Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2020 Sep 28;266:113415. doi: 10.1016/j.jep.2020.113415

Atractylodis Rhizoma: A review of its traditional uses, phytochemistry, pharmacology, toxicology and quality control

Wen-jin Zhang a,b, Zhen-yu Zhao a, Li-kun Chang c, Ye Cao d, Sheng Wang a, Chuan-zhi Kang a, Hong-yang Wang e, Li Zhou a, Lu-qi Huang a,∗∗, Lan-ping Guo a,
PMCID: PMC7521906  PMID: 32987126

Abstract

Ethnopharmacological relevance

Atractylodis Rhizoma (AR), mainly includes Atractylodes lancea (Thunb.) DC. (A. lancea) and Atractylodes chinensis (DC.) Koidz. (A. chinensis) is widely used in East Asia as a diuretic and stomachic drug, for the treatment of rheumatic diseases, digestive disorders, night blindness, and influenza as it contains a variety of sesquiterpenoids and other components of medicinal importance.

Aim of the review

A systematic summary on the botany, traditional uses, phytochemistry, pharmacology, toxicology, and quality control of AR was presented to explore the future therapeutic potential and scientific potential of this plant.

Materials and methods

A review of the literature was performed by consulting scientific databases including Google Scholar, Web of Science, Baidu Scholar, Springer, PubMed, ScienceDirect, CNKI, etc. Plant taxonomy was confirmed to the database “The Plant List”.

Results

Over 200 chemical compounds have been isolated from AR, notably sesquiterpenoids and alkynes. Various pharmacological activities have been demonstrated, especially improving gastrointestinal function and thus allowed to assert most of the traditional uses of AR.

Conclusions

The researches on AR are extensive, but gaps still remain. The molecular mechanism, structure-activity relationship, potential synergistic and antagonistic effects of these components need to be further elucidated. It is suggested that further studies should be carried out in the aspects of comprehensive evaluation of the quality of medicinal materials, understanding of the “effective forms” and “additive effects” of the pharmacodynamic substances based on the same pharmacophore of TCM, and its long-term toxicity in vivo and clinical efficacy.

Keywords: Atractylodis rhizoma, Phytochemistry, Pharmacology, Quality control, Toxicology

Graphical abstract

Image 1

Abbreviations

AR

Atractylodis Rhizoma

AM

Atractylodis Macrocephalae Rhizoma

ATL-I

atractylenolide I

CCA

cholangiocarcionma

MPO

myeloperoxidase

MCP

monocyte chemoattractant protein

HMC-1

human mast cell line

TLR4

toll like receptor 4

COX-2

cyclooxygenase-2

ASNA

adrenal sympathetic nerve activity

IAV

influenza A virus

TLR7

toll-like receptor 7

IFN-β

interferon-β

CNS

central nervous system

TCM

traditional Chinese medicine

IUPAC

international Union of Pure and Applied Chemistry

PTPLB

protein tyrosine phosphatase-like, member b

TNF-α

necrosis factor-α; interleukin-8 (IL-8)

PGE2

prostaglandin E2

JNK

c-Jun N-terminal kinase

NQO1

NAD(P)H-quinone oxidoreductase 1

HUVEC

human umbilical vein endothelial cells

AMPK

AMP-activated protein kinase

ACC

acetyl-CoA carboxylase; carboxylase

MCP

monocyte chemoattractant protein

HA

Hubei type AR

MA

maoshan type AR

CRF

corticotropin-releasing factor

IEC-6

intestinal epithelial

5-LOX

5-lipoxygenase

GLUT-4

glucose transporter type 4

CYP3A

cytochrome P450 3A

NLRP3

nucleotide-binding domain-like receptor protein 3

TSLP

thymic stromal lymphopoietin

Bcl-2

B-cell lymphoma 2

p-PKB

phospho-protein kinase B

p-ERK

phospho-extracellular regulated protein kinases

p-JAK2

phospho-janus kinase 2

p-STAT3

phospho-signal transducer and activator of transcription 3

MMP-2

matrix metalloproteinase-2

Mcl-1

myeloid cell leukemia 1

Bcl-xL

B-cell lymphoma-extra large

CDK1

cyclin-dependent kinase 1

Cdc25c

cell division cycle 25C

G-CSF

granulocyte-colony stimulating factor

TRPC1

transient receptor potential canonical 1

PLC-γ1

phospholipase C-γ1

EGF

epidermal growth factor

TFF2

trefoil factor 2

TRPC1

transient receptor potential canonical 1

TLC

thin layer chromatography

ICAM-1

intercellular adhesion molecule-1

MIP-2

macrophage inflammatory protein 2

DOI

1-(2, 5-dimethoxy-4-iodophenyl)-2-aminopropane

TRPA1

transient receptor potential cation channel subfamily a member 1

ATR

Atractyloside

1. Introduction

Genus Atractylodes (Compositae) comprises eight species of perennial herbs distributed in East Asia (Hiraoka, 1993) and is classified into two major groups, Cangzhu and Bai-zhu (common Chinese names). Dried rhizomes of Atractylodes plants, including Atractylodes lancea (Thunb.) DC. (A. lancea), Atractylodes chinensis (DC.) Koidz. (A. chinensis), Atractylodes japonica Koidz. (A. japonica), and Atractylodes macrocephala Koidz. (A. macrocephala) etc, have been used as important crude drugs prescribed in Chinese, Japanese, Korean and Thai traditional medicine. A. lancea and A. chinensis are perennial plants, known to be widely distributed in China, are prescribed in the Chinese, Japanese and Korean Pharmacopoeia as the botanical origins of the crude drug, Cangzhu (Sojutsu in Japanese) and collectively termed Atractylodis Rhizoma (AR) in present paper.

A. japonica and A. macrocephala are prescribed in the Japanese and Korean Pharmacopoeia as the botanical origin of the crude drug, Baizhu (Byakujutsu in Japanese and Baekchul in Korean). Traditional Chinese medicine (TCM) theory states that both groups have a similar mode of action for invigorating the spleen and eliminating dampness. However, it has been reported that the former crude drug has anti-sudorific activity and the latter exhibits diaphoretic activity (Mizukami et al., 1996), and they have been used for different clinical purposes. AR has been used as an important crude drug for the treatment of rheumatic diseases, digestive disorders, night blindness, and influenza in TCM (Xiao et al., 2002) and was listed in Shennong Bencao Jing, the first Chinese Pharmacopoeia. In the Korean and Japanese Pharmacopoeias, the AR has been prescribed in traditional medicine as a diuretic and stomachic drugs (Kitajima et al., 2003c). In Thai traditional medicine, the main use of A. lancea is for the treatment of fever and cold (Kesara et al., 2017). These ethnopharmacological uses of AR either as a single extract or as a component in herbal medicine formula are explained by its properties in eliminating dampness, invigorating spleen, and expelling wind according to the theory of TCM. However, “Zhu” medicine materials have a long history of application and have always been confused. The Latin names of AR are miscellaneous. The multiple components contained in the AR contributed to its beneficial effects including a series of sesquiterpenoids, alkynes, triterpenoids, aromatic glycosides, oligosaccharides, and polysaccharides (Jun et al., 2018a). But the available literature is not sufficient to explain its medical claims. And the corresponding structures have not been fully introduced and summarized yet. Although recent researches reveal that the extracts from AR could also exert improving gastrointestinal function, anti-tumor activity, immunomodulatory activity, anti-inflammatory activity, antibacterial activity (Jun et al., 2018). The findings have not yet been established links between traditional uses and modern scientific knowledge of AR. And the toxicity and safety of the chemicals have not been defined (Zhu et al., 2018). On the other hand, the drugs distributed in the market have hugely varied in contents of the major essential oil compounds, posing a challenge for the cultivation of AR with constant quality (Nishikawa et al., 1975). A systematic review of A. macrocephala has been previously reported (Zhu et al., 2018). This paper reviews the available recent literature on botany, traditional use, phytochemistry, pharmacological activity and its possible mechanisms, toxicity of AR with an emphasis on the biological activity and its application in TCM, quality control, and to critically analyse the reported studies. Its other possible clinical applications and future research directions also were proposed to provide a valuable and comprehensive compilation of research findings on AR to improve its medical value and use.

2. Botany

Genus Atractylodes is the main source of Atractylodis Rhizoma (known as “Cangzhu” in China, “Khod-Kha-Mao” in Thailand and “Sojutsu” in Japan) (Ahmed et al., 2016) and Atractylodis Macrocephalae Rhizoma (AM, known as “Baizhu” in China, “Byakujutsu” in Japan and “Baekchul” in Korea) in China, Japan, and Korea (Table 1 ), both being long used as a stomachic. In the Japanese Pharmacopoeia, Atractylodes Lancea Rhizome is the rhizome of Atractylodes lancea De Candolle, Atractylodes chinensis Koidzumi or their interspecific hybrids. Atractylodes Rhizome is the rhizome of Atractylodes japonica Koidzumi ex Kitamura (Wa-byakujutsu) or Atractylodes macrocephala Koidzumi (Atractylodes ovata De Candolle) (Kara-byakujutsu) (Shimato et al., 2018). Atractylodes Rhizome is the rhizome of Atractylodes lancea De Candlle or of Atractylodes chinensis Koidzumi. Atractylodes Rhizome White is the rhizome of Atractylodes japonica Koidzumi or Atractylodes macrocephala Koidzumi in Korean Pharmacopoeia. However, A. japonica is not included in the Pharmacopoeia of the People's Republic of China (Since, 2015 Edition) and Taiwan Herbal Pharmacopoeia (Committee, 2015).

Table 1.

Atractylodis rhizoma and atractylodis macrocephalae rhizoma in pharmacopoeia of China, Japan and korea.

Pharmacopoeia Crude drug Scientific names
Pharmacopoeia of the People's Republic of China Atractylodis Rhizoma Atractylodes lancea (Thunb.) DC. or Atractylodes chinensis (DC.) Koidz.
Atractylodis Macrocephalae Rhizoma Atractylodes macrocephala Koidz.
Japanese Pharmacopoeia Atractylodes Lancea Rhizome Atractylodes lancea De Candolle or Atractylodes chinensis Koidzumi or their interspecific hybrids
Atractylodes Rhizome Atractylodes japonica Koidzumi ex Kitamura (Wa-byakujutsu) or Atractylodes macrocephala Koidzumi (Atractylodes ovata De Candolle) (Kara-byakujutsu)
Korean Pharmacopoeia Atractylodes Rhizome Atractylodes lancea De Candlle or Atractylodes chinensis Koidzumi
Atractylodes Rhizome White Atractylodes japonica Koidzumi or Atractylodes macrocephala Koidzumi

A comparison of the components in the rhizomes of Chinese Atractylodes has been made by thin layer chromatography (TLC) and gas-liquid chromatography (GLC). The results were in accordance with their morphological features and pharmaceutical merits. A. macrocephala, with its leaves pinnately incised, is characterized by the presence of rich atractylon and absence or lack of atractylodin. As for A. lancea and A. chinensis with their leaves not incised or only lobed, are characterized by high contents of atractylodin, β-eudesmol, and hinesol, but poor in atractylon (Fu et al., 1981). In addition, A. lancea, A. chinensis, and A. macrocephala are the three most widely used medicinal species of the Atractylodes genus. Their similar morphological features cause disagreement as whether they are three unique species, leading to their frequent misuses in medical products (Wang et al., 2020). A. macrocephala is distinguishable from other Atractylodes plants by RAPD analysis (Kohjyouma et al., 1997), and the phylogenetic relationship between A. lancea and A. japonica is suggested to be closer than that between A. japonica and A. macrocephala (Kitajima et al., 2003b). further examinated the water-soluble portion of A. macrocephala to determine the chemotaxonomic relationships among them. And confirmed that the chemotaxonomic relationship between A. lancea and A. japonica can be considered to be close. The basic term “zhu” was the only one used when atractylodes was first recorded in the ancient Shennong Bencao Jing (ca. 100 A.D.) (Chang et al., 2015); the division between these two related herb materials first occurred in the Mingyi Bielu (500 A.D.). At that time, the tuber-like rhizomes of these plants were specified as either Baizhu (bai = white) and Chizhu (chi = red), referring to the color observed in the sliced rhizomes, the red being due to spots of accumulated oils. Later, Chizhu was renamed Cangzhu (cang = gray or black), which refers to the appearance of the outer skin of the rhizome, a dark gray-black color. “Baizhu” in the Song Dynasty gradually turned into a cultivated product. After the Yuan (1271–1368 A.D.) and Ming Dynasties (1368–1644 A.D.), “Cangzhu” and “Baizhu” were divided into two herbs with different effects. After the Ming and Qing Dynasties (1636–1912 A.D.), “Mao Cangzhu” was recommended in Jiangsu province, while AM highly praised wild species in Zhejiang Province. Later it was found that this herb includes two species, A. lancea and A. chinensis, separately in China and people have used these together as AR (ECoNCMMBC, 1999). According to “The Plant List” (www.theplantlist.org), Atractylodes amurensis (Freyn ex Kom.) H.S.Pak, Atractylodes carlinoides (Hand.-Mazz.) Kitam., Atractylodes japonica Koidz. ex Kitam., Atractylodes koreana (Nakai) Kitam., Atractylodes lancea (Thunb.) DC., is accepted names for the plant, with more than ten synonyms. Generally, cultivated or wild A. lancea, also known as Mao Cangzhu in Chinese, is mainly produced in Jiangsu, Hubei, Zhejiang, Anhui, and other places. A. lancea is irregularly curved, cylindrical rhizome, 3 cm–10 cm in length and 10 mm–25 mm in diameter. The external surface is dark grayish-brown to dark yellow-brown. A transverse section reveals nearly orbicular, with pale brown to red-brown secretes as fine points. Often white cotton-like crystals are produced on its surface if A. lancea stored long time with fragrant smelling and sweet tasty. A. chinensis (Bei Cangzhu) is mainly produced in Hebei, Shanxi, Inner Mongolia, Liaoning, and other places. A. chinensis is irregularly cylindrical rhizome, 4 cm–9 cm in length and 10 mm–40 mm in diameter. The external surface is black-brown to yellow-brown. The fragrance is light, pungent, and bitter (Fig. 1 ) (Committee, 2018). Due to the different origins of A. lancea and A. chinensis, the leaf shapes and active ingredients are different. So it is appropriate to treat it differently and consider “Bei Cangzhu” as a variant of “Mao Cangzhu” (Fu et al., 1981).

Fig. 1.

Fig. 1

The above-ground portion (a), medicinal portion (b), and commercial herbal pieces (c) of (1) A. chinensis and (2) A. lancea. Distribution of Atractylodis Rhizoma resources (3).

3. Traditional uses

3.1. Medicinal history of AR

Beginning from the ancient times, the traditional use of AR constantly develops and changes with the deepening of cognition. AR is listed as a top grade in Shennong Bencao Jing (Gu, 1955), in which it was recorded as “removing wind-cold and resolving dampness, dead muscle spasm, and jaundice. The decoctions are taken for a long time, lighting body and prolonging life without hunger.” AR is pungent and fragrant. It can relax muscle and induce perspiration. It can dispel the cold and pathogenic wind on the surface of the skin. However, the effect of sweating is not obvious. AR is most suitable for treating cold and dampness because it is good at dehumidification. According to the Yaopin Hua Yi (Jia, 2015), AR is the main drug from the treatment of the syndrome of dampness, turbidity, and stagnation of middle-jiao (the upper part of the abdominal cavity). Mingyi Bielu (Tao, 2013) documented “curing headache, eliminating phlegm and water-retention, removing the swelling of wind and water between the skin, eliminating heart disease and vomiting in cholera, warming the stomach and eliminating food”. AR could invigorate spleen-Qi, and activate the spleen and stomach. The Yang is clear and rising, nourishing and clearing orifices, leading to a clear-sight. The Taiping Shenghui Fang (Wang et al., 1958) recorded it as “treating night blindness” and Jingyan Fang recorded “treating internal and external barriers to the eyes”. AR eliminates dampness with aromatics by avoiding filth and pestilence. Bencao Qiuyuan (Zhao, 2009) added the treatment of “injury from food, heat, and diarrhea, spleen dampness and blood”. The records of doctors from all dynasties reflected the main treatment scopes of AR. Therefore, the Materia Medica in the Ming and Qing Dynasties summarized the functions of AR as drying dampness and strengthening the spleen, eliminating pollution and turbidity, eliminating wind dampness, relieving symptoms, and clarifying the eyes.

3.2. Compatibility and application of AR

These functions also reflected in the application of ancient herbal medicine containing AR in typhoid fever, internal and external barriers of the eyes, diarrhea, arthralgia, beriberi, and edema. Jiang et al. (2016) searched the database management system of Puji Fang, and obtained 951 pieces of AR compound, which was used for 347 kinds of diseases. The compound containing AR has the most treatments for deficiency syndromes, including tonifying asthenia and Qi, strengthening and tonifying Yuanyang, and ear asthenia. In addition, AR is mainly indicated for typhoid fever (including miscellaneous treatment of typhoid fever, moderate cold, and the common cold), internal and external barriers (including eye blindness and bird blindness), spleen and stomach weakness (including spleen and stomach deficiency and food fatigue), diarrhea, and paralysis, jaundice, cold deficiency, toothache, vomiting, cough, hemorrhoids, phlegm, spasm, bleeding, dystocia, constipation, chancre disease, rhinorrhea, endless lochia, thirst, heatstroke, and palpitation were also indicated. The indications were classified into 43 categories in total. When AR is used for treating diseases, syndromes, and symptoms, it is compatible with different herbs. It is often compatible with Phellodendri Chinensis Cortex and Coptidis Rhizoma for clearing heat and drying dampness and with Magnoliae Officinalis Cortex and Citri Reticulatae Pericarpium for fortifying the spleen and drying dampness (Zhao et al., 2019). The compatible application of AR mainly includes drying dampness and activating spleen, dispersing dampness, removing blockage, exorcising evils, relieving depression, nourishing the stomach and supplementing deficiency, and enriching the liver to brighten the eyes. AR must be used to trap the spleen with dampness (Zhuang et al., 2015). For example, Pingwei Powder comes from AR, Magnoliae Officinalis Cortex and Citri Reticulatae Pericarpium, and it is used to remove dampness and regulate Qi. In the treatment of diarrhea with dampness, AR is associated with Citri Reticulatae Pericarpium, Poria or Plantaginis Herba, and Liuyi Powders. Zingiberis Rhizoma or Cinnamomi Cortex is also added for cold-dampness. If heat is converted into dampness stagnancy, AR is combined with Paeoniae Radix Alba, Coptidis Rhizoma and Scutellariae Radix. For people who have experience dampness and evil trapped in the spleen, abdominal pain, and diarrhea. AR harmonizes the liver and spleen when combined with Paeoniae Radix Alba, Scutellariae Radix, Cinnamomi Cortex, and Saposhnikoviae Radix. AR is good at dispersing cold and dampness and benefiting the joints, especially for dampness arthralgia (Zhuang et al., 2015). AR can induce sweat and dehumidification and dispel the evil wind. It is also compatible with Notopterygii Rhizoma et Radix and Saposhnikoviae Radix, such as Nine-Ingredient Notopterygium Decoction. For example, the extracts with single AR are used to treat severe pain and joint swelling, and they can be combined with AM and Notopterygii Rhizoma et Radix; AM, Ephedrae Herba, and Cinnamomi Ramulus; or AM, Poria, and Alismatis Rhizoma. Ermiao Powder is composed of AR and Phellodendri Chinensis Cortex. It is effective in treating phlegm arthralgia, foot dampness, and leg sores. Baihu Jia Cangzhu Decoction treats damp-temperature disease with AR being compatible with Gypsum Fibrosum and Anemarrhenae Rhizoma. However, the functions of AR in eliminating filth, promoting Qi and relieving depression have not been appreciated. AR has a fragrant smell and good nature but it is not conservative. If used to prevent diseases, AR can be worn or fumigated in a medicine bag made of AR and Acori Tatarinowii Rhizoma and Artemisiae Argyi Folium. Yueju Pill is mainly made of AR, and it can invigorate the spleen. If the spleen is healthy, the other four viscera are healthy, and the melancholia resolves itself. The Chinese traditional incense consisting mainly of AR, Phellodendri Chinensis Cortex, and Artemisiae Argyi Folium has demonstrated the advantages of Chinese medicine in “avoid poisonous gas” in the prevention of the novel coronavirus COVID-19 (Zhang et al., 2020). “AR could warm the stomach, transport food and increase appetite” when combined with Galli Gigerii Endothelium Corneum, and it can promote appetite and increase the effect of Galli Gigerii Endothelium Corneum on relieving food and guiding stagnation (Tao, 2013). The combination of AR and Poria can strengthen the spleen and stomach, promote the clearance and reduction in turbidity, regulate the acquired constitution, generate vigorous Qi and blood, nourish the viscera and limbs, and resolve the deficiency. AR can be used for the treatment of blindness, freckles, dry eyes, and other eye diseases. Ground AR and Ephedrae Herba are combined into powder, mixed with wine, and used to treat dry eyes. In Zhengzhi Zhunsheng (Wang, 1993), single AR was used to treat night blindness, while stir-fried AR combined with AM, Cicadae Periostracum, and Coptidis Rhizoma, was used in powder form to treat painful eyes and corneal opacity.

In conclusion, most of the traditional clinical functions of AR are still currently being used. However, the core viewpoints of AR dehumidification are not unified. AR is generally believed to be good at removing the dampness in upper Jiao (chest above the diaphragm, including heart, lungs, head, face), while Li Gao, the founder of “Spleen Stomach Theory” of TCM, believed that AR is an essential medicine for treating flaccidity syndrome (Li, 2007). AR could rule the dampness in the three parts. If the dampness is in the upper Jiao, dampness phlegm could be easily produced to dry the dampness and conduct phlegm. If the dampness is in the middle Jiao, the stagnation of Qi is used for diarrhea, to broaden the middle Jiao and strengthen the spleen. If the dampness is in the lower part, the foot and knee are flaccid and soft. Phellodendri Chinensis Cortex, which can strengthen the foot and knee, can be used to treat this flaccidity. AR's Qi is strong and vigorous, and the effect of sweating is optimal. The effect of invigorating the middle Jiao is weaker than that of AM. However, the research on treating flaccidity by removing the dampness of the lower Jiao is insufficient. The clinical dosage of AR is also related to the difference in the diseases and types of syndrome and symptoms. Zhao et al. (2019) collected ancient books and clinical experience and concluded that the AR dosage should be 5–25 g when drying dampness, fortifying the spleen, enriching Yin and supplying Yang to treat gynecological diseases (such as infertility, polycystic ovary syndrome, and primary dysmenorrhea). It is 12–15 g when diffusing impediment and unblocking the collaterals, clearing heat, and draining dampness to treat rheumatoid arthritis and gouty arthritis.

4. Phytochemistry

4.1. Terpenoids and their glycosides

AR is rich in volatile oil and other small polar components. The reported volatile oil components were mainly identified using GC-MS. The identified compounds in eight samples accounted for 89.36%–95.79% of the total volatile oil. Sesquiterpene is the main component of relatively high boiling point in AR volatile oil, with strong aroma and biological activity. The chemical profiles of the essential oil from A. lancea showed that wild grown plants produced mostly significant amounts of sesquiterpenes, with the top three being hinesol (68.5%), β-eudesmol (13.1%) and elemol (6.2%) (Jia et al., 2004).

Guaiacane sesquiterpenes are isolated and identified earlier from AR, and they belong to azulene derivatives, an aromatic skeleton compound composed of five and seven-membered rings. Wang et al. (2008) obtained sesquiterpenes (1–5) from A. lancea, and confirmed that the compounds have no cytotoxic activity on P388 and A549 cells through cytotoxic experiments. Kitajima et al. (2003b) isolated various water-soluble guaiacum sesquiterpenoids from the rhizomes of A. lancea and A. japonica, and analyzed the structure of the compounds (Table 2 , 7–16). Compounds 13 and 14 were isolated from A. japonica, and compounds 18–21 were isolated from A. lancea for the first time. Pharmacological experiments showed that compounds 18 and 20 have no significant inhibitory effect on protein tyrosine phosphatase-like, member b (PTPLB) and β-glucosidase. Wang et al. (2008) isolated and identified cineole sesquiterpenes from AR. Duan et al. (2008) analyzed the structure of compound 25. Endo and Hikino (1979) determined the absolute configuration of compounds 33 and 34 via circular dichroism. Endo et al. (1979) isolated atractylenolide II (37) from A. japonica for the first time and analyzed its structure. Atractylolide Ⅲ (38) was isolated from A. Japonica for the first time by Yosioka et al. (1959). And both atractylolide Ⅲ and atractylone had acaricidal activity (Meng et al., 2010). Kitajima et al. (2003a) isolated a series of eucalyptosyl sesquiterpene glycosides from AR and analyzed the chemical structure of compounds (42–50), among which compounds (43, 44, 46) were first isolated from A. lancea methanol extract, as shown in Table 2. Compounds 54–64 were isolated from 30% n-butanol fraction of A. lancea and their structures were analyzed (Xu, 2017). At present, 14 species of spirosane sesquiterpenes (Table 2, 65–78) have been found in A. lancea. Compounds 77 and 78 are irimophenol alkane sesquiterpenes. The triterpenoid saponins in AR are mainly tetracyclic triterpenoids, pentacyclic triterpenoids, and tetracyclic triterpenoids. Six triterpenoids (Table 2, 79–84) were isolated from A. lancea by Duan et al. (2008), and their chemical structures were analyzed. The information of the sesquiterpenoid biosynthesis is largely unknown. Ahmed et al. (2016) investigated the transcriptome of different tissues of A. lancea. All the enzymes that took part in the terpenoid biosynthesis and five different known sesquiterpenoids were identified via cytosolic mevalonic acid and plastidal methylerythritol phosphate pathways. Besides, 6864 simple sequence repeats were found to be great potential markers in A. lancea. Jia et al. (2016) demonstrated that endophytic bacterium-triggered ROS could directly oxidize oxygen-free sesquiterpenoids and increase the oxygenous sesquiterpenoid content and diversity in A. lancea. Takeda et al. (1996) transplanted the rhizomes of A. lancea, A. chinensis and A. koreana growing in China from 18 populations in the same experimental field. Main essential oil components: the sesquiterpenes and the polyacetylene of atractylodin were determined. The analytical data of 306 cultivated plants were compared with plants collected in their habitat. A. lancea varied significantly in the contents of the components after cultivation. Compared to this, A. chinensis had constant content values. Three types of A. lancea and two types of A. chinensis, which are distinguished by the characteristics of the components in the wild conditions, were statistically recognized after cultivation. From these results, it was determined that the geographical variation in the components of these species mainly reflects genetic variability.

Table 2.

The terpenoids and their glycosides from Atractylodis Rhizoma.

Chemical compounds Nos. Compounds name Structure Origin and Ref.
Guaiacane sesquiterpenes and their glycosides 1 4α,7α-epoxyguaiane-10α,11-diol Image 2 A. lancea (Wang et al., 2008)
2 7α,10α-epoxyguaiane-4α,11-diol Image 3
3 10β,11β-epoxyguaiane-1α, 4α-diol Image 4
4 10β,11β-epoxyguaiane-1α,4α,7α-triol Image 5
5 1-patchoulene-4α,7α-diol Image 6
6 eremanthin Image 7 A. japonica (Toda et al., 2017)
7 atractyloside A [(1S,4S,5R,7R,10R)-4,10,11,14-tetrahydroxyguai-3-one 11-O-β-D-glucopyranoside] Image 8 A. lancea (Kitajima et al., 2003b)
A. japonica (Kitajima et al., 2003a)
8 atractyloside B [(1S,3S,4S,
5R,7R,10R)-guai-3,4,10,11,14-pentol 11-O-β-D-glucopyranoside]
Image 9
9 (1S,4S,5S,7R,10R)-10,11,14-trihydroxyguai-3-one 11-O-β-D-glucopyranoside Image 10
10 (1S,4S,5R,7R,10R)-11,14-dihydroxyguai-3-one11-O-β-D-glucopyranoside Image 11
11 (1S,5R,7R,10R)-secoatractylolactone11-O-β -D-glucopyranoside Image 12
12 atractyloside A 14-O-β-D-fructofuranoside Image 13
13 10-epi-atractylosideA [(1S,4S,5R,7R,10S)-4,10,11,14-tetrahydroxyguai-3-one 11-O-β -D-glucopyranoside] Image 14
14 (1S,4S,5S,7R,10R)-10.11,14-trihydroxyguai-3-one11-O-β-D-glucopyranoside Image 15
15 (1S,4S,5S,7R,10S)-10,11,14-trihydroxyguai-3-one 11-O-β-D-glucopyranoside Image 16
16 (1S,5R,7R,10R)-secoatractylolactone11-O-β-D-glucopyranoside Image 17
17 3,4,11,14-tetrahydroxyguai-9-en-11-O-β-D-glucopyranoside Image 18 A. lancea (Chen, 2007)
18 (3R,4R,7R,10R)-2-hydroxypancherione-11-O-β-D-glucopyranoside Image 19 A. lancea (Xu et al., 2016a)
19 (1S,7R,10R)-11,15-dihydroxy-4-guaien-3-one 11-O-β-D-glucopyranoside Image 20
20 (1R,7R,10S)-10,11-dihydroxy-4-guaien-3-one 11-O-β-D-glucopyranoside Image 21
21 (5R,7R,10S)-3-O-β-D-glucopyranosylisopterocarpolone-11-O-β-D-apiofur-anosyl-(1 → 6)-β-D-glucopyranoside Image 22 A. lancea (Jiang et al., 2018)
Cineole sesquiterpenes and their glycosides 22 eudesm-4 (15)-ene-7α,11-diol Image 23 A. lancea (Wang et al., 2008)
23 eudesm-4 (15),7-diene-9α,11-diol Image 24
24 eudesm-4 (15),7-diene-11-ol-9-one Image 25
25 β-eudesmol Image 26 A. lancea (Duan et al., 2008)
26 14-hydroxy-isopterocarpolone Image 27 A. lancea (Kamauchi et al., 2015)
27 kudtdiol Image 28
28 (11R)-2,11,12-trihydroxy-β-selinene Image 29
29 2,11,13-trihydroxy-β-selinene Image 30
30 3α-hydroxy-pterocarpol Image 31
31 4 (15),11-eudesmadien Image 32 A. lancea (Van Minh et al., 2004)
32 pterocarpol [(2S,4aS,6R,8aS)-6-(2-hydroxypropan-2-yl)-8a-methyl-4-methylidene-1,2,3,4a,5,6,7,8-octahydronaphthalen-2-ol] Image 33 A. lancea (Yahara et al., 1989)
33 eudesma-4(14),7(11)-dien-8-one Image 34 A. japonica (Endo and Hikino, 1979)
34 (+) eudesma-4(14),7(11)-dien-8-one Image 35
35 atractylon [(4aS,8aR)-3,8a-Dimethyl-5-methylene-4,4a,5,6,7,8,8a,9-octahydronaphtho [2,3-b]furan] Image 36 A. lancea (Resch et al., 1998)
36 atractylenolide Ⅰ Image 37 A. lancea (Resch et al., 1998); A. chinensis (Meng et al., 2010)
A. japonica (Endo et al., 1979)
37 atractylenolide II Image 38
38 atractylenolide III Image 39 A. japonica (Yosioka et al., 1959); A. lancea (Hikino et al., 1964)
39 atractylenolide IV Image 40 A. lancea (Hikino et al., 1964)
40 eudesm-7 (11)-en-4-ol Image 41 A. japonica (Hong et al., 2012)
41 α-bisabolol Image 42 A. lancea (Takeda et al., 2001)
42 atractyloside C Image 43 A. lancea (Kitajima et al., 2003b)
43 atractyloside G Image 44
44 atractyloside G 2-O-β-D-glucopyranoside Image 45
45 atractyloside D Image 46
46 atractyloside I Image 47
47 atractyloside E Image 48
48 (5R,7R,10S)-isopterocarpolone-β-D-glucopyranoside [(5R,7R,10S)-11-hydroxyeudesm-3-en-2-one11-O-β-D-glucopyranoside] Image 49
49 cis-atractyloside I Image 50
50 (2R,3R,5R,7R,10S)-Atractyloside G 2-O-β-Glucopyranoside Image 51
51 (3S)-3-hydroxyatractylenolide III 3-O-D-glucopyranoside [(3S,5R,8R,10R)-3,8-dihydroxyeudesma-4(15),7(11)-diene-8,12-olide 3-O-β-D-glucopyranoside] Image 52 A. japonica (Kitajima et al., 2003a)
52 officinoside C Image 53
53 atractyloside F Image 54 A. lancea (Yahara et al., 1989)
54 (3S,4R,5S,7R)-13-hydroxylhinesolone-11-O-β-D-glucopyranoside Image 55 A. lancea (Xu, 2017)
55 (5R,7R,10S)-3-hydroxylisopterocarpolone-3-O-β-D-glucopyranoside Image 56
56 (5R,7R,10S)-6″-O-β-D-apiofuranosylatractyloside I Image 57
57 (5R,7R,10S)-6″-O- acetylatractyloside I Image 58
58 (5R,7R,10S)-6′-O- acetylatractyloside I Image 59
59 (5R,7R,10S)- isopterocarpolone-11-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 60
60 (5R,7R,10S)-14-hydroxylisopterocarpolone-11-O-β-D-glueopyranoside Image 61
61 (5R,7R,10S)-14-carboxylisopterocarpolone-11-O-β-D-glucopyranoside Image 62
62 (2S,7R,10S)-2-hydroxylcarissone-11-O-β-D-glucopyranoside Image 63
63 (2R,7R,10S)-2-hydroxylcarissone-11-O-β-D-glucopyranoside Image 64
64 (1R,7R,10R)-1-hydroxylcarissone-11-O-β-D-glucopyranoside Image 65
Spirosane sesquiterpenes and irimophenone sesquiterpenes of vetiver and their glycosides 65 hinesol Image 66 A. lancea (Hashimoto et al., 1999)
66 hinesolone Image 67 A. lancea (Long et al., 2018)
67 2-oxo-12-hydroxy-hinesol Image 68 A. lancea (Kamauchi et al., 2015)
68 2-oxo-15-hydroxy-hinesol Image 69
69 (7R)-3,4-dehydrohinesolone-11-O-β-D-glucopyranoside Image 70 A. lancea (Xu et al., 2016a)
70 (7R)-3,4-dehydrohinesolone-11-O-β-D-glucopyranosideosideyranoside-hydroxyeud Image 71
71 (5R,7R)-14-hydroxy-3,4-dehydrohinesolone-11-O-β-D-glueopyranoside Image 72
72 (5R,7R)-14-hydroxy-3,4-dehydrohinesolone-11-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 73
73 (5R,7R)-14-hydroxy-3,4-dehydrohinesolone-14-O-β-D-xylopyranoside Image 74
74 (4S,5S,7R)-15-hydroxylhinesolone-15-O-β-D-xylopyranoside Image 75
75 (4S,5S,7R)-14-hydroxylhinesolone-14-O-β-D-xylopyranoside Image 76
76 (3S,4R,5S,7R)-13-hydroxylhinesolone-11-O-β-D-glucopyranoside Image 77
77 (3S,4R,5R,7R)-3,11-dihydroxy-11,12-dihydronootkatone-11-O-β-D-glucopyranoside Image 78
78 (3S,4R,5S,7R)-3,4,11-trihydroxy-11,12-dihydronootkatone-11-O-β-D-glucopyranoside Image 79
Triterpenoids and steroids 79 stigmasterol 3-O-β-D-glucopyranoside Image 80 A. lancea (Duan et al., 2008)
80 daucosterol [3β-(β-D-glucopyranosyloxy)stigmast-5-en] Image 81
81 stigmasterol [(3S,8S,9S,10R,13R,14S,17R)-17-[(2R,5R)-5-Ethyl-6-methyl-2-heptanyl]-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta [a]phenanthren-3-ol] Image 82
82 β-sitosterol [(3S,8S,9S,10R,13R,14S,17R)-17-[(E,2R,5S)-5-ethyl-6-methylhept-3-en-2-yl]-10,13-dimethyl-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1H-cyclopenta [a]phenanthren-3-ol] Image 83
83 traxerol acetate [(3S,4aR,6aR,8aR,12aR,12bS,14aR,14bR)-4,4,6a,8a,11,11,12b,14b-Octamethyl-1,2,3,4,4a,5,6,6a,8,8a,9,10,11,12,12a,12b,13,14,14a,14b-icosahydro-3-picenyl acetate] Image 84
84 ϕ-traxasteryl acetate (heterolupeol) Image 85
85 oleanolic acid [(3β)-3-hydroxy-olean-12-en-28-oic acid] Image 86

4.2. Alkynes and their glycosides

Alkynes include unsaturated alcohols, ketones, acids, esters, benzene, furans and other functional groups in addition to unsaturated triple bonds and double bonds. The triple and double bonds are relatively stable, and thus they can be extracted, separated, and identified by conventional phytochemical techniques. The polyacetylenes were classified by their chemical backbones and the species of genus Atractylodes where the compounds were contained were presented. The polyacetylenes of diene-diyne types were classified into furan-ring-attached, alcohol-attached, acetyl-attached, and two more functional groups-attached compounds. A part of atractylodin-related acetylenes, which contained a furan-ring at the terminal carbon, consisted of nine carbons. Among them, atractylodin was the most reported and it is contained in A. lancea, A. chinensis, A. japonica and A. koreana (Kim, 2016). Other polyacetylenes comprised 14 carbons with double bonds at C-6 and C-12, and triple bonds at C-8 and C-10. Diacetyl atractylodiol (100) comprised 13 carbons and (2E, 8E)-decadiene-4,6-diyne-1,10-diol-1-O-β-D-glucopyranoside (101) was the only polyacetylene consisted of 10 carbons and glucosamine. However, no diene-diyne types were reported to be contained in AM (Table 3 , 86–122). As shown in Table 3, most compounds of triene-diyne type polyacetylenes (123–162) originated from A. lancea and possessed 14 carbons with triple bonds at C-4 and C-6, double bonds at C-2, C-8, and C-12. Senecioyloxy, isovaleryloxy, methylpropionyl, methylbutyryl, and acetoxy were the main functional groups of these compounds and were positioned at C-12 or C-14. Another 14 carbon polyacetylenes with triple bonds at C-8 and C-10, double bonds at C-4, C-6, and C-12. And the main functional groups were isovaleryloxy, acetoxy, methylbutyryloxy, or senecioyloxy. Monoene-diyne type polyacetylenes and their glycosides were reported to be a single double bond and two triple bonds, consisting of 9, 10, 12, and 13 carbons, with furan ring or sugars attached (Table 3, 163–175). Kitajima et al. (2003b) obtained enyne-derived glycoside (Table 3, 169) from the water-soluble components of AR for the first time and determined their skeleton structure. Besides, several polyacetylenes with C10 chain alkyne, C13 chain alkyne, C14 chain alkyne, furan cycloalkyne, and thiophene cycloalkyne were isolated from 30% ethanol fraction of the n-butanol fraction of Cangzhu 80% ethanol extracts (Xu, 2017). Resch et al. (1998) showed that compounds 184 and 185 have 5-lipoxygenase and cyclooxygenase activities. Chen et al. (2012) revealed that compounds 86, 88107, and 187188 have anti-Escherichia coli, Staphylococcus aureus, Bacillus subtilis, and Candida albicans activities. A. lancea essential oil data were first collected from the literature referring to the bioinformatic methods to study the variation rules and classify the chemotype of A. lancea essential oil. Then, the essential oil among different populations or individuals in the same population was compared. (1) Large varieties in A. lancea essential oil, which involved components and their content, were found among populations or even among the individuals in the same population. (2) The total essential oil and the six major components in it (elemol, atractylon, hinesol, β-eudesmol, selina-4(14), 7(11)-dienone and atractylodin) varied continuously in different geographical populations. (3) The total essential oil of A. lancea decreased from South China to North China. Two chemotypes of A. lancea essential oil were mainly found in China, one was Hubei chemotype (HBA), which has hinesol and β-eudesmol as the main components, with a little atractylodin and almost without atractylon and selina-4(14), 7(11)-dienone. The other type was Mt. Maoshan chemotype (MA), which is characterized by atractylon and atractylodin as main components with little hinesol, β-eudesmol and other components (Guo et al., 2008).

Table 3.

The alkynes and their glycosides from Atractylodis Rhizoma.

Chemical compounds Nos. Compounds name Structure Origin and Ref.
Polyacetylenes of diene-diyne types and their glycosides 86 atractylodin
2-[(1E,7E)-nona-1,7-dien-3,5-diynyl]furan
Image 87 A. lancea (Takeda, 1994);
A. chinensis (Nishikawa et al., 1975);
A. lancea (Kawanishi, 1994);
A. koreana (Kawanishi, 1994)
87 (1Z)-atractylodin
2-[(1Z,7E)-nona-1,7-dien-3,5-diynyl]furan
Image 88 A. lancea (Resch et al., 2001)
88 9-nor-atractylodin Image 89 A. lancea (Chen et al., 2012)
89 atractylodinol [(2E,8E)-9-(furan-2-yl)nona-2,8-dien-4,6-diyn-1-ol] Image 90 A. lancea, A. chinensis (Nishikawa et al., 1976)
90 (1Z)-atractylodinol [(2E,8Z)-9-(furan-2-yl)nona-2,8-dien-4,6-diyn-1-ol] Image 91 A. chinensis (Meng et al., 2010)
91 acetylatractylodinol
[(2E,8E)-9-(furan-2-yl)nona-2,8-dien-4,6-diynyl] acetate
Image 92 A. lancea, A. chinensis (Nishikawa et al., 1976)
92 (1Z)-acetylatractylodinol
[(2E,8Z)-9-(furan-2-yl)nona-2,8-dien-4,6-diynyl] acetate
Image 93 A. lancea (Resch et al., 2001)
93 (6E,12E)-tetradecadiene-8,10-diyne-1,3-diol Image 94 A. chinensis (Meng et al., 2010);
A. japonica (Jeong et al., 2010);
Atractylodis Rhizoma (Sakurai et al., 1994b)
94 (6E,12Z)-tetradecadiene-8,10-diyne-1,3-diol Image 95 A. chinensis (Meng et al., 2011)
95 (6Z,12Z)-tetradecadiene-8,10-diyne-1,3-diol Image 96
96 (6E,12E)-tetradecadiene-8,10-diyne-1,3-diol diacetate Image 97 A. chinensis (Meng et al., 2010)
97 (6E,12E)-3-acetoxytetradeca-6,12-dien-8,10-diyn-1-ol Image 98
98 (6E,12E)-1-acetoxytetradeca-6,12-dien-8,10-diyn-3-ol Image 99
99 1,4-acetoxytetradeca-6,12-diene-8,10-diyne Image 100 Atractylodis Rhizoma (Sakurai et al., 1994a)
100 diacetyl atractylodiol [(5E,11E)-3-acetyloxytrideca-5,11-dien-7,9-diynyl] acetate Image 101 A. japonica (Yosioka et al., 1974)
101 (2E,8E)-decadiene-4,6-diyne-1,10-diol 1-O-β -D-glucopyranoside Image 102 A. lancea (Kitajima et al., 2003b)
102 atractylodinol [1-(2-Furyl)-(1E,7E)-nonadiene-3,5-diyne-9-ol] Image 103 A. lancea (Chen et al., 2012)
103 1-(2-Furyl)-(1E,7E)-nonadiene-3,5-diyne-9-al Image 104
104 1-(2-Furyl)-(1E,7Z)-nonadiene-3,5-diyne-9-ol Image 105
105 1-(2-Furyl)-(1E,7E)-nonadiene-3,5-diyne-9-yl benzoate Image 106
106 1-(2-Furyl)-(1E,7E)-nonadiene-3,5-diyne-9-yl 4-methylbenzoate Image 107
107 1-(2-Furyl)-(1E,7E)-nonadiene-3,5-diyne-9-acid Image 108
108 (1Z)-atractylodinol 1-(2-Furyl)-(1Z,7E)-nonadiene-3,5-diyne-9-ol Image 109 A. lancea (Resch et al., 2001)
109 (1Z)-acetylatractylodinol 1-(2-Furyl)-(1Z,7E)-nonadiene-3,5-diyne-9-yl acetate Image 110
110 (4E,10E)-dodeca-4,10-dien-6,8-diyne-1,3-diyl diacetate Image 111 A. lancea (Meng et al., 2010)
111 (6E,12E)-aetradecadiene-8,10-diyne-1,3-diol Image 112
112 (2E,8Z)-deca-2,8-diene-4,6-diyne-1,10-diol-1- O-β-D-glucopyranoside Image 113 A. lancea (Xu, 2017)
113 (2E,8E)-deca-2,8-diene-4,6-diyne-1,10-diol-1-O-β-D-apiofuranosyl-(1 → 6) -β-D-glucopyranoside Image 114
114 (2E,8Z)-deca-2,8-diene-4,6-diyne-1,10-diol-1-O-β-D-apiofuranosyl-(1 → 6) -β-D-glucopyranoside Image 115
115 (8R,9S)-2E,10Z-tridecadiene-4,6-diyne-8,9,12,13-triol-9-O-β-D-glucopyranoside Image 116
116 (8R,9S)-2E,10Z-tridecadiene-4,6-diyne-8,9,12,13-triol-8-O-β-D-glucopyranoside Image 117
117 (8R,9S)-2E,10Z-tridecadiene-4,6-diyne-8,9,12,13-triol Image 118
118 (2Z,8E)-deca-2,8-diene-4,6-diyne-1,10-diol-1-O-β-D-glucopyranoside Image 119
119 (2E,8E,10R)-tridecatriene-4,6-diyne-1,10,11,12,13-pentol-10-O-β-D-glucopyranoside Image 120
120 (2E,8E,10R)-tridecane-2,8-diene-4,6-diyne-1,10, 13-di-O-β-D-xylopyranoside Image 121
121 (3R,8E,10E)-tetradecadiene-4,6-diyne-3,12,14-triol-3-O-β-D-glucopyranoside Image 122
122 (2E,8E,12S)-tetradecadiene-4,6-diyne-1,10,14-triol-1-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 123
Triene-diyne types of atractylodes polyacetylenes and their glycosides 123 (1,3Z,11E)-Tridecatriene-7,9-diyne-5-hydroxyl-6-O-β-D-glucopyranoside Image 124 A. lancea (Ji et al., 2010)
124 atractyloyne [(3S,4E,6E,12E)-1-isovaleryloxy-tetradeca-4,6,12-triene-8,10-diyne-3,14-diol Image 125 A. chinensis (Nakai et al., 2005); Atractylodis Rhizoma (Kim, 2016)
125 12,14- diacetate- 2E,8E,10E- trien- 4,6-diyn-1-ol Image 126 Atractylodis Rhizoma (Kim, 2016)
126 (4E,6E,12E)-tetradecatriene-8,10-diyne-1,3-diyl diacetate Image 127 A. lancea (Resch et al., 2001)
127 (4E,6E,12E)-Tetradecatriene-8,10-diyne-1,3-diol Image 128
128 (4E,6E,12E)-Tetradecatrien-8,10-diyn-1-ol Image 129
129 (4E,6E,12E)-3-isovaleryloxy-tetradeca-4,6,12-triene-8,10-diyne-1,14-diol Image 130
130 erythro-(1,3Z,11E)-tridecatriene-7,9-diyne-5,6-diyl-diacetate Image 131
131 erythro-(1,5E,11E)-tridecatriene-7,9-diyne-3,4-diacetate Image 132 A. lancea (Lehner et al., 1997)
132 threo-(1,5E,11E)-tridecatriene-7,9-diyne-3,4-diacetate Image 133
133 (1,5E,11E)-trideca-1,5,11-trien-7,9-diyne-3,4-diacetate Image 134 Atractylodis Rhizoma (Kim, 2016)
134 (3Z,5E,11E)-tridecatriene-7,9-diynyl-1-O-(E)-ferulate Image 135 A. lancea (Resch et al., 2001)
135 (3E,5E,11E)-tridecatriene-7,9-diyne-l,2-diacetate Image 136 A. lancea (Lehner et al., 1997)
136 (3Z,5E,11E)-tridecatriene-7,9-diyne-1,2-diacetate Image 133 A. lancea (Resch et al., 2001)
137 (3E,5Z,11E)-tridecatriene-7,9-diyne-1,2-diacetate Image 138 A. lancea (Lehner et al., 1997)
138 (2Z,4E,10E)-trideca-2,4,10-trien-6,8-diynyl acetate Image 139 Atractylodis Rhizoma (Kim, 2016)
139 (1,5E,11E)-tridecatriene-7,9-diyne-3,4-diacetate Image 140 A. lancea (Resch et al., 2001)
140 (3E,5Z,11E)-tridecatriene-7,9-diyne-1,2-diyl diacetate Image 141
141 (4E,6E,12E)-aetradecatrien-8,10-diyn-1-ol Image 142 A. lancea (Meng et al., 2010)
142 (4E,6E,12E)-tetradeca-4,6,12-trien-8,10-diyne-1,3,14-triol Image 143 Atractylodis Rhizoma (Kim, 2016)
143 (4E,6E,12E)-tetradecadiene-triene-8,10-diol Image 144 A. lancea (Meng et al., 2010)
144 (5E,11E)-trideca-1,5,11- trien-7,9-diyne-3,4- diyldiacetate Image 145 Atractylodis Rhizoma (Kim, 2016)
145 (4E,6E,12E)-1-acetoxy-3-isovaleryloxy-4,6,12-trien-8,10-diyn-14-ol Image 146
146 (4E,6E,12E)-1-acetoxy-3-(2-methylbutyryloxy)-4,6,12-trien-8,10-diyn-14-ol Image 147
147 (8S,9R)-2E,10Z,12-tridecadiene-4,6-diyne-1,8,9-triol-8-O-β-D-glucopyranoside Image 148 A. lancea (Xu, 2017)
148 (8S,9R)-2E,10Z,12-tridecadiene-4,6-diyne-1,8,9-triol-8-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 149
149 (8S,9R)-2E,10Z,12-tridecadiene-4,6-diyne-1,8,9-triol-9-O-β-D-glucopyranoside Image 150
150 (8S,9R)-2E,10Z,12-tridecadiene-4,6-diyne-1,8,9-triol-9-O-β-D-apiofuranosyl-(1 → 6) -β-D-glucopyranoside Image 151
151 (10R,11R)-2R,8E,12-tridecatriene-4,6-diyne-1,10,11-triol-10-O-β-D-glucopyranoside Image 152
152 (10R,11R)-2E,8E,12-tridecatriene-4,6-diyne-1,10,11-triol-1-O-β-D-glucopyranoside Image 153
153 (10R,11R)-2E,8E,12-tridecatriene-4,6-diyne-1,10,11-triol-1,10-O-di-β-D-glucopyranoside Image 154
154 (10R,11R)-2E,8E,12-tridecatriene-4,6-diyne-1,10,11-triol-10-O-β-D- apiofuranosyl-(1 → 6) -β-D-glucopyranoside Image 155
155 (10R,11S)-2R,8E,12-tridecatriene-4,6-diyne-1,10,11-triol-10-O-β-D-glucopyranoside Image 156
156 (10R,11S)-2E,8E,12-tridecatriene-4,6-diyne-1,10,11-triol-10-O-β-D- apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 157
157 (10S,11R)-2E,8E,12-tridecatriene-4,6-diyne-1,10,11-triol-10-O-β-D-glucopyranoside Image 158
158 (2E,8E,10E,12R)-tridecatriene-4,6-diyne-1,12,13-triol-1,12-di-O-β-D-glucopyranoside Image 159
159 (2E,8E,10E,12R)-tridecatriene-4,6-diyne-1,12,13-triol-1-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 160
160 (8E,10E)-tetradecadiene-4,6-diyne-3,12,14-triol-3-O-β-D-glucopyranoside Image 161
161 (2E,8E,10E,12R)-tetradeca-2,8,10-triene-4,6-diyne-1,2,14-triol-1-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 162
162 (2E,8E,10E,12R)-tetradeca-2,8,10-triene-4,6-diyne-1,12,14-triol-1-O-β-D-glucopyranoside Image 163
Monoene-diyne types of aractylodes polyacetylenes and their glycosides 163 1-(2-Furyl)-(7E)-nonene-3,5-diyne-1,2-diacetate Image 164 A. lancea (Lehner et al., 1997); Atractylodis Rhizoma (Kim, 2016)
164 (2E)-decene-4,6-diyne-1,8-diol8-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 165 A. lancea (Kitajima et al., 2003b)
165 (2E,8R)-decane-4,6-diyne-1,8-diol-8-O-β-D-glucopyranoside Image 166 A. lancea (Xu, 2017)
166 (2E,8S)-decane-4,6-diyne-1,8-diol-8-O-β-D-glucopyranoside Image 167
167 (2E,8R)-decene-4,6-diyne-1,8-diol-1-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 168
168 (2E,8R)-decane-4,6-diyne-1,8-diol-O-di-β-D-glucopyranoside Image 169
169 (E)-deca-2-ene-4,6-diyne-1,10-diol-1-O-β-D-glucopyranoside Image 170
170 (E)-deca-2-ene-4,6-diyne-1,10-dio1-1-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 171
171 (2E,10S)-tridecane-2-ene-4,6-diyne-1,10,13-triol-1,13-di-O-β-D-xylopyranoside Image 172
172 (2E,10S)-tridecatriene-4,6-diyne-1,12,13-triol-13-O-β-D-glucopyranoside Image 173
173 (2E,10S)-tridecatriene-4,6-diyne-1,12,13-triol-10-O-β-D-glucopyranoside Image 174
174 (8R,9R)-8,9-dihydroxylatractylodinol-8-O-β-D-glucopyranoside Image 175
175 (8R,9R)-8,9-dihydroxylatractylodinol-9-O-β-D-glucopyranoside Image 176
176 (2E,10S)-tetradecadiene-4,6-diyne-1,10,14-triol-10-O-β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 177
177 1-(2-furyl)-(7E)-non-ene-3,5-diyne-l,2-diacetate Image 178
Other alkynes and their glycosides 178 (10R)-atracthioenyneside A Image 179 A. lancea (Xu, 2017)
179 (10R)-atracthioenyneside B Image 180
180 (10S,11R)-atracthioenyneside C Image 181
181 (10S,11R)-atracthioenyneside D Image 182
182 (10R,11S)-atracthioenyneside E Image 183
183 (8S)-decane-4,6-diyne-1,8-diol-O-β-D-glucopyranoside Image 184
184 2,8-dimethyl-6-hydroxy-2-(4-methyl-3-pentenyl)-2H-chromene Image 185 A. lancea (Resch et al., 1998)
185 2-[(2E)-3,7-dimethyl-2,6-octadienyl]-6-methyl-2,5-cyclohexadiene-1,4-dione Image 186
186 2-[(2′E)-3′,7′-dimethyl-2′,6′-octadienyl]-4-methoxy-6-methylphenol Image 187
187 bis (5-[(1E,7E)-nona-1,7-dien-3,5-diyn-1-yl)]furan-2-yl)methane Image 188 A. lancea (Chen et al., 2012)
188 bis (5-[(1E,7E)-nona-1,7-dien-3,5-diyn-1-yl)]furan-2-yl)isopropane Image 189
189 (4E,6E,12E)-1-acetoxy-3-senecloyloxytetradeca-4,6,12-trien-8,10-diyn-14-ol Image 190 Atractylodis Rhizoma (Kano et al.)

4.3. Aromatic glycosides and acyl sugar compounds

Naturally occurring carbohydrates with aromatic aglycones are a class of important compounds, such as antibiotics (e.g., vancomycin and chromomycin), and some plant-derived phenolic hydroxyl compounds are also glycosylated (e.g., arbutin, sennoside A, and glucoside). Kitajima et al. (2003b) isolated compounds (Table 4 , 190–198) from A. lancea and analyzed its structure. Some known compounds (201–203) of aromatic glycosides were identified from A. lancea by Feng et al. (2018). A. lancea also contains acyl sugar derivatives (Table 5 ).

Table 4.

The aromatic glycosides and its structure of Atractylodis Rhizoma.

Chemical compounds Nos. Compounds name Structure Origin and Ref.
Aromatic glycosides 190 4-hydroxy-3-methoxyphenol-β-D-glucopyranoside Image 191 A. lancea (Kitajima et al., 2003b)
191 seguinoside B,4-hydroxyphenyl 1-O-β-D - apiopyranosyl-(1 → 6)-β-D-glucopyranoside] Image 192
192 syringin [4-[(1E)-3-Hydroxyprop-1-en-1-yl]-2,6-dimethoxyphenyl-β-D-glucopyranoside] Image 193
193 4-hydroxy-3-methoxyphenol-β-D-apiopyranosyl-(1 → 6)-D-glucopyranoside Image 194
194 4-hydroxy-3-methoxyphenyl-β-xylopyranosyl-(1 → 6)-β-glucopyranoside Image 195
195 icariside F2 [benzyl β-D-apiopyranosyl-(1 → 6)-β-D-glucopyranoside] Image 196
196 icariside D1 [phenethyl β-D-apiopyranosyl-(1 → 6)-β-D-glucopyranoside] Image 197
197 phenethyl α-L-rhamnopyranosyl-(1 → 6)-β-D-glucopyranoside Image 198
198 scopoletin-D-xylopyranosyl-(1 → 6)-D-glucopyranoside Image 199
199 p-hydroxybenzoic acid-4-O-β-D-glucopyranosyl-(1 → 3)-α-L-rhamnopyranoside Image 200 A. lancea (Xu et al., 2016b)
200 vanillic acid-4-O-β-D-glucopyranosyl-(1 → 3)-α-L-rhamnopyranoside Image 201
201 1,3-di-O-caffeoylquinic acid Image 202 A. lancea (Feng et al., 2018)
202 chlorogenic acid (1S,3R,4R,5R)-3-[3-(3,4-dihydroxyphenyl)prop-2-enoyloxy]-1,4,5-trihydroxycyclohexanecarboxylic acid Image 203
203 5-O-feruloylquinic acid (1R,3R,4S,5R)-1,3,4-trihydroxy-5-[(2E)-3-(4-hydroxy-3-methoxyphenyl)-prop-2-enoyl]-oxycyclohexane-1-carboxylic acid Image 204

Table 5.

The acyl sugar compounds and its structure of Atractylodis Rhizoma.

Chemical compounds Nos. Compounds name Structure Origin and Ref.
Acyl sugar compounds 204 2,6,3′,6′-tetra (3-methylbutanoyl) sucrose Image 205 A. lancea (Ina, 2009)
205 2,4,3′,6′-tetra (3-methylbutanoyl) sucrose Image 206
206 2,6,3′,4′-tetra (3-methylbutanoyl) sucrose Image 207
207 2,4,3′,4′-tetra (3-methylbutanoyl) sucrose Image 208
208 2,1′,3′,6′-tetra (3-methylbutanoyl) sucrose Image 209
209 3′,4′,6′-tris (3-methylbutanoyl)-1'-(2-methylbutanoyl) sucrose Image 210

4.4. Other compounds

AR also contains other compounds (Table 6 ), including osthol (210), amino acids (212, 213), and other water-soluble compounds. Duan et al. (2008) first isolated compound (219) from A. lancea. Some new compounds were also isolated from A. lancea, including lignans, phenolic acids, and pyrazines. An intestinal immune system modulating arabino-3,6-galactan, containing shorter (AF-GN-1 and 3), and longer (AF-GN-2) oligosaccharide fractions were isolated from A. lancea (Taguchi et al., 2004). AF-GN-3 mainly contains eight oligosaccharides (oligo-1–8) and the monosaccharides (Ara and Gal). AF-GN-2 contains oligosaccharides (oligo-9–15) and small proportions of oligo-3–8. It also contains a large amount of the fraction of longer oligosaccharides. An acidic polysaccharide (ALP-3) and a neutral polysaccharide (ALP-1) were purified from A. lancea (Qin et al., 2019) and the results showed that ALP-1 exhibits a linear backbone composed of (2 → 1)-linked β-D-fructofuranose. The backbone of ALP-3 was elucidated as →4)-GalAp-(1 → 3, 4)-Rhap-(1→, and with branch chain substituted at O-3 position of →3, 4)-Rhap-(1 → . The branch chain consists of →3, 5)-Araf-(1→, →5)-Araf-(1→, and Araf-(1 → . A series of oligo-saccharides DP 3–15 were separated from A. lancea and identified as FOS, which consisted of linear β-(2, 1)-linked fructofuranosyl units mostly carry a terminal single α-(1–2)-linked α-glucopyranosyl unit (Zhuang et al., 2019). The ACPS was extracted via ultrasonic method in enzymolysis A. lancea. ACPS structure can be expressed as α-D-Glcp-(1 → 2)-[β-D-Fruf-(1 → 2)-β-D-Fruf-]n-(1 → 2)-β-D-Fruf. Atractylis polysaccharide could inhibit the proliferation of human ovarian cancer, liver cancer in cellular level (Xu, 2015).

Table 6.

The other compounds and its structure of Atractylodis Rhizoma.

Chemical compounds Nos. Compounds name Structure Origin and Ref.
Other compounds 210 osthol Image 211 A. lancea (Resch et al., 1998)
211 5-hydroxymethyl furaldehyde Image 212 A. lancea (Meng et al., 2010)
212 L-phenylalanine Image 213 A. lancea (Kitajima et al., 2003b)
213 D-tryptophan [(R)-α-amino-3-indolepropionic Acid] Image 214
214 wogonin [5,7-dihydroxy-8-methoxyflavone] Image 215 A. chinensis (Jin, 2002)
215 3-methoxy-4-hydroxybenzoic acid Image 216 A. japonica (Li et al., 2002)
216 3,5-dimethoxy-4-hydroxybenzoic acid Image 217
217 palmitic acid Image 218 A. lancea (Resch et al., 2001)
218 atractylochromene [2,8-dimethyl-2-(4-methyl-3-penten-1-yl)-2H-chromen-6-ol] Image 219 A. lancea (Resch et al., 1998)
219 trans-2-hydroxyisoxypropyl-3-hydroxy-7-isopentene-2,3-dihydrobenzofuran-5-carboxylic acid Image 220 A. lancea (Duan et al., 2008)
220 2-[(2′E)-3′,7′-dimethyl-2′,6′-octadienyl]-4-methoxy-6-methylphenol Image 221 A. lancea (Resch et al., 2001)
221 5-hydroxymethyl furaldehyde Image 222 A. lancea (Meng et al., 2010)
222 vanillic acid (4-Hydroxy-3-methoxy-benzoic acid) Image 223 A. lancea, A. chinensis (Liang et al., 2002)
223 diethyl phthalate Image 224 A. lancea (Yang, 2007)
224 3-methyl-2-butenyl β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 225 A. lancea (Kitajima et al., 2003b)
225 isopropyl β-D-apiofuranosyl-(1 → 6)-β-D-glucopyranoside Image 226
226 Oligo-1 β- D-Galp-(1 → 6)-D-Galp A. lancea (Taguchi et al., 2004)
227 Oligo-2 β- D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-D-Galp
228 Oligo-3 β- D-Galp-(1 → 6)- β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp
229 Oligo-4 β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)- β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-D-Galp
230 Oligo-5 β- D-Galp-(1 → 6)-β-D-Galp-(1 → 6)- β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-D-Galp
231 Oligo-6 Not determined
232 Oligo-7 β- D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β- D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-D-Galp
233 Oligo-8 β- D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)- β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-β-D-Galp-(1 → 6)-D-Galp
234 ALP-1 linear backbone composed of (2 → 1)-linked β-D-fructofuranose A. lancea (Qin et al., 2019)
235 ALP-3 →4)-GalAp-(1 → 3,4)-Rhap-(1→, and with branch chain substituted at O-3 position of →3,4)-Rhap-(1 → . The branch chain consists of →3,5)-Araf-(1→, →5)-Araf-(1→, and Araf-(1→
236 FOS linear β-(2,1)-linked fructofuranosyl units mostly carry a terminal single α-(1–2)-linked α-glucopyranosyl unit Atractylodis Rhizoma (Zhuang et al., 2019)
237 ACPS α-D-Glcp-(1 → 2)-[β-D-Fruf-(1 → 2)-β-D-Fruf-]n-(1 → 2)-β-D-Fruf A. lancea (Xu et al., 2016)

Overall, more than 200 compounds have been isolated and identified from AR, and the majority of them are volatile oil. Some volatile components of AR were identified only via GC-MS alone by comparison with the databases which were only tentative identification and could not give unambiguous structures. These kinds of compounds were not listed in the tables. Transcriptomic analyses were furthermore performed via RNA-Seq to reveal the molecular mechanism regulating sesquiterpenoid formation of A. lancea. A total of 857 unigenes were identified to be involved in sesquiterpenoid formation via analysis of differentially expressed genes. A putative sesquiterpenoid biosynthetic pathway was proposed, including mevalonate, 2-C-methyl-d-erythritol-4-phosphate, sesquiterpene synthase, cytochrome P450, and the peroxisome pathway (Chen et al., 2017). However, increasing studies showed that macromolecular substances (e.g., polysaccharides and proteins) play an indispensable role in the quality formation of TCM (Zhang et al., 2020). The biomacromolecules in AR have wide bioactivity-related efficacy but yet to be fully appreciated compared with small molecule components.

5. Pharmacology

Modern pharmacological studies supported the broad pharmacological effects of AR on various diseases. The pharmacological activities of A. lancea have previously been reviewed (Jun et al., 2018; Koonrungsesomboon et al., 2014). In vitro or vivo studies on animal models demonstrated promising activities of the crude extracts (ethanolic, water, benzene, and methanol) of A. lancea and its major constituents (β-eudesmol, atractylodin, atractylon, and hinesol) on cancers, inflammation, fever and pain, gastrointestinal system, nervous system, cardiovascular system, and various microorganisms (Staphylococcus aureus, E. coli, C. albicans, Saccharomyces cerevisiae and Rhodotorula glutinis). The pharmacological activities of AR and its constituents on the gastrointestinal system (delay of gastric emptying, stimulation of intestinal motility, inhibition of gastric secretion, and antiulcer property) strongly supported their clinical use for the alleviation of digestive symptoms in traditional medicine (Kimura and Sumiyoshi, 2012; Plengsuriyakarn et al., 2012).

5.1. Improving gastrointestinal function

In the system of TCM, spleen deficiency plays an important role in the development of gastrointestinal dysfunction. Ancient Chinese doctors believed that AR can be used for dampness blocking, abdominal distention, and diarrhea. A. japonica has been commonly used to treat the gastrointestinal (GI) disorders in Korean traditional medicine. Modern researches also found that AR could protect the intestinal tract and promote intestinal movement and has anti-diarrhea activity through anti-inflammation (Deng et al., 2016). AR extracts have been shown to delay gastric emptying and stimulate small intestinal motility in a dose-dependent manner. The extract (1000 mg/kg) could inhibit the intestinal dyskinesia caused by atropine, and the extract (500 mg/kg) could block the gastric emptying and intestinal dyskinesia caused by 5-hydroxytryptamine (5-HT) (Kimura and Sumiyoshi, 2012). The n-butanol extract of AR has a broad-spectrum anti-ulcer effect and could inhibit protease activity and eliminate gastric acid. Choi et al. (2011) suggested that ethyl acetate extracts of A. japonica (AJEA) may specifically act on the distal colon longitudinal muscles (DCLM) among gastrointestinal smooth muscles, and AJEA-induced DCLM contraction is likely mediated, at least, by activation of ChAT and acetylcholinergic muscarinic receptors. Aqueous extract of A. lancea was also shown to increase the levels of gastric hormone motilin and gastrin while decreasing the levels of somatostatin and corticotrophin-releasing factor, which results in improving the gastric emptying condition. And polyacetylenic compounds contributed to its activity (Nakai et al., 2003). Essential oil extracted from A. lancea delayed gastric emptying are preformed mainly via inhibition of the release of central corticotropin-releasing factor and activation of the vagal pathway, which is also involved in the release of gastrointestinal hormones such as motilin, gastrin, and somatostatin (Zhang et al., 2008).

AR decoction has a curative effect on histamine-induced gastric acid hypersecretion and ulcer with a mucosal lesion as the main factor (Piao and Piao, 1996). Intestinal epithelial (IEC-6) cells play a major role in gastrointestinal disease to accelerate wounds and mucosal ulcer healing. A recent study found that atractylenolide I (ATL-I) could promote IEC-6 cell proliferation and migration by increasing cytosolic free Ca2+ concentration and enhancing transient receptor potential canonical 1 (TRPC1) and phospholipase C-γ1 (PLC-γ1) activity in IEC-6 cells (Song et al., 2017). Satoh et al. (2000) found that hinesol could resist gastric ulcer by inhibiting H+, K+-ATPase and Mg2+-ATPase and Ca2+-ATPase activity. AR exerted anti-gastric ulcer effect by decreasing inflammatory mediators [i.e. tumor necrosis factor-α (TNF-α), prostaglandin E2 (PGE2), and interleukin-8 (IL-8), IL-6)] and increasing trefoil factor 2 (TFF2) and epidermal growth factor (EGF) in the rat model (Yu et al., 2015). A series of oligosaccharides (DP 3–15) were isolated from A. lancea and identified as FOS. They could selectively stimulate the growth and/or activity of intestinal microflora and provide a health benefit to the host (Zhuang et al., 2019). It is increasingly clear that FOS could reduce or prevent gastroenteritis, inflammatory bowel disease, reduce the risk of colon cancer, and potential pathogenic gastrointestinal bacteria (Pöhnl et al., 2017). Atractylodiol and diacetyl-atractylodiol from A. japonica could stimulate the contractility of the distal colon in rats by inhibiting nitrergic-purinergic relaxation. Atractylodin, acetylatractylodinol, atractylodinol, and 4, 6, 12- tetradecatriene-8, 10-diyne-1, 3, 14-triol from A. lancea could promote delayed gastric emptying (Kim, 2016). These acetylenes, even though the traditional therapeutic properties cannot be entirely explained, are considered key elements for pharmacological accounts of the effects of AR that have been used to treat gastrointestinal disorders. These activities are consistent with the AR quality of natural and cultured products.

5.2. Immunomodulatory activity

AR is commonly used as a TCM for strengthening spleen. The spleen, a vital secondary lymphoid organ, works in innate immune response owing to its participation in immune memory and phagocytosis. AR and AM pharmaceutical uses differ in traditional Japanese Kampo medicine and TCM, with less apparent scientific evidence. Shimato et al. (2018) compared the immunomodulatory activity between Byakujutsu (A. macrocephala and A. japonica) and Sojutsu (A. chinensis, A. lancea, and the hybrids). The extract of A. japonica significantly induced granulocyte-colony stimulating factor (G-CSF) secretion from MCE301 cells in a concentration-dependent manner and had significantly higher activity than A. macrocephala. However, these effects of Byakujutsu samples were not significantly different from those of Sojutsu samples. The methanol extract of A. lancea could inhibit nitric oxide (NO) production in lipopolysaccharide-stimulated murine macrophage-like RAW264.7 cells and induce G-CSF secretion in murine normal colonic epithelial MCE301 cells. A neutral polysaccharide (ALP-1) and an acidic polysaccharide (ALP-3) were purified from A. lancea. ALP-3 exhibited macrophage proliferation in a dose-dependent manner, and stimulated phagocytic, NO and cytokines production on RAW264.7 cells. In addition, ALP-1 and ALP-3 could activate T cells in Peyer patch cells and promote the production of CSF. ALP-3 is better than ALP-1 in regulating the intestinal immune system (Qin et al., 2019). An intestinal immune system modulating arabino-3, 6-galactan (ALR-5IIa-1-1) has been found in rhizomes of A. lancea and the active clusters of polysaccharides were investigated (Taguchi et al., 2004; Yu et al., 1998).

5.3. Anti-tumor activity

Several conventional anticancer drugs for patients were and are extracted from plants. These include vincristine, paclitaxel, and camptothecin, all of which have been approved by the U.S. Food and Drug Administration (Dholwani et al., 2008). Moreover, some TCMs have been proved to have good anticancer activity in laboratory and clinical trials. The potential anticancer and antiangiogenesis properties of A. lancea extract and its major constituents have been demonstrated in various cancer, for examples, murine blastoma cells Hela (human cervical cells), SGC-7901 (human gastric cancer cells), BEL-7402 (human liver cancer cells), H33, S180, HL-60 leukemic cells, and gastric cancer (Masuda et al., 2015; Zhao et al., 2014). In recent years, A. lancea especially as an alternative treatment for cholangiocarcinoma (CCA) patients, seems to be a promising candidate herbal medicine. Results from a series of studies (Na-Bangchang et al., 2017) confirm anti-CCA potential and safety profiles of both the crude extract and the finished product (oral pharmaceutical formulation of the standardized A. lancea extract). Phases I and II clinical trials of the product to confirm tolerability and efficacy in healthy subjects and patients with advanced-stage CCA will be carried out soon. AR ethanol extract exhibited an inhibitory effect on opisthorchis viverrini (OV)-induced CCA animal without obvious toxicity after oral administration for 30 days (Ohara et al., 2018).

Atractylenolide I (ATL-I) has an antitumor effect on bladder cancer in vitro and in vivo. The results showed that ATL-I increased the level of p21 and decreased cyclin B1, cyclin-dependent kinase 1 (CDK1), and cell division cycle 25C (Cdc25c) levels and inhibiting bladder cancer cell proliferation. Further study indicated that ATL-I induced G2/M cell cycle arrest. Besides, ATL-I could promote apoptosis by inhibiting PI3K/Akt/mTOR. Animal experiments proved that ATL-I has an inhibitory effect on tumor growth and no obvious toxicity (Zhao et al., 2014). Huang et al. (2016) found that ATL-I could inhibit CML and AML leukemia cell proliferation and growth and induce apoptosis. Fu et al. (2018) found that ATL-I could inhibit melanoma cell migration by suppressing phospho-janus kinase 2 (p-JAK2), phospho-signal transducer, and activator of transcription 3 (p-STAT3), matrix metalloproteinase-2 (MMP-2) and MMP-9. The clinical study of ATL-I in the treatment of gastric cancer cachexia has been carried out. Atractylenolide II (ATL-II) treatment showed a dose-dependent manner inhibitory effect on p-STAT3, myeloid cell leukemia 1 (Mcl-1) and B-cell lymphoma-extra large (Bcl-xL) in B16 and A375 cells. Among them, STAT3 could promote melanoma. A further animal study showed that ATL-II suppressed STAT3 activation and tumor growth (Fu et al., 2014). ATL-II could significantly inhibit cell proliferation, motility and induce apoptosis in a dose and time-dependent manner by modulating Akt/ERK signaling pathway (Tian and Yu, 2017). Atractylenolide III (ATL-III) possessed antitumor activity on human mast cells stimulated by thymic stromal lymphopoietin (TSLP), human mast cell line-1 (HMC-1) by inhibiting mast cell proliferation, phosphorylated signal transducer, IL-13, and activator of transcription (Yoou et al., 2017). It was indicated that hinesol inhibited human leukemia HL-60 cells by reducing nuclear fragmentation and DNA fragmentation, activating c-Jun N-terminal kinase (JNK) (Masuda et al., 2015). β-eudesmol had anti-tumor activity in multiple tumor cell models (CCA, Hep-G2, A549, HT29, HeLa, SGC-7901, and BEL-7402, U266 cells) mainly through regulating apoptosis (Kotawong et al., 2018) and growth factors (Ma et al., 2008) in a time-and dose-dependent manner (Srijiwangsa et al., 2018). Moreover, β-eudesmol (2.5–5 mg/kg) treatment significantly inhibited tumor growth in H22 and S180 mice (Ma et al., 2008). Phosphorylation of ERK1/2 and angiogenesis in mice subcutaneous matrix gel plug and mice adjuvant granuloma were inhibited by high concentration of β-eudesmol, suggesting that β-eudesmol can be used as a candidate drug to treat angiogenic diseases (Tsuneki et al., 2005). Pongsakorn et al. (2018) investigated cytotoxic interactions between the three major constituents (β-eudesmol, hinesol and atractylodin) of A. lancea. Cytotoxic activities against the human CCA cells CL-6 of the dual and triple combinations with different concentration ratios were evaluated by MTT assay. The mixture of the three compounds produced synergistic interaction with combination index values of 0.519 ± 0.10 and 0.65 ± 0.17 (mean ± SD) at the concentrations that inhibit cell growth at the 50% and 90% leveled, respectively.

5.4. Anti-inflammatory activity

Shimato et al. (2018) compared the anti-inflammatory properties between Byakujutsu and Sojutsu. They found that methanol extract could inhibit the production of NO in macrophage-like RAW264.7 cells stimulated by lipopolysaccharide. The inhibitory effects of Byakujutsu on NO production were significantly higher than those of Sojutsu. This activity of A. japonica rhizome was significantly higher than that of A. macrocephala and A. lancea. The activity of A. chinensis was significantly higher than that of A. lancea. Chen et al. (2016) prepared n-hexane extracts of the three species (A. japonica, A. chinensis and A. macrocephala) and evaluated their anti-inflammatory effects on lipopolysaccharide (LPS)-induced RAW 264.7 cells. Among them, A. japonica most strongly inhibited NO production in LPS-induced RAW 264.7 cells.

Atractylon significantly inhibited NO and prostaglandin E2 production as well as inducible NO synthase and cyclooxygenase-2 expression in LPS-induced RAW 264.7 cells. Atractylon (40 mg/kg) also significantly reduced the acetic-acid-induced writhing response, carrageenan-induced paw edema, and hot-plate latent pain response in mice. The traditional use of A. lancea for anti-inflammatory effect is demonstrated to be due mainly to interference with the enzymes in the inflammatory pathway, that is, 5- lipoxygenase (5-LOX) and cyclooxygenase-1 (COX-1) (Resch et al., 1998). The current research showed that atractylodin could inhibit the inflammatory response induced by lipopolysaccharide (LPS). Namely, atractylodin inhibited the wet-to-dry weight ratio of lung, myeloperoxidase (MPO) activity, inflammatory cell infiltration, protein leakage, TNF-α, IL-1β, IL-6, and monocyte chemoattractant protein (MCP-1) secretion. Its mechanism was that atractylodin could down-regulate nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome and activate toll-like receptor 4 (TLR4) (Tang et al., 2018). Another study reported that atractylodin exhibited anti-inflammatory activity and ameliorated simultaneous dyskinesia in rats with constipation and diarrhea by reducing plasma proinflammatory cytokines (TNF-α, IL-1β, and IL-6) (Yu et al., 2017). Zhang et al. (2015) indicated that ATL-I could protect acute lung injury by inhibiting TLR4 expression and NF-κB activation. Ji et al. (2016) found that ATL-III inhibited the production of NO, PGE2, IL-6, and TNF-α in a dose-dependent manner and had an anti-inflammatory activity on LPS triggered macrophages of RAW264.7 mice. The high concentration of ATL-III (100 μM) could inhibit the expression of cyclooxygenase-2 (COX-2) by inhibiting the activation of NF-κB and ERK1/2. Antwi et al. (2017) indicated that stigmasterol could inhibit eosinophils, peribronchiolar, lymphocytes, monocytes proliferation, alveolar infiltration of inflammatory cells, perivascular, vascular cell adhesion molecule-1, ovalbumin (OVA)-specific immunoglobulin E expression and thus inhibit OVA-induced airway inflammatory damage in guinea pigs after intraperitoneal injection. Atractylone inhibited allergic inflammation in OVA-induced animal model while decreased rub scores, IL-1, IL-4, IL-5, IL-6, IL-13, COX-2, intercellular adhesion molecule-1 (ICAM-1), and macrophage inflammatory protein (MIP)-2 expression (Kim et al., 2016). The results exhibited that atractylone could inhibit histidine decarboxylase activity and expression, tryptase and histamine release in PMACI-induced HMC-1 cells and inhibit filamentous actin formation and morphological alteration in stem cell factor-stimulated peritoneal mast cells (RPMCs) animal model (Han et al., 2016). Stigmasterol and β-sitosterol had inhibitory effects on colitis induced by dextran sodium sulfate in C57BL/6J male mice. They critically suppressed colon shortening, colitis, and distal colon via the inactivation of NF-κB and decreased fecal hemoglobin content (Feng et al., 2017). A recent study showed that β-sitosterol had an anti-inflammatory activity based on the rat pleurisy assay and rat paw edema test (Paniagua-Pérez et al., 2017). Polyacetylenes exhibited anti-inflammatory activity with structure-activity relationships: the introduction of an acyl group into a compound increased the inhibitory effect against NO production. Deacylated product of 14-β-methylbutyryltetradeca-2E, 8E, 10E-trien-4, 6-diyn-1-ol, due to boiling extraction with water, results in diminished anti-inflammatory activity (Kim, 2016).

5.5. Antibacterial activity

Yin et al. (2000) carried out a systematic multi gradient external antibacterial experiment on the extract of AR. The results showed that AR had different degrees of inhibition on 15 kinds of fungi, and the effect was stronger than that of Hibiscus cortex, Phellodendron bark, and other traditional Chinese medicines. Stigmasterol exerted the synergistic antibiotic effect (suppressed colony counts, yielding 98.7%) as an adjuvant of ampicillin to gram-negative and gram-positive bacteria. However, the inhibition of ampicillin and stigmasterol alone was ineffective (Yenn et al., 2017). The aqueous extract of A. lancea has the activity of anti-monilial infection. Oral administration of 140 mg/kg/d could significantly prolong the life span of mice infected with candida (Inagaki et al., 2001).

5.6. Other activities

AR or its compounds have certain biological activities on the nervous system, lung, glucose, and lipid metabolism. The pharmacological activity of the AR on the central nervous system has been demonstrated in various animal models with regards to its effects on the general behavior and spontaneous movement, potentiation of hypnotic action of hexobarbital sodium, and anti-electroshock convulsion. Murayama et al. (2014) indicated that water extract of A. japonica showed a stronger inhibitory effect on 1-(2, 5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI)-induced head-twitch response than that of A. lancea corresponding to their major constituents of atractylenolide III and β-eudesmol, and suggested that the major constituents should be active constituents contributing to the antihallucination effects of Byaku- and So-jutsu. Besides, the part B–C ring (butenolide) in ATL III was found to be similar to the structure of serotonin and suggested that the B–C ring may partially play role in antagonistic activity against serotonin receptors. Thus, a rational design of butenolide-related compounds may produce potential lead compounds for new drug development (Murayama et al., 2014). ATL-III presented a neuroprotective effect on learning and memory impairment rats by decreasing ROS production and protein kinase C level (Zhao et al., 2015). The neurological activity of β-eudesmol, including its antagonistic effect on neostigmine-induced neuromuscular failure, was demonstrated in previous studies on the accelerating effect of desensitization of the nicotinic acetylcholine receptor and blockage action during the closed state of the receptor channel or nerve-evoked contraction at the neuromuscular junction (Chiou and Chang, 1992). Ohara et al. (2018) showed that β-eudesmol could inhibit the efferent activity of the adrenal sympathetic nerve in rats through transient receptor potential cation channel subfamily a member 1 (TRPA1) and afferent vagus nerve. Cheng et al. (2016) found that atractylone (40 mg/kg for 5 days) could decrease influenza A virus (IAV)-induced pulmonary injury in mice by inhibiting IL-6, TNF-α, and IL-1β, but promoting TLR7, IFN-β, myeloid differentiation primary response 88 (MYD88) expression. Stigmasterol possessed anti-diabetic activity in vitro and in vivo. Which could reduce and oral glucose tolerance by decreasing blood glucose levels and blood lipid indexes, and alleviating insulin resistance in vivo while increasing glucose transporter type 4 (GLUT-4) translocation and expression in vitro (Wang et al., 2017). A water extract of the oriental crude drug Atractylodes Japonica Rhizomes showed hypoglycemic activity in mice. The extract was fractionated by monitoring the pharmacological activity to obtain three glycans, atractans A, B and C. These constituents exerted significant hypoglycemic actions in normal and alloxan-induced hyperglycemic mice. The A. lancea ethanol extracts exhibited an inhibitory effect on lipase with the IC50 9.06 μg/mL and anti-obesity effect on the obesity mice was presented on the high concentration (500 mg/kg) (Jiao et al., 2014). Water extracts from A. lancea reduced triptolide-induced toxicity via down-regulated hepatic cytochrome P450 3A (CYP3A) expression and improved anti-inflammatory activity of triptolide. This suggested that A. lancea could inhibit toxicity and increase the therapeutic activity of triptolide (Yenn et al., 2017). Additionally, ATL-I could inhibit 5-lipoxygenase (5-LOX) production to decrease allergic responses (Lim et al., 2012). The pharmacological interaction between the constituents of Keishi-ka-zyutubu-tō (composed of seven crude drugs, namely Glycyrrhizae Radix et Rhizoma, Cinnamomi Cortex, Zingiberis Rhizoma Recens, Atractylodis Rhizoma, etc.) was investigated in in-situ sciatic nerve-gastrocnemius muscle preparations by genetically diabetic KK-CAy mice. When administered intra-arterially, the drug caused a degree of inhibition of the diabetic neuromuscular junction 280 times greater than normal. When further combined with Paeoniae Radix Alba and Glycyrrhizae Radix et Rhizoma, the potency ratio was increased 4.9 times for Cinnamomi Cortex, 5.9 times for Zingiberis Rhizoma Recens, and 8.7 times for Atractylodis Rhizoma. In combination with the representative compounds, the potency ratio of paeoniflorin and glycyrrhizin was increased 6.6 times for cinnamaldehyde, 6.1 times for 8-gingerol and 14 times for β-eudesmol in diabetic muscles. The single effect of β-eudesmol or 8-gingerol was 5.2 times or 3.7 times more potent, respectively, on diabetic muscles than on normal muscles. These results indicated that the preferentially more potent neuromuscular blocking effects of Keishi-ka-zyutubu-tō in diabetic muscles were mainly attributable to β-eudesmol, a major component of Atractylodis Rhizoma (Kimura et al., 1987).

As seen in Table 7 , AR has been used as TCM for several thousand years, and current studies indicated that AR and some of its compounds (most of which are volatile oil) housed various biological activities. However, the in-depth molecular mechanism remains elusive. Many reports are available on sesquiterpenes, alkynes, and glycosides in AR, which are common in Compositae plants, and alkynes in safflower. They are important secondary metabolites formed by the long-term coevolution of plants and insects. Most of the main components could produce cytotoxic effects on cancer cells, especially melanoma. However, a certain gap still exists between the current pharmacological activities and its clinical uses. For instance, the role of AR in the treatment of eyesight and flaccidity-syndrome has not been explored. In addition, the existing pharmacological activity studies were mainly based on cell models and supplemented by animal models. The dosage of AR compounds with AR extracts considerably varies. Although no potential safety hazards were shown by the existing evidence using in clinical settings remain underdeveloped. In some studies, the type, weight, sex, and other factors of the animal models are not clear, and the reference value of drug doses ignoring the actual factors of experimental animals is of little value. Meanwhile, AR is traditionally used in decoction clinically, and previous studies showed that the activity of water solution and volatile oil in AR are basically the same in terms of strengthening spleen and stomach (Liu et al., 2012), anti-inflammatory activities, liver protection (Ta et al., 2011), and other pharmacological aspects (Xu et al., 2015). Therefore, the water-soluble material basis of AR must be studied for enhanced guidance on the application of AR in Chinese patent medicine. In addition, the pharmacological activity and mechanism of compound AR and the interaction of effective components need to be further studied.

Table 7.

Dosage, experimental model, pharmacological action and mechanism of Atractylodis Rhizoma extracts.

Pharmacology Pharmacological effects Substances and dosages Experimental model Mechanisms Ref.
Improving gastrointestinal function Stimulating gastric emptying or small intestinal motility The extract (500 or 1000 mg/kg) and β-eudesmol (50 or 100 mg/kg) Atropine-, dopamine-, 5-hydroxytryptamine-treated mice Inhibiting the dopamine D2 and 5-HT3 receptor Kimura and Sumiyoshi (2012)
Inhibiting of gastric ulcer n-butanol extract
0.18 g/kg
Rats Improving blood circulation in ulcer, promoting DNA, RNA and protein synthesis Piao and Piao (1996)
Anti-gastrointestinal mucosal injury Atractylenolide I (ATL-I) 5 and 10 μM IEC-6 cell Promoting IEC-6 cell proliferation and migration; increasing polyamines content, and enhancing TRPC1 and PLC-γ1 mRNA and protein expression Song et al. (2017)
Resisting gastric ulcer Hinesol
IC50: 5.8*105 M
Porcine gastric membrane vesicles Inhibiting H+, K+-ATPase and Mg2+-ATPase and Ca2+-ATPase activity Satoh et al. (2000)
Anti-gastric ulcer A. lancea 2.5 g/kg Acetic acid imitated rats The gastroprotective effects were mediated by up-regulating trefoil factor 2 and epidermal growth factor Yu et al. (2015)
Immunomodulatory activity Immunostimulatory effects The boiling water extract
1.0 mg/mL
Murine normal colonic epithelial MCE301 cells Inducing secretion of granulocyte colony-stimulating factor Shimato et al. (2018)
Immunomodulatory activity Acidic polysaccharide (ALP-3) 50, 100, 250, 500, 1000, 2000 μg/mL Murine RAW264.7 macrophage cell line Stimulating macrophage proliferation, and stimulating phagocytic, NO and cytokines production on RAW264.7 cells Qin et al. (2019)
Intestinal immune system modulating activity ALP-1 and ALP-3 50, 100, 250, 500, 1000, 2000 μg/mL Peyer's patch cells Activating T cells in Peyer patch cells and promoting the production of CSF Qin et al. (2019)
Intestinal immune system modulating activity Polysaccharides
100 μg/mL
Peyer's patch cells Regulating intestinal immune system Yu et al. (1998)
Anti-tumor activity Inhibitory effect on human gastric cancer cells Water extract of the A. lancea 0.0625, 0.125, 0.25, 0.5, and 1 mg/mL BGC-823 and SGC-7901 cells Inhibiting the growth of BGC-823 and SGC-7901 cells Zhao et al. (2014)
Anti-leukemia Atractylenolide I (ATL-I) 3.13–100 μg/mL Human K562 CML, U937 AML and Jurkat T lymphoma cells Inducing apoptosis and differentiation of macrophage lineage Huang et al. (2016)
Anti-melanoma effects Atractylenolide I (ATL-I) 40, 60, 80, 100, 120, 150 μM Human A375, Hs294T and SK-MEL-5 melanoma cells Inhibiting phospho-janus kinase 2, phospho-signal transducer and activator of transcription 3, matrix metalloproteinase-2 and -9 Fu et al. (2018)
Antimelanoma effects Atractylenolide II (ATL-II) 12.5, 25 mg/kg B16 xenograft mouse Suppressing STAT3 activation and tumor growth Fu et al. (2014)
Antigastric carcinoma ATL-II 50, 100, 200, 400 μM Human gastric carcinoma cell lines HGC-27 and AGS Modulating Akt/ERK signaling pathway to inhibit cell proliferation, motility and inducing apoptosis Tian and Yu (2017)
Antitumor activity Atractylenolide III (ATL-III) 1, 10, 100 μM HMC-1 cell Inhibiting mast cell proliferation, phosphorylated signal transducer, IL-13, proinflammatory cytokines and activator of transcription Yoou et al. (2017)
Antitumor activity ATL-III 100 μM LAD2 human mast cell Inhibiting mast cell proliferation and the production of inflammatory cytokine Yoou et al. (2017)
Antitumor activity Hinesol 5, 10, 25, 50, 100 μM Human leukemia HL-60 cells Hinesol induced apoptosis through the JNK signaling pathway in HL-60 cells Masuda et al. (2015)
Anti-cholangiocarcinoma β-eudesmol
IC50: 39.33 mg/mL
The CCA cell lines, and normal human cell line Promoting cell cycle arrest at G1 phase, and inducing cell apoptosis through activation of caspase-3/7 Kotawong et al. (2018)
Anti-hepatocellular carcinoma β-eudesmol
IC50: 16.51 ± 1.21–24.57 ± 2.75 μg/mL
B16–F10 and HepG2 cell lines Inducing tumor cell death by caspase-mediated apoptosis pathways Bomfim et al. (2013)
Anti-multiple myeloma cells β-sitosterol 0, 6.25, 12.5, 25, 50, 100 mM Human multiple myeloma U266 and MM1S cell Increasing the sub-G1 apoptotic population, activating caspase-9 and -3 Sook et al. (2014)
Anti-tumor activity β-eudesmol 10–100 mM HeLa, SGC-7901, and BEL-7402 Inhibiting angiogenesis by suppressing CREB activation in the growth factor signaling pathway Ma et al. (2008)
Inhibiting of Cholangiocarcinoma β-eudesmol (0, 1, 10, 30, 100 μM), dicoumarol (1 μM), 5-FU (0, 3, 10, 30, 100 μM), DOX (0, 0.1, 0.01, 1, 10 μM) Human CCA cell line, KKU-100 Enhancing chemotherapeutic effects of 5-fluorouracil and doxorubicin in the high NQO1-expressing human CCA cell line, NQO1-KKU-100 Srijiwangsa et al. (2018)
Inhibiting of lung and colon cancer β-eudesmol 5–100 μM Human lung (A549) cells, colon (HT29 and Caco-2) cells Inhibiting proliferation of tumor cells and superoxide production; inhibiting adhesion and migration of A549 and HT29 cell Sghaier et al. (2016)
Anti-angiogenic mechanism and anti-tumor activity β-eudesmol (2.5–5 mg/kg) Implanting H22 and S180 mice tumor cells into oxters of 7-week-old KM mice Suppressing CREB activation in growth factor signaling pathway Ma et al. (2008)
Inhibiting angiogenesis β-eudesmol 10–100 μM Porcine brain microvascular endothelial cells, human dermal microvascular endothelial cells The blockade of the ERK signaling pathway Tsuneki et al. (2005)
Anti-inflammatory activity Anti-inflammatory effects in vitro AR methanol extract 100 μg/mL Murine macrophage-like cell line RAW264.7 cells Inhibiting the production of NO Shimato et al. (2018)
Anti-inflammatory effect ATL-III 1–100 μM, LPS 1 μg/mL The murine macrophage cell line RAW264.7 Suppressing the release of NO, PGE2, TNF-α and IL-6 related to the NF-κB- and MAPK signaling pathways Ji et al. (2016)
Improving intestinal inflammation and cooccurring dysmotility Atractylodin 10.0 mg/kg Diarrheaprominent rats were established by acetic acid and restraint stress; constipation-prominent rats were established by cool water Reducing pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 in the plasma and inhibiting the expression of inflammatory mediators iNOS and NF-κB in jejunal segments in both CP and DP rats. Yu et al. (2017)
Anti-inflammatory effects Water extracts of A. lancea rhizomes 100, 200 mg/kg Male C57BL/6 mice Inducing the hepatic expression of CYP3A Yuan et al. (2017)
Alleviating LPS-induced inflammatory responses Atractylodin 40 or 80 mg/kg Lipopolysaccharide (LPS)-induced inflammatory rates Suppression of nucleotide-binding domain-like receptor protein 3 inflammasome and toll-like receptor 4 activation Tang et al. (2018)
Ameliorate colitis β-sitosterol and stigmasterol 0.4% Dextran sulfate sodium (DSS)-induced colitis in C57BL/6J male mice Suppressing the activation of inflammatory master regulator NF-κB. Stigmasterol significantly lowered colonic inflammation score and the expression of cyclooxygenase-2 and colony stimulating factor-1 Feng et al. (2017)
Anti-bacterial activity Antimycotic effect AR 0.5–10.0% Trichophyton tonsuraus, Microsporum gypseum et al. This drug had apparent fungi action Yin et al. (2000)
Combating multidrug resistant strains Stigmasterol 1 μg/disc, or in combination (1 μg/disc ampicillin + 1 μg/disc stigmasterol) 2 g-positive bacteria and 2 g-negative bacteria Synergistic effects of stigmasterol and ampicillin against β-lactamase producing clinical isolates Yenn et al. (2017)
Other activities Protective effects of acute lung injury LPS + AO-I (5, 10 and 20 mg/kg), LPS + VGX-1027 (0.5 mg/mouse) LPS-induced acute lung injury mouse model Inhibiting of TLR4 expression and NF-κB activation Zhang et al. (2015)
Antiasthmatic properties Stigmasterol 10–100 mg/kg Ovalbumin-induced Guinea pigs Reducing the proliferation of eosinophils, lymphocytes, and monocytes while reducing peribronchiolar, perivascular, and alveolar infiltration of inflammatory cells Antwi et al. (2017)
Treatment of hallucinations of dementia Atractylenolide III 0.03, 0.15, 0.75 mg/kg;
β-eudesmol 0.4, 2.0, 10 mg/kg
1-(2,5-Dimethoxy-4-iodophenyl)-2-aminopropane (DOI)-induced head-twitch mice Active constituents contributing to the anti-hallucination effects of Byaku- and So-jutsu against serotonin receptors Murayama et al. (2014)
Neuroprotective activity Atractylenolide III 0.6, 1.2 and 2.4 mg/kg/day Chronic high-dose homocysteine administration induced learning and memory impairment in rats Decreasing homocysteine-induced reactive oxygen species formation and restored homocysteine-induced decrease of phosphorylated protein kinase C expression level Zhao et al. (2015)
Autonomic nerve activity β-eudesmol 1.6, 1.8, 2.3, 2.4 ng/mL Male Wistar rats (300–350 g) Suppressing ASNA, partly through TRPA1 and the afferent vagus nerve Ohara et al. (2018)
Influenza A virus treatment Atractylon treatment at doses of 10–40 mg/kg Influenza a virus (IAV) caused pneumonia in mice Upregulating the expression of Τoll-like receptor 7, MyD88, tumor necrosis factor receptor-associated factor 6 and IFN-β mRNA but downregulating NF-κB p65 protein expression in the lung tissues of IAV-infected mice Cheng et al. (2016)
Anti-diabetic activity Metformin 200 mg/kg, stigmasterol 50, 100 mg/kg Rat L6 cells Enhancing the GLUT4 expression in L6 cells, skeletal muscle and white adipose tissue Wang et al. (2017)
Anti-obesity effect the ethanol extract of Atractylodes lancea rhizome (250, 500 mg/kg) A high-fat diet-induced obesity mice Atractylodin showed the highest lipase inhibitory activity Jiao et al. (2014)
Anti-allergic activities AR ethanol extracts 200 mg/day Male NC/Nga mice Inhibiting 5-LOX from RBL-1 cells Lim et al. (2012)

6. Toxicity

According to TCM, the adverse reactions of AR are mainly manifested in “dryness”. The “dryness” of AR is not only a pharmacological action for treating diseases but also a factor causing adverse reactions. Unprocessed AR has strong “dryness” and is easy to consume Yin fluid. The adverse reactions include impairment of spleen Yin, dysfunction of spleen in transportation, distress in the epigastrium, mouth parching and tongue scorching, and constipation. Therefore, Yixue Rumen indicated AR to “mistakenly take blood consumption, yin fail to nourish the body, hyperactive the fire due to yin deficiency”. After processing, the “dryness” of AR is slowed down, and it could eliminate dampness and activate spleen and stomach by entering the middle Jiao. Taking AR orally does not cause swelling and could eliminate dampness blocking and abdominal distention. In the case of Yin deficiency and heat injury to the body, the “dryness” of AR is particularly prominent. Therefore, Bencao Jingshu recorded that “AR should not be taken for diseases that belong to Yin deficiency, blood deficiency, essence deficiency, tidal fever, dry mouth and lips, cough and spit, spitting blood, sore nose and throat, and constipation, stagnation and Qi deficiency in the liver and kidney.” Bencao Zheng also recorded that “those who have a deficiency of internal heat and Yin and loose sweating on the surface should not take AR.” The “dryness” of AR is related to its volatile oil. The Compendium of Materia Medica (Bencao Gangmu) recorded, “because of the dryness of AR, processing would be taken that its oil is soaked in glutinous rice swill, and it is used for slicing and baking.” The results showed that the volatile oil content of AR decreased after being fried with bran and rice swill (Zhao, 2009). Whether AR should be processed to reduce the content of the essential oil in clinical medication should be based on the needs of the disease. For instance, raw AR is suitable for cold syndrome and cold dampness arthralgia, while processed AR is suitable for dampness resistance and middle Jiao and flaccidity syndrome. In addition, the deficiency of the element body Yin should be used with caution. Studies on modern toxicology and pharmacology of AR verified the safety profiles in different animal models. Following administration of ethanolic A. lancea extract at the high dose level (5000 mg/kg body weight) in rats and mice, no significant toxicity was observed except for gastric stimulation and central nervous system inhibition symptoms (diminished response to touch and balance and decreased alertness and motor ability) (Plengsuriyakarn et al., 2012). It is important however for the knowledge of potential deleterious effects of A. lancea on cardiovascular system. The antiplatelet aggregation of A. lancea is possibly mediated through suppression of collagen-induced signaling pathway which is upstream of the release of thromboxane A2 from platelets (Nasu et al., 2009). Care should be taken when using A. lancea extract or its active constituents in patients with platelet disorders or coagulopathy. The results of acute and subacute toxicity tests in rats and mice indicated that A. lancea was safe at a wide range of dose levels (1000–5000 mg/kg body weight) (Koonrungsesomboon et al., 2014). An observational study conducted in China showed a safety profile of Fufang Cangzhu Tang, a Chinese herbal formula containing 15 g of AR decocted into 300 mL of liquor and separately administered orally two times a day for 8 weeks in 32 senile patients with obesity or overweight complicated with impaired glucose tolerance (Shi et al., 2006). In addition, animal experiments showed that the volatile oil of AR has a sedative effect in small dose, hyperreflexia in spinal cord, and inhibition effect in large dose, which eventually leads to respiratory paralysis and death (Wu, 1999). Its main inhibitory components are β-eucalyptol and atractylol, thus, it is harmful to organisms. Therefore, AR should be used with caution in patients with nervous system diseases because of its various effects on the nervous system. Meanwhile, it can discharge sodium and potassium; thus, electrolyte balance should be given attention in long-term applications. Several clinical studies of AR have been conducted in patients with different diseases/symptoms by using AR in various formulations (Liu et al., 2008). Atractyloside (ATR) is a diterpenoid glycoside and occurs naturally in Asteraceae plants (A. macrocephala, A. lancea and Xanthium sibiricum), many of which are used in foods and ethnomedicines. The toxicity of ATR has caused fatal renal proximal tubule necrosis and/or centrilobular hepatic necrosis in man and farm animals. And the degradation of ATR is a way to increase pharmaceutical safety for the Chinese medicinal industries (Zhu, 2012). However, no clinical study using AR extract or its main components alone has been conducted, signifying the need for further clinical trials to prove its clinical efficacy and safety for the human body. Although clinical research directly supporting its safety in the human body is lacking, the available information showed no serious adverse event.

7. Quality control

There is evidence that the sesquiterpenoids found in AR have pharmacological properties of improving gastrointestinal function, anti-tumor and anti-inflammatory, so the presence of these compounds may be a key indicator of the quality of the extract (Yang et al., 2012). The processing of AR also influenced the physicochemical properties and absorption of polyacetylenes. The extraction efficiencies of atractylodin and (4E, 6E, 12E)-tetradecatriene-8, 10-diyne-1, 3-diyl diacetate from AR were decreased when the rhizomes were stir-fried with bran compared to crude rhizomes (Liu et al., 2013). However, the processing of AR could increase the absorption of atractylodin. Stirfrying of AR with wheat bran could promote and accelerate the absorption of (4E, 6E, 12E)-tetradecatrinen-8,10-diyne-1, 3-diyl diacetate and its concentration was highest in the spleen, possibly increasing the spleen-tonifying effect according to the traditional theory (Kim, 2016). Stability is another concerning issue as the occurrences of bioactive polyacetylenes can be changed under certain circumstances, like drying of the rhizomes under the sun. Atractylodin, atractylodinol, and acetylatractylodinol were reported to have 1-cis isomers, (1Z)-atractylodin, (1Z)-atractylodinol, and (1Z)- acetylatractylodinol. The proportions of those cis isomers were less than trans isomers as the cis isomers could be formed during the drying process of the AR under the sun. For example, (1E)-acetylatractylodinol is rapidly isomerized to (1Z)-acetylatractylodinol. On the one hand, the stabilities of 1-cis isomers were weaker than those of 1-trans isomers: (1Z)-acetylatractylodinol more expeditiously disappeared in n-hexane solution compared to (1E)-acetylatractylodinol dissolved in same solvent in the freezer. On the other hand, 1-cis isomer of atractylodin was relatively increased up to a similar ratio with its parent compound, atractylodin, in freezer storage, indicating that atractylodin was considerably unstable than its isomer and different from those two polyacetylenes, (1E)-atractylodinol and (1Z)-atractylodinol, which showed the same degree of instability (Resch et al., 2001).

According to the TCM theory, genuine (Dao-di) herb is a high-quality medicine produced from specific areas, with a long history of traditional use and excellent efficacy (Wang et al., 2018). Mao Cangzhu is considered a fine-quality good beginning from Hongjing Tao in the Northern and Southern Dynasties (386–589 A.D.). It has been used for more than 1500 years and it is found in Maoshan in Jiangsu Province. Cangzhu always takes Maoshan as geo-authentic habitats. The volatile oil components of AR vary considerably and are distributed continuously. According to the composition of the volatile oil of AR, it can be divided into two chemical types. One is represented by “Hubei Cangzhu”, which is mainly found in Hubei, Anhui, Shaanxi, Southern Henan and other places. The total amount of the volatile oil in Hubei type (HBA) is high, and mainly composed of hinesol and β-cineole, with or without very small amounts of atractylone and atractylodin. The other type, Maoshan type (MA), is mainly represented in Maoshan, Jiangsu Province, including Jiangsu, Shandong, Hebei and northern Henan. Its total volatile oil is low, and mainly composed of atractylone and atractylodin. In the volatile oil, atractylone, hinesol,β-eudesmol,atractylodin showed a specific ratio. In MA, their ratio is 0.70–2.00: 0.04–0.35: 0.09–0.40: 1 (Guo et al., 2008). These results showed that 91% of the measured values are in this range. The clustering results of HPLC fingerprint of the water extract of AR were similar to those of the volatile components. The main components of “white frost” in AR are citronellol and β-cineole. The content of these two components in Mao Cangzhu is much higher than that in unfamous-region AR. The content of atractylone, which is the main component of the cinnabar point of Mao Cangzhu, is 25.92% of the total volatile oil of AR. The average content of atractylone in other production areas is only 0.69% of the total volatile oil, but this component was detected in some samples (Guo et al., 2002). Thus, atractylone is essential in reflecting the genuine characteristics of AR, suggesting that the efficacy of AR is more similar to that of AM. The strong ability of AR in drying dampness and strengthening the spleen is the reason why it has become a genuine medicine compared with the AR from other places of origin. Generally, phenotypic variation is a product of both genetic and environmental variation (O'Reilly-Wapstra et al., 2013). The qualities of AR samples are closely associated with their habitat, and the contents of curative components in AR vary considerably based on geographical factors. However, it remains unclear whether the geographical variations are based on genetic or environmental factors. At the colony levels, as a genuine medicinal material of AR, the phenotype (volatile oil) of AR has obvious differentiation, but at the same time, its genetic differentiation is not significant. It can be seen that the larger phenotypic variation and the smaller genetic variation of AR are inconsistent, and the correlation between them is poor. All of these suggest that in addition to genetic factors, the environment is even a very important factor in the formation of herbal medicine of AR. Japanese scholars also confirmed this point by transplanting AR. Takeda et al. (1996) found that the volatile oil composition of A. lancea, A. chinensis and A. koreana was closer to that of 2–3 years after it was transplanted to Nanjing Institute of Botany. The climate of Maoshan area is characterized by high temperatures, short dry season and sufficient rainfall. The unique ratio of volatile oil of AR may be the result of the specific metabolism of AR caused by a specific environment. The meteorological conditions in October have the greatest impact on the volatile oil components. The annual average and the meteorological conditions in February and September have a greater impact on the volatile oil components. High temperature is the limiting factor for the growth and development of AR, while several meteorological factors related to temperature in Maoshan area are the highest in the whole distribution area (Guo, 2005). The formation of AR has an obvious adverse effect. Some studies have reported that the activation of phytohormones such as jasmonic acid and abscisic acid through symbiosis with several endophytes induces the production of the essential oil compounds in A. lancea (Wang et al., 2015). Yuan et al. (2009) found that soil acidity stimulated the accumulation of essential oils in A. lancea, particularly β-eudesmol, by influencing the concentrations of phytohormones such as abscisic acid. These results suggested that the contents of essential oil compounds in A. lancea could vary depending on environmental factors such as biological and abiotic stresses. In contrast, Takeda et al. (1996) suggested that the contents of essential oil compounds are influenced largely by genetic factors based on a comparative study of the essential oil compounds between wild and cultivated A. lancea. To examine genotype-environment (G × E) interaction and the effects of environmental factors, Tsusaka et al. (2019) cultivated six clones in different years and different locations (Hokkaido, Ibaraki), and determined contents of β-eudesmol, hinesol, atractylon, and atractylodin in the clones. Subsequently, genetic variances, environmental variances, and G × E interaction variance based on the compound contents were evaluated. It is demonstrated that the contents of β-eudesmol, hinesol, atractylon, and atractylodin in A. lancea are influenced mainly by genetic factors. In addition, these essential oil compounds could be selected regardless of rhizome yields in the course of A. lancea cultivar development. Consequently, A. lancea cultivars with high yields and high contents of the essential oil compounds could be developed and a further investigation on the effects of environment factors on the compound contents is a must.

The quality evaluation of AR is obviously based on the chemical composition and relative content of volatile oil as evaluation indicators. However, the lack of non-volatile parts limited this study. From Shennong Bencao Jing, who recorded that AR is used for “long-term use of AR would get longevity” to Bencao Gangmu, who recorded that “people often burn AR to avoid evil spirits in this disease and New Year's day”. “Decocting baits” mainly used non-volatile parts, while “avoid poisonous gas” mainly used volatile oil parts. Enhancing the understanding on the quality evaluation of AR demands taking the volatile components as the index while regarding the non-volatile components. In the case of the unclear material basis of TCM, the overall effect of fingerprint is better than that of individual index components in terms of quality control. In addition, biomacromolecules play considerable roles in Chinese medicine, as their biological activities are related to TCM efficacy and their biological functions of the relationship between primary metabolism and the quality formation of TCM (Zhang et al., 2020). Understanding the functional properties and mechanisms of biological macromolecules could help demystify the drug properties and health benefits of TCM.

Further, The blood concentrations of the pharmacodynamic substances (mainly the active ingredients) of TCMs are usually very low. How can they exert pharmacological actions, in which forms (original form, metabolite or the both) do they exert the actions? The total chemical components of multi-sources TCMs from different plants are roughly the same, but the types and contents of chemical components are often obviously different (Kim et al., 2018). Is it reasonable for plants from different origins to be used as the same traditional Chinese medicine clinically? A new concept includes that the aggregate or summation of “effective forms” of pharmacodynamic substances of TCM is the core material base of the efficacy of TCM, and the “additive effect” of the blood concentrations of different “effective forms” is one part of the action mechanism (Xu et al., 2014). The “additive effect” of the different “effective forms” of a TCM means an “additive effect” of numerous constituents or/and metabolites on a same target, and therefore the efficacy brought by the addition of the concentrations of all these compounds, which differ from the “synergy effect” of multi-constituents on multi-targets and is a beneficial complement. Some constituents can be converted to each other in vivo and some metabolites are bioactive. These compounds having the similar structure are likely to have the same pharmacological effects on the same target, which could provide experimental evidence for the concept of “effective forms” and the hypothesis of “additive effect”.

8. Conclusions and future perspectives

The botany, traditional uses, phytochemistry, pharmacology, toxicity, and quality control of AR have been summarized in the present review. More than 200 compounds have been isolated and identified from the herb. Among them, sesquiterpenoids, and alkynes are the major active ingredients. And improving gastrointestinal activity supported the traditional use of AR. Nevertheless, there is still a lack of sufficient research and further investigations need to be done in AR in the future. The related researches on Genus Atractylodes are mainly focused on A. lancea and A. macrocephala. As far as their active chemical components are concerned, other species also have important value, which is also a beneficial supplement for resource utilization of Atractylodes. The traditional uses of AR need to be further studied in the prevention and treatment of plague and night blindness and should be subjected to modern pharmacology to verify this idea and/or clarify its potential mechanism. The effective components, pharmacological activities, and mechanisms of TCM should be fully considered in the “effective forms”, “additive effect” and synergistic effect. We suggested that the “effective forms” and “additive effects” of the pharmacodynamic substances based on the same pharmacophore of TCM should be extensively investigated to further understand the pharmacodynamic substances and action mechanism of TCM. A. lancea is expected to be one of the candidate drugs for the treatment of cholangiocarcinoma. Further study on its biological activities in vivo and in vitro should be combined with toxicity study, and the appropriate dosages, models, and medication cycles should be selected according to the actual application. We should follow the quality control characteristics of TCM (integrity, processing, genuine) and combine with the clinical application, and a deep analysis of volatile oil and water-soluble components and their relationship is a must.

Author's contributions

Wen-jin Zhang and Zhen-yu Zhao collated documents and wrote the manuscript; Li-kun Chang and Ye Cao helped perform the arrangement of tables and figures; Sheng Wang, Chuan-zhi Kang and Li Zhou contributed significantly to analysis and manuscript preparation; Hong-yang Wang assistance to the revision of the manuscript; Lu-qi Huang provided valuable idea; Lan-ping Guo provided financial supports and valuable discussion. All authors read and approved the final manuscript.

Declaration of competing interest

The authors declare that they do not have any conflict of interest.

Acknowledgments

This research was funded by the major projects of the National Natural Science Foundation of China (81891014); Special fund project of modern agricultural industrial technology system (CARS-21).

References

  1. Ahmed S., Zhan C., Yang Y., Wang X., Yang T., Zhao Z., Zhang Q., Li X., Hu X. The transcript profile of a traditional Chinese medicine, Atractylodes lancea, revealing its sesquiterpenoid biosynthesis of the major active components. PloS One. 2016;11(3) doi: 10.1371/journal.pone.0151975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Antwi A.O., Obiri D.D., Osafo N. Stigmasterol modulates allergic airway inflammation in Guinea pig model of ovalbumin-induced asthma. Mediat. Inflamm. 2017;2017:1–11. doi: 10.1155/2017/2953930. 2953930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Chang Z., Jia D., Bare J. PMPH-USA; 2015. Chinese Materia Medica. [Google Scholar]
  4. Chen F., Wei Y.X., Zhang J.M., Sang X.M., Dai C.C. Transcriptomics analysis investigates sesquiterpenoids accumulation pattern in different tissues of Atractylodes lancea (Thunb.) DC. plantlet. Plant. Cell. Tiss. Org. 2017;130(1):73–90. [Google Scholar]
  5. Chen L.G., Jan Y.S., Tsai P.W., Norimoto H., Michihara S., Murayama C., Wang C.C. Anti-inflammatory and antinociceptive constituents of Atractylodes japonica Koidzumi. J. Agric. Food Chem. 2016;64(11):2254–2262. doi: 10.1021/acs.jafc.5b05841. [DOI] [PubMed] [Google Scholar]
  6. Chen J. Nanjing Agricultural University; 2007. Studies on the Chemical Constituents of Atractylodes Lancea and Angelica Chinensis. [Google Scholar]
  7. Chen Y., Wu Y., Wang H., Gao K. A new 9-nor-atractylodin from Atractylodes lancea and the antibacterial activity of the atractylodin derivatives. Fitoterapia. 2012;83(1):199–203. doi: 10.1016/j.fitote.2011.10.015. [DOI] [PubMed] [Google Scholar]
  8. Cheng Y., Mai J.Y., Hou T.L., Ping J., Chen J.J. Antiviral activities of atractylon from atractylodis rhizome. Mol. Med. Rep. 2016;14(4):3704–3710. doi: 10.3892/mmr.2016.5713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Chiou L.C., Chang C.C. Antagonism by β-eudesmol of neostigmine-induced neuromuscular failure in mouse diaphragms. Eur. J. Pharmacol. 1992;216(2):199–206. doi: 10.1016/0014-2999(92)90361-7. [DOI] [PubMed] [Google Scholar]
  10. Choi K.H., Jeong S.I., Lee J.H., Hwang B.S., Lee S., Choi B.K., Jung K.Y. Acetylene compound isolated from Atractylodes japonica stimulates the contractility of rat distal colon via inhibiting the nitrergic–purinergic relaxation. J. Ethnopharmacol. 2011;134(1):104–110. doi: 10.1016/j.jep.2010.11.059. [DOI] [PubMed] [Google Scholar]
  11. Committee F.O.C.E. Missouri Botanical Garden and Harvard University Herbaria; 2018. Flora of China. [Google Scholar]
  12. Committee N.P. 2015. Pharmacopoeia of the People's Republic of China; pp. 188–189. Part 1. [Google Scholar]
  13. Deng A., Li Y., Wu Z., Liu T., Kang L., Nan T., Zhan Z., Guo L. Advances in studies on chemical compositions of Atractylodes lancea and their biological activities. China J. Chin. Mater. Med. 2016;41(21):3904–3913. doi: 10.4268/cjcmm20162104. [DOI] [PubMed] [Google Scholar]
  14. Dholwani K., Saluja A., Gupta A., Shah D. A review on plant-derived natural products and their analogs with anti-tumor activity. Indian J. Pharmacol. 2008;40(2):49. doi: 10.4103/0253-7613.41038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Duan J.A., Wang L., Qian S., Su S., Tang Y. A new cytotoxic prenylated dihydrobenzofuran derivative and other chemical constituents from the rhizomes of Atractylodes lancea DC. Arch Pharm. Res. (Seoul) 2008;31(8):965–969. doi: 10.1007/s12272-001-1252-z. [DOI] [PubMed] [Google Scholar]
  16. ECoNCMMBC H. Shanghai Science and Technology Publisher; 1999. Editorial Committee of National Chinese Medical Manage Bureau Chinese Herb. [Google Scholar]
  17. Endo K., Hikino H. Sesquiterpenoids. LIV. Absolute configuration of eudesma-4 (14), 7 (11)-dien-8-one. Bull. Chem. Soc. Jpn. 1979;52(8):2439–2440. [Google Scholar]
  18. Endo K., Taguchi T., Taguchi F., Hikino H., Yamahara J., Fujimura H. Antiinflammatory principles of Atractylodes rhizomes. Chem. Pharm. Bull. 1979;27(12):2954–2958. doi: 10.1248/cpb.27.2954. [DOI] [PubMed] [Google Scholar]
  19. Feng S., Dai Z., Liu A., Wang H., Chen J., Luo Z., Yang C.S. β-Sitosterol and stigmasterol ameliorate dextran sulfate sodium-induced colitis in mice fed a high fat Western-style diet. Food. Funct. 2017;8(11):4179–4186. doi: 10.1039/c7fo00375g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Feng Z.M., Xu K., Wang W., Du N., Zhang J.H., Yang Y.N., Jiang J.S., Zhang P.C. Two new thiophene polyacetylene glycosides from Atractylodes lancea. J. Asian Nat. Prod. Res. 2018;20(6):531–537. doi: 10.1080/10286020.2018.1458841. [DOI] [PubMed] [Google Scholar]
  21. Fu S.M., Fang H.J., Liu G.S., Xiao P.G. A study on the medicinal plants of the genus Atractylodes. J. Systemat. Evol. 1981;19(2):195–202. [Google Scholar]
  22. Fu X.Q., Chou G.X., Kwan H.Y., Tse A.K.W., Zhao L.H., Yuen T.K., Cao H.H., Yu H., Chao X.J., Su T. Inhibition of STAT 3 signalling contributes to the antimelanoma action of atractylenolide II. Exp. Dermatol. 2014;23(11):855–857. doi: 10.1111/exd.12527. [DOI] [PubMed] [Google Scholar]
  23. Fu X.Q., Chou J.Y., Li T., Zhu P.L., Li J.K., Yin C.L., Su T., Guo H., Lee K.W., Hossen M.J. The JAK 2/STAT 3 pathway is involved in the anti‐melanoma effects of atractylenolide I. Exp. Dermatol. 2018;27(2):201–204. doi: 10.1111/exd.13454. [DOI] [PubMed] [Google Scholar]
  24. Gu G. People’s Medical Publishing House; Peking, China: 1955. Shen Nong's Herbal Classic. [Google Scholar]
  25. Guo L.P. China Academy of Chinese Medical Sciences; 2005. Research on Ecological Factors Affecting the Quality of Atractylodes Lancea. [Google Scholar]
  26. Guo L.P., Huang L.Q., Hu J., Shao A.J. Variation rules and chemotype classification of Atractylodes lancea essential oil based on bio-information science. Resour. Sci. 2008;30(5):770–777. [Google Scholar]
  27. Guo L.P., Liu J.Y., Ji L., Huang L.Q. Analysis of the volatile oil composition characteristics of authentic herbs of Atractylodes lancea. Chinese Journal of Traditional Chinese Medicine. 2002;27(11):814–819. [PubMed] [Google Scholar]
  28. Han N.R., Moon P.D., Nam S.Y., Ryu K.J., Yoou M.S., Choi J.H., Hwang S.Y., Kim H.M., Jeong H.J. Inhibitory effects of atractylone on mast cell-mediated allergic reactions. Chem. Biol. Interact. 2016;258:59–68. doi: 10.1016/j.cbi.2016.08.015. [DOI] [PubMed] [Google Scholar]
  29. Hashimoto T., Noma Y., Kato S., Tanaka M., Takaoka S., Asakawa Y. Biotransformation of hinesol isolated from the crude drug Atractylodes lancea by Aspergillus niger and Aspergillus cellulosae. Chemical And Pharmaceutical Bulletin-Tokyo. 1999;47:716–717. doi: 10.1248/cpb.47.716. [DOI] [PubMed] [Google Scholar]
  30. Hikino H., Hikino Y., Yosioka I. Studies on the constituents of atractylodes. IX. structure and autoxidation of atractylon. Chem. Pharm. Bull. 1964;12(7):755–760. doi: 10.1248/cpb.12.755. [DOI] [PubMed] [Google Scholar]
  31. Hiraoka N. Vitro Culture and the Evaluation of Micropropagated Plants for Sesquiterpenes and Acetylenic Compounds, Medicinal and Aromatic Plants V. Springer; 1993. Atractylodes spp; pp. 79–91. [Google Scholar]
  32. Hong M.M.J., Kim E., Kim J. Sesquiterpenes from Atractylodes japonica and their inhibitory activities on nitric oxide production in macrophage RAW264. 7 cells. Planta Med. 2012;78(5):74. [Google Scholar]
  33. Huang H.L., Lin T.W., Huang Y.L., Huang R.L. Induction of apoptosis and differentiation by atractylenolide-1 isolated from Atractylodes macrocephala in human leukemia cells. Bioorg. Med. Chem. Lett. 2016;26(8):1905–1909. doi: 10.1016/j.bmcl.2016.03.021. [DOI] [PubMed] [Google Scholar]
  34. Ina A. Structure elucidation of acylsucrose derivatives from Atractylodes lanceae rhizome and Atractylodes rhizome. Nat. Prod. Commun. 2009;4(8):1095. [PubMed] [Google Scholar]
  35. Inagaki N., Komatsu Y., Sasaki H., Kiyohara H., Yamada H., Ishibashi H., Tansho S. Acidic polysaccharides from rhizomes of Atractylodes lancea as protective principle in candida-lnfected mice. Planta Med. 2001;67(5):428–431. doi: 10.1055/s-2001-15822. [DOI] [PubMed] [Google Scholar]
  36. Jeong S.I., Kim S.Y., Kim S.J., Hwang B.S., Kwon T.H., Yu K.Y., Hang S.H., Suzuki K., Kim K.J. Antibacterial activity of phytochemicals isolated from Atractylodes japonica against methicillin-resistant Staphylococcus aureus. Molecules. 2010;15(10):7395–7402. doi: 10.3390/molecules15107395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Ji G.Q., Chen R.Q., Wang L. Anti-inflammatory activity of atractylenolide III through inhibition of nuclear factor-κB and mitogen-activated protein kinase pathways in mouse macrophages. Immunopharmacol. Immunotoxicol. 2016;38(2):98–102. doi: 10.3109/08923973.2015.1122617. [DOI] [PubMed] [Google Scholar]
  38. Ji Y., Feng X., Xiao C.C., Dong Y.F., Wang Q.Z., Wang M., Zhao Y.Y. A new polyacetylene glycoside from the rhizomes of Atractylodes lancea. Chin. Chem. Lett. 2010;21(7):850–852. [Google Scholar]
  39. Jia C., Mao D., Zhang W., Sun X. Studies on chemical constituents in essential oil from wild Atractylodes lancea in dabie mountains. J. Chin. Med. Mater. 2004;27(8):571–574. [PubMed] [Google Scholar]
  40. Jia S.X. China Press of Traditional Chinese Medicine; 2015. YaoPin Huayi. [Google Scholar]
  41. Jiang J.S., Xu K., Feng Z.M., Yang Y.N., Zhang P.C. Four new sesquiterpenes from Atractylodes lancea. Phytochem. Lett. 2018;26:88–92. [Google Scholar]
  42. Jiao P., Tseng-Crank J., Corneliusen B., Yimam M., Hodges M., Hong M., Maurseth C., Oh M., Kim H., Chu M. Lipase inhibition and antiobesity effect of Atractylodes lancea. Planta Med. 2014;80(7):577–582. doi: 10.1055/s-0034-1368354. [DOI] [PubMed] [Google Scholar]
  43. Jin W. Studies on the chemical constituents of Atractylodes chinensis (DC.) Koidz. J. Shenyang Pharm. Univ. 2002;19(3):178–179. [Google Scholar]
  44. Jun X., Fu P., Lei Y., Cheng P. Pharmacological effects of medicinal components of Atractylodes lancea (Thunb.) DC. Chin. Med-Uk. 2018;13(1):59. doi: 10.1186/s13020-018-0216-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Kamauchi H., Kinoshita K., Takatori K., Sugita T., Takahashi K., Koyama K. New sesquiterpenoids isolated from Atractylodes lancea fermented by marine fungus. Tetrahedron. 2015;71(13):1909–1914. [Google Scholar]
  46. Kawanishi F. Comparison of the outer morphologies, growth and the components in the rhizomes of Atractylodes plants cultivated in Kyoto and Bejing. Nat. Med. 1994;48:1–10. [Google Scholar]
  47. Kim J.H., Doh E.J., Lee G. Chemical differentiation of genetically identified Atractylodes japonica, A. macrocephala, and A. chinensis Rhizomes using high-performance liquid chromatography with chemometric analysis. Evid-Based. Compl. Alt. 2018;2018:1–16. doi: 10.1155/2018/4860371. Article ID 4860371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Kim H.Y., Nam S.Y., Hwang S.Y., Kim H.M., Jeong H.J. Atractylone, An active constituent of KMP6, attenuates allergic inflammation on allergic rhinitis in vitro and in vivo models. Mol. Immunol. 2016;78:121–132. doi: 10.1016/j.molimm.2016.09.007. [DOI] [PubMed] [Google Scholar]
  49. Kim J.H. Polyacetylenic compounds from Atractylodes rhizomes. The Korea Journal of Herbology. 2016;31(5):25–39. [Google Scholar]
  50. Kimura M., Kimura I., Muroi M., Yoshizaki M., Hikino H. Pharmacological evidence for an interaction between constituents (Blend effect) of the Japanese-Sino medicine "Keishi-ka-zyutubu-tö" in neuro-muscular blockade in diabetic mice. Phytother Res. 1987;1(3):107–113. [Google Scholar]
  51. Kimura Y., Sumiyoshi M. Effects of an Atractylodes lancea rhizome extract and a volatile component β-eudesmol on gastrointestinal motility in mice. J. Ethnopharmacol. 2012;141(1):530–536. doi: 10.1016/j.jep.2012.02.031. [DOI] [PubMed] [Google Scholar]
  52. Kitajima J., Kamoshita A., Ishikawa T., Takano A., Fukuda T., Isoda S., Ida Y. Glycosides of Atractylodes japonica. Chem. Pharm. Bull. 2003;51(2):152–157. doi: 10.1248/cpb.51.152. [DOI] [PubMed] [Google Scholar]
  53. Kitajima J., Kamoshita A., Ishikawa T., Takano A., Fukuda T., Isoda S., Ida Y. Glycosides of Atractylodes lancea. Chem. Pharm. Bull. 2003;51(6):673–678. doi: 10.1248/cpb.51.673. [DOI] [PubMed] [Google Scholar]
  54. Kohjyouma M., Nakajima S., Namera A., Shimizu R., Mizukami H., Kohda H. Random amplified polymorphic DNA analysis and variation of essential oil components of Atractylodes plants. Biol. Pharm. Bull. 1997;20(5):502–506. doi: 10.1248/bpb.20.502. [DOI] [PubMed] [Google Scholar]
  55. Koonrungsesomboon N., Na-Bangchang K., Karbwang J. Therapeutic potential and pharmacological activities of Atractylodes lancea (Thunb.) DC. Asian. Pac. J. Trop. Med. 2014;7(6):421–428. doi: 10.1016/S1995-7645(14)60069-9. [DOI] [PubMed] [Google Scholar]
  56. Kotawong K., Chaijaroenkul W., Muhamad P., Na-Bangchang K. Cytotoxic activities and effects of atractylodin and β-eudesmol on the cell cycle arrest and apoptosis on cholangiocarcinoma cell line. J. Pharmacol. Sci. 2018;136(2):51–56. doi: 10.1016/j.jphs.2017.09.033. [DOI] [PubMed] [Google Scholar]
  57. Lehner M.S., Steigel A., Bauer R. Diacetoxy-substituted polyacetylenes from Atractylodes lancea. Phytochemistry. 1997;46(6):1023–1028. [Google Scholar]
  58. Li X., Wang J.H., Li X., Liang D.L. Study on the chemical constituents of Atractylodes lancea Ⅰ. J. Shenyang Pharm. Univ. 2002;19(3):178–179. [Google Scholar]
  59. Lim H., Lee J.H., Kim J., Kim Y.S., Kim H.P. Effects of the rhizomes of Atractylodes japonica and atractylenolide I on allergic response and experimental atopic dermatitis. Arch Pharm. Res. (Seoul) 2012;35(11):2007–2012. doi: 10.1007/s12272-012-1118-3. [DOI] [PubMed] [Google Scholar]
  60. Li G.Y. Liaoning science and Technology Press; 2007. Essentials of Spleen and Stomach. [Google Scholar]
  61. Liu Y., Jia T., Cai Q. Determination of three components in stir-frying Atractylodes Rhizoma with and without bran by HPLC. Chinese Traditional Patent Medicine. 2013;35(1):131–135. [Google Scholar]
  62. Liang D.L., L Li X., Li X. Study on the chemical constituents of Chinese herbal medicine Atractylodes. Shandong Pharmaceutical Industry. 2002;21(5):2–4. [Google Scholar]
  63. Liu Y., Jia Z., Dong L., Wang R., Qiu G. A randomized pilot study of atractylenolide I on gastric cancer cachexia patients. Evid-Based. Compl. Alt. 2008;5(3):337–344. doi: 10.1093/ecam/nem031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Liu Y.J., Chen W.W., Zeng M., Xu K. Pharmacodynamics study of water extract before and after preparation of Atractylodes lancea. Chinese Journal of Traditional Chinese Medicine. 2012;(15):2276–2279. [PubMed] [Google Scholar]
  65. Long L., Wang L., Qi S., Yang Y., Gao H. New sesquiterpenoid glycoside from the rhizomes of Atractylodes lancea. Nat. Prod. Res. 2018;1–8 doi: 10.1080/14786419.2018.1553170. [DOI] [PubMed] [Google Scholar]
  66. Ma E.L., Li Y.C., Tsuneki H., Xiao J.F., Xia M.Y., Wang M.W., Kimura I. β-Eudesmol suppresses tumour growth through inhibition of tumour neovascularisation and tumour cell proliferation. Nat. Prod. Res. 2008;10(2):159–167. doi: 10.1080/10286020701394332. [DOI] [PubMed] [Google Scholar]
  67. Masuda Y., Kadokura T., Ishii M., Takada K., Kitajima J. Hinesol, a compound isolated from the essential oils of Atractylodes lancea rhizome, inhibits cell growth and induces apoptosis in human leukemia HL-60 cells. J. Nat. Med-Tokyo. 2015;69(3):332–339. doi: 10.1007/s11418-015-0897-5. [DOI] [PubMed] [Google Scholar]
  68. Meng H., Li G.Y., Dai R.H., Ma Y.P., Zhang K., Zhang C., Li X., Wang J.H. Two new polyacetylenic compounds from Atractylodes chinensis (DC. Koidz. J. Asian. Nat. Prod. Res. 2011;13(4):346–349. doi: 10.1080/10286020.2011.557662. [DOI] [PubMed] [Google Scholar]
  69. Meng H., Li G., Dai R., Ma Y., Zhang K., Zhang C., Li X., Wang J. Chemical constituents of Atractylodes chinensis (DC. Koidz. Biochem. Syst. Ecol. 2010;38(6):1220–1223. [Google Scholar]
  70. Mizukami H., Shimizu R., Kohda H., Kohjyouma M., Kawanishi F., Hiraoka N. Restriction fragment length polymorphisms of rDNA and variation of essential oil composition in Atractylodes plants. Biol. Pharm. Bull. 1996;19(4):577–580. doi: 10.1248/bpb.19.577. [DOI] [PubMed] [Google Scholar]
  71. Murayama C., Wang C.C., Michihara S., Norimoto H. Pharmacological effects of “jutsu” (Atractylodis Rhizome and Atractylodis lanceae rhizome) on 1-(2, 5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI)-induced head twitch response in mice (I) Molecules. 2014;19(9):14979–14986. doi: 10.3390/molecules190914979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Na-Bangchang K., Plengsuriyakarn T., Karbwang J. Research and development of Atractylodes lancea (Thunb) DC. as a promising candidate for cholangiocarcinoma chemotherapeutics. Evid-Based. Compl. Alt. 2017;2017:1–16. doi: 10.1155/2017/5929234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Nakai Y., Kido T., Hashimoto K., Kase Y., Sakakibara I., Higuchi M., Sasaki H. Effect of the rhizomes of Atractylodes lancea and its constituents on the delay of gastric emptying. J. Ethnopharmacol. 2003;84(1):51–55. doi: 10.1016/s0378-8741(02)00260-x. [DOI] [PubMed] [Google Scholar]
  74. Nakai Y., Sakakibara I., Hirakura K., Terabayashi S., Takeda S. A new acetylenic compound from the rhizomes of Atractylodes chinensis and its absolute configuration. Chem. Pharm. Bull. 2005;53(12):1580–1581. doi: 10.1248/cpb.53.1580. [DOI] [PubMed] [Google Scholar]
  75. Nasu Y., Iwashita M., Saito M., Fushiya S., Nakahata N. Inhibitory effects of Atractylodis lanceae rhizoma and Poria on collagen-or thromboxane A2-induced aggregation in rabbit platelets. Biol. Pharm. Bull. 2009;32(5):856–860. doi: 10.1248/bpb.32.856. [DOI] [PubMed] [Google Scholar]
  76. Nishikawa Y., Watanabe Y., Seto T. Studies on the evaluation of crude drugs (I) : comparative studies on the components of Atractylodes Rhizomes. Jpn. J. Pharmacogn. 1975;29:139–146. [Google Scholar]
  77. Nishikawa Y., Yasuda I., Watanabe Y., Seto T. Studies on the evaluation of crude drugs. II. Identification of the ingredients of Atractylodes by thin-layer chromatography, gas chromatography and gas chromatography-mass spectrometry, and the physical and chemical evaluation. Jpn. J. Pharmacogn. 1976;30:132–137. [Google Scholar]
  78. O'Reilly-Wapstra J.M., Miller A.M., Hamilton M.G., Williams D., Glancy-Dean N., Potts B.M. Chemical variation in a dominant tree species: population divergence, selection and genetic stability across environments. PloS One. 2013;8(3) doi: 10.1371/journal.pone.0058416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Ohara K., Katayama M., Nagai K. β-Eudesmol, an oxygenized sesquiterpene, affects efferent adrenal sympathetic nerve activity via transient receptor potential ankyrin 1 in rats. Neurosci. Lett. 2018;684:18–24. doi: 10.1016/j.neulet.2018.06.057. [DOI] [PubMed] [Google Scholar]
  80. Paniagua-Pérez R., Flores-Mondragón G., Reyes-Legorreta C., Herrera-López B., Cervantes-Hernández I., Madrigal-Santillán O., Morales-González J.A., Álvarez-González I., Madrigal-Bujaidar E. Evaluation of the anti-inflammatory capacity of beta-sitosterol in rodent assays. Afr. J. Tradit. Complem. 2017;14(1):123–130. doi: 10.21010/ajtcam.v14i1.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Piao S.H., Piao H.S. Anti-ulcer effect of n-butanol extract of Atractylodes japonica. Chinese Herbal Medicine. 1996;27(7):410–413. [Google Scholar]
  82. Plengsuriyakarn T., Viyanant V., Eursitthichai V., Picha P., Kupradinun P., Itharat A., Na-Bangchang K. Anticancer activities against cholangiocarcinoma, toxicity and pharmacological activities of Thai medicinal plants in animal models. BMC Compl. Alternative Med. 2012;12(1):23. doi: 10.1186/1472-6882-12-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Pöhnl T., Böttcher C., Schulz H., Stürtz M., Widder S., Carle R., Schweiggert R.M. Comparison of high performance anion exchange chromatography with pulsed amperometric detection (HPAEC-PAD) and ultra-high performance liquid chromatography with evaporative light scattering (UHPLC-ELSD) for the analyses of fructooligosaccharides in onion (Allium cepa L.) J. Food Compos. Anal. 2017;63:148–156. [Google Scholar]
  84. Qin J., Wang H.Y., Zhuang D., Meng F.C., Zhang X., Huang H., Lv G.P. Structural characterization and immunoregulatory activity of two polysaccharides from the rhizomes of Atractylodes lancea (Thunb.) DC. Int. J. Biol. Macromol. 2019;136:341–351. doi: 10.1016/j.ijbiomac.2019.06.088. [DOI] [PubMed] [Google Scholar]
  85. Resch M., Heilmann J., Steigel A., Bauer R. Further phenols and polyacetylenes from the rhizomes of Atractylodes lancea and their anti-inflammatory activity. Planta Med. 2001;67(5):437–442. doi: 10.1055/s-2001-15817. [DOI] [PubMed] [Google Scholar]
  86. Resch M., Steigel A., Chen Z.L., Bauer R. 5-Lipoxygenase and cyclooxygenase-1 inhibitory active compounds from Atractylodes lancea. J. Nat. Prod. 1998;61(3):347–350. doi: 10.1021/np970430b. [DOI] [PubMed] [Google Scholar]
  87. Sakurai T., Sugawara H., Saito K.I., Kano Y. Effects of the acetylene compound from Atractylodes rhizome on experimental gastric ulcers induced by active oxygen species. Biol. Pharm. Bull. 1994;17(10):1364–1368. doi: 10.1248/bpb.17.1364. [DOI] [PubMed] [Google Scholar]
  88. Sakurai T., Yamada H., Saito K., Kano Y. On the evaluation of the preparation of Chinese medicinal prescriptions VIII. Comparative studies on the acetylenes in Atractylodes rhizomes by 3D-HPLC. Nat. Med. 1994;48:291–296. [Google Scholar]
  89. Satoh K., Nagai F., Kano I. Inhibition of H+, K+-ATPase by hinesol, a major component of So-jutsu, by interaction with enzyme in the E1 state. Biochem. Pharmacol. 2000;59(7):881–886. doi: 10.1016/s0006-2952(99)00399-8. [DOI] [PubMed] [Google Scholar]
  90. Sghaier M.B., Mousslim M., Pagano A., Ammari Y., Luis J., Kovacic H. β-eudesmol, a sesquiterpene from Teucrium ramosissimum, inhibits superoxide production, proliferation, adhesion and migration of human tumor cell. Environ. Toxicol. Pharmacol. 2016;46:227–233. doi: 10.1016/j.etap.2016.07.019. [DOI] [PubMed] [Google Scholar]
  91. Shi J., Hu Y., Wang Q. Fufang cangzhu tang for treatment of senile obesity or overweight complicated with impaired glucose tolerance-a clinical observation in 32 cases. J. Tradit. Chin. Med. 2006;26(1):33–35. [PubMed] [Google Scholar]
  92. Shimato Y., Ota M., Asai K., Atsumi T., Tabuchi Y., Makino T. Comparison of byakujutsu (Atractylodes rhizome) and sojutsu (Atractylodes lancea rhizome) on anti-inflammatory and immunostimulative effects in vitro. J. Nat. Med. 2018;72(1):192–201. doi: 10.1007/s11418-017-1131-4. [DOI] [PubMed] [Google Scholar]
  93. Song H.P., Hou X.Q., Li R.Y., Yu R., Li X., Zhou S.N., Huang H.Y., Cai X., Zhou C. Atractylenolide I stimulates intestinal epithelial repair through polyamine-mediated Ca2+ signaling pathway. Phytomedicine. 2017;28:27–35. doi: 10.1016/j.phymed.2017.03.001. [DOI] [PubMed] [Google Scholar]
  94. Srijiwangsa P., Ponnikorn S., Na-Bangchang K. Effect of β-Eudesmol on NQO1 suppression-enhanced sensitivity of cholangiocarcinoma cells to chemotherapeutic agents. BMC Pharmacol. Toxico. 2018;19(1):32. doi: 10.1186/s40360-018-0223-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Ta X.S., Zhang J., Hang Y.F., Han A.P., Ding X.S. A comparative study on the hepatoprotective effect of aqueous extracts and polysaccharides before and after the processing of Atractylodes chinensis. Modern Chinese Medicine Research and Practice. 2011;25(3):45–47. [Google Scholar]
  96. Taguchi I., Kiyohara H., Matsumoto T., Yamada H. Structure of oligosaccharide side chains of an intestinal immune system modulating arabinogalactan isolated from rhizomes of Atractylodes lancea DC. Carbohydr. Res. 2004;339(4):763–770. doi: 10.1016/j.carres.2004.01.003. [DOI] [PubMed] [Google Scholar]
  97. Takeda O. Variation of essential oil components of Atractylodes lancea growing in Mt. Maoshan area in Jiangsu province, China. Nat. Med. 1994;48:11–17. [Google Scholar]
  98. Takeda O., Miki E., Terabayashi S., Okada M., Lu Y., He H.S., He S.A. A comparative study on essential oil components of wild and cultivated Atractylodes lancea and A. chinensis. Planta Med. 1996;62(5):444–449. doi: 10.1055/s-2006-957936. [DOI] [PubMed] [Google Scholar]
  99. Tang F., Fan K., Wang K., Bian C. Atractylodin attenuates lipopolysaccharide-induced acute lung injury by inhibiting NLRP3 inflammasome and TLR4 pathways. J. Pharmacol. Sci. 2018;136(4):203–211. doi: 10.1016/j.jphs.2017.11.010. [DOI] [PubMed] [Google Scholar]
  100. Tao H.J. edition. China traditional Chinese Medicine Press; 2013. Medical Records of Famous Doctors. [Google Scholar]
  101. Tian S., Yu H. Atractylenolide II inhibits proliferation, motility and induces apoptosis in human gastric carcinoma cell lines HGC-27 and AGS. Molecules. 2017;22(11):1886. doi: 10.3390/molecules22111886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Toda Y., Shigemori H., Ueda J., Miyamoto K. Isolation and identification of polar auxin transport inhibitors from Saussurea costus and Atractylodes japonica. Acta Agrobot. 2017;70(3) doi: 10.5586/aa.1700. [DOI] [Google Scholar]
  103. Tsuneki H., Ma E.L., Kobayashi S., Sekizaki N., Maekawa K., Sasaoka T., Wang M.W., Kimura I. Antiangiogenic activity of β-eudesmol in vitro and in vivo. Eur. J. Pharmacol. 2005;512(2–3):105–115. doi: 10.1016/j.ejphar.2005.02.035. [DOI] [PubMed] [Google Scholar]
  104. Tsusaka T., Makino B., Ohsawa R., Ezura H. Genetic and environmental factors influencing the contents of essential oil compounds in Atractylodes lancea. PloS One. 2019;14(5) doi: 10.1371/journal.pone.0217522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Van Minh C., Van Kiem P., Huong H.T., Lee J.J., Kim Y.H. Terpenoids and coumarin from Atractylodes lancea growing in vietnam. J. Chem. Neuroanat. 2004;42(4):499–502. [Google Scholar]
  106. Wang C., Zhao H., Yan Y., Shi J., Chen S., Tan M., Chen J., Liu Z., Chen C., Zou L. Thoughts of omics research on quality formation in Dao-di herbs. China J. Chin. Mater. Med. 2018;43(11):2407–2412. doi: 10.19540/j.cnki.cjcmm.20180307.010. [DOI] [PubMed] [Google Scholar]
  107. Wang H., Wang Y., Deng Q. People’s Health Publishing House; Beijing: 1958. Peaceful Holy Benevolent Prescriptions. [Google Scholar]
  108. Wang H.X., Liu C.M., Liu Q., Gao K. Three types of sesquiterpenes from rhizomes of Atractylodes lancea. Phytochemistry. 2008;69(10):2088–2094. doi: 10.1016/j.phytochem.2008.04.008. [DOI] [PubMed] [Google Scholar]
  109. Wang J., Huang M., Yang J., Ma X., Zheng S., Deng S., Huang Y., Yang X., Zhao P. Anti-diabetic activity of stigmasterol from soybean oil by targeting the GLUT4 glucose transporter. Food Nutr. Res. 2017;61(1):1364117. doi: 10.1080/16546628.2017.1364117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Wang L., Zhang H., Wu X., Wang Z., Fang W., Jiang M., Chen H., Huang L., Liu C. Phylogenetic relationships of Atractylodes lancea, A. chinensis and A. macrocephala, revealed by complete plastome and nuclear gene sequences. PloS One. 2020;15(1) doi: 10.1371/journal.pone.0227610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Wang K.T. Ancient Chinese Medicine Publishing House; 1993. Criterion of Syndrome and Treatment Eye Collection. [Google Scholar]
  112. Wang X.M., Yang B., Ren C.G., Wang H.W., Wang J.Y., Dai C.C. Involvement of abscisic acid and salicylic acid in signal cascade regulating bacterial endophyte‐induced volatile oil biosynthesis in plantlets of Atractylodes lancea. Physiol. Plantarum. 2015;153(1):30–42. doi: 10.1111/ppl.12236. [DOI] [PubMed] [Google Scholar]
  113. Wu J.F. The effect of rice simmering and bran frying on the volatile oil content of Atractylodes lancea. Shanghai J. Tradit. Chin. Med. 1999;4:46. [Google Scholar]
  114. Xiao P.G., Li D.P., Yang S.L. Chemical Industry Press; 2002. Modern Chinese Materia Medica. [Google Scholar]
  115. Xu F., Yang D.H., Shang M.Y., Wang X., Cai S.Q. Effective forms, additive effect, and toxicities scattering effect of pharmacodynamic substances of TCMs some reflections evoked by the study on the metabolic disposition of Traditional Chinese Medicines (TCM) World Science and Technology: Modernization of Traditional Chinese Medicine. 2014;(4):688–703. [Google Scholar]
  116. Xu J. Tianjin Medical University; 2015. Extraction, Properties and Anti-tumor Activity of Polysaccharides from Atractylodes Lancea. [Google Scholar]
  117. Xu J., Chen D., Liu C., Wu X.Z., Dong C.X., Zhou J. Structural characterization and anti-tumor effects of an inulin-type fructan from Atractylodes chinensis. Int. J. Biol. Macromol. 2016;82:765–771. doi: 10.1016/j.ijbiomac.2015.10.082. [DOI] [PubMed] [Google Scholar]
  118. Xu K. Peking Union Medical College; 2017. Chemical Constituents and Pharmacological Activities of Atractylodes Lancea. [Google Scholar]
  119. Xu K., Jiang J.S., Feng Z.M., Yang Y.N., Li L., Zang C.X., Zhang P.C. Bioactive sesquiterpenoid and polyacetylene glycosides from Atractylodes lancea. J. Nat. Prod. 2016;79(6):1567–1575. doi: 10.1021/acs.jnatprod.6b00066. [DOI] [PubMed] [Google Scholar]
  120. Xu K., Yang Y.N., Feng Z.M., Jiang J.S., Zhang P.C. Six new compounds from Atractylodes lancea and their hepatoprotective activities. Bioorg. Med. Chem. Lett. 2016;26(21):5187–5192. doi: 10.1016/j.bmcl.2016.09.069. [DOI] [PubMed] [Google Scholar]
  121. Xu W.D., Rui M.J., Ouyang Z., Zhao M., Peng H.S. Study on the inhibitory effect of water extracts of the genus Zhu on the proliferation of gastric cancer SGC-7901 cells. Shi Zhen Guo Yao Guo Yao. 2015;26(12):2861–2864. [Google Scholar]
  122. Yahara S., Higashi T., Iwaki K., Nohara T., Marubayashi N., Ueda I., Kohda H., Goto K., Izumi H., Nuno M. Studies on the constiutents of Atractylodes lancea. Chem. Pharm. Bull. 1989;37(11):2995–3000. [Google Scholar]
  123. Yang E., Zhong Y., Feng Y. Advance on the chemical constituents and pharmacological effects of Atractylodes macrocephala Koidz. J. Guangdong. Pharm. Univ. 2012;28(2):219–221. [Google Scholar]
  124. Yang L. Jiangsu University; 2007. Study on the Essential Oil from Atractylodes Lancea. [Google Scholar]
  125. Yenn T.W., Khan M.A., Syuhada N.A., Ring L.C., Ibrahim D., Tan W.N. Stigmasterol: an adjuvant for beta lactam antibiotics against beta-lactamase positive clinical isolates. Steroids. 2017;128:68–71. doi: 10.1016/j.steroids.2017.10.016. [DOI] [PubMed] [Google Scholar]
  126. Yin X., Pu Z., Wang B., Zhao S., Zhao L. A preliminary study of Rhizoma atractylodis in lts antimycotic effect. J. Beihua Univ. (Nat. Sci.). 2000;1(6):493–494. [Google Scholar]
  127. Yoou M.S., Nam S.Y., Jin M.H., Lee S.Y., Kim M.S., Roh S.S., Choi I.H., Woo N., Lim S., Kim D.H. Ameliorative effect of atractylenolide III in the mast cell proliferation induced by TSLP. Food Chem. Toxicol. 2017;106:78–85. doi: 10.1016/j.fct.2017.05.042. [DOI] [PubMed] [Google Scholar]
  128. Yosioka I., Takahashi S., Hikino H., Sasaki Y. Studies on the constituents of atractylodes III. separation of atractylol into eudesmol and hinesol. Chem. Pharm. Bull. 1959;7(3):319–323. [Google Scholar]
  129. Yosioka I., Tani T., Hirose M., Kitagawa I. Diacetyl-atractylodiol, a new acetylenic compound from Atractylodes japonica Koidzumi. Chem. Pharm. Bull. 1974;22(8):1943–1945. doi: 10.1248/cpb.22.1943. [DOI] [PubMed] [Google Scholar]
  130. Yu C., Xiong Y., Chen D., Li Y., Xu B., Lin Y., Tang Z., Jiang C., Wang L. Ameliorative effects of atractylodin on intestinal inflammation and co-occurring dysmotility in both constipation and diarrhea prominent rats. KOREAN J. PHYSIOL. PHARMACOL. 2017;21(1):1–9. doi: 10.4196/kjpp.2017.21.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Yu K.W., Kiyohara H., Matsumoto T., Yang H.C., Yamada H. Intestinal immune system modulating polysaccharides from rhizomes of Atractylodes lancea. Planta Med. 1998;64(8):714–719. doi: 10.1055/s-2006-957564. [DOI] [PubMed] [Google Scholar]
  132. Yu Y., Jia T.Z., Cai Q., Jiang N., Ma M.Y., Min D.Y., Yuan Y. Comparison of the anti-ulcer activity between the crude and bran-processed Atractylodes lancea in the rat model of gastric ulcer induced by acetic acid. J. Ethnopharmacol. 2015;160:211–218. doi: 10.1016/j.jep.2014.10.066. [DOI] [PubMed] [Google Scholar]
  133. Yuan Y., Liu Y., Huang L., Cui G., Fu G. Soil acidity elevates some phytohormone and β-eudesmol contents in roots of Atractylodes lancea. Russ. J. Plant. Physl. 2009;56(1):133–137. [Google Scholar]
  134. Zhang H., Han T., Sun L.N., Huang B.K., Chen Y.F., Zheng H.C., Qin L.P. Regulative effects of essential oil from Atractylodes lancea on delayed gastric emptying in stress-induced rats. Phytomedicine. 2008;15(8):602–611. doi: 10.1016/j.phymed.2008.02.005. [DOI] [PubMed] [Google Scholar]
  135. Zhang J.L., Huang W.M., Zeng Q.Y. Atractylenolide I protects mice from lipopolysaccharide-induced acute lung injury. Eur. J. Pharmacol. 2015;765:94–99. doi: 10.1016/j.ejphar.2015.08.022. [DOI] [PubMed] [Google Scholar]
  136. Zhang W.J., Wang S., Kang C.Z., Lv C.G., Zhou L., Huang L.Q., Guo L.P. Pharmacodynamic material basis of traditional Chinese medicine based on biomacromolecules: A review. Plant Methods. 2020;16(1):1–28. doi: 10.1186/s13007-020-00571-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Zhao H., Ji Z.H., Liu C., Yu X.Y. Neuroprotection and mechanisms of atractylenolide III in preventing learning and memory impairment induced by chronic high-dose homocysteine administration in rats. Neuroscience. 2015;290:485–491. doi: 10.1016/j.neuroscience.2015.01.060. [DOI] [PubMed] [Google Scholar]
  138. Zhao M., Wang Q., Ouyang Z., Han B., Wang W., Wei Y., Wu Y., Yang B. Selective fraction of Atractylodes lancea (Thunb.) DC. and its growth inhibitory effect on human gastric cancer cells. Cytotechnology. 2014;66(2):201–208. doi: 10.1007/s10616-013-9559-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Zhao Q.G. Guangdong science and Technology Press; 2009. Seeking the Origin of Materia Medica: the Second. [Google Scholar]
  140. Zhao Q.H. Chinese Academy of Traditional Chinese Medicine; 2009. Effect of Bran Stir Frying on Volatile Oil of Atractylodes Lancea and its Relationship with Authenticity. [Google Scholar]
  141. Zhu B., Zhang Q.L., Hua J.W., Cheng W.L., Qin L.P. The traditional uses, phytochemistry, and pharmacology of Atractylodes macrocephala Koidz.: a review. J. Ethnopharmacol. 2018;226:143–167. doi: 10.1016/j.jep.2018.08.023. [DOI] [PubMed] [Google Scholar]
  142. Zhu J H. National Taiwan University; 2012. Study on Reducing Toxicity of Atractylodes Macrocephala, Atractylodes Lancea and Xanthium Sibiricum. [Google Scholar]
  143. Zhuang D., Qin J., Wang H.Y., Zhang Y., Liu C.Y., Ding Q.Q., Lv G.P. Oligosaccharide-based quality evaluation of Atractylodis Rhizome and a strategy for simplifying its quality control. BMC. Chem. 2019;13(1):92. doi: 10.1186/s13065-019-0605-8. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Ethnopharmacology are provided here courtesy of Elsevier

RESOURCES