioxidant, and detoxifying phase 2 enzymes. Oxidative stress and ligands for tumor necrosis factor receptors (TNFRs) and toll-like receptors (TLRs) activate upstream Ik-B kinases (IKKs), ensuing phosphorylation of IkB that is generally bound to the inactive NF-kB dimer in the cytoplasm. After that, IkB is targeted for proteasomal degradation and NF-kB, then it moves into the nucleus where it induces inflammatory cytokine expression in addition to the genes encoding proteins like superoxide dismutase (SOD) 2 and B cell chronic lymphocytic leukemia (CLL)/lymphoma 2 (Bcl2) involved in adaptive stress response (Figure 7). The bioactive molecules of A. calamus can inhibit NF-kB in inflammatory immune cells, while other phytoconstituents may activate NF-kB in neuronal cells to improve stress resistance.” A. calamus phytoconstituents regulate NF-kB, LOX, and COX-2 activity. These compounds dose-dependently suppress the production of inflammatory factors like NO, TNF-α, IL-6, IL-1β, and JNK signaling, acting as anti-inflammatory agents. In addition, it was also noted that the inflammation induced by various chemicals was inhibited by bioactive constituents through suppression of IkB/NF-kB and JNK/AP-1 signaling pathways. Thus, over several studies, it has been reported that asarone compounds have a potential against neurodegenerative diseases.
PPAR gene and C/EBP are involved in the differentiation process. PPAR-δ and PPAR-γ promote adipogenesis. In the same way, amino acids and glucose react with C/EBP- δ and C/EBP-β. If low levels of glucose induce gadd153, the inactive dimer is formed, with C/EBP-β inhibiting the progress of adipocyte development. C/EBP delta activates C/EBP-α. This is mainly involved in the formation of mature adipocytes and lipid accumulation in adipose tissue. In 3T3-L1 preadipocytes, α-asarone and β-asarone inhibited adipocyte differentiation and reduced the intracellular lipid accumulation, and also decreased the expression levels of adipogenic transcription factors (PPARγ and C/EBPα). These phytochemicals significantly promoted adenosine monophosphate-activated protein kinase (AMPK), which is known to suppress adipogenesis. It was also found that pretreatment with α-asarone and β-asarone, a typical inhibitor of AMPK, attenuated the inhibitory effect of asarone on AMPK phosphorylation. The asarone-induced AMPK activation leads to a decrease in adipogenic transcription factor expression, and suppresses adipogenesis.

9. Perspectives and Future Directions

The present review provides a plethora of information apropos ethnomedicinal uses, marketed formulations, geographical distribution, chemical constituents, pharmacological activities of crude, n-hexane, ethyl acetate, methanolic, ethanolic, hydro-alcoholic, aqueous extracts along with pure compounds, and clinical trials related to A. calamus.
Investigations on extracts and compounds of A. calamus suggested antidiabetic, anti-obesity, antihypertensive, anti-inflammatory, antioxidant, anticonvulsant, antidepressant, neuroprotective, and cardioprotective potentials with distinct underlying signaling pathways. The biological potential and mechanisms of action of some of the chemical constituents (α-asarone, β-asarone, eugenol) are known. However, other compounds need to be scientifically explored for their bioactivities and molecular modes of action, which could provide a lead for further development into therapeutics. More systematic, well-designed, and multi-center clinical studies are warranted to evaluate standardized extracts of A. calamus therapeutically and to identify the pharmacokinetic-dynamic roles of pharmacologically active biomolecules. There is scarce data from experimental and clinical reports on hypertension, diabetes, and atherosclerosis, and less supporting evidence is available on the use of A. calamus to treat hypertension and diabetes. Based on the available data, it is suggested that this plant could be used as an adjuvant to the established targeted drugs for neurological and metabolic disorders.
In 1974, United States food & drug administration (USFDA) banned A. calamus due to its carcinogenic effects following animal studies. They reported β-asarone as a carcinogenic agent, but the study was conducted on the calamus oil which consists of β-asarone in about 80%, while its different genotype in Europe and India contains β-asarone in lower concentrations. A. calamus cultivated in various geographical regions may have different chemical compositions along with therapeutic properties challenging quality control, toxicity, and safety concerns of A. calamus. In addition, the heavy metal, mycotoxin, and pesticide concentrations are required to be addressed in all toxicity studies.

10. Conclusions

Compelling in vitro, in vivo and clinical evidence suggests that the potential role of A. calamus rhizomes for modulating metabolic and neurological disorders could be due to their richness in several classes of active phytoconstituents. The predominant compounds present in rhizomes and leaves responsible for expression of potent bioactivities include α-asarone, β-asarone, eugenol, and calamine. The present report is expected to fill the gaps in the existing knowledge and could provide a lead for researchers working in the areas of phytomedicine, ethnopharmacology, and clinical research.

Author Contributions

R.S. and V.S. conceived the idea and wrote the manuscript. D.S.G., K.K., E.N., and N.M. edited and proofread the document. The entire team approved the submission of the final manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This paper was supported by the UHK Excellence project.

Acknowledgments

The authors express their sincere gratitude to Bharat Ratna Mahamana Pandit Madan Mohan Malviya, the founder of the Banaras Hindu University, Varanasi, for his services to humanity, great vision, and blessings. This work was also supported by University of Hradec Kralove (Faculty of Science, VT2019-2021) [KK, EN].

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Report. Available online: https://www.who.int/whr/2001/media_centre/press_release/en/ (accessed on 4 October 2019).
  2. Toniolo, A.; Cassani, G.; Puggioni, A.; Rossi, A.; Colombo, A.; Onodera, T.; Ferrannini, E. The diabetes pandemic and associated infections: Suggestions for clinical microbiology. Rev. Med. Microbiol. 2019, 30, 1–17. [Google Scholar] [CrossRef]
  3. Younossi, Z.M. Non-alcoholic fatty liver disease-A global public health perspective. J. Hepatol. 2019, 70, 531–544. [Google Scholar] [CrossRef] [Green Version]
  4. Després, J.P. Is visceral obesity the cause of the metabolic syndrome. Ann. Med. 2006, 38, 52–63. [Google Scholar] [CrossRef]
  5. Farooqui, A.A.; Farooqui, T.; Panza, F.; Frisardi, V. Metabolic syndrome as a risk factor for neurological disorders. Cell. Mol. Life Sci. 2012, 69, 741–762. [Google Scholar] [CrossRef]
  6. Tilg, H.; Hotamisligil, G.S. Nonalcoholic fatty liver disease: Cytokine-adipokine interplay and regulation of insulin resistance. Gastroenterology 2006, 131, 934–945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Suzanne, M.; Tong, M. Brain metabolic dysfunction at the core of Alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 548–559. [Google Scholar]
  8. Quraishi, A.; Mehar, S.; Sahu, D.; Jadhav, S.K. In vitro mid-term conservation of Acorus calamus L. via cold storage of encapsulated microrhizome. Braz. Arch. Biol. Technol. 2017, 60, 1–9. [Google Scholar] [CrossRef] [Green Version]
  9. Balakumbahan, R.; Rajamani, K.; Kumanan, K. Acorus calamus: An overview. J. Med. Plant Res. 2010, 4, 2740–2745. [Google Scholar]
  10. Sharma, V.; Singh, I.; Chaudhary, P. Acorus calamus (The Healing Plant): A review on its medicinal potential, micropropagation and conservation. Nat. Prod. Res. 2014, 28, 1454–1466. [Google Scholar] [CrossRef]
  11. Singh, R.; Sharma, P.K.; Malviya, R. Pharmacological properties and ayurvedic value of Indian buch plant (Acorus calamus): A short review. Adv. Biol. Res. 2011, 5, 145–154. [Google Scholar]
  12. Global Biodiversity Information Facility. Available online: https://www.gbif.org/ (accessed on 10 February 2020).
  13. Kingston, C.; Jeeva, S.; Jeeva, G.M.; Kiruba, S.; Mishra, B.P.; Kannan, D. Indigenous knowledge of using medicinal plants in treating skin diseases in Kanyakumari district, Southern India. Indian J. Tradit. Knowl. 2009, 8, 196–200. [Google Scholar]
  14. Pradhan, B.K.; Badola, H.K. Ethnomedicinal plant use by Lepcha tribe of Dzongu valley, bordering Khangchendzonga Biosphere Reserve, in north Sikkim India. J. Ethnobiol. Ethnomed. 2008, 4, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Sharma, P.K.; Chauhan, N.S.; Lal, B. Observations on the traditional phytotherapy among the inhabitants of Parvati valley in western Himalaya, India. J. Ethnopharmacol. 2004, 92, 167–176. [Google Scholar] [CrossRef] [PubMed]
  16. Dwivedi, S.N.; Dwivedi, S.; Patel, P.C. Medicinal plants used by the tribal and rural people of Satna district, Madhya Pradesh for the treatment of gastrointestinal diseases and disorders. Nat. Prod. Rad. 2006, 5, 60–63. [Google Scholar]
  17. Usher, G. Spilanthes Acmella, a Dictionary of Plants Used by Man; CBS Publishers and Distributers: New Delhi, India, 1984; p. 38. [Google Scholar]
  18. Ghosh, A. Ethnomedicinal plants used in West Rarrh region of West Bengal. Nat. Prod. Rad. 2008, 7, 461–465. [Google Scholar]
  19. Natarajan, B.; Paulsen, B.S.; Korneliussen, V. An ethnopharmacological study from Kulu District, Himachal Pradesh, India: Traditional knowledge compared with modern biological science. Pharm. Biol. 2000, 38, 129–138. [Google Scholar] [CrossRef]
  20. Nisha, M.C.; Rajeshkumar, S. Survey of crude drugs from Coimbatore city. Indian J. Nat. Prod. Resour. 2010, 1, 376–383. [Google Scholar]
  21. Ragupathy, S.; Steven, N.G.; Maruthakkutti, M.; Velusamy, B.; Ul-Huda, M.M. Consensus of the ‘Malasars’ traditional aboriginal knowledge of medicinal plants in the Velliangiri holy hills, India. J. Ethnobiol. Ethnomed. 2008, 4, 8–16. [Google Scholar] [CrossRef] [Green Version]
  22. Tomar, A. Folk medicinal uses of plant roots from Meerut district, Uttar Pradesh. Indian J. Tradit. Knowl. 2009, 8, 298–301. [Google Scholar]
  23. Rajith, N.P.; Ramachandran, V.S. Ethnomedicines of Kurichyas, Kannur district, Western Ghats, Kerala. Indian J. Nat. Prod. Resour. 2010, 1, 249–253. [Google Scholar]
  24. Barbhuiya, A.R.; Sharma, G.D.; Arunachalam, A.; Deb, S. Diversity and conservation of medicinal plants in Barak valley, Northeast India. Indian J. Tradit. Knowl. 2009, 8, 169–175. [Google Scholar]
  25. Kadel, C.; Jain, A.K. Folklore claims on snakebite among some tribal communities of Central India. Indian J. Tradit. Knowl. 2008, 7, 296–299. [Google Scholar]
  26. Boktapa, N.R.; Sharma, A.K. Wild medicinal plants used by local communities of Manali, Himachal Pradesh, India. Ethnobot. Leafl. 2010, 3, 259–267. [Google Scholar]
  27. Kingston, C.; Nisha, B.S.; Kiruba, S.; Jeeva, S. Ethnomedicinal plants used by indigenous community in a traditional healthcare system. Ethnobot. Leafl. 2007, 11, 32–37. [Google Scholar]
  28. Jain, A.; Roshnibala, S.; Kanjilal, P.B.; Singh, R.S.; Singh, H.B. Aquatic /semi-aquatic plants used in herbal remedies in the wetlands of Manipur, Northeastern India. Indian J. Tradit. Knowl. 2007, 6, 346–351. [Google Scholar]
  29. Yabesh, J.M.; Prabhu, S.; Vijayakumar, S. An ethnobotanical study of medicinal plants used by traditional healers in silent valley of Kerala, India. J. Ethnopharmacol. 2014, 154, 774–789. [Google Scholar] [CrossRef]
  30. Sher, Z.; Khan, Z.; Hussain, F. Ethnobotanical studies of some plants of Chagharzai valley, district Buner, Pakistan. Pak. J. Bot. 2011, 43, 1445–1452. [Google Scholar]
  31. Poonam, K.; Singh, G.S. Ethnobotanical study of medicinal plants used by the Taungya community in Terai Arc Landscape, India. J. Ethnopharmacol. 2009, 123, 167–176. [Google Scholar] [CrossRef]
  32. Shrestha, P.M.; Dhillion, S.S. Medicinal plant diversity and use in the highlands of Dolakha district Nepal. J. Ethnopharmacol. 2003, 86, 81–96. [Google Scholar] [CrossRef]
  33. Khatun, M.A.; Harun-Or-Rashid, M.; Rahmatullah, M. Scientific validation of eight medicinal plants used in traditional medicinal systems of Malaysia: A review. Am. Eurasian J. Sustain. Agric. 2011, 5, 67–75. [Google Scholar]
  34. Dastur, J.F. Medicinal Plants of India and Pakistan; D. B. Taraporevala Sons and Co. Ltd: Bombay, India, 1951; p. 12. [Google Scholar]
  35. Satyavati, G.V.; Raina, M.K.; Sharmal, M. Medicinal Plants of India; Indian Council of Medical Research: New Delhi, India, 1976; Volume I, pp. 14–16. [Google Scholar]
  36. Jain, S.K. Medicinal Plants; National Book Trust: New Delhi, India, 1968.
  37. Malhi, B.S.; Trivedi, V.P. Vegetable antifertility drugs of India. Q. J. Crude Drug Res. 1972, 12, 19–22. [Google Scholar]
  38. Singh, M.P.; Malla, S.B.; Rajbhandari, S.B.; Manandhar, A. Medicinal plants of Nepal retrospect’s and prospects. Econ. Bot. 1979, 33, 185–198. [Google Scholar] [CrossRef]
  39. Kirtikar, K.R.; Basu, B.D. Indian Medicinal Plants; M/S. Bishen Singh Mahendra Pal Singh: Dehradun, India, 1975; Volume IV. [Google Scholar]
  40. Lama, S.; Santra, S.C. Development of Tibetan plant medicine. Sci. Cult. 1979, 45, 262–265. [Google Scholar] [PubMed]
  41. Burang, T. Cancer therapy of Tibetan healers. Comp. Med. East West 1979, 7, 294–296. [Google Scholar] [CrossRef] [PubMed]
  42. Wallnofer, H.; Rottauscher, A. Chinese Folk Medicine and Acupuncture; Bell Publishing Co, Inc: New York, NY, USA, 1965. [Google Scholar]
  43. Agarwal, S.L.; Dandiya, P.C.; Singh, K.P.; Arora, R.B. A note on the preliminary studies of certain pharmacological actions of Acorus calamus. J. Am. Pharm. Assoc. 1956, 45, 655–656. [Google Scholar] [CrossRef]
  44. Duke, J.A.; Ayensu, E.S. Medicinal Plants of China; Reference Publications, Inc: Algonac, MI, USA, 1985. [Google Scholar]
  45. Perry, L.M.; Metzger, J. Medicinal Plants of East and Southeast Asia; MIT Press: Cambridge, UK, 1980. [Google Scholar]
  46. Boissya, C.L.; Majumder, R. Some folklore claims from the Brahmaputra Valley (Assam). Ethnomedicine 1980, 6, 139–145. [Google Scholar]
  47. Dragendorff, G. Die Heilpflanzen der Verschie Denen Volker und Zeiten; F. Enke: Stuttgart, Germany, 1898. [Google Scholar]
  48. Li, H.L. Hallucinogenic plants in Chinese herbals. Harv. Univ. Bot. Mus. Leafl. 1977, 25, 161–177. [Google Scholar]
  49. Shih-Chen, L. Chinese Medicinal Herbs; Georgetown Press: San Francisco, CA, USA, 1973. [Google Scholar]
  50. Hirschhorn, H.H. Botanical remedies of the former Dutch East Indies (Indonesia) I: Eumycetes, Pteridophyta, Gymnospermae, Angiospermae (Monocotyledones only). J. Ethnopharmacol. 1983, 7, 123–156. [Google Scholar] [CrossRef]
  51. Wren, R.C. Potter’s New Cyclopaedia of Botanical Drugs and Preparations; Sir Isaac Pitman and Sons, Ltd: London, UK, 1956. [Google Scholar]
  52. Grieve, M. A Modern Herbal; Dover Publications, Inc: New York, NY, USA, 1971; Volume II. [Google Scholar]
  53. Wheelwright, E.G. Medicinal Plants and Their Stor; Dover Publications, Inc: New York, NY, USA, 1974. [Google Scholar]
  54. Moerman, D.E. Geraniums for the Iroquois; Reference Publications, Inc: Algonac, MI, USA, 1981. [Google Scholar]
  55. Jochle, W. Menses-inducing drugs: Their role in antique, medieval and renaissance gynecology and birth control. Contraception 1974, 10, 425–439. [Google Scholar] [CrossRef]
  56. Watt, J.M.; Breyer-Brandwijk, M.G. The Medicinal and Poisonous Plants of Southern and Eastern Africa; E. & S. Livingstone Ltd.: London, UK, 1962. [Google Scholar]
  57. Kantor, W. Quack abortifacients and declining birth rate. Therap. Monatsh. 1916, 30, 561–568. [Google Scholar]
  58. Herrmann, G. Therapy with medicinal plants in present medicine. Med. Monatsschr. Pharm. 1956, 10, 79. [Google Scholar]
  59. Burkill, I.H. Dictionary of the Economic Products of the Malay Peninsula; Ministry of Agriculture and Cooperatives: Kuala Lumpur, Malaysia, 1966; Volume 1.
  60. Motley, T.J. The Ethnobotany of Sweet Flag, Acorus calamus (Araceae). Econ. Bot. 1994, 48, 397–412. [Google Scholar] [CrossRef]
  61. Krochmal, A.; Krochmal, C. A Guide to the Medicinal Plants of the United States; Quadrangle/The New York Times Book Co: New York, NY, USA, 1975. [Google Scholar]
  62. El’Yashevych, O.H.; Cholii, R. Some means of treatment in the folk medicine of L’Vov. Farmatsevtychnyi Zhurnal 1972, 27, 78. [Google Scholar]
  63. Barton, B.H.; Castle, T. The British Flora Medica; Chatto and Windus: Piccadilly, London, UK, 1877. [Google Scholar]
  64. Mokkhasamit, M.; Ngarmwathana, W.; Sawasdimongkol, K.; Permphiphat, U. Pharmacological evaluation of Thai medicinal plants. (Continued). J. Med. Assoc. Thail. 1971, 54, 490–504. [Google Scholar]
  65. Harris, B.C. The Complete Herbal; Barre Publishers: Barre, MA, USA, 1972. [Google Scholar]
  66. Lindley, J. Flora Medica; Paternoster-Row: London, UK, 1838. [Google Scholar]
  67. Caius, J.F. The Medicinal and Poisonous Plants of India; Scientific Publishers: Jodhpur, India, 1986. [Google Scholar]
  68. Clymer, R.S. Nature’s Healing Agents; Dorrance and Company: Philadelphia, PA, USA, 1963. [Google Scholar]
  69. Manfred, L. Siete Mil Recetas Botanicas a Base de Mil Trescientas Plantas; Edit Kier: Buenos Aires, Argentina, 1947. [Google Scholar]
  70. Dobelis, I.N. Magic and Medicine of Plants; The Reader’s Digest Association, Inc.: Pleasantville, New York, NY, USA, 1986. [Google Scholar]
  71. Kumar, H.; Song, S.Y.; More, S.V.; Kang, S.M.; Kim, B.Y. Traditional Korean East Asian Medicines and Herbal Formulations for Cognitive Impairment. Molecules 2013, 18, 14670–14693. [Google Scholar] [CrossRef]
  72. Napagoda, M.T.; Sundarapperuma, T.; Fonseka, D.; Amarasiri, S.; Gunaratna, P. Traditional Uses of Medicinal Plants in Polonnaruwa District in North Central Province of Sri Lanka. Scientifica 2019, 2019, 1–12. [Google Scholar] [CrossRef]
  73. Chaudhury, S.S.; Gautam, S.K.; Handa, K.L. Composition of calamus oil from calamus roots growing in Jammu and Kashmir. Indian J. Pharm. Sci. 1957, 19, 183–186. [Google Scholar]
  74. Mukherjee, P.K. Quality Control of Herbal Drugs: An Approach to Evaluation of Botanicals; Business Horizons: New Delhi, India, 2002; pp. 692–694. [Google Scholar]
  75. Soledade, M.; Pedras, C.; Zheng, Q. The Chemistry of Arabidopsis thaliana. Comp. Nat. Prod. 2010, 3, 1297–1315. [Google Scholar]
  76. Sharma, J.D.; Dandiya, P.C.; Baxter, R.M.; Kandel, S.I. Pharmacodynamical effects of asarone and β-asarone. Nature 1961, 192, 1299–1300. [Google Scholar] [CrossRef]
  77. Sharma, P.K.; Dandiya, P.C. Synthesis and some pharmacological actions of asarone. Indian J. Appl. Chem. 1969, 32, 236–238. [Google Scholar]
  78. Nigam, M.C.; Ateeque, A.; Misra, L.N. GC-MS examination of essential oil of Acorus calamus. Indian Perfum. 1990, 34, 282–285. [Google Scholar]
  79. Matejić, J.; Šarac, Z.; Ranđelović, V. Pharmacological activity of sesquiterpene lactones. Biotech. Biotechnol. Equip. 2010, 24, S95–S100. [Google Scholar] [CrossRef]
  80. Benaiges, A.; Guillén, P. Botanical Extracts. Anal. Cosmet. Prod. 2007, 345–363. [Google Scholar] [CrossRef]
  81. Sparg, S.; Light, M.E.; Van Staden, J. Biological activities and distribution of plant saponins. J. Ethnopharmacol. 2007, 94, 219–243. [Google Scholar] [CrossRef]
  82. Rai, R.; Siddiqui, I.R.; Singh, J. Triterpenoid Saponins from Acorus calamus. ChemInform 1998, 29, 473–476. [Google Scholar]
  83. Rai, R.; Gupta, A.; Siddiqui, I.R.; Singh, J. Xanthone Glycoside from rhizome of Acorus calamus. Indian J. Chem. 1999, 38, 1143–1144. [Google Scholar]
  84. Kumar, S.S.; Akram, A.S.; Ahmed, T.F.; Jaabir, M.M. Phytochemical analysis and antimicrobial activity of the ethanolic extract of Acorus calamus rhizome. Orient. J. Chem. 2010, 26, 223–227. [Google Scholar]
  85. Wu, H.S.; Li, Y.Y.; Weng, L.J.; Zhou, C.X.; He, Q.J.; Lou, Y.J. A Fraction of Acorus calamus L. extract devoid of β-asarone Enhances adipocyte differentiation in 3T3-L1 cells. Phytother. Res. 2007, 21, 562–564. [Google Scholar] [CrossRef]
  86. Vashi, I.G.; Patel, H.C. Chemical constituents and antimicrobial activity of Acorus calamus Linn. Comp. Physiol. Ecol. 1987, 12, 49–51. [Google Scholar]
  87. Weber, M.; Brändle, R. Dynamics of nitrogen-rich compounds in roots, rhizomes, and leaves of the Sweet Flag (Acorus calamus L.) at its natural site. Flora 1994, 189, 63–68. [Google Scholar] [CrossRef]
  88. Asif, M.; Siddiqi, M.T.A.; Ahmad, M.U. Fatty acid and sugar composition of Acorus calamus Linn. Fette Seifen Anstrichm. 1984, 86, 24–25. [Google Scholar] [CrossRef]
  89. Lee, M.H.; Chen, Y.Y.; Tsai, J.W.; Wang, S.C.; Watanabe, T.; Tsai, Y.C. Inhibitory effect of β-asarone, a component of Acorus calamus essential oil, on inhibition of adipogenesis in 3T3-L1 cells. Food Chem. 2011, 126, 1–7. [Google Scholar] [CrossRef]
  90. Padalia, R.C.; Chauhan, A.; Verma, R.S.; Bisht, M.; Thul, S.; Sundaresan, V. Variability in rhizome volatile constituents of Acorus calamus L. from Western Himalaya. J. Essent. Oil Bear. Plants 2014, 17, 32–41. [Google Scholar] [CrossRef]
  91. Kumar, S.N.; Aravind, S.R.; Sreelekha, T.T.; Jacob, J.; Kumar, B.D. Asarones from Acorus calamus in combination with azoles and amphotericin b: A novel synergistic combination to compete against human pathogenic candida species In-vitro. Appl. Biochem. Biotech. 2015, 175, 3683–3695. [Google Scholar] [CrossRef]
  92. Srivastava, V.K.; Singh, B.M.; Negi, K.S.; Pant, K.C.; Suneja, P. Gas chromatographic examination of some aromatic plants of Uttar Pradesh hills. Indian Perfum. 1997, 41, 129–139. [Google Scholar]
  93. Özcan, M.; Akgül, A.; Chalchat, J.C. Volatile constituents of the essential oil of Acorus calamus L. grown in Konya province (Turkey). J. Essent. Oil Res. 2002, 14, 366–368. [Google Scholar] [CrossRef]
  94. Kim, W.J.; Hwang, K.H.; Park, D.G.; Kim, T.J.; Kim, D.W.; Choi, D.K.; Lee, K.H. Major constituents and antimicrobial activity of Korean herb Acorus calamus. Nat. Prod. Res. 2011, 25, 1278–1281. [Google Scholar] [CrossRef]
  95. Patra, A.; Mitra, A.K. Constituents of Acorus calamus: Structure of acoramone. Carbon-13 NMR spectra of cis-and trans-asarone. J. Nat. Prod. 1981, 44, 668–669. [Google Scholar] [CrossRef]
  96. Saxena, D.B. Phenyl indane from Acorus calamus. Phytochemistry 1986, 25, 553–555. [Google Scholar] [CrossRef]
  97. Radušienė, J.; Judžentienė, A.; Pečiulytė, D.; Janulis, V. Essential oil composition and antimicrobial assay of Acorus calamus leaves from different wild populations. Plant Genet. Resour. 2007, 5, 37–44. [Google Scholar] [CrossRef]
  98. Haghighi, S.R.; Asadi, M.H.; Akrami, H.; Baghizadeh, A. Anti-carcinogenic and anti-angiogenic properties of the extracts of Acorus calamus on gastric cancer cells. Avicenna J. Phytomed. 2017, 7, 145. [Google Scholar]
  99. Nawamaki, K.; Kuroyanagi, M. Sesquiterpenoids from Acorus calamus as germination inhibitors. Phytochemistry 1996, 43, 1175–1182. [Google Scholar] [CrossRef]
  100. Zaugg, J.; Eickmeier, E.; Ebrahimi, S.N.; Baburin, I.; Hering, S.; Hamburger, M. Positive GABAA receptor modulators from Acorus calamus and structural analysis of (+)-dioxosarcoguaiacol by 1D and 2D NMR and molecular modeling. J. Nat. Prod. 2011, 74, 1437–1443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Yamamura, S.; Iguchi, M.; Nishiyama, A.; Niwa, M.; Koyama, H.; Hirata, Y. Sesquiterpenes from Acorus calamus L. Tetrahedron 1971, 27, 5419–5431. [Google Scholar] [CrossRef]
  102. Li, J.; Zhao, J.; Wang, W.; Li, L.; Zhang, L.; Zhao, X.F.; Li, S.X. New Acorane-Type Sesquiterpene from Acorus calamus L. Molecules 2017, 22, 529. [Google Scholar] [CrossRef] [Green Version]
  103. Zhou, C.X.; Qiao, D.; Yan, Y.Y.; Wu, H.S.; Mo, J.X.; Gan, L.S. A new anti-diabetic sesquiterpenoid from Acorus calamus. Chin. Chem. Lett. 2012, 23, 1165–1168. [Google Scholar] [CrossRef]
  104. Yao, X.; Ling, Y.; Guo, S.; Wu, W.; He, S.; Zhang, Q.; Zou, M.; Nandakumar, K.S.; Chen, X.; Liu, S. Tatanan A from the Acorus calamus L. root inhibited dengue virus proliferation and infections. Phytomedicine 2018, 42, 258–267. [Google Scholar] [CrossRef]
  105. Prisilla, D.H.; Balamurugan, R.; Shah, H.R. Antidiabetic activity of methanol extract of Acorus calamus in STZ induced diabetic rats. Asian Pac. J. Trop. Biomed. 2012, 2, S941–S946. [Google Scholar] [CrossRef]
  106. Prashanth, D.; Ahmed, F.Z. Evaluation of hypoglycemic activity of methanolic extract of Acorus calamus (linn). roots in alloxan induced diabetes rat model. Int. J. Basic Clin. Pharmacol. 2017, 6, 2665–2670. [Google Scholar]
  107. Wu, H.S.; Zhu, D.F.; Zhou, C.X.; Feng, C.R.; Lou, Y.J.; Yang, B.; He, Q.J. Insulin sensitizing activity of ethyl acetate fraction of Acorus calamus L. In-vitro and in-vivo. J. Ethnopharmacol. 2009, 123, 288–292. [Google Scholar] [CrossRef]
  108. Liu, Y.X.; Si, M.M.; Lu, W.; Zhang, L.X.; Zhou, C.X.; Deng, S.L.; Wu, H.S. Effects and molecular mechanisms of the antidiabetic fraction of Acorus calamus L. on GLP-1 expression and secretion in-vivo and In-vitro. J. Ethnopharmacol. 2015, 166, 168–175. [Google Scholar] [CrossRef] [PubMed]
  109. Si, M.M.; Lou, J.S.; Zhou, C.X.; Shen, J.N.; Wu, H.H.; Yang, B.; Wu, H.S. Insulin releasing and alpha-glucosidase inhibitory activity of ethyl acetate fraction of Acorus calamus In-vitro and in-vivo. J. Ethnopharmacol. 2010, 128, 154–159. [Google Scholar] [CrossRef] [PubMed]
  110. Parab, R.S.; Mengi, S.A. Hypolipidemic activity of Acorus calamus L. in rats. Fitoterapia 2002, 73, 451–455. [Google Scholar] [CrossRef]
  111. D’Souza, T.; Mengi, S.A.; Hassarajani, S.; Chattopadhayay, S. Efficacy study of the bioactive fraction (F-3) of Acorus calamus in hyperlipidemia. Indian J. Pharmacol. 2007, 39, 196–200. [Google Scholar]
  112. Kumar, G.; Nagaraju, V.; Kulkarni, M.; Kumar, B.S.; Raju, S. Evaluation of Antihyperlipidemic Activity of Methanolic Extract of Acorus Calamus in fat diet Induced Rats. Asian J. Med. Pharm. Sci. 2016, 4, 71–76. [Google Scholar]
  113. Arun, K.S.; Augustine, A. Hypolipidemic Effect of Methanol Fraction of Acorus calamus Linn. in Diet-Induced Obese Rats. In Prospects in Bioscience: Addressing the Issues; Springer, Springer Science & Business Media: New Delhi, India, 2012; pp. 399–404. [Google Scholar]
  114. Athesh, K.; Jothi, G. Pharmacological screening of anti-obesity potential of Acorus calamus linn. In high fat cafeteria diet fed obese rats. Asian J. Pharm. Clin. Res. 2017, 10, 384–390. [Google Scholar]
  115. Patel, P.; Vaghasiya, J.; Thakor, A.; Jariwala, J. Antihypertensive effect of rhizome part of Acorus calamus on renal artery occlusion induced hypertension in rats. Asian Pac. J. Trop. Dis. 2012, 2, S6–S10. [Google Scholar] [CrossRef]
  116. Shah, A.J.; Gilani, A.H. Blood pressure-lowering and vascular modulator effects of Acorus calamus extract are mediated through multiple pathways. J. Cardiovasc. Pharmacol. 2009, 54, 38–46. [Google Scholar] [CrossRef] [Green Version]
  117. Sundaramahalingam, M.; Ramasundaram, S.; Rathinasamy, S.D.; Natarajan, R.P.; Somasundaram, T. Role of Acorus calamus and alpha-asarone on hippocampal dependent memory in noise stress exposed rats. Pak. J. Biol. Sci. 2013, 16, 770–778. [Google Scholar] [CrossRef]
  118. Jain, D.K.; Gupta, S.; Jain, R.; Jain, N. Anti-inflammatory Activity of 80% Ethanolic Extract of Acorus calamus Linn. Leaves in Albino Rats. Res. J. Pharm. Tech. 2010, 3, 882–884. [Google Scholar]
  119. Manikandan, S.; Devi, R.S. Antioxidant property of α-asarone against noise-stress-induced changes in different regions of rat brain. Pharmacol. Res. 2005, 52, 467–474. [Google Scholar] [CrossRef] [PubMed]
  120. Devi, S.A.; Ganjewala, D. Antioxidant activities of methanolic extracts of sweet-flag (Acorus calamus) leaves and rhizomes. J. Herbs Spices Med. Plants 2011, 1, 1–11. [Google Scholar] [CrossRef]
  121. Acuña, U.M.; Atha, D.E.; Ma, J.; Nee, M.H.; Kennelly, E.J. Antioxidant capacities of ten edible North American plants. Phytother. Res. 2002, 16, 63–65. [Google Scholar] [CrossRef] [PubMed]
  122. Palani, S.; Raja, S.; Kumar, R.P.; Parameswaran, P.; Kumar, B.S. Therapeutic efficacy of Acorus calamus on acetaminophen induced nephrotoxicity and oxidative stress in male albino rats. Acta Pharm. Sci. 2010, 52, 89–100. [Google Scholar]
  123. Venkatramaniah, C.; Praba, A.M.A. Effect of Beta Asarone–The Active Principle of Acorus Calamus in Neuroprotection and Nerve Cell Regeneration on the Pyramidal Region of Hippocampus in Mesial Temporal Lobe Epileptic Rat Models. J. Neurosci. 2019, 5, 19–24. [Google Scholar]
  124. Jayaraman, R.; Anitha, T.; Joshi, V.D. Analgesic and anticonvulsant effects of Acorus calamus roots in mice. Int. J. PharmTech Res. 2010, 2, 552–555. [Google Scholar]
  125. Kaushik, R.; Jain, J.; Yadav, R.; Singh, L.; Gupta, D.; Gupta, A. Isolation of β-Asarone from Acorus calamus Linn. and Evaluation of its Anticonvulsant Activity using MES and PTZ Models in Mice. Pharmacol. Toxicol. Biomed. Rep. 2017, 3, 21–26. [Google Scholar] [CrossRef] [Green Version]
  126. Chandrashekar, R.; Adake, P.; Rao, S.N. Anticonvulsant activity of ethanolic extract of Acorus calamus rhizome in swiss albino mice. J. Sci. Innov. Res. 2013, 2, 846–851. [Google Scholar]
  127. Yende, S.R.; Harle, U.N.; Bore, V.V.; Bajaj, A.O.; Shroff, K.K.; Vetal, Y.D. Reversal of neurotoxicity induced cognitive impairment associated with phenytoin and phenobarbital by Acorus calamus in mice. J. Herb. Med. Toxicol. 2009, 3, 111–115. [Google Scholar]
  128. Pawar, V.S.; Anup, A.; Shrikrishna, B.; Shivakumar, H. Antidepressant–like effects of Acorus calamus in forced swimming and tail suspension test in mice. Asian Pac. J. Trop. Biomed. 2011, 1, S17–S19. [Google Scholar] [CrossRef]
  129. Pushpa, V.H.; Padmaja, S.K.; Suresha, R.N.; Vaibhavi, P.S.; Kalabharathi, H.L.; Satish, A.M.; Naidu, S. Antidepressant Activity of Methanolic Extract of Acorus Calamus Leaves in Albino Mice. Int. J. Pharm. Tech. 2013, 5, 5458–5465. [Google Scholar]
  130. Shashikala, G.H.; Prashanth, D.; Jyothi, C.H.; Maniyar, I.; Manjunath, H. Evaluation of antidepressant activity of aqueous extract of roots of acorus calamus in albino mice. World J. Pharm. Res. 2015, 4, 1357–1365. [Google Scholar]
  131. De, A.; Singh, M.S. Acorus calamus linn. Rhizomes extract for antidepressant activity in mice model. Adv. Res. Pharm. Biol. 2013, 3, 520–525. [Google Scholar]
  132. Tripathi, A.K.; Singh, R.H. Experimental evaluation of antidepressant effect of Vacha (Acorus calamus) in animal models of depression. Ayu 2010, 31, 153–158. [Google Scholar] [CrossRef] [PubMed]
  133. Pandy, V.; Jose, N.; Subhash, H. CNS activity of methanol and acetone extracts of Acorus calamus leaves in mice. J. Pharmacol. Toxicol. 2009, 4, 79–86. [Google Scholar] [CrossRef] [Green Version]
  134. Tiwari, N.; Mishra, A.; Bhatt, G.; Chaudhary, A. Isolation of Principle Active Compound of Acorus Calamus. In-vivo assessment of pharmacological activity in the treatment of neurobiological disorder (stress). J. Med. Clin. Res. 2014, 2, 2201–2212. [Google Scholar]
  135. Muthuraman, A.; Singh, N. Neuroprotective effect of saponin rich extract of Acorus calamus L. in rat model of chronic constriction injury (CCI) of sciatic nerve-induced neuropathic pain. J. Ethnopharmacol. 2012, 142, 723–731. [Google Scholar] [CrossRef]
  136. Muthuraman, A.; Singh, N. Attenuating effect of Acorus calamus extract in chronic constriction injury induced neuropathic pain in rats: An evidence of anti-oxidative, anti-inflammatory, neuroprotective and calcium inhibitory effects. BMC Complement. Altern. Med. 2011, 11, 1–14. [Google Scholar] [CrossRef] [Green Version]
  137. Vengadesh Prabu, K.; George, T.; Vinoth Kumar, R.; Nancy, J.; Kalaivani, M.; Vijayapandi, P. Neuromodulatory effect of Acrous calamus leaves extract on dopaminergic system in mice. Int. J. PharmTech Res. 2009, 1, 1255–1259. [Google Scholar]
  138. Hazra, R.; Guha, D. Effect of chronic administration of Acorus calamus on electrical activity and regional monoamine levels in rat brain. Biog. Amines 2003, 17, 161–170. [Google Scholar] [CrossRef]
  139. Shukla, P.K.; Khanna, V.K.; Ali, M.M.; Maurya, R.; Khan, M.Y.; Srimal, R.C. Neuroprotective effect of Acorus calamus against middle cerebral artery occlusion–induced ischaemia in rat. Hum. Exp. Toxicol. 2006, 25, 187–194. [Google Scholar] [CrossRef] [PubMed]
  140. Fathima, A.; Patil, H.V.; Kumar, S. Suppression of elevated reactive oxygen species by acorus calamus (vacha) a sweet flag in drosophila melanogaster under stress full conditions. Int. J. Pharm. Sci. Res. 2014, 5, 1431–1439. [Google Scholar]
  141. Kumar, M.S.; Hiremath, V.S.M.A. Cardioprotective effect of Acorus calamus against doxorubicin-induced myocardial toxicity in albino Wistar rats. Indian J. Health Sci. Biomed. Res. 2016, 9, 225–234. [Google Scholar]
  142. Shah, A.J.; Gilani, A.H. Bronchodilatory effect of Acorus calamus (Linn.) is mediated through multiple pathways. J. Ethnopharmacol. 2010, 131, 471–477. [Google Scholar] [CrossRef] [PubMed]
  143. Thakare, M.M.; Surana, S.J. β-Asarone modulate adipokines and attenuates high fat diet-induced metabolic abnormalities in Wistar rats. Pharmacol. Res. 2016, 103, 227–235. [Google Scholar] [CrossRef] [PubMed]
  144. Karthiga, T.; Venkatalakshmi, P.; Vadivel, V.; Brindha, P. In-vitro anti-obesity, antioxidant and anti-inflammatory studies on the selected medicinal plants. Int. J. Toxicol. Pharmacol. Res. 2016, 8, 332–340. [Google Scholar]
  145. Singh, D.K.; Kumar, N.; Sachan, A.; Lakhani, P.; Tutu, S.; Shankar, P.; Dixit, R.K. An experimental study to see the antihypertensive effects of gymnema sylvestre and acorus calamus in wistar rats and its comparison with amlodipine. Asian J. Med. Sci. 2017, 8, 11–15. [Google Scholar] [CrossRef] [Green Version]
  146. Tanaka, S.; Yoichi, S.; Ao, L.; Matumoto, M.; Morimoto, K.; Akimoto, N.; Zaini bin Asmawi, M. Potential immunosuppressive and anti-inflammatory activities of Malaysian medicinal plants characterized by reduced cell surface expression of cell adhesion molecules. Phytother. Res. 2001, 15, 681–686. [Google Scholar] [CrossRef]
  147. Kim, H.; Han, T.H.; Lee, S.G. Anti-inflammatory activity of a water extract of Acorus calamus L. leaves on keratinocyte HaCaT cells. J. Ethnopharmacol. 2009, 122, 149–156. [Google Scholar] [CrossRef]
  148. Ahmed, S.; Gul, S.; Zia-Ul-Haq, M.; Stanković, M.S. Pharmacological basis of the use of Acorus calamus L. in inflammatory diseases and underlying signal transduction pathways. Bol. Latinoam. Caribe Plantas Med. Aromát. 2014, 13, 38–46. [Google Scholar]
  149. Loying, R.; Gogoi, R.; Sarma, N.; Borah, A.; Munda, S.; Pandey, S.K.; Lal, M. Chemical Compositions, In-vitro Antioxidant, Anti-microbial, Anti-inflammatory and Cytotoxic Activities of Essential Oil of Acorus calamus L. Rhizome from North-East India. J. Essent. Oil Bear. Plants 2019, 22, 1299–1312. [Google Scholar] [CrossRef]
  150. Bahukhandi, A.; Rawat, S.; Bhatt, I.D.; Rawal, R.S. Influence of solvent types and source of collection on total phenolic content and antioxidant activities of Acorus calamus L. Natl. Acad. Sci. Lett. 2013, 36, 93–99. [Google Scholar] [CrossRef]
  151. Manju, S.; Chandran, R.P.; Shaji, P.K.; Nair, G.A. In-vitro free radical scavenging potential of Acorus Calamus L. rhizome from Kuttanad Wetlands, Kerala, India. Int. J. Pharm. Pharm. Sci. 2013, 5, 376–380. [Google Scholar]
  152. Barua, C.C.; Sen, S.; Das, A.S.; Talukdar, A.; Hazarika, N.J.; Barua, A.G.; Barua, I. A comparative study of the In-vitro antioxidant property of different extracts of Acorus calamus Linn. J. Nat. Prod. Plant Resour. 2014, 4, 8–18. [Google Scholar]
  153. Elayaraja, A.; Vijayalakshmi, M.; Devalarao, G. In-vitro free radical scavenging activity of various root and rhizome extracts of Acorus calamus Linn. Int. J. Pharm. Biol. Sci. 2010, 1, 301–304. [Google Scholar]
  154. Govindarajan, R.; Agnihotri, A.K.; Khatoon, S.; Rawat, A.K.S.; Mehrotra, S. Pharmacognostical evaluation of an antioxidant plant-Acorus calamus Linn. Nat. Prod. Sci. 2003, 9, 264–269. [Google Scholar]
  155. Sujitha, R.; Bhimba, B.V.; Sindhu, M.S.; Arumugham, P. Phytochemical Evaluation and Antioxidant Activity of Nelumbo nucifera, Acorus calamus and Piper longum. Int. J. Pharm. Chem. Sci. 2013, 2, 1573–1578. [Google Scholar]
  156. Shukla, R.; Singh, P.; Prakash, B.; Dubey, N.K. Efficacy of Acorus calamus L. essential oil as a safe plant-based antioxidant, A flatoxin B 1 suppressor and broad-spectrum antimicrobial against food-infesting fungi. Int. J. Food Sci. Tech. 2013, 48, 128–135. [Google Scholar] [CrossRef]
  157. Ahmeda, F.; Urooja, A.; KS, R. In-vitro antioxidant and anticholinesterase activity of Acorus calamus and Nardostachys jatamansi rhizomes. J. Pharm. Res. 2009, 2, 830–833. [Google Scholar]
  158. Bhat, S.D.; Ashok, B.K.; Acharya, R.N.; Ravishankar, B. Anticonvulsant activity of raw and classically processed Vacha (Acorus calamus Linn.) rhizomes. Ayu 2012, 33, 119–122. [Google Scholar] [CrossRef]
  159. Patel, S.; Rajshree, N.; Shah, P. Evaluation of antidepressant activity of herbomineral formulation. Int. J. Pharm. Pharm. Sci. 2016, 8, 145–147. [Google Scholar]
  160. Rauniar, G.P.; Deo, S.; Bhattacharya, S.K. Evaluation of anxiolytic activity of tensarin in mice. Kathman. Univ. Med. J. 2007, 5, 188–194. [Google Scholar]
  161. Naderi, G.A.; Khalili, M.; Karimi, M.; Soltani, M. The effect of oral and intraperitoneal administration of Acorus calamus L. extract on learning and memory in male rats. J. Med. Plant 2010, 2, 46–56. [Google Scholar]
  162. Vohora, S.B.; Shah, S.A.; Dandiya, P.C. Central nervous system studies on an ethanol extract of Acorus calamus rhizomes. J. Ethnopharmacol. 1990, 28, 53–62. [Google Scholar] [CrossRef]
  163. Singh, B.K.; Pillai, K.K.; Kohli, K.; Haque, S.E. Isoproterenol-Induced Cardiomyopathy in Rats: Influence of Acorus calamus Linn. Cardiovasc. Toxicol. 2011, 11, 263–271. [Google Scholar] [CrossRef] [PubMed]
  164. Shah, A.J.; Gilani, A.H. Aqueous-methanolic extract of sweet flag (Acorus calamus) possesses cardiac depressant and endothelial-derived hyperpolarizing factor-mediated coronary vasodilator effects. J. Nat. Med. 2012, 66, 119–126. [Google Scholar] [CrossRef] [Green Version]
  165. Hasheminejad, G.; Caldwell, J. Genotoxicity of the alkenylbenzenes α− and β-asarone, myristicin and elemicin as determined by the UDS assay in cultured rat hepatocytes. Food Chem. Toxicol. 1994, 32, 223–231. [Google Scholar] [CrossRef]
  166. Cartus, A.T.; Schrenk, D. Metabolism of the carcinogen alpha-asarone in liver microsomes. Food Chem. Toxicol. 2016, 87, 103–112. [Google Scholar] [CrossRef]
  167. Cartus, A.T.; Stegmuller, S.; Simson, N.; Wahl, A.; Neef, S.; Kelm, H.; Schrenk, D. Hepatic metabolism of carcinogenic betaasarone. Chem. Res. Toxicol. 2015, 28, 1760–1773. [Google Scholar] [CrossRef]
  168. Cartus, A.T.; Schrenk, D. Metabolism of carcinogenic alpha-asarone by human cytochrome P450 enzymes. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 213–223. [Google Scholar] [CrossRef]
  169. Pandit, S.; Mukherjee, P.K.; Ponnusankar, S.; Venkatesh, M.; Srikanth, N. Metabolism mediated interaction of α-asarone and Acorus calamus with CYP3A4 and CYP2D6. Fitoterapia 2011, 82, 369–374. [Google Scholar] [CrossRef] [PubMed]
  170. Muthuraman, A.; Singh, N. Acute and sub-acute oral toxicity profile of Acorus calamus (Sweet flag) in rodents. Asian Pac. J. Trop Biomed. 2012, 2, S1017–S1023. [Google Scholar] [CrossRef]
  171. Areekul, S.; Sinchaisri, P.; Tigvatananon, S. Effects of Thai plant extracts on the oriental fruit fly III. Nat. Sci. 1988, 22, 160–164. [Google Scholar]
  172. Shah, P.D.; Ghag, M.; Deshmukh, P.B.; Kulkarni, Y.; Joshi, S.V.; Vyas, B.A.; Shah, D.R. Toxicity study of ethanolic extract of Acorus calamus rhizome. Int. J. Green Pharm. 2012, 6, 29–35. [Google Scholar] [CrossRef]
  173. Bhat, S.D.; Ashok, B.K.; Acharya, R.; Ravishankar, B. A comparative acute toxicity evaluation of raw and classically processed rhizomes of Vacha (Acorus calamus Linn.). Indian J. Nat. Prod. Resour. 2012, 3, 506–511. [Google Scholar]
  174. Keller, K.; Stahl, E. Composition of the essential oil from beta-Asarone free calamus. Planta Med. 1983, 47, 71–74. [Google Scholar] [CrossRef]
  175. JECFA (Joint FAO/WHO Expert Committee on Food Additives). Monograph on -asarone. In WHO Food Additive Series No. 16; WHO Food Additives Series; JECFA, WHO Press: Geneva, Switzerland, 1981. [Google Scholar]
  176. Opdyke, D.L.J. Monographs on fragrance raw materials. Food Cosmet. Toxicol. 1973, 11, 855–876. [Google Scholar] [CrossRef]
  177. Jenner, P.M.; Hagan, E.C.; Taylor, J.M.; Cook, E.L.; Fitzhugh, O.G. Food flavourings and compounds of related structure I. Acute oral toxicity. Food Cosmet. Toxicol. 1964, 2, 327–343. [Google Scholar] [CrossRef]
  178. Singh, A.K.; Ravishankar, B.; Sharma, P.P.; Pandaya, T. Clinical study of anti-hyperlipidaemic activity of vacha (Acorus calamus linn) w.s.r to sthaulya. Int. Ayurvedic Med. J. 2017, 5, 1–8. [Google Scholar]
  179. Tajadini, H.; Saifadini, R.; Choopani, R.; Mehrabani, M.; Kamalinejad, M.; Haghdoost, A.A. Herbal medicine Davaie Loban in mild to moderate Alzheimer’s disease: A 12-week randomized double-blind placebo-controlled clinical trial. Complement. Ther. Med. 2015, 23, 767–772. [Google Scholar] [CrossRef]
  180. Bhattacharyya, D.; Sur, T.K.; Lyle, N.; Jana, U.; Debnath, P.K. A clinical study on the management of generalized anxiety disorder with Vaca (Acorus calamus). Indian J. Tradit. Knowl. 2011, 10, 668–671. [Google Scholar]
  181. Soni1, P.; Sharma, C. A clinical study of Vachadi Churna in the management of obesity. Int. J. Ayurveda Allied Sci. 2012, 1, 179–186. [Google Scholar]
  182. Kulatunga, R.D.H.; Dave, A.R.; Baghel, M.S. Clinical efficacy of Guduchyadi Medhya Rasayana on senile memory impairment. Ayu 2012, 33, 202–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Pande, D.N.; Mishra, S.K. Vacha (Acorus Calamus) as an ayurvedic premedicant. Ayu 2009, 30, 279–283. [Google Scholar]
  184. Mishra, J.; Joshi, N.P.; Pandya, D.M. A comparative study of Shankhapushpyadi Ghana Vati and Sarpagandhadi Ghana Vati in the management of “Essential Hypertension”. Ayu 2012, 33, 54–61. [Google Scholar] [CrossRef] [Green Version]
  185. Ramu, M.G.; Senapati, H.M.; Janakiramaiah, N.; Shankara, M.R.; Chaturvedi, D.D.; Murthy, N.N. A pilot study of role of brahmyadiyoga in chronic unmada (schizophrenia). Anc. Sci. Life 1983, 2, 205–207. [Google Scholar]
  186. Appaji, R.R.; Sharma, R.D.; Katiyar, G.P.; Sai, P.A. Clinical study of the Immunoglobululin Enhancing Effect of “Bala compound” on Infants. Anc. Sci. Life 2009, 28, 18–22. [Google Scholar]
  187. Pawar, M.; Magdum, P. Clinical study of assessment of therapeutic potential of Vachadi ghrita, a medicated ghee formulation on healthy individual’s cognition. Int. J. Pharm. Sci. Res. 2018, 9, 3408–3413. [Google Scholar]
  188. Mishra, D.; Tubaki, B.R. Effect of Brahmi vati and Sarpagandha Ghana vati in management of essential hypertension–A randomized, double blind, clinical study. J. Ayurveda Integr. Med. 2019, 10, 269–276. [Google Scholar] [CrossRef]
  189. Sharma, Y.; Upadhyay, A.; Sharma, Y.K.; Chaudhary, V. A randomized clinical study to evaluate the effect of Tagaradi yoga in the management of insomnia. Indian J. Tradit. Knowl. 2017, 16, S75–S80. [Google Scholar]
  190. Paradkar, S.R.; Pardhi, S.N. Clinical evaluation of lekhaniya effect of vacha (acorus calamus) and musta (cyperus rotundus) in medoroga wsr to obesity: A comparative study. Res. Rev. J. Pharmacogn. 2019, 3, 1–8. [Google Scholar]
  191. Mamgain, P.; Singh, R.H. Control clinical trial of the lekhaniya drug vaca (Acorus calamus) in case of ischemic heart diseases. J. Res. Ayurveda Siddha 1994, 15, 35–51. [Google Scholar]
  192. 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, 1159–1166. [Google Scholar] [CrossRef]
  193. 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, 2097–2101. [Google Scholar] [CrossRef] [PubMed]
  194. 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 M adopar. Clin. Exp. Pharmacol. Physiol. 2014, 41, 685–690. [Google Scholar] [PubMed]
  195. 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, 634–643. [Google Scholar] [CrossRef]
  196. 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, 20658. [Google Scholar]
  197. Liu, S.J.; Yang, C.; Zhang, Y.; Su, R.Y.; Chen, J.L.; Jiao, M.M.; Quan, S.J. Neuroprotective effect of β-asarone against Alzheimer’s disease: Regulation of synaptic plasticity by increased expression of SYP and GluR1. Drug Des. Dev. Ther. 2016, 10, 1461. [Google Scholar] [CrossRef] [Green Version]
  198. Li, C.; Xing, G.; Dong, M.; Zhou, L.; Li, J.; Wang, G.; Niu, Y. 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, 96–102. [Google Scholar] [CrossRef]
  199. Xue, Z.; Guo, Y.; Zhang, S.; Huang, L.; He, Y.; Fang, R.; Fang, Y. Beta-asarone attenuates amyloid beta-induced autophagy via Akt/mTOR pathway in PC12 cells. Eur. J. Pharmacol. 2014, 741, 195–204. [Google Scholar] [CrossRef]
  200. Yang, Q.Q.; Xue, W.Z.; Zou, R.X.; Xu, Y.; Du, Y.; Wang, S.; Chen, X.T. β-Asarone rescues Pb-induced impairments of spatial memory and synaptogenesis in rats. PLoS ONE 2016, 11, e0167401. [Google Scholar] [CrossRef] [PubMed]
  201. Guo, J.H.; Chen, Y.; Wei, G.; Nei, H.; Zhou, Y.; Cheng, S. Effects of active components of Rhizoma Acori Tatarinowii and their compatibility at different ratios on learning and memory abilities in dementia mice. Tradit. Chin. Drug Res. Clin. Pharmacol. 2012, 23, 144–147. [Google Scholar]
  202. Li, J.; Li, Z.X.; Zhao, J.P.; Wang, W.; Zhao, X.F.; Xu, B.; Li, S.X. A Novel Tropoloisoquinoline Alkaloid, Neotatarine, from Acorus calamus L. Chem. Biodivers. 2017, 14, e1700201. [Google Scholar] [CrossRef] [PubMed]
  203. He, X.; Cai, Q.; Li, J.; Guo, W. Involvement of brain-gut axis in treatment of cerebral infarction by β-asaron and paeonol. Neurosci. Lett. 2018, 666, 78–84. [Google Scholar] [CrossRef]
  204. Gao, E.; Zhou, Z.Q.; Zou, J.; Yu, Y.; Feng, X.L.; Chen, G.D.; Gao, H. Bioactive Asarone-derived phenylpropanoids from the rhizome of Acorus tatarinowii Schott. J. Nat. Prod. 2017, 80, 2923–2929. [Google Scholar] [CrossRef]
  205. Zhang, S.; Gui, X.H.; Huang, L.P.; Deng, M.Z.; Fang, R.M.; Ke, X.H.; Fang, Y.Q. Neuroprotective effects of β-asarone against 6-hydroxy dopamine-induced parkinsonism via JNK/Bcl-2/Beclin-1 pathway. Mol. Neurobiol. 2016, 53, 83–94. [Google Scholar] [CrossRef]
  206. Liang, S.; Ying, S.S.; Wu, H.H.; Liu, Y.T.; Dong, P.Z.; Zhu, Y.; Xu, Y.T. A novel sesquiterpene and three new phenolic compounds from the rhizomes of Acorus tatarinowii Schott. Bioorg. Med. Chem. Lett. 2015, 25, 4214–4218. [Google Scholar] [CrossRef]
  207. Xu, F.; Wu, H.; Zhang, K.; Lv, P.; Zheng, L.; Zhao, J. Pro-neurogenic effect of β-asarone on RSC96 Schwann cells in vitro. In Vitro Cell. Dev. Biol. Anim. 2016, 52, 278–286. [Google Scholar] [CrossRef]
  208. Deng, M.; Huang, L.; Ning, B.; Wang, N.; Zhang, Q.; Zhu, C.; Fang, Y. 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, 188–194. [Google Scholar] [CrossRef]
  209. Yang, Y.; Xuan, L.; Chen, H.; Dai, S.; Ji, L.; Bao, Y.; Li, C. Neuroprotective Effects and Mechanism of β-Asarone against Aβ1–42-Induced Injury in Astrocytes. Evid.-Based Complement. Altern. Med. 2017, 2017, 8516518. [Google Scholar] [CrossRef] [Green Version]
  210. 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, 5634–5649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Liu, H.; Song, Z.; Liao, D.G.; Zhang, T.Y.; Liu, F.; Zhuang, K.; Lei, J.P. Anticonvulsant and sedative effects of eudesmin isolated from Acorus tatarinowii on mice and rats. Phytother. Res. 2015, 29, 996–1003. [Google Scholar] [CrossRef] [PubMed]
  213. Tian, J.; Tian, Z.; Qin, S.L.; Zhao, P.Y.; Jiang, X. Anxiolytic-like effects of α-asarone in a mouse model of chronic pain. Metab. Brain Dis. 2017, 32, 2119–2129. [Google Scholar] [CrossRef] [PubMed]
  214. Miao, J.K.; Chen, Q.X.; Li, C.; Li, X.W.; Wu, X.M.; Zhang, X.P. Modulation Effects of α-Asarone on the GABA homeostasis in the Lithium-Pilocarpine Model of Temporal Lobe Epilepsy. Pharmacology 2013, 9, 24–32. [Google Scholar]
  215. Wang, Z.J.; Levinson, S.R.; 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. [Google Scholar] [CrossRef] [Green Version]
  216. Chen, L.; Liao, W.P. Changes of amino acid content in hippocampus of epileptic rats treated with volatile oil of Acorus tatarinowii. Zhongguo ZhongYao ZaZhi 2004, 29, 670–673. [Google Scholar]
  217. Jo, M.J.; Kumar, H.; Joshi, H.P.; Choi, H.; Ko, W.K.; Kim, J.M.; Kim, K.T. Oral administration of α-Asarone promotes functional recovery in rats with spinal cord injury. Front. Pharmacol. 2018, 9, 445. [Google Scholar] [CrossRef] [Green Version]
  218. Lam, K.Y.; Yao, P.; Wang, H.; Duan, R.; Dong, T.T.; Tsim, K.W. Asarone from Acori Tatarinowii Rhizome prevents oxidative stress-induced cell injury in cultured astrocytes: A signaling triggered by Akt activation. PLoS ONE 2017, 12, e0179077. [Google Scholar] [CrossRef] [Green Version]
  219. Wu, Q.D.; Yuan, D.J.; Wang, Q.W.; Wu, X.R. Effects of volatile oil of Rhizoma Acori Tatarinowii on morphology and cell viability in cultured cardiac myocytes. Zhong Yao Cai 2009, 32, 242–245. [Google Scholar]
  220. Yong, H.Y.F.Y.J.; Shuying, L.Y.W. In-vitro Observation of β-asarone for Counteracting Arteriosclerosis. J. Guangzhou Univ. Tradit. Chin. Med. 2008, 3, 14. [Google Scholar]
  221. Yang, Y.X.; Chen, Y.T.; Zhou, X.J.; Hong, C.L.; Li, C.Y.; Guo, J.Y. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Das, B.K.; Choukimath, S.M.; Gadad, P.C. Asarone and metformin delays experimentally induced hepatocellular carcinoma in diabetic milieu. Life Sci. 2019, 230, 10–18. [Google Scholar] [CrossRef] [PubMed]
  223. Lee, S.H.; Kim, K.Y.; Ryu, S.Y.; Yoon, Y.O.O.S.I.K.; Hahm, D.H.; Kang, S.A.; Lee, H.G. Asarone inhibits adipogenesis and stimulates lipolysis in 3T3-L1 adipocytes. Cell. Mol. Biol. 2010, 56, 1215–1222. [Google Scholar]
Figure 1. Pathophysiology of insulin resistance, metabolic malfunction, and progression to a neurological disorder. TNF, tumor necrosis factor; IL, interleukin.
Figure 1. Pathophysiology of insulin resistance, metabolic malfunction, and progression to a neurological disorder. TNF, tumor necrosis factor; IL, interleukin.
Jcm 09 01176 g001
Figure 2. Photographs of Acorus calamus: (A) Natural habitat; (B) Fresh rhizome; (C) Dried rhizome.
Figure 2. Photographs of Acorus calamus: (A) Natural habitat; (B) Fresh rhizome; (C) Dried rhizome.
Jcm 09 01176 g002
Figure 3. Flowchart of the selection process.
Figure 3. Flowchart of the selection process.
Jcm 09 01176 g003
Figure 4. Distribution of A. calamus worldwide and in India.
Figure 4. Distribution of A. calamus worldwide and in India.
Jcm 09 01176 g004
Figure 5. Chemical structures of isolated compounds from A. calamus.
Figure 5. Chemical structures of isolated compounds from A. calamus.
Jcm 09 01176 g005aJcm 09 01176 g005bJcm 09 01176 g005c
Figure 6. Illustration of role of A. calamus mechanisms in the treatment of neurological and metabolic disorders. AChE, acetylcholinesterase; APP, amyloid precursor protein; Bcl-2, B-cell lymphoma 2; CHOP, C/EBP homologous protein; CCAAT (cytosine-cytosine-adenosine-adenosine-thymidine)-enhancer-binding protein homologous protein; C/EBP, CCAAT enhancer-binding protein; GABAA, γ-Aminobutyric acid type A; GRP78, 78-kDa glucose-regulated protein; HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A; iNOS, inducible nitric oxide synthase; JNK, c-Jun NH2-terminal kinase; LC3b, microtubule-associated proteins 1A/1B light chain 3B; MCP, modified citrus pectin; MDA, malondialdehyde; MIP, macrophage inflammatory protein; p-PERK, phospho-protein kinase RNA-like ER kinase; PPARγ, peroxisome proliferator-activated receptor gamma; ERK1/2, extracellular signal-regulated protein kinase.
Figure 6. Illustration of role of A. calamus mechanisms in the treatment of neurological and metabolic disorders. AChE, acetylcholinesterase; APP, amyloid precursor protein; Bcl-2, B-cell lymphoma 2; CHOP, C/EBP homologous protein; CCAAT (cytosine-cytosine-adenosine-adenosine-thymidine)-enhancer-binding protein homologous protein; C/EBP, CCAAT enhancer-binding protein; GABAA, γ-Aminobutyric acid type A; GRP78, 78-kDa glucose-regulated protein; HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A; iNOS, inducible nitric oxide synthase; JNK, c-Jun NH2-terminal kinase; LC3b, microtubule-associated proteins 1A/1B light chain 3B; MCP, modified citrus pectin; MDA, malondialdehyde; MIP, macrophage inflammatory protein; p-PERK, phospho-protein kinase RNA-like ER kinase; PPARγ, peroxisome proliferator-activated receptor gamma; ERK1/2, extracellular signal-regulated protein kinase.
Jcm 09 01176 g006
Figure 7. The role of the Nrf-2, NF-κB, PI3K/AKT, Ras/MAPK, and PPARγ signaling pathways as affected by phytoconstituents of Acorus calamus to upregulate antioxidant, neuroprotective, detoxifying enzymes and suppress inflammation. Ub, ubiquitin; NEMO, NF-kB essential modulator; ARE, antioxidant response element; Maf, musculoaponeurotic fibrosarcoma oncogene homolog; NLS, nuclear localization signal; CAT, catalase; GPX, glutathione peroxidase; Trk, tyrosine kinase receptor; LPS, lipopolysaccharide; TLRs, toll-like receptors; PI3K, phosphatidylinsoitol-3-kinase; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycin; ERK, extracellular signal-regulated kinases; Nrf2, nuclear factor e2-related factor 2; Keap-1, kelch-like ECH-associated protein-1; MEK, mitogen-activated protein kinase; JNK, c-Jun N-terminal kinase;NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor-kappa B; IkB, inhibitor of kB; IKK, inhibitor of kB kinases.
Figure 7. The role of the Nrf-2, NF-κB, PI3K/AKT, Ras/MAPK, and PPARγ signaling pathways as affected by phytoconstituents of Acorus calamus to upregulate antioxidant, neuroprotective, detoxifying enzymes and suppress inflammation. Ub, ubiquitin; NEMO, NF-kB essential modulator; ARE, antioxidant response element; Maf, musculoaponeurotic fibrosarcoma oncogene homolog; NLS, nuclear localization signal; CAT, catalase; GPX, glutathione peroxidase; Trk, tyrosine kinase receptor; LPS, lipopolysaccharide; TLRs, toll-like receptors; PI3K, phosphatidylinsoitol-3-kinase; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycin; ERK, extracellular signal-regulated kinases; Nrf2, nuclear factor e2-related factor 2; Keap-1, kelch-like ECH-associated protein-1; MEK, mitogen-activated protein kinase; JNK, c-Jun N-terminal kinase;NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor-kappa B; IkB, inhibitor of kB; IKK, inhibitor of kB kinases.
Jcm 09 01176 g007
Table 1. Ethnomedicinal use of A. calamus in various countries.
Table 1. Ethnomedicinal use of A. calamus in various countries.
CountryAilment/UsePart Used/Dosage FormRoute of AdministrationReferences
IndiaEczemaThe paste of A. calamus rhizomes are given with the paste of Curcuma aromatica rhizomes and Azadirachta indica leavesOral[13]
Skin diseasesRhizomes paste A. calamus and C. aromatica are applied with the seed paste of Argemone Mexicana
Cough, stuttering, ulcer, fever, dermatitis, scab, soresRhizomes[14]
Cold, cough, and feverRhizomes paste of A. calamus is given to children with mother’s milk, Myristica fragrance, and Calunarejan spinosa fruits[15]
Two teaspoonfuls of herbal powder containing A. calamus rhizomes, Boerhaavia diffusa roots, Calonyction muricutum flower pedicles, Ipomoea muricate seeds, Senna leaves, Cassia fistula fruits pulp, Curcuma longa rhizomes, Helicteres isora fruits, and Mentha arvensis leaves, black pepper is taken with lukewarm water[16]
Gastric disordersA. calamus rhizomes paste is given with cow milk[17]
Carminative, flavoring, tonic, and head lice infestationInfusion of a dried rhizomes (collected and stored in the autumn season)[17,18,19]
Epilepsy, dysentery, mental illnesses, diarrhea, kidney and liver disordersA. calamus rhizomes paste is given with honey[20]
Wounds, fever, body painRhizomes[21,22]
DysenteryFresh ground rhizomes is mixed with hot water and given for 3 days[23]
StimulantDry powder of A. calamus is given with honey[24]
InjuriesExternal application of the A. calamus rhizomes pasteDermal[25]
StomachacheAsh of the A. calamus rhizomes paste[26]
Otitis externaA. calamus roots paste is given with coconut husk juice[27]
LotionFresh leaves of A. calamus[28]
Cough, cancer, and feverA. calamus roots juice is given with honey and MyristicaDactyloidesOral[29]
AnalgesicA. calamus rhizomes are given with cinchona bark[30]
Gastrointestinal, respiratory, emmenagogue, antihelminticRhizomes
Prolonged laborRhizomes is applied with saffron and horse milk
Paralysis, arthritisRhizomes ash is applied with castor oil
Neurological disorder, gastrointestinal, respiratory, increases menstrual flow, analgesic, contraceptiveRhizomesOral[31,32,33]
Herpangina, analgesic, neurological disorder, gastrointestinal, respiratory[34]
PakistanColic and diarrheaWhole plant[35]
NepalBlood pressureRoots infusion of A. calamus[36]
Cough, headache, snake bite, sore throat, and painRhizomes[37]
DysenteryRhizomes juice is given with hot water
Neurological, respiratoryRhizomes[38]
MalaysiaRheumatism, diarrhea, dyspepsia, and hair lossWhole plant[39]
TibetFever, gastrointestinalDried rhizomes is given with Saussurea lappa, Ferula foetida, Terminalia chebula, Cuminum cyminum, Inula racemosa, and Zingiber officinale[40]
CancerRhizomes[41]
ChinaGastrointestinal, respiratory, neuroprotective, analgesic, contraceptive, cancerRhizomes[42,43,44]
Antipyretic and ear-related diseaseRhizomes given with squeezed Coccinia cordifolia stems along with water[45]
DetoxificationRhizomes with vinegar, Alpinia galanga, Zingiber purpureum
AnalgesicHerbal baths of the rhizomeExternal
HemorrhageRhizomes paste[46]
AphrodisiacRhizomesOral[47]
HallucinationRhizomesare mixed with Indian hemp and Podophyllum pleianthum[48]
Fair skinLeaves of A. calamus are given with Artemisia vulgarisDermal[49]
IndonesiaGastrointestinalRhizomesOral[50]
EnglandRhizomes blended with chalk and magnesium oxide[51]
Gastrointestinal, antibacterial, analgesicRhizomes[52]
Neurological, dysentery, and chronic catarrhRhizomesare given with Gentiana campestris L.
MalariaRhizomes[53]
EuropeObesity, influenza, gastrointestinal, respiratory[54,55]
Republic of South AfricaTooth powder, gastrointestinal, tonic, aphrodisiac[56]
SwedenLiquor[57]
GermanyIncreases menstrual flow, gastrointestinal[58,59]
JavaLactation[60]
LithuaniaChest pain, diarrheaRhizomes and leaves are taken with sugar[52]
Relieves pain, gout, rheumatismLeaves decoctionExternal[61]
New GuineaMiscarriageRhizomesOral[62]
PhilippinesGastrointestinal, rheumatism[56]
RussiaTyphoid, syphilis, baldness, fever, cholera[63]
ThailandBlood purifier, fever[64]
TurkeyWound healing, cough, tuberculosisExternal and oral[61]
GastrointestinalOral[65,66]
Arab countriesGastrointestinal, tuberculosis[67,68]
BrazilDestroys parasitic worms[68]
ArgentinaDysmenorrhea[69]
United StatesGastrointestinal, abortifacient, stimulant, tonic, respiratory disorderRhizomes[70]
KoreaImproves memory and life span[71]
Sri LankaCough, worm infestationRhizomes paste are given with milk[72]
Table 2. Pharmaceutical products of A. calamus available in the market.
Table 2. Pharmaceutical products of A. calamus available in the market.
Medicine/FormulationsIndications/UseManufacturers
Pilochek tabletsHemorrhoidsDabur India Limited
Brahm RasayanNervine tonic
Mahasudarsan ChurnaMalaria
Janma Ghunti HoneyBabies growth, Constipation, Diarrhea
Brahmi Pearls capsulesBrain NourisherKerala Ayurveda
GT capsulesOsteoarthritis, osteoporosis, hyperlipidemia
Histantin tabletsAnti-allergic
Santhwanam oilAntioxidant, rejuvenate
Mahathikthaka Ghrita capsulesSkin disease, malabsorption syndrome
Calamus root tinctureStimulates the digestive systemFlorida Herbal Pharmacy
Vacha capsulesFood supplementsDR Wakde’s Natural Health Care, London
Mentat tablets and syrupNervine tonicHimalaya Herbal Healthcare
AbanaCardiovascular disorders, hyperlipidemia, dyslipidemia
Mentat tablets and SyrupAnxiety, depression, insomnia
Muscle & Joint RubBackaches, muscular sprains, pain
AnxocareAnxiety
Erina-EPEctoparasites
Himpyrin, Himpyrin VetAnalgesic and anti-inflammatory
Scavon VetAnti-bacterial, anti-fungal
Vacha powderBrain tonic, improves digestion, and prevents nauseaBixa Botanical
AmalthHerbal supplementsMcnow Biocare Private Limited
Sunarin capsulesAnal fissures, piles, rectal inflammation, congestionSG Phyto Pharma
Dr Willmar Schwabe India Acorus calamus mother tinctureIntestinal worms and stomach disorders, fever, nauseaDr Willmar Schwabe India Pvt Ltd.
Himalayan calamus root essential oilPain relief and calm mindNaturalis Essence of Nature
Calamus oilBody, skin care, hair growthKazima Perfumers
Calamus root powderMental health problemsHeilen Biopharm
Winton tablets and syrupReduce tension, stress, and anxietyScortis Healthcare
Chesol syrupMuscular aches and pains, chest colds, and bronchitisJ & J Dechane Laboratories Private Limited
Enzo FastAcidity, gastritis, flatulence, indigestionNaturava
Dark Forest Vekhand powderAbdomen pain, worms (infants)Simandhar Herbal Pvt. Ltd.
NervocareInsomniaDeep Ayurveda
Antress tabletsAnxiety and stress disordersAyursun Pharma
Grapzone syrupMental wellnessAlna Biotech Pvt Ltd.
Memoctive syrupImproves memory powerAayursh Herbal India
Smrutihills capsulesStress, anxiety, adaptogenicAyush Arogyam
Gastrin capsulesGastritis, dyspepsiaSarvana Marundhagam
Pigmento tabletsLeukoderma or vitiligoCharak Pharma
Paedritone dropsDigestive functions
Vacha ChurnaBrain tonic, digestion, nauseaSadvaidyasala
Alert capsulesImmunomodulator, anxietyVasu Healthcare
Brento tabletsIncreasing cognitive functionsZandu Realty Limited
Livotrit ForteHepatitis, jaundice
ZanduzymeIndigestion and dyspepsia
Vedic SlimAnti-obesityVedic Bio-Labs Pvt. Ltd.
Hinguvachaadi GulikaAnorexia, indigestion, appetite lossNagarajuna Pvt. Ltd.
Nilsin capsulesSinusitis and allergic rhinitisPhytomarketing
Norbeepee tabletHypertensionAVN Formulations
Sooktyn tabletAntacid, antispasmodicAlarsin Pharma Pvt. Ltd.
Deonac oilPain reliving oilDoux Healthcare Pvt. Ltd.
Smrutisagar RasaMemory enhancerShree Dhootpapeshwar Limited
Yogaraj GuggulVitiligo, anorexia, indigestion, loss of appetite
Kankayan BatiGastritis, flatulence, dyspepsiaBaidyanath Pvt. Ltd.
Brahmi GhritaInsanity and memory issues
Fat GoControls high cholesterol levelJolly Healthcare
Divya Medha VatiImproves memory powerPatanjali Ayurveda
Divya Mukta VatiHigh blood pressure
Table 3. Chemical compounds isolated from different botanical parts of A. calamus.
Table 3. Chemical compounds isolated from different botanical parts of A. calamus.
ClassificationCompound No.Chemical IngredientMethods of CharacterizationParts/ExtractReferences
Phenylpropanoids1 α-AsaroneGC-FID, GC-MSRhizomes/n-hexane, aqueous, methanol, ethanol[74,78,84,89,90,91]
2 β-Asarone
3 γ-Asarone
4 Eugenyl acetateGC-MSRhizomes/aqueous extract[74,78,91]
5 Eugenol
6 Isoeugenol
7 Methyl eugenolRhizomes/n-hexane,
ethyl acetate
[92]
8 Methyl isoeugenolRhizomes/hexane[74,78,91,94]
9 CalamolRhizomes/aqueous extract[74,78,91]
10 Azulene
11 Eugenol methyl ether
12 Dipentene
13 Asaronaldehyde
14 Terpinolene
15 1,8-cineole
16 (E)-isoeugenol acetateGC-FID, GC-MS[89]
17 (E)-methyl isoeugenol
18 Cis-methyl isoeugenolRhizomes/n-hexane, ethyl acetate[92]
19 Euasarone
20 Cinnamaldehyde
21 CyclohexanoneGC-MSRhizomes/hexane[94]
22 AcorinNMRRhizomes/chloroform[95]
23 Isoasarone
24 Safrole
25 Z-3-(2,4,5-trimethoxyphenyl)-2-propenalFTIR, NMRRhizomes/ethanol[96]
26 2,3-dihydro-4,5,7-trimethoxy-1-ethyl-2-methyl-3 (2,4,5-trimethoxyphenyl) indene
27 (Z)-asaroneGC-MSLeaves/n-hexane[97]
28 (E)-caryophyllene
29 EstragoleRhizomes/aqueous[98]
30 Carvacrol
31 2-cyclohexane-1-one
32 Naphthalene
33 γ-Cadinene
34 Aristolene
35 1(5),3-aromadenedradiene
36 5-n-butyltetraline
37 4,5-dehydro-isolongifolene
38 Calarene
39 Isohomogenol
40 Zingiberene
41 α-Calacorene
42 5,8-dimethyl isoquinoline
43 Cyclohexane methanol
44 Longifolene
45 Isoelemicin
Sesquiterpenoids46 Calamene[74,78,91]
47 Calamenenol
48 Calameone
49 Preisocalamendiol
50 1,4-(trans)1,7(trans)-acorenone[93]
51 1,4-(cis)-1,7-(trans)-acorenone
52 2,6 diepishyobunone
53 α-Gurjunene
54 β-Gurjunene
55 α-Cedrene[98]
56 β-Elemene
57 β-Cedrene[93]
58 β-Caryophyllene
59 Valencene
60 Viridiflorene
61 α-SelineneGC-FID, GC-MS[89,93]
62 δ-CadineneGC-MS[93]
63 α-Curcumene
64 Shyobunone[84,93,99,100]
65 Isoshyobunone[93,99,101]
66 Caryophyllene oxide[93]
67 Humulene oxide IIGC-FID, GC-MS[89,93]
68 ElemolGC-MS[93]
69 Cedrol
70 Spathulenol
71 Acorenone
72 α-Cadinol
73 Humulene epoxide IIGC-FID, GC-MS[89]
74 α-Bisabolol
75 AsaronaldehydeNMRRhizomes/chloroform[95]
76 CalamusenoneGLC, IR, NMRRhizomes/petroleum ether[99]
77 Isocalamendiol
78 Dehydroxyiso-calamendiol
79 Epishyobunone
80 AcoroneNMRRhizomes/hydro alcoholic[100]
81 Neo-acorane ARhizomes/ethanol[102]
82 Acoric acid
83 Calamusin D
84 1β,5α-Guaiane-4β,10α-diol-6-one[103]
85 DioxosarcoguaiacolHPLCRhizomes/petroleum ether[101]
86 7-tetracycloundecanol,4,4,11,11-tetramethylGC-MSRhizomes/ethanol[84]
87 4α,7-Methano-4α-naphth[1,8a-b] oxirene,
88 SpathulenolRhizomes/aqueous[98]
89 Vulgarol B
90 Tatanan AHPLC, NMRRhizomes/95% ethanol[104]
91 Acoramone
92 2-hydroxyacorenone
93 4-(2-formyl-5-methoxymethyl
pyrrol-1-yl) butyric acid methyl ester
94 2-acetoxyacorenone
95 Acoramol
96 N-transferuloyl
tyramine
97 Tatarinoid A
98 Tatarinoid B
99 Acortatarin A
Monoterpenes100 α-PineneGC-MSRhizomes, roots/aqueous[74,78,91,93]
101 β-Pinene
102 Camphene[74,78,91,93,98]
103 o-Cymol[98]
104 p-CymeneGC-FID, GC-MS[89,93,98]
105 γ-TerpineneGC-MS[98]
106 α-Terpinolene
107 Anethole
108 Thymol
109 Isoaromadendrene epoxide
110 CamphorRhizome, leaves, roots/aqueous, hexane[93,97]
111 SabineneRoots/aqueous[93]
112 2-hexenal
113 Limonene[93,98]
114 Cis-linaloloxide[93]
115 Cis-sabinene hydrate
116 Trans-linalol oxide
117 Linalool[93,97]
118 Terpinen-4-ol[93]
119 α-Acoradiene
120 β-Acoradiene
121 α-Terpineol
122 IsoborneolLeaves/hexane[97]
Xanthone glycosides123 4,5,8-trimethoxy-xanthone-2-O-β-D-glucopyranosyl (1-2)-O-β-D-galactopyranosideNMRRhizome/ethanol[83]
Triterpenoid saponins124 1β,2α,3β, 19α-Tetrahydroxyurs-12-en-28-oic acid-28-O- {(β-D-glucopyranosyl (1-2)}-β-D galactopyranoside[82]
125 3-β, 22-α-24,29-Tetrahydroxyolean-12-en-3-O-(β-Darabinosyl (1,3)}-β-D-arabinopyranoside
Alkaloids126 Trimethoxyamphetamine,2,3,5GC-MS[84]
127 Pyrimidin-2-one,4-[N-methylureido]-1-[4methyl amino carbonloxy methy]
Triterpene glycoside128 22-[(6-deoxy-α-L-rhamnopyranosyl) oxy]-3,23-dihydroxy-, methyl ester, (3β,4β,20α,22β)NMRRoot, Rhizomes/ethyl acetate[85]
Steroids/Sterols129 β-daucosterol
Amino acids130 ArginineHPLCRoots/ethanol[86,87]
131 Lysine
132 Phenylalanine
133 Threonine
134 Tryptophan
135 α-alanine
136 Asparagine
137 Aspartic acid
138 Norvaline
139 Proline
140 Tyrosine
141 Glutamic acid
Fatty acids142 Palmitic acidGLCRhizome/petroleum ether[88]
143 Myristic acid
144 Palmitoleic acid
145 Stearic acid
GC-FID, gas chromatography – flame ionization detector; GC-MS, gas chromatography – mass spectrometry; NMR, nuclear magnetic resonance; FTIR, Fourier-transform infrared spectroscopy; GLC, gas liquid chromatography; IR, infrared spectroscopy; HPLC, high-performance liquid chromatography.
Table 4. Preclinical claims of A. calamus in neurological and metabolic disorders.
Table 4. Preclinical claims of A. calamus in neurological and metabolic disorders.
ActionParts of PlantExtract/CompoundAnimal ModelDosageResultsReferences
Antidiabetic effectsRhizomesMethanolSTZ-induced50, 100, and 200 mg/kg, p.o. to rats↓ Lipid profile and blood glucose, while ↑ levels of plasma insulin, tissue glycogen, and G6PD[105]
Alloxan-induced150 and 200 mg/kg, p.o. to rat↓ Blood glucose level[106]
Ethyl acetateGenetically obese diabetic C57BL/Ks db/db mice100 mg/kg, p.o.↓ Levels of triglycerides and serum glucose[107]
GLP-1 expression and secretion with STZ-induced100 mg/kg, i.g.↑ Secretion of GLP-1 and ↓ blood glucose levels[108]
In vitro HIT-T15 cell line and alpha-glucosidase enzyme6.25, 12.5, and 25 µg/mL↑ Insulin secretion in HIT-T15 cells[109]
Glucose tolerance400 and 800 mg/kg, p.o. to mice↓ Serum glucose, and abolished the ↑ level of blood glucose
Anti-obesity effectsEthanol and aqueousHFD-induced100 and 200 mg/kg to rats↓ Levels of serum cholesterol and triglycerides, ↑ lipoprotein fraction[110]
Diethyl etherHFD-induced20 and 40 mg/kg, p.o. to rats↓ Total cholesterol and low-density lipoprotein levels, ↑ plasma fibrinogen levels[111]
MethanolTriton-X-100-induced hyperlipidemic250 and 500 mg/kg to ratsDose-dependent anti-hyperlipidemic effect[112]
HFD-induced250 and 500 mg/kg, p.o. to rats↓ Level of total cholesterol, triglycerides, and LDL, ↑ HDL cholesterol[113]
AqueousHFD-induced100, 200, and 300 mg/kg, p.o. to rats↓ Levels of serum glucose, leptin, and insulin along with ↓ triglyceride, low-density lipoprotein, very LDL cholesterol, total cholesterol, phospholipids, and free fatty acid increased levels[114]
Antihypertensive effectsEthyl acetateClamping the left kidney artery for 4 h250 mg/kg, p.o. to rats↓SBP and DBP, blood urea nitrogen, creatinine and LPO, ↑ level of nitric oxide, SOD, CAT, GPX[115]
Crude extract, ethyl acetate and n-hexaneBlood pressure lowering effect in normotensive10, 30, and 50 mg/kg to anesthetized ratsRelaxant effects mediated through Ca+2 antagonism and NO pathways[116]
Ethanol and α-asaroneDimethyl sulfoxide-induced noise stress to rats100 and 9 mg/kg, p.o. to rats↓ Destructive effect of stress enlightening the morphological changes of hippocampus[117]
Anti-inflammatory effectsLeavesEthanolCarrageenan-induced paw edema100 and 200 mg/kg to rats↓ Histamine, 5-HT, and kinins[118]
Antioxidant effectsRhizomesα-asaroneNoise stress induced to rats3, 6, and 9 mg/kg, i.p. to rats↑ SOD and LPO, decreased ↓ CAT, GPX, GSH, vitamins C and E, and protein thiol levels[119]
Leaves and rhizomesEthyl acetate and methanolDPPH radical scavenging chelating ferrous ions, FRAP200, 100, 80, 60, 40, 20, 10, and 5 μg/mLProminent DPPH scavenging activity, chelating ferrous ions, and reducingpower[120,121]
RhizomesEthanolAcetaminophen-induced250, 500 mg/kg, p.o. to rats↓ MDA and ↑ SOD, CAT, GPX, GSH levels[122]
Anticonvulsant effectsRootsEthanol and β-asaroneKainic acid-induced convulsion35 and 20 mg/kg↓ Epileptic seizure, neuroprotective, and regenerative ability[123]
MethanolPTZ-induced convulsion100 and 200 mg/kg, p.o. to mice↑ Latency period and ↓ PTZ-induced seizure time[124]
RhizomesCalamus oilMES, PTZ, and MCS model30, 100, and 300 mg/kg, p.o. to miceCalamus oil is found stable[125]
EthanolMES and PTZ-induced convulsion250, 500 mg/kg, p.o. to mice↓ Hind limb extension and tonic flexion of forelimbs[126]
MethanolMES and PTZ-induced250 and 150 mg/kg, p.o. to rats↓ Immobility time at 250 mg/kg; however, ineffective at 150 mg/kg[127]
Antidepressant effectsTST and FST50 and 100 mg/kg, i.p. to mice↓ Immobility time in a dose-dependent manner[128]
LeavesTST and FST50 and 100 mg/kg↓ Immobility time[129]
RootsAqueousTST and FST100, 150, 200 mg/kg, p.o. to mice↓ Immobility time[130]
RhizomesHydro-alcoholic extractTST and FST75 and 150 mg/kg, p.o. to mice↓ Corticosteroid levels[131]
EthanolOFB and HPM test72 mg/kg, p.o.No stimulation of postsynaptic 5-HT1A receptors[132]
Methanol and acetoneBehavioral despair test5, 20, and 50 mg/kg, p.o.↓ Spontaneous locomotor activity[133]
β-asaroneEPM and FST25, 50, and 100 mg/kg, p.o.↓ Immobility time[134]
Neuroprotective effectsHydro-alcoholicCCI of sciatic nerve-induced neuropathic pain10 mg/kg to ratsSignificantly ameliorated CCI-induced nociceptive pain[135]
CCI of sciatic nerve-induced peripheral neuropathy100 and 200 mg/kg to ratsPrevented CCI-induced neuropathy through ↓ oxidation and inflammation[136]
LeavesMethanol and acetoneApomorphine-induced stereotypy and haloperidol-induced catalepsy20 and 50 mg/kg to miceReversed stereotypy induced by apomorphine and significantly potentiated catalepsy induced by haloperidol[137]
RhizomesEthanolSpontaneous electrical activity and monoamine levels of the brain200 and 300 mg/ kg to ratsDepressive response by altering electrical activity, including changing brain monoamine levels[138]
Hydro-alcoholicMCAo-produced brain ischemia25 mg/kg to ratsImprovement in neurobehavioral performance, ↓ levels of GSH, SOD, and ↑ LPO level[139]
EthanolMethotrexate-induced stress5, 10, 15, 20, 25 ppm concentration to fruit flies↓ Elevated ROS, SOD, CAT, and GPX levels[140]
Cardioprotective effectsWhole plantDOX-induced myocardial toxicity100 and 200 mg/kg to rats↓ Serum enzyme levels and protected the myocardium from the toxic effect of DOX[141]
RhizomesCrude, n-hexane, ethyl acetateGuinea pig tracheal segments0.01 mg/mL↓ Force and rate of contractions at higher concentrations[142]
CAT, catalase; CCI, chronic constriction injury; COX, cyclooxygenase; DBP, diastolic blood pressure; DOX, doxorubicin; DPPH, 2,2-diphenyl-1-picrylhydrazyl radical; EPM, elevated plus maze; FRAP, ferric reducing antioxidant power; FST, forced swim test; GLP-1, glucagon-like peptide-1; GPX, glutathione peroxidase; GR, glutathione reductase; GSH, reduced glutathione; HDL, high-density lipoproteins; HFD, high-fat diet; HPM, high plus maze; i.g., intragastric; i.p., intraperitoneal; LDL, low-density lipoprotein; LPO, lipid peroxides; MCAo, middle cerebral artery occlusion; MCS, minimal clonic seizure; MDA, malondialdehyde; MES, maximal electroshock; NO, nitric oxide; OFB, open field behavior; p.o., per oral; PTZ, pentylenetetrazol; ROS, reactive oxygen species; SBP, systolic blood pressure; SOD, superoxide dismutase; STZ, streptozotocin; TST, tail suspension test.
Table 5. Clinical claims of A. calamus in neurological and metabolic disorders.
Table 5. Clinical claims of A. calamus in neurological and metabolic disorders.
Formulations/Dosage forms A. calamusSubjectsStudy DesignIntervention Primary EndpointOutcomeEvidence QualityReference
A. calamus rhizome powder24 patients of both sexes with hyperlipidemiaRandomized single-blind controlled study500 mg twice daily after meal for 1 monthBMI, body perimeter, skinfold depthSignificant reduction in skinfold depth, fatigue, and excessive hungerIII[178]
Davaie Loban capsules (A. calamus, nut grass, incense, ginger, and black pepper)24 patients of both sexes with Alzheimer’s diseaseDouble-blind randomized clinical study500 mg capsule thrice daily for 3 monthsADAS-cog and CDR-SOB scoresAt 4 weeks and 12 weeks: significant reduction in the ADAS-cog and CDR-SOB scoresIII[179]
70% hydro-alcoholic extract of A. calamus33 patients of both sexes (20 male and 13 female) with anxiety disorderNon-randomized, open-label, single-arm study500 mg extract of one capsule twice daily after meal for 2 monthsBPRS scoreSignificant reduction of anxiety and stress-related disorderIII[180]
Vachadi Churna (A. calamus, Cyperus rotundus, Cedrus deodara, ginger, Aconitum Heterophyllum, T. chebula)30 obese patients of both sexes aged 14–50 yearsNon-randomized, open-label, single-arm study3 g powder twice daily with lukewarm water before meal for 1 monthBMI, girth measurements of mid-thigh, abdomen, hip, chestSignificant improvement in extreme sleep, body heaviness, fatigue, and excessive hungerIII[181]
Guduchyadi Medhya Rasayana, (A. calamus, Tinospora cordifolia, Achyranthes aspera, Embelia ribes, Convolvulus pluricaulis, T. chebula, S. lappa, Asparagus racemosus, cow ghee, and sugar)138 patients of both sexes aged 55–75 years with senile memory impairmentRandomized, two-parallel-group study3 g granule thrice daily after meal for 3 monthsMini–Mental State Examination, BPRS score, and estimation of serum acetylcholinesteraseSignificant improvement in terms of recall memory, cognitive impairment, amnesia, concentration ability, depression, and stressIII[182]
Dried aqueous extract of A. calamus40 healthy volunteers, both sexes aged 18–50 years with a premedicant for anesthesiaOpen-label randomized, two- parallel-group study90 min before anesthesia;
In the control group:
0.2 mg intramuscular (IM) glycopyrrolate and a 0.2 mg IM 50 mg tablet of promethazine hydrochloride with water;
In the second group: 0.2 mg IM glycopyrrolate and 100 mg A. calamus extract
Pulse rate, blood pressure, respiratory rate, body temperatureThe dried aqueous extract exhibited anti-hyperthermic and sedative effect without producing
any respiratory depression
III[183]
Shankhapushpyadi Ghana Vati (A. calamus, C. pluricaulis, Bacopa monnieri, T. cordifolia, C. fistula, A. indica, S. lappa, Tribulus terrestris)20 hypertensive patients of both sexesRandomized single-blind controlled
study
1 g twice daily after meal for 2 monthsSBP and DBPSignificant relief in raised SBP and DBPIII[184]
Brahmyadiyoga (A. calamus, Centella asiatica, Rauvolfia serpentina, Saussurea lappa, Nardostachys jatamansi)10 schizophrenia patients of both sexes aged 18–40 yearsNon-randomized,
open-label, single-
arm study
4 tablets thrice daily for three months after mealSymptoms rating scaleSignificant effect as a brain tonic, tranquillizer, hypnotic, and sedativeIII[185]
Bala compound (A. calamus, Emblica officinalis, E. ribes, T. cordifolia, Piper longum, Glycyrrhiza glabra, C. rotundus, A. heterophyllum)24 neonates, both sexes, 2.5–3 kg body weightRandomized single-blind controlled
study
5 oral drops twice daily for 6 monthsChange in serum immunoglobulins (IgG, IgM, and IgA) levelsSignificant improvement in immunoglobulin levels after 6 monthsIb[186]
Vachadi Ghrita (A. calamus, T. cordifolia, Hedychium spicatum, C. pluricaulis, E. ribes, ginger, A. aspera, T. chebula, and cow ghee)90 healthy individuals of both sexes aged 40–50 years for assessment of cognitionNon-randomized
positive-controlled study
10 g twice daily for 1 month with lukewarm waterPost Graduate Institute Memory Scale (PGIMS) testSignificant change in the mental balance score, holding
of like and different pairs, late-immediate memory, and also improved digestion
III[187]
Bramhi Vati (A. calamus, B. monnieri, C. pluricaulis, Onosma bracteatum, copper pyrite, iron pyrite, mercuric sulphide, Piper nigrum, N. jatamansi)68 essential hypertension patients of both sexes aged 20–70 yearsRandomized, double-blind, parallel-group comparative study500 mg tablets twice daily for 1 monthHamilton anxiety rating scale, SBP and DBP, and MAPSignificant improvement in the Hamilton anxiety rating scale, SBP and DBP, and MAPIII[188]
Tagaradi Yoga (A. calamus, Valeriana wallichii, N. jatamansi)24 insomnia patients of both sexes aged 18–75 yearsNon-randomized positive-controlled study500 mg hydro-alcoholic extract capsule twice daily after meal for 15 daysSleep duration, initiating time of sleep, quality of sleepSignificant improvement in sleep duration, in the initiating time of sleep, and in quality of sleepIII[189]
Acorus calamus rhizome powder20 obese patients of both sexesRandomized single-blind study250 mg rhizome powder twice daily for 1 monthBody weight, height according to age, waist-hip ratio, and BMISignificant improvement in extreme sleep, body heaviness, fatigue, and excessive hungerIII[190]
Acorus calamus rhizome powder45 ischemic heart disease patientsNon-randomized positive-controlled study3 gm rhizome powder twice daily for 3 monthsECG, serum cholesterol levelImprovement of chest pain, dyspnea on effort, reduction of the body mass index, improved ECG: reduced serum cholesterol, reduced serum LDL, and increased serum HDLIb[191]
ADAS-cog, alzheimer’s disease assessment scale–cognitive subscale; BMI, body mass index; BPRS, brief psychiatric rating scale; CDR-SOB, clinical dementia rating scale sum of boxes; DBP, diastolic blood pressure; ECG, electrocardiogram; Ib, evidence from at least one randomized study with control; HDL, high-density lipoprotein; Ig, immunoglobulin; III, evidence from well-performed nonexperimental descriptive studies, as well as from comparative studies, correlation studies, and case studies; LDL, low-density lipoprotein; MAP, mean arterial pressure; SBP, systolic blood pressure.
Table 6. Mechanistic role of phytochemicals of A. calamus in the treatment of neurological and metabolic disorders.
Table 6. Mechanistic role of phytochemicals of A. calamus in the treatment of neurological and metabolic disorders.
StudyCompoundModelIncreased LevelDecreased LevelReferences
Anti-Parkinsonβ-Asarone6-OHDA parkinsonianBcl-2 expressionGRP78, p-PERK, CHOP, and Beclin-1 expression[192]
6-OHDA parkinsonian-mRNA levels of GRP78 and CHOP and p-IRE1and XBP1[193]
Dopamine in the striatumTH plasma concentrationsStriatal COMT levels[194]
6-OHDA parkinsonianL-DOPA, DA, DOPAC, and HVA levelsP-gp, ZO-1, occludin, actin, and claudin-5[195]
Alzheimer’sAβ25-35-induced inflammationBcl-2 levelTNF-α, IL-1β, IL-6, Beclin-1, and LC3B level[196]
NG108 cells-Upregulated SYP and GluR1 expression[197]
PC12 cells-Aβ-induced JNK activation, Bcl-w and Bcl-xL levels, cytochrome c release, and caspase-3 activation[198]
Aβ-induced cytotoxicityCell viability, p-Akt and p-mTORNSE levels, Beclin-1 expression[199]
NeuroprotectivePb-induced impairmentsNR2B protein expression along with Arc/Arg3.1 and Wnt7a mRNA levels-[200]
β-Asarone, eugenolScopolamine-inducedImprovement of neuron organelles and synaptic structureAPP expression[201]
NeotatarineMTT reduction assay-Aβ25-35–induced PC12 cell death[202]
β-asarone, paeonolMCAo modelCholecystokinin and NF-κB signalingTNF-α, IL-1β, IL-6 production[203]
β-AsaroneCultured rat astrocytesNGF, BDNF, and GDNF expression-[204]
SN4741 cellsp62, Bcl-2 expressionJNK, p-JNK and Beclin-1 expressions[205]
TatarinolactonehSERT-HEK293 cell line-SERTs activity[206]
β-AsaroneRSC96 Schwann cellsGDNF, BDNF, and CNTF expression-[207]
Aβ-inducedp-mTOR and p62 expressionAChE and Aβ42 levels, p-Akt, Beclin-1, and LC3B expression, APP mRNA and Beclin-1 mRNA levels[208]
Aβ1–42-induced injury-GFAP, AQP4, IL-1β, and TNF-α expression[209]
Anti-depressionChronic unpredictable mild stressBDNF expressionBlocked ERK1/2-CREB signaling[210]
α-AsaroneNoradrenergic and serotonergic neuromodulators in TSTα1 and α2 adrenoceptors and 5-HT1A receptors-[211]
Anticonvulsant and sedativeEudesminMES and PTZGABA contents, expressions of GAD65, GABAA, and Bcl-2Glu contents and ratio of Glu/GABA, caspase-3[212]
Anti-anxietyα-AsaroneBLA or CFA-inducedDown-regulation of GABAA receptorsUp-regulation of GluR1-containing AMPA, NMDA receptors[213]
Anti-epilepsyTemporal lobe epilepsyLevels of GABA, GAD67, and GABAAR-mRNA expressionGABA-T[214]
Mitral cellsDown-regulation of GABAA receptorsNa+ channel blockade[215]
β-AsaroneKA-inducedGABAGlu[216]
Anti-inflammatoryα-AsaroneSpinal cord injuryIL-4, IL-10, and arginase 1 levelsTNF-α, IL-1β, IL-6, MCP-1, MIP-2, iNOS levels[217]
Cytoprotectiveβ-AsaronetBHP-induced astrocyte injuryGST, GCLM, GCLC, NQO1, Akt phosphorylation-[218]
CardioprotectiveCultured neonate rat cardiac myocytesViability of cardiac myocytesPulse frequency[219]
ArteriosclerosisECV304 cell strainApoptotic rate of ECV304 cellsApoptotic rate of MMP, stabilized MMP and VSMC proliferation[220]
Anti-adipogenic3T3-L1 preadipocytes-C/EBPβ, C/EBPα, and PPARγ expression levels, ERK1/2 phosphorylation[89]
AntioxidantCerebral artery occlusionAntioxidant activityFocal cerebral ischemic/reperfusion injury[221]
Anti-diabeticα-Asarone + β-asarone + metformin HClSTZ-inducedInsulin levelGlucose, glycosylated hemoglobin level, liver dysfunction, and tumor biomarkers[222]
Asarone3T3-L1 preadipocytesHormone-sensitive lipase phosphorylationIntracellular triglyceride levels, down-regulation of PPARγ and C/EBPα[223]
6-OHDA, 6-hydroxydopamine; Ox-LDL, oxidized low-density lipoprotein; BDNF, brain-derived neurotrophic factor; NGF, nerve growth factor; GDNF, glial derived neurotrophic factor; SERTs, serotonin transporters; MCAo, middle cerebral artery occlusion; Aβ, β-amyloid; NSE, neuron specific enolase; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; NMDA, NR2A-containing N-methyl-D-aspartate; GABAA, γ-aminobutyric acid A; BLA, basolateral amygdala; CFA, complete Freund’s adjuvant; CNTF, ciliary neurotrophic factor; COMT, catechol-O-methyltransferase; TH, tyrosine hydroxylase; DA, dopamine; DOPAC, 3,4-dihydroxyphenylacetic acid; HVA, homovanillic acid; P-gp, P-glycoprotein; ZO-1, zonula occludens-1; SYP, synaptophysin; GluR1, glutamatergic receptor 1; GABA-T, GABA transaminase; TST, tail suspension test; KA, kainic acid; MCP-1, monocyte chemoattractant protein 1; MIP-2, macrophage inflammatory protein 2; iNOS, inducible nitric oxide synthase; GST, glutathione S-transferase; GCLM, glutamate-cysteine ligase modulatory subunit; GCLC, glutamate-cysteine ligase catalytic subunit; NQO1, NAD(P)H quinone oxidoreductase; GFAP, glial fibrillary acidic protein; AQP, aquaporin; VSMC, vascular smooth muscle cells; MMP, mitochondrial membrane potential; C/EBP, CCAAT enhancer-binding protein; PPARγ, peroxisome proliferator-activated receptor gamma; ERK1/2, extracellular signal-regulated protein kinase; XBP1, x-box binding protein; IRE1, inositol-requiring enzyme 1; Aβ1-42, amyloid β peptide; mTOR, mammalian target of rapamycin; MTT, 3-(4,5-dimethythiazol-. 2-yl)-2,5-diphenyl tetrazolium bromide; CREB, cAMP response element-binding protein; GABAAR, gamma-aminobutyric acid type-A receptor, tBHP, t-butyl hydroperoxide.

Share and Cite

MDPI and ACS Style

Sharma, V.; Sharma, R.; Gautam, D.S.; Kuca, K.; Nepovimova, E.; Martins, N. Role of Vacha (Acorus calamus Linn.) in Neurological and Metabolic Disorders: Evidence from Ethnopharmacology, Phytochemistry, Pharmacology and Clinical Study. J. Clin. Med. 2020, 9, 1176. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm9041176

AMA Style

Sharma V, Sharma R, Gautam DS, Kuca K, Nepovimova E, Martins N. Role of Vacha (Acorus calamus Linn.) in Neurological and Metabolic Disorders: Evidence from Ethnopharmacology, Phytochemistry, Pharmacology and Clinical Study. Journal of Clinical Medicine. 2020; 9(4):1176. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm9041176

Chicago/Turabian Style

Sharma, Vineet, Rohit Sharma, DevNath Singh Gautam, Kamil Kuca, Eugenie Nepovimova, and Natália Martins. 2020. "Role of Vacha (Acorus calamus Linn.) in Neurological and Metabolic Disorders: Evidence from Ethnopharmacology, Phytochemistry, Pharmacology and Clinical Study" Journal of Clinical Medicine 9, no. 4: 1176. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm9041176

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop