Ophiopogon Japonicus

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 77

Author’s Accepted Manuscript

Ophiopogon japonicus—A phytochemical,


ethnomedicinal and pharmacological review

Min-Hui Chen, Xiao-Jia Chen, Mei Wang, Li-Gen


Lin, Yi-Tao Wang

www.elsevier.com/locate/jep

PII: S0378-8741(16)30037-X
DOI: http://dx.doi.org/10.1016/j.jep.2016.01.037
Reference: JEP9948
To appear in: Journal of Ethnopharmacology
Received date: 25 July 2015
Revised date: 30 December 2015
Accepted date: 22 January 2016
Cite this article as: Min-Hui Chen, Xiao-Jia Chen, Mei Wang, Li-Gen Lin and
Yi-Tao Wang, Ophiopogon japonicus—A phytochemical, ethnomedicinal and
pharmacological review, Journal of Ethnopharmacology,
http://dx.doi.org/10.1016/j.jep.2016.01.037
This is a PDF file of an unedited manuscript that has been accepted for
publication. As a service to our customers we are providing this early version of
the manuscript. The manuscript will undergo copyediting, typesetting, and
review of the resulting galley proof before it is published in its final citable form.
Please note that during the production process errors may be discovered which
could affect the content, and all legal disclaimers that apply to the journal pertain.
Ophiopogon japonicus—A phytochemical, ethnomedicinal and pharmacological review

Min-Hui Chen 1†, Xiao-Jia Chen 1, 2†, Mei Wang 2, Li-Gen Lin 1*, Yi-Tao Wang 1*

1
State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences,

University of Macau, Macao, China

2
Sino-Dutch Center for Preventive and Personalized Medicine/Leiden Amsterdam Center for Drug Research

(LACDR), Leiden University, Leiden, The Netherlands

† The authors contributed equally to this work.

*Corresponding authors. Tel: +853-88228041 (L.G. Lin), +853-8822 4691 (Y.T. Wang); Fax:

+853-28841358; E-mail addresses: ligenl@umac.mo (L.G. Lin), ytwang@umac.mo (Y.T. Wang).

1
Contents
1. Introduction ........................................................................................................................................ 6
2. Botanical characterization and distribution ....................................................................................... 7
3. Chemical constituents ........................................................................................................................ 8
3.1 Steroidal saponins .................................................................................................................... 8
3.2 Homoisoflavonoids .................................................................................................................. 9
3.3 Polysaccharides ........................................................................................................................ 9
3.4 Other compounds ..................................................................................................................... 9
4. Quality control ................................................................................................................................. 10
5. Traditional use and ethnopharmacology .......................................................................................... 12
6. Pharmacological activities ............................................................................................................... 13
6.1 Bioactivities of crude extracts ................................................................................................ 14
6.1.1 Anti-thrombosis........................................................................................................... 14
6.1.2 Anti-inflammation ....................................................................................................... 15
6.2 Bioactivities of saponins ........................................................................................................ 15
6.2.1 Protection of the cardiovascular system ..................................................................... 15
6.2.2 Anti-inflammation ....................................................................................................... 17
6.2.3 Anticancer ................................................................................................................... 18
6.2.4 Anti-oxidation ............................................................................................................. 19
6.2.5 Antitussive activity ..................................................................................................... 20
6.2.6 Immunomodulation ..................................................................................................... 20
6.3 Bioactivities of homoisoflavonoids ....................................................................................... 20
6.4 Bioactivities of polysaccharides ............................................................................................ 21
6.4.1 Anti-myocardial ischemia ........................................................................................... 22
6.4.2 Anti-diabetes ............................................................................................................... 22
6.4.3 Anti-oxidation ............................................................................................................. 24
6.4.4 Immunomodulation ..................................................................................................... 25
6.5 Bioactivities of other components ......................................................................................... 26
6.5.1 Anti-oxidation ............................................................................................................. 26
6.5.2 Antimicrobial .............................................................................................................. 26
6.5.3 Anticancer ................................................................................................................... 26
6.6 Relationship between traditional uses and modern pharmacological activities ..................... 26
6.7 Summary of pharmacological effects .................................................................................... 28
7. Toxicity ............................................................................................................................................ 29
8. Clinical reports on the therapeutic use of O. japonicus ................................................................... 29
9. Conclusion ....................................................................................................................................... 32
References ............................................................................................................................................ 35

2
Abstract

Ethnopharmacological relevance: Ophiopogonis Radix (Maidong in Chinese), the root of

Ophiopogon japonicus, is widely used in local medicines of China, Japan and some south-eastern

Asian countries. According to the traditional Chinese medicine (TCM) principle, Ophiopogonis

Radix nourishes the yin, promotes body fluid production, moistens the lung, eases the mind and

clears away heart fire. This review summarizes the achievements of the investigations in botany,

phytochemistry, quality control, traditional uses, pharmacological activities and clinical studies on

O. japonicus; this review also describes the shortcomings of studies on this herbal drug and thus

serves as the basis of further scientific research and development of this traditional herbal drug.

Materials and methods: O. japonicus-related information was collected from various resources,

including books on Chinese herbs and the Internet databases, such as Google Scholar, SciFinder,

Web of Science, Elsevier, ACS, PubMed and China Knowledge Resource Integrated (CNKI).

Results: O. japonicus is widely distributed in East Asia, especially in China. Numerous compounds

were identified from this plant. The main components of O. japonicus include steroidal saponins,

homoisoflavonoids and polysaccharides, which exhibited various pharmacological activities, such

as cardiovascular protection, anti-inflammation, anticancer, anti-oxidation, immunomodulation,

cough relief, antimicrobial, and anti-diabetes.

Conclusions: O. japonicus is a common traditional Chinese herbal drug used as the main ingredient

in many prescriptions. Modern researches verified that O. japonicus can be used either as a healthy

food or a therapeutic agent for disease prevention and treatment. The molecular mechanisms and

chemical principles of this herbal medicine should be further explored.

3
Keywords: Ophiopogon japonicus; Phytochemistry; Quality control; Ethnopharmacology;

Pharmacology

Abbreviations

ABTS, 2,2'-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid); ADR, adverse reaction; ALT, alanine

transaminase; AST, aspartate transaminase; BBMV, intestinal brush border membrane vesicles;

CCR5, C-C chemokine receptor type 5; DOX, Doxorubicin; DPPH, 2,2-diphenyl-1-picrylhydrazyl;

DTH, delayed-type hypersensitivity; DT-13, ruscogenin

1-O-[β-D-glucopyranosyl-(1→2)]-[β-D-xylopyranosyl-(1→3)]-β-D-fucopyranoside; ELSD,

evaporating light scattering detector; eNOS, endothelial nitric oxide synthase; GSK, glycogen

synthase kinase; HDL-C, high density lipoprotein cholesterol; HKCMMS, Hong Kong Chinese

Materia Medica Standards; HPLC, high performance liquid chromatography; HUVEC, human

umbilical vein endothelial cells; ICAM-1, intercellular adhesion molecule-1; IDH, intradialytic

hypotension; IFN-γ, interferon-γ; IL, interleukin; iNOS, inducible nitric oxide synthase; InsR,

insulin receptor; IRS-1, insulin receptor substrate-1; LDL-C, low density lipoprotein cholesterol;

LPS, lipopolysaccharide; MCAO, middle cerebral artery occlusion; MMP, matrix

metalloproteinase; MNT, mouse bone marrow micronucleus test; MPO, myeloperoxidase; MS,

mass spectrometry; NF-κB, nuclear factor-κB; NSCLC, non-small cell lung cancer; OGTT, oral

glucose tolerance test; OJL, O. japonicus lectin; OOJ, oligosaccharides of O. japonicus; OPL,

Ophiopogon polysaccharide liposome; PCl, picryl chloride; PI3K, phosphoinositide 3-kinase;

PMA, phorbol-12-myristate-13-acetate; ROJ-ext, 70% ethanol fraction from aqueous extract of O.

4
japonicus root; ROS, reactive oxygen species; RUS, ruscogenin; SGMI, Shengmai injection; SMC,

Shengmai capsule; SMG, submandibular gland; SMI, Shenmai injection; SMS, Shengmai San;

SPE, solid-phase extraction; TC, total cholesterol; TCM, traditional Chinese medicine; TF, tissue

factor; TG, triglycerides; TNF-α, tumor necrosis factor-α; UV, ultraviolet; VEGF, vascular

endothelial growth factor.

5
1. Introduction

Ophiopogonis Radix (Maidong in Chinese), the root of Ophiopogon japonicus (Thunb.)

Ker-Gawl (Liliaceae), is a widely used traditional Chinese herb (Pharmacopoeia Commission of

PRC, 2015). According to traditional Chinese medicine (TCM) theory, Ophiopogonis Radix

nourishes yin, promotes body fluid production, moistens the lung, eases the mind and clears away

heart fire. Ophiopogonis Radix is listed as an edible Chinese medicine by the Chinese Ministry of

Public Health because of its efficiency, high availability and safety (Liang et al., 2012).

The chemical constituents and pharmacological activities of O. japonicus have been extensively

investigated. Phytochemical studies have revealed various biologically active compounds,

including steroidal saponins, homoisoflavonoids, and polysaccharides, which possess therapeutic

effects against acute and chronic inflammation, diabetes, cardiovascular diseases, and other

disorders (Xiao, 2002). In addition, quality control is crucial to ensure the safety and efficacy of O.

japonicus in clinical applications.

Although several reviews on the chemical and biological activities of O. japonicus have been

published, these reviews are published in Chinese and are not comprehensive. The current review

aims to systematically present and analyze recently published literatures related to the botanical

characteristics, chemical constituents, quality control, traditional uses, pharmacological activities

and clinical trials of O. japonicus. This work also emphasizes the relationship between the

traditional uses and biological activities of O. japonicus. The major achievements, shortcomings

and prospects of O. japonicus research are also discussed. This work intends to provide information

as a basis of further development and utilization of this herbal resource.

6
2. Botanical characterization and distribution

O. japonicus is a perennial tuft-forming and herbaceous plant with creeping stolons. Its root is

moderately thick and tuberous near its middle or tip. The leaf bases, generally 10–50 cm long and

1.5–3.5 mm wide, are fasciculate, tufted, sessile and garss like; these bases bear 3–7 veins and

exhibit serrulate margins. Its Inflorescence is characterized by a reduced panicle with a length of 2–

5 cm, and this part contains several to more than 10 flowers. Bracts are lanceolate, and basal ones

may measure a length of 7–8 mm. These flowers grow solitarily or in pair and are usually nodding;

the pedicel reaches up to 3–4 mm long and is articulate near its middle. Tepals are white or purplish,

lanceolate, and approximately 5 mm long. Morever, filaments are very short, and anthers are 2.5–3

mm long. The style is narrowly conical with a length of 4 mm, and its basal is moderately thick and

wide. The seeds are globose, with a diameter of 7–8 mm (Fig.1) (Editorial Board of Flora of China,

1978).

This plant is widely cultivated in East Asian countries, such as China, Korea and Japan. O.

japonicus is mainly distributed in moist and shady places, in lowland and foothills, forests, or dense

scrub at altitudes of 200–2,800 m (Editorial Board of Flora of China, 1978).

As a traditional Chinese herbal drug, this plant is challenged by some adulterants, including 23

species and 3 varieties from the genera Ophiopogon and Liriope. Ophiopogon japonicus (Thunb.)

Ker Gawl. (Liliaceae) is indicated as the indigenous herbal origin of Maidong in Chinese

Pharmacopoeia (Pharmacopoeia Commission of PRC, 2015). O. japonicus is mainly produced in

Sichuan and Zhejiang provinces in China, where this plant is called Chuanmaidong or Zhemaidong

7
respectively. Liriopes Radix (Shanmaidong in Chinese), the root of Liriope spicata (Thunb.) Lour.

var. prolifera Y.T. Ma or Liriope muscari (Decne.) Baily, is clinically used as a substitute for O.

japonicus in traditional Chinese herbal drug and is mainly distributed in Hubei and Fujian

provinces in China (Pharmacopoeia Commission of PRC, 2015; Yu and Xu, 1995; Yu et al., 1991).

3. Chemical constituents

Numerous compounds, including steroidal saponins, homoisoflavonoids and polysaccharides,

have been isolated from different parts of O. japonicus. Steroidal saponins and homoisoflavonoids,

which exhibit multiple pharmacological activities, are considered as the main active ingredients of

O. japonicus. This section describes and structurally characterizes the identified constituents of this

plant.

3.1 Steroidal saponins

Steroidal saponins are one of the main characteristic components of O. japonicus. This group of

compounds exhibit broad biological activities such as cardiovascular protection (Guan et al., 2013;

Lan et al., 2013), anti-inflammation (Song et al., 2010; Tian et al., 2011), anticancer (Chen et al.,

2013b; Zhang et al., 2012b; Zhao et al., 2013), anti-oxidation (Qian et al., 2010; Xiong et al., 2012),

immunomodulation (Xiong et al., 2012) and antitussive activity (Ishuibashi et al., 2001). Thus far,

approximately 75 steroidal saponins have been isolated from O. japonicus. Steroidal saponins are

classified into spirostanol saponins and furostanol saponins on the basis of the differences in

aglycone. Spirostanol saponins contain a hexacyclic ABCDEF-ring system as a core structure, these

saponins are further classified into ruscogenin (RUS) -type and diosgenin-type depending on the

8
structure of the ring F (Table 1 and Fig. 2). By contrast, furostanol saponins possess a pentacyclic

ABCDE-ring system with a five-carbon side chain (Table 2 and Fig. 2). The sugar moieties are

attached to the hydroxyl groups at C-1 or C-3 in the ordinary steroidal saponins.

3.2 Homoisoflavonoids

Homoisoflavonoids, a unique type of flavonoid containing one additional carbon atom between

B and C rings, mainly exist in Liliaceae and Fabaceae families (Lin et al., 2014).

Homoisoflavonoids, which exhibit anti-oxidative and anti-inflammatory activities, are another kind

of main ingredients of O. japonicus (Hung et al., 2010; Li et al., 2012a; Zhou et al., 2008a). The

homoisoflavonoids of O. japonicus are classified into two groups on the basis of the saturation of

the C2-C3 bond; one group contains a saturated C2-C3 bond and the other group comprises a

double bond at the same position (Table 3).

3.3 Polysaccharides

O. japonicus is rich in polysaccharides, which are possibly responsible for this plant’s

biological activities, such as anti-myocardial ischemia (Wang et al., 2010; Zheng et al., 2009),

anti-diabetes (Ding et al., 2012; Wang, 2013), anti-oxidation (Wang et al., 2012b; Xiong et al.,

2011) and immunomodulation (Wang et al., 2007; Xiong et al., 2011). Eleven polysaccharides have

been isolated and identified from the water extract of O. japonicus. Their structural characteristics

are summarized in Table 4.

3.4 Other compounds

Other components have also been isolated from O. japonicus, including 13 organic acids:

salicylic acid, p-hydroxybenzoic (Iqbal et al., 2004a), vanillic acid, p-hydroxybenzaldenhyde,

9
trans-p-coumaric acid, oleanolic acid, azelaic acid, n-tricosanoic acid (Cheng et al., 2005a),

tianshic acid (Cheng et al., 2005b), L-pyroglutamic acid (Dai and Mei, 2005), 9,12-octadecadienoic

acid, hexadecanoic acid and 6-octadecenoic acid (Shen et al., 2008); 4 glycosides:

ophiopogonoside A (Cheng et al., 2004), L-borneol-β-D-glucopyranoside (Cheng et al., 2005a),

3,4-dihydroxy-allylbenzene-4-O-α-L-rhamnopyranosy(1 ൺ 6)-β-D-glucopyranoside and

ophiopojaponin D (Dai and Mei, 2005); 2 cyclodipetides: cyclo-(Phe-Tyr) and cyclo-(Leu-Ile)

(Cheng et al., 2005b); and N-(2-(4-hydroxyphenyl)ethyl)-4-hydroxy-cinnamide, α-humulene

(Nakanishi and Kameda, 1987), β-sitosterol, stigmasterol, β-sitosterol-β-D-glucoside (Kato et al.,

1968), chrysophenol and emodin (Cheng et al., 2005a).

4. Quality control

Pharmacological studies have shown that steroidal saponins and homoisoflavonoids are the

main active constituents of O. japonicus. The quality control of O. japonicus thus mostly focused

on these two components. According to Chinese Pharmacopoeia (Pharmacopoeia Commission of

PRC, 2015), the total saponins in O. japonicus are determined by UV spectrophotometry with RUS

as the reference standard, and the content of total saponins in O. japonicus should not be less than

0.12% (calculated as RUS). In the Hong Kong Chinese Materia Medica Standards (HKCMMS),

ophiopogonin D is assayed by high performance liquid chromatography (HPLC) equipped with

evaporating light scattering detector (ELSD); the content of ophiopogonin D in O. japonicus should

not be less than 0.01%. HPLC, a conventional method of analyzing non-volatile compounds, has

been widely used to qualitatively and quantitatively analyze O. japonicus because of its high

10
resolution, powerful reproducibility and favorable maneuverability (Table 5). However,

pre-concentration through liquid-liquid extraction (Liu et al., 2010; Liu et al., 2007), solid-phase

extraction (SPE) (Li et al., 2013a; Qi et al., 2010; Xie et al., 2012) or macroporous resin column

chromatography (Che et al., 2008; Wang et al., 2011a; Wu et al., 2006b) are often necessary to

enrich the analytes prior to analysis because of the low contents of saponins and homoisoflavonoids

in O. japonicus. Moreover, UV detector is commonly employed for homoisoflavanoids and ELSD

for saponins on the basis of their different UV absorption spectra. Mass spectrometry (MS) can

reveal unambiguous information about the analyte’s molecular weight and structure; this technique

has been utilized to identify different components of O. japonicus (Qi et al., 2010; Wang et al.,

2011a; Xie et al., 2012) and to quantitatively analyze the components (Wang et al., 2013b; Wu et

al., 2014; Ye et al., 2005b). 1H-NMR fingerprinting is applied to characterize the components of O.

japonicus cultivated in different areas (Jiang et al., 2012). Near-infrared spectroscopy is also

employed to determine total flavonoids and total saponins in O. japonicus (Zhang and Wang, 2012).

In addition, chemometric approaches, such as hierarchical cluster analysis (Li et al., 2013a; Liu et

al., 2010; Wu et al., 2014), principle component analysis (Li et al., 2013a) and fingerprinting (Jiang

et al., 2012; Li et al., 2013a; Liu et al., 2010; Liu et al., 2007) are used to authenticate, to

differentiate, or to evaluate the quality of O. japonicus obtained from different geographical areas

and its adulterants.

O. japonicus can be distinguished from other potential product substitutes by considering the

corresponding chemical characteristics for authentication. Determination results have revealed that

saponins are the major components of Ophiopogonis Radix and Liriopes Radix; by contrast,

11
homoisoflavonoids are mainly found in Ophiopogonis Radix and hardly detected in Liriopes Radix;

this finding indicates that homoisoflavonoids can be used to distinguish these herbal drugs from each

other (Wu et al., 2014). Hyphenated techniques, such as HPLC-UV-ELSD, which is efficient for

components with and without chromophores, can be considered as a routine approach for species

identification and quality evaluation.

5. Traditional use and ethnopharmacology

O. japonicus roots, which are characterized by a wide spectrum of therapeutic effects, have

been used in TCM for many centuries. According to TCM theory, O. japonicus nourishes yin,

regenerates body fluid, moistens the lung and clears away the heart-fire. In addition, this herb is

used to treat lung dryness (cough, pharyngitis and hemoptysis) caused by yin deficiency, and

emaciation-thirst caused by fever and anxiety. O. japonicus was initially recorded in Shennong

Bencao Jing (the first Chinese Materia Medica in China during the Eastern Han Dynasty,

25AD-220AD) for the cure of heart-qi stagnation. Other well-known Materia Medica, such as

Kaibao Bencao and Bencao Mengquan (State Administration of Traditional Chinese Medicine of

PRC, 1999), have also indicated that this herb nourishes the yin of stomach, spleen, heart, and lungs,

clears heat, and eliminates irritability. Mingyi Bielu (Han Dynasty, 219 AD) has described that O.

japonicus is used to treat xerostomia and polydipsia. Bencao Shiyi (Tang Dynasty, 741 AD) has

demonstrated that this herb is used as an expectorant to relieve sore throat. Furthermore, O.

japonicus is used to treat insufficient menstruation and lactation in Yixue Qiyuan (Song Dynasty,

1127 AD). In addition, O. japonicus is used to treat pulmonary abscesses according to Yao Xing

12
Lun (Tang Dynasty, 741 AD). O. japonicus is also used to treat asthenia of the heart and the lungs,

as indicated in Bencao Yanyi (Song Dynasty, 1116 AD).

O. japonicus is combined with other herbs in TCM formulas to improve clinical effect.

Shengmai San (SMS), one commonly used formula, comprises Panax ginseng, Schisandra

chinensis and O. japonicus. SMS is used to treat viral myocarditis and coronary atherosclerotic

cardiopathy and to relieve dry cough, heat and phlegm in the lungs because of bacterial infection

(Liang et al., 2012; Xia et al., 2009; Yao et al., 2008). Table 6 shows the subsets of Chinese patent

medicines containing O. japonicus in different formulations.

In Japan, O. japonicus is traditionally used as an antiphlogistic agent to relieve sore throat and

to inhibit physiological thirst (Izawa, 1967; Kishima, 1963). O. japonicus is also a natural herbicide

used to control weeds. Furthermore, O. japonicus exerts allelopathic activity by producing plant

growth inhibitors (Iqbal et al., 2004b).

In the local medicine of Vietnam, O. japonicus also serves as a medicinal plant because of its

antitussive, expectorant and tonic effects (Do, 1991).

O. japonicus is also considered as a functional food in China and other East Asian countries. It

tastes aromatic, sweet, and mucilaginous. Although O. japonicus has been used to treat certain

diseases in ethnomedicines for thousands of years, this herbal medicine should be comprehensively

understood and recent information should be obtained for its clinical use.

6. Pharmacological activities

O. japonicus exerts various pharmacological activities, including cardiovascular protection,

13
anti-inflammation, anticancer, anti-oxidation, and anti-diabetes. To explain the nature of the active

compounds of O. japonicus, we summarize the pharmacological effects and potential mechanisms

of this plant on the basis of different types of components (Fig. 3 and Table 7).

6.1 Bioactivities of crude extracts

6.1.1 Anti-thrombosis

Thromboembolic diseases can be effectively treated by protecting the endothelium or by

preventing the adhesion of leukocytes to endothelial cells. 70% ethanol fraction of O. japonicus

root aqueous extract (ROJ-ext), which probably contained saponins as the main constituent, was

found to inhibit venous thrombosis by protecting the human umbilical vein endothelial cell line

(HUVEC) ECV304 cells and by inhibiting the adhesion of the human pro-myelocytic leukemia cell

strain HL-60 cells to ECV304 cells (Kou et al., 2005a). Moreover, ROJ-ext also inhibited tail

thrombus formation in mice injected with carrageenan (12.5 and 25.0 mg/kg) and reduced

thrombosis induced by arterial-venous shunt (silk thread) (6.25 and 12.5 mg/kg) in rats (Kou et al.,

2006).

6.1.2 Anti-inflammation

ROJ-ext (25 and 50 mg/kg) also exerted anti-inflammatory activity by inhibiting ear swelling,

paw edema, pleural leukocyte migration, peritoneal total leukocyte and neutrophil migration, as

well as reducing adhesion of HL-60 cells to ECV304 cells in mouse and rat inflammatory models

(Kou et al., 2005b).

6.2 Bioactivities of saponins

Saponins are a diverse group of natural compounds occurring in plants and foods. Studies

14
recently showed that the saponins of O. japonicus exhibited protective effects on the cardiovascular

system and exert other bioactivities including anti-inflammation, anticancer, anti-oxidation,

antitussive, and immunomodulation (Chen et al., 2013b; Xiong et al., 2012; Zhang et al., 2015).

6.2.1 Protection of the cardiovascular system

The total saponins of O. japonicus root (10 mg/kg) inhibited ventricular arrhythmias in dogs

(Chen et al., 1990). Ruscogenin1-O-[β-D-glucopyranosyl-(1→2)]-[β-D-xylopyranosyl-

(1→3)]-β-D-fucopyranoside (DT-13) (0.1 µM) from O. japonicus reduced cardiac intracellular

Ca2+ and the reperfusion damage associated with Ca2+ overload in the cardiovascular system (Tao et

al., 2005). Two glycosides isolated from O. japonicus, namely ophiopogonin D and ruscogenin

1-O-[2-O-acetyl-α-L-rhamnopyranosyl(1→2)]-β-D-xylopyranosyl (1→3)-β-D-fucopyranoside (10

and 100 μg/ml, respectively), improved the tube formation in human myocardial microvascular

endothelial cell (Lan et al., 2013). Moreover, ophiopogonin D (1 μM) attenuated the DOX-induced

autophagy by reducing the LC3-II/LC3-I ratio and inhibiting activation of c-Jun N-terminal kinase

and extracellular signal-regulated kinase in H9c2 cells (Zhang et al., 2015). RUS exerted protective

effect on ischemic stroke caused by intraluminal middle cerebral artery occlusion (MCAO) by

inhibiting the expression of nuclear factor-κB (NF-κB), intercellular adhesion molecule-1

(ICAM-1), inducible nitric oxide synthase (iNOS), cyclooxygenase, tumor necrosis factor (TNF)-α,

and interleukin (IL)-1β (Guan et al., 2013). In addition, a furostanol glycoside isolated from O.

japonicus, named ophiopogonin J, inhibited fatty acid synthase with IC50 of 76 ± 2 μM, indicating

its potential function in regulating hyperlipidemia (Duan et al., 2012).

15
6.2.2 Anti-inflammation

RUS exerted anti-inflammatory activity by suppressing the expression of ICAM-1, TNF-α and

NF-κB pathway (Song et al., 2010,Sun et al., 2012,Lu et al., 2014). RUS also attenuated

pulmonary inflammation by inhibiting the endothelial cell apoptosis and upregulating the

expression of endothelial cell membrane proteins such as eNOS, caveolin-1, and CD31 (Bi et al.,

2013). In vitro study showed that ophiopogonin C exerted inhibitory effect on the inflammatory

activities induced by leukocyte-endothelial adhesion via anti-histamine or by inhibiting the protein

kinase C activation (Tian et al., 2011).

6.2.3 Anticancer

DT-13 exerted anti-proliferative activities in the breast cancer cell line MDA-MB-435, and it

reduced tumor cell adhesion and migration, inhibited expression of tumor αvβ3 integrin, TF, early

growth response gene-1, matrix metalloproteinase (MMP)-2/9, vascular endothelial growth factor

(VEGF), C-C chemokine receptor type 5 (CCR5) mRNAs and CCR5 protein (Sun et al., 2010;

Zhang et al.,2012b; Zhao et al., 2014). DT-13 (0.01, 0.1, and 1 μM) also exhibited anti-angiogenic

activity by inhibiting tube formation and HUVEC migration and by reducing the level of p-Akt and

p-extracellular signal-regulated kinase 1/2 and p-vascular endothelial growth factor receptor 2

(Zhao et al., 2013).

In addition, Ophiopogonin B displayed cytotoxicity on a panel of non-small cell lung cancer

(NSCLC) cell lines. In NCI-H157 and H460 cells, ophiopogonin B (IC50 of 2.86 and 4.61 μM)

induced autophagy and inhibited PI3K/Akt/mTOR/p70S6K pathway (Chen et al., 2013b).

16
6.2.4 Anti-oxidation

The steroidal saponins of O. japonicus demonstrated free radical-scavenging activity. They

exhibited potent scavenging rate for 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical and hydroxyl

radical at the concentration of 5 mg/ml and 2 mg/ml, respectively (Xiong et al., 2012).

Ophiopogonin D played a protective role against H2O2-induced endothelial cell injury. In

addition, ophiopogonin D directly inhibited H2O2-induced mitochondrial ROS generation,

inflammatory responses and redox-sensitive signal transduction at the dosage of 0.6 to 60.0 μM

(Qian et al., 2010). Ophiopogonin D also showed anti-osteoporosis effect by reducing oxidative

stress. It also reduced ROS generation in mouse pre-osteoblast MC3T3-E1 subclone 4 cells and

RAW264.7 macrophage cells via the FoxO3a-β-catenin signaling pathway at 1–100 μM.

Furthermore, ophiopogonin D reduced osteoclast-specific markers and reversed the effect of

ovariectomy in vivo at dose of 5-25 mg/kg body weight (Huang et al., 2015). Nolinospiroside F

also exerted an anti-aging effect. This compound significantly extended the replicative lifespan of

K6001 yeast and increased yeast survival at doses of 1, 3, and 10 μM; this property was possibly

attributed to its anti-oxidative effect (Sun et al., 2013).

A steroidal glycoside isolated from O. japonicus, named ruscogenin 1-O-α-L-

rhamnopyranosyl-(1→2)-4-O-sulfo-α-L-arabinopyranoside-3-O-β-D-glucopyranoside, exerted

inhibitory activity against neutrophil respiratory burst stimulated by PMA with IC50 of 35.90 ± 3.00

μM as shown by a chemiluminescence assay (Qi et al., 2015a).

6.2.5 Antitussive activity

Ophiopogonis Radix is the main component of Mai-men-dong-tang, a traditional Chinese

17
medicinal prescription used to treat severe dry cough in patients with pharyngitis and bronchitis.

Ishuibashi et al. (2001) suggested that ophiopogonin D is one of the main antitussive components

of O. japonicus. Ophiopogonin D (10 μM) selectively activated K+ channels and reduced the

parasympathetic control of airway functions by inhibiting the depolarizing action of acetylcholine

in rat paratracheal ganglion neurons.

6.2.6 Immunomodulation

The saponins (100, 200 and 400 μg/ml) from O. japonicus exhibited macrophage-modulating

activities by dose-dependently promoting phagocytic capacity, macrophage viability rate, NO, and

IL-1β production (Xiong et al., 2012). Recent research found that DT-13 effectively treated liver

injury induced by delayed-type hypersensitivity (DTH) to picryl chloride (PCl) in mice. DT-13 and

RUS also improved the immunological liver injury mainly by rendering the liver-infiltrating cells

dysfunctional in a time-dependent behavior. Compared with RUS that showed slight inhibition,

DT-13 (10 or 20 mg/kg) remarkably inhibited the levels of serum alanine transaminase and

aspartate transaminase when administered during the effector phase of DTH in ICR-mice with

PCl-DTH liver-injury (Wu et al., 2000).

6.3 Bioactivities of homoisoflavonoids

The homoisoflavanoids of O. japonicus displayed anti-inflammatory and anti-oxidative

activities in vitro. Ophiopogonanone G from O. japonicus roots (25.0 μM) were beneficial against

allergic diseases by suppressing the release of the inflammatory chemokine eotaxin stimulated by

IL-4 and TNF-α in bronchial epithelial BEAS-2B cells (Hung et al., 2010). Ophiopogonanone H

isolated from O. japonicus also inhibited LPS-induced NO production in the murine microglial

18
BV-2 cell line with IC50 values of 20.1 μM (Li et al., 2012a).

Methylophiopogonanone A (2.5 and 5.0 mg/kg) attenuated blood-brain barrier disruption by

regulating the tight junction proteins and MMP-9 in MCAO and reperfusion rats, which is

associated with oxidative stress attenuation and leukocyte/EC adhesion inhibition (Lin et al., 2015).

5-Hydroxy-7,8-dimethoxy-6-methyl-3-(3',4'-dihydroxybenzyl)chroman-4-one, a homoisoflavonoid

isolated from O. japonicus, exhibited oxygen free radicals scavenging activity with IC50 of 4.52 μM

(Zhou et al., 2008a).

6.4 Bioactivities of polysaccharides

Many polysaccharides isolated from different natural sources represent a diverse class of

macromolecules, and their biological relevance was studied. The polysaccharides isolated from the

roots of O. japonicus show a variety of biological activities in vitro and in vivo, including

anti-myocardial ischemia, anti-diabetes, anti-oxidation and immunomodulation.

6.4.1 Anti-myocardial ischemia

MDG-1 (0.4–10 mM), a water-soluble β-D-fructan extracted from O. japonicus, protected

cardiomyocyte and human microvascular endothelial HMEC-1 cells from ischemia-induced cell

damage by inducing sphingosine 1-phosphate receptor 1 and basic fibroblast growth factor (Wang

et al., 2010). FOJ-5 (1-100 µg/ml) restored coronary blood flux and heart contraction and restrained

the increasing heart rate caused by ischemia–reperfusion in Langendorff model, and FOJ-5 (20 and

40 mg/kg) also increased the ST segment shift of electrocardiogram and lactate dehydrogenase

level by protecting cell membrane in blood plasma in isoprenaline-induced acute myocardial

ischemic model (Zheng et al., 2009).

19
6.4.2 Anti-diabetes

The polysaccharide-rich extract of O. japonicus (0.06–240 mg/ml) inhibited glucose absorption

into the intestinal brush border membrane vesicles, reduced the activity of α-glucosidase and

improved the activity of NIT-1 cells damaged by streptozotocin, which was associated with the

inhibition of carbohydrate digestion and absorption, as well as protection of the pancreatic islet

cells (Ding et al., 2012). The Ophiopogon polysaccharides (125, 250, and 500 mg/kg) could reduce

the fasting blood glucose and serum insulin levels, as well as improve the adiponectin mRNA level

in fat tissue and placenta of gestational diabetic mellitus rat (Wang, 2013).

MDG-1 (300 mg/kg) exhibited hypoglycemic, hypolipidemic and insulin-sensitizing activity

(Xu et al., 2011;Wang et al., 2012a;Wang et al., 2014; Zhu et al., 2014; Li et al., 2014; Shi et al.,

2015). MDG-1 improved oral glucose tolerance and reducin serum insulin level and triglyceride

content in the liver of ob/ob mice (Xu et al., 2011); upregulated the phosphoinositide 3-kinase

(PI3K) p85 subunit, Akt, insulin receptor (InsR), insulin receptor substrate-1 (IRS-1), and Glut-4

expression but downregulated glycogen synthase kinase (GSK) 3β expression (Wang et al., 2012a);

attenuated plasma lipid profiles, leptin secretion and hepatic lipid accumulation, as well as

increased the expression levels of genes related to energy and lipid metabolism in the liver (Wang

et al., 2014); suppressed intestinal glucose absorption, liver glycogenolysis, promoted liver

glycogenesis and GLP-1 secretion in KKAy mice (Zhu et al., 2014); ameliorated body weight gain,

fed blood glucose, oral glucose tolerance test (OGTT), and insulin resistance in obesity mice (Li et

al., 2014); reduced the Firmicutes/Bacteroidetes ratio, altered the abnormal gut microbiota and the

metabolic profiles in C57BL/6 mice (Shi et al., 2015).

20
OJP1 (150, 300 mg/kg), a water-soluble polysaccharide from O. japonicus, reduced blood

glucose level, increased insulin level and remediated destruction of pancreatic islets and pancreatic

β-cell damage in streptozotocin induced diabetic rats (Chen et al., 2011). Moreover, OJP1 (150 and

300 mg/kg) also protected lipid metabolism, the liver and kidney, from the injurious effects of

diabetes, as well as reduced the development of diabetic complications (Chen et al., 2013c).

The oligosaccharides of O. japonicus (OOJ) (225 and 450 mg/kg) exerted anti-diabetic effect

by increasing the body weight, improving oral glucose tolerance and reducing fasting blood glucose

level, as well as the organ related weights of liver and kidney in experimental type 2 diabetes

mellitus rats (Li et al., 2012b).

6.4.3 Anti-oxidation

Four sulfated heteropolysaccharide fractions (OJP-1, OJP-2, OJP-3 and OJP-4) isolated from

the tubers of O. japonicus exhibited anti-oxidative activity by scavenging DPPH radical (with IC50

of 145.26, 34.21, 11.12, and 7.52 mg/ml, respectively) and hydroxyl radical (with IC50 of 0.04, 8.65,

5.89, and 2.07 mg/ml, respectively) (Xiong et al., 2011). POJ-U1a (8 mg/ml), a polysaccharide of

O. japonicus, also exhibited anti-oxidative activity through its DPPH radical, hydrogen radical and

superoxide anion scavenging activities (Wang et al., 2012b).

6.4.4 Immunomodulation

The O. japonicus polysaccharides (0.05 and 0.1 g/kg) displayed beneficial effects against

Sjogren’s syndrome by increasing the salivary flow and body weight, as well as reducing the water

intake, submandibular gland (SMG) index, spleen index, interferon-γ (IFN-γ) level, and IFN-γ/IL-4

ratio of the SMG autoantigen-immunized C57BL/6 mice (Wang et al., 2007). OJP-1, OJP-2, OJP-3

21
and OJP-4 (100–400 µg/ml) exhibited macrophage-activating capability and immunomodulation

activity by promoting phagocytosis, energy metabolism rate, and NO and IL-1β production (Xiong

et al., 2011).

Ophiopogon polysaccharide liposome (OPL) could improve the immune-enhancing activity of

O. japonicus polysaccharides after encapsulation with liposome (Fan et al., 2014; Fan et al., 2015).

OPL (7.813-125 µg/ml) induced NO, iNOS, IL-6 and IL-12 secretion, and improved the expression

of the costimulatory molecules CD80 and CD86 in Kupffer cells (Fan et al., 2014), promoted

cytokine secretion and phagocytosis of macrophages, increased lymphocyte proliferation and

antibody titer, as well as improved the protective effect of the herbal drug against Newcastle

disease vaccine (Fan et al., 2015).

6.5 Bioactivities of other components

6.5.1 Anti-oxidation

Phenolic compounds, such as phenolic acids, flavonoids and tannins, were usually used as

anti-oxidants owing to their phenolic hydroxyl groups, which could act as an electron or hydrogen

donor (Li et al., 2012c). Lin et al. (2010) reported that the anti-oxidative activity of O. japonicus

was greatly influenced by the phenolics composition instead of the homoisoflavonoid

concentration.

6.5.2 Antimicrobial

O. japonicus lectin (OJL), a mannose-binding lectin purified from O. japonicus rhizome,

exhibited anti-virus activity by inhibiting the herpes simplex virus type II with EC50 of 3.93 μg/ml,

as well as exhibited antifungal activity against the phytopathogenic fungi, Gibberella saubinetii and

22
Rhizoctonia solani at the dosage of 0.06 and 0.05 mg/ml (Tian et al., 2008).

6.5.3 Anticancer

OJL exerted inhibitory effect on the growth of MCF-7 cells with IC50 of 22 µg/ml. In addition,

OJL exhibited anti-proliferative activity, as well as induced apoptosis in human breast cancer

MCF-7 cells via a caspase-dependent pathway (Liu et al., 2009).

6.6 Relationship between traditional uses and modern pharmacological activities

O. japonicus has been used in TCM as a tonic herb to nourish the yin. According to TCM theory,

yin deficiency is an internal pathogenic factors that causes diseases, leading to qi stagnation,

phlegm coagulation, blood stagnation and toxic heat accumulation. Pharmacological activities

indicate that the extracts or compounds from O. japonicus can prevent or treat disorders, especially

cardiovascular system diseases, diabetes and inflammation related diseases. To a certain extent,

these bioactivities validated the correlations between ethnomedical uses and modern scientific

evaluations.

The pathological concept of diabetes appeared in modern medical theories; however, diabetes has

a long history in TCM. In the ancient Chinese medical monograph “Huangdi Neijing,” diabetes was

described as “XiaoKe”, which is caused by pathogens factors including deficiency of yin and dryness

heat. The traditional use of O. japonicus in nourishing the yin indicates that this herb is likely to treat

diabetes. Sjogren’s syndrome is a chronic inflammatory autoimmune disease, affecting mainly the

salivary and lacrimal grands; this disease falls into the TCM categories of “dryness syndrome” and

“dry-Bi,” which result from yin depletion. O. japonicus has been used for centuries in TCM to treat

dryness-associated disorders. In terms of physiological function, the proliferative response of

23
immune cells upon antigenic challenge pertains to yin. Thus, the pathogenesis consistency, as well as

the symptomatic use of O. japonicus strengthens the ethnopharmacological relevance of its

anti-diabetes and immunomodulation activities.

In addition, the potential mechanisms of the anti-inflammatory and antimicrobial activities of O.

japonicus may be related to its ability to nourish the yin and to eliminate dry heat. Further studies

are necessary to validate this relationship.

6.7 Summary of pharmacological effects

In summary, O. japonicus exhibits a wide spectrum of pharmacological activities (Table 7).

However, no sufficient systematic data is available to explore the chemical constituents of O.

japonicus and their corresponding pharmacological activities. Therefore, the pharmacological

effects and the possible mechanisms of the pharmacological activities of O. japonicus must be

urgently explored. In addition, studies have focused on the micromolecular constituents of O.

japonicus (saponins and homoisoflavonoids), whereas investigations on the macromolecular

compounds (polysaccharides) of the plant remains insufficient. Only 11 polysaccharides have so far

been identified from this plant, and investigations on the pharmacological effects of

polysaccharides have mostly focused on the bioactivities of MDG-1. Thus, further investigations on

the pharmacological effects of the other polysaccharides of O. japonicus are a worthy endeavor.

DT-13 exhibits a remarkable pharmacological effects, including anticancer and cardioprotective

activities. Interestingly, DT-13 is only isolated from the roots of Liriope muscari (Decne.) Baily, a

substitute for O. japonicus (Ma et al., 2011; Yu et al., 1990). Whether DT-13 exists in O. japonicus

remains unproven. However, a recent study has confirmed using the LC-MS method that a low

24
amount of DT-13 can be found in O. japonicus (Wu et al., 2014). Moreover, numerous studies are

still investigating the pharmacological effects of the crude extracts; thus determining which

components in the extracts are the exact molecules that render the pharmacological effects of the

plant remains challenging.

7. Toxicity

Although O. japonicus is widely known as a functional food, as well as a medicinal herb, the

toxicity and safety evaluations for this plant remain insufficient. Only few toxicity studies on O.

japonicus decoction were performed. The genetic toxicology test for O. japonicus decoction was

performed based on the mouse lymphoma assay and mouse bone marrow micronucleus test (MNT).

In MNT, the decoction (3.34, 6.68, and 13.35 g/kg, oral gavage) did not show inhibitory effect on

the bone marrow of mice, and the number of micronucleated polychromatic erythrocytes induced in

each group did not obviously increase compared with the negative control. The results

demonstrated that O. japonicus exerted no damaging effect on the chromosome of bone marrow

cells in ICR mice and had no genotoxicity in vivo after metabolic activation (Hu et al., 2009).

Zhang et al. (2010b) investigated the potential development of toxicity caused by O. japonicus

decoction in SD rats. No significant effects was observed on the maternal body weight, fetus weight

and viability, incidences of fetal malformation and variation in SD rats administered with O.

japonicus decoction (26.9 g/kg). The results indicated that O. japonicus exerted no detectable

adverse effects to the treated female SD rats or to the fetuses.

25
8. Clinical reports on the therapeutic use of O. japonicus

So far, no clinical study reported on the sole use of O. japonicus. According to TCM theory, O.

japonicus is usually used with other herbs in TCM formulas, such as SMS (consisting of Panax

ginseng, O. japonicus and Schisandra chinensis), and Shendong Yin (consisting of Panax ginseng

and O. japonicus), which can benefit the qi, nourish the yin, replenish body fluids and prevent

exhaustion. Currently, Shengmai injection (SGMI) and Shengmai capsule (SMC) derived from

SMS, as well as Shenmai injection (SMI) derived from Shendong Yin, are the common

complementary treatments involving Chinese patent medicine that are applied in modern therapies.

A series of clinical studies including adverse reactions (ADR) and case analysis on these medicines

have been conducted in several hospitals.

In China, SMS is usually used as a complementary treatment for ischemiac heart disease and

heart failure, which are caused by yin and qi deficiencies. Twelve randomized controlled trials and

two cross-over trials of SMS in the treatment of patients with heart failure have been conducted. A

total of 858 Chinese patients (52.2% were male and 43.1% were female; age range: 23–85 years old)

participated in these trials. Some positive effects of SMS were observed resulting upgrading New

York Heart Association function classification and better performances in exercise test, ejection

fraction, cardiac output, cardiac index, left ventricular end-systolic volume, stroke volume and

myocardial contractility. Six patients from the high-dose (60 ml/day) SMS group suffered from

mild adverse effects (mild asomnia, dry mouth, fidgety responses, stomach discomfort and

hypoglycemia), and the rate of adverse effects of SMS was 0.70% (Zhou et al., 2014).

Ten randomized clinical trials involving 437 participants were performed to assess the

26
effectiveness and safety of SGMI as an adjunct therapy for intradialytic hypotension (IDH). SGMI

(40–60 ml) was administered intravenously. Compared with the conventional therapy, the SGMI

adjunct therapy showed significant effects in improving the clinical effectiveness rate (P < 0.01), in

increasing diastolic blood pressure (P < 0.01), in reducing the incidence of IDH episode (P < 0.01),

and in the frequency of nursing interventions (P < 0.01). Four studies have reported these adverse

events, although no serious adverse effects were reported in any of the included trials. Therefore,

SGMI adjunct therapy appears is a potential effective IDH treatment and is generally safe (Chen et

al., 2013a).

A clinical trial was designed to assess the effect of long-term application of SMC in coronary

heart disease treatment. A total of 60 patients (44–75 years old) were assigned into two groups for a

6-month treatment. The patients in the treatment group were administered with SMC [three

capsules (1.32 g/capsule)] each time thrice a day. SMC alleviated angina pectoris, improved

electrocardiogram, and reduced consumption of nitroglycerin of patients in the SMC group

compared with control group (Niu et al., 2000).

SMI, consisting of Panax ginseng and O. japonicus, has been commercially available and

widely used in the clinical treatment of heart failure (coronary or pulmonary heart diseases),

cerebral infarction, and malignant tumor in China since 1995. Zhang et al. (2010a) analyzed the

SMI-associated ADR. They found that 146 out of 1828 clinical studies of SMI reported a total of

576 ADR cases. The most common ADR reported in clinical studies was headache or dizziness,

and the most common SMI dosage was 40–60 ml. Moreover, 80.90% ADR cases occurred in

first-time medication, mainly in the first 30 min after injection. Several trials have suggested that

27
SMI exhibited a therapeutic effect on chronic pulmonary heart disease. A total of 33 randomized

clinical trials (2617 participants) were conducted to evaluate the effects of SMI. Compared with the

conventional medical treatment alone, the SMI combined with conventional medical treatment

demonstrated a significant improvement in the classification of clinical status in the New York

Heart Association (odds ratio 0.24; 95% confidence interval 0.19 – 0.30). Among these trials, five

studies have reported ADR cases without any serious adverse effects (Li et al., 2011).

9. Conclusion

This review summarizes the phytochemistry, quality control, traditional uses, pharmacological

activities, toxicity and clinical reports on O. japonicus. The phytochemistry results indicated that

the major components of O. japonicus are steroidal saponins, homoisoflavonoids and

polysaccharides, which exhibit multiple pharmacological effects. Homoisoflavonoids may be

developed into anti-inflammatory and anti-oxidative agents to treat neurodegenerative diseases. In

addition, ophiopogonin C and D (saponins) are potential candidates to treat cardiovascular diseases,

such as hyperlipidemia, stroke and cerebral ischemia. The traditional use of O. japonicus in TCM

to nourish the yin has been verified by therapies for various diseases, such as diabetes and Sjogren’s

syndrome. However, other traditional uses of O. japonicus have yet to be well validated by modern

pharmacological investigations. Thus, the correlations of the traditional uses of O. japonicus with

its bioactive components and their corresponding pharmacological activities should be confirmed to

optimize the development and utilization of this herb.

Although investigations on the phytochemistry and pharmacology of O. japonicus have shown

28
a remarkable success, several points should be noted to enhance our understanding of this herb.

First, although the tuberous roots of O. japonicus have been used as a medicinal component and

have been investigated for many years worldwide, only few studies have reported on the chemical

constituents and pharmacological effects of the fibrous root and the aerial parts, which also possess

potential value as a drug and functional food. For the sustainable utilization of O. japonicus, further

research should be conducted through phytochemical and pharmacological techniques. Second,

various adulterants of O. japonicus exist and are used in folk clinical treatments. The contents of

effective constituents are significantly influenced by many factors, such as geographical area,

growth condition, harvest season, and processing method. The quality control for O. japonicus to

ensure the safety and efficacy of medication remains a great challenge. However, the identification

and assay of O. japonicus differ between the Hong Kong Chinese Materia Medica Standards and

Chinese Pharmacopoeia. For instance, sample preparation methods and reference standards are

different. Hence, a common international standard should be established for the quality control of O.

japonicus. Third, O. japonicus is commonly used in combination with other herbs in conventional

therapies. The molecular mechanisms and potential interactions between O. japonicus and the

components of other herbs should be further verified. Fourth, only few relevant data on the clinical

trials of O. japonicus have been reported in Western research or have been published in English

because most studies on this subject were mainly conducted in China and published in local

publications. As a consequence, Western doctors cannot easily understand relevant data; the limited

evaluation of O. japonicus outside China reduces its external validity. In addition, most clinical

trials use a relatively small sample size and insufficient information; thus, inadequate conclusion

29
and doubtful validity of their statistical analyses have been reported. Further detailed clinical

studies should be conducted to evaluate the side effects and toxicity of O. japonicus to target

organs. Further investigations may also provide additional evidence to develop the medicinal

resources of O. japonicus.

Acknowledgments

This work was supported by the Science and Technology Development Fund, Macao S.A.R

(FDCT) (074/2012/A3) and the Research Fund of University of Macau

(MRG013/WYT/2013/ICMS, MYRG2015-00153-ICMS-QRCM and CPG2014-00012-ICMS).

Conflict of Interests

The authors declare that they have no conflict of interests.

30
References

Adinolfi, M., Parrilli, M., Zhu, Y., 1990. Terpenoid glycosides from Ophiopogon japonicus roots.

Phytochemistry 29, 1696-1699.

Asano, T., Murayama, T., Hirai, Y., Shoji, J., 1993a. Comparative studies on the constituents of

ophiopogonis tuber and its congeners. VII. Studies on the homoisoflavonoids of the subterranean

part of Ophiopogon japonicus Ker-Gawler cv. Chemical and Pharmaceutical Bulletin 41, 391-393.

Asano, T., Murayama, T., Hirai, Y., Shoji, J., 1993b. Comparative studies on the constituents of

ophiopogonis tuber and its congeners. VIII. Studies on the glycosides of the subterranean part of

Ophiopogon japonicus Ker-Gawler cv. Nanus. Chemical and Pharmaceutical Bulletin 41, 566-570.

Bi, L.-Q., Zhu, R., Kong, H., Wu, S.-L., Li, N., Zuo, X.-R., Zhou, S.-M., Kou, J.-P., Yu, B.-Y.,

Wang, H., Xie, W.-P., 2013. Ruscogenin attenuates monocrotaline-induced pulmonary hypertension

in rats. International immunopharmacology 16, 7-16.

Chang, J., Shen, C., Huang, Y., Chien, M., Ou, J., Shieh, B., Chen, C., 2002. Five new

homoisoflavonoids from the tuber of Ophiopogon japonicus. Journal of Natural Products 65,

1731-1733.

Che, R.G., Sheng, H.W., Shang, X.M., Fu, S.P., Jin, F.X., Yu, H.S., 2008. Study on the purification

of Ophiopogon japonicus total saponins with absorption resin. Lishizhen Medicine and Materia

Medica Research 19, 2068-2070.

Chen, C.Y., Lu, L.Y., Chen, P., Ji, K.T., Lin, J.F., Yang, P.L., Tang, J.F., Wang, Y., 2013a. Shengmai

injection, a traditional chinese patent medicine, for intradialytic hypotension: a systematic review

and meta-analysis. Evidence-based complementary and alternative medicine 2013, 1.

31
Chen, J.J., Zhu, Z.L., Luo, S.D., 2000. Cixi-ophiopogon A and B C27 steroidal glycosides from

Ophiopogon japonicus. Plant Diversity and Resources 22, 97–102.

Chen, M., Du, Y., Qui, M., Wang, M., Chen, K., Huang, Z., Jiang, M., Xiong, F., Chen, J., Zhou, J.,

Jiang, F., Yin, L., Tang, Y., Ye, L., Zhan, Z., Duan, J., Fu, H., Zhang, X., 2013b. Ophiopogonin

B-induced autophagy in non-small cell lung cancer cells via inhibition of the PI3K/Akt signaling

pathway. Oncology reports 29, 430-436.

Chen, M., Yang, Z.W., Zhu, J.T., Xiao, Z.Y., Xiao, R., 1990. Antiarrhythmic effects and

electrophysiological properties of Ophiopogon total saponins. Zhong Guo Yao Li Xue Bao 11, 161–

165.

Chen, X., Jin, J., Tang, J., Wang, Z., Wang, J., Jin, L., Lu, J., 2011. Extraction, purification,

characterization and hypoglycemic activity of a polysaccharide isolated from the root of

Ophiopogon japonicus. Carbohydrate Polymers 83, 749-754.

Chen, X., Tang, J., Xie, W., Wang, J., Jin, J., Ren, J., Jin, L., Lu, J., 2013c. Protective effect of the

polysaccharide from Ophiopogon japonicus on streptozotocin-induced diabetic rats. Carbohydrate

Polymers 94, 378-385.

Cheng, Z., Wu, T., Annie, B.S., Bashall, A., Yu, B., 2004. cis-Eudesmane Sesquiterpene Glycosides

from Liriope muscari and Ophiopogon japonicus. Journal of Natural Products 67, 1761-1763.

Cheng, Z.H., Wu, T., Li, L.Z., Liu, N., Yu, B.Y., Xu, L.S., 2005a. Studies on the liposoluble

components from tuber of Ophiopogon japonicus. Zhongguo yao xue za zhi 5, 337–341.

Cheng, Z.H., Wu, T., Yu, B.Y., 2005b. Chemical constituents in the tubers of Ophiopogon japonicus.

Natural Product Research and Development 17, 1-3.

32
Cheng, Z.H., Wu, T., Yu, B.Y., 2006. Steroidal glycosides from tubers of Ophiopogon japonicus.

Journal of Asian natural products research 8, 555-559.

Dai, H., Mei, W., 2005. Ophiopojaponin D, a new phenylpropanoid glycoside from Ophiopogon

japonicus Ker-Gawl. Archives of Pharmacal Research 28, 1236-1238.

Dai, H.F., Deng, S.M., Tan, N.H., Zhou, J., 2005. A new steroidal glycoside from Ophiopogon

japonicus (Thunb.) KerGawl. Journal of Integrative Plant Biology 47, 1148–1152.

Dai, H.F., Zhou, J., Ding, Z.T., Xiong, J., Tan, N.H., 2000. Two new steroidal glycosides from

Ophiopogon japonicus. Chinese Chemical Letters 11, 901-904.

Ding, L., Li, P., Lau, C.B., Chan, Y.W., Xu, D., Fung, K.P., Su, W., 2012. Mechanistic studies on

the antidiabetic activity of a polysaccharide-rich extract of Radix Ophiopogonis. Phytotherapy

Research 26, 101-105.

Do, T.L., 1991. Glossary of Vietnamese Medicinal Plants. Science and Technics Publication, Hanoi,

Vietnam pp, 793.

Duan, C.L., Kang, Z.Y., Lin, C.R., Jiang, Y., Liu, J.X., Tu, P.F., 2009. Two new homoisoflavonoids

from the fibrous roots of Ophiopogon japonicus (Thunb.) Ker-Gawl. Journal of Asian Natural

Products Research 11, 876-879.

Duan, C.L., Li, Y.J., Li, P., Jiang, Y., Liu, J., Tu, P., 2010a. Spirostanol saponins from the fibrous

roots of Ophiopogon japonicus (Thunb.) Ker-Gawl. Helvetica Chimica Acta 93, 227–232.

Duan, C.L., Ma, X.F., Jiang, Y., Liu, J.X., Tu, P.F., 2010b. Two new furostanol glycosides from the

fibrous root of Ophiopogon japonicus (Thunb.) Ker-Gawl. Journal of Asian Natural Products

Research 12, 745-751.

33
Duan, C.L., Wang, Y., Ma, X.F., Jiang, Y., Liu, J.X., Tu, P.F., 2012. A new furostanol glycoside with

fatty acid synthase inhibitory activity from Ophiopogon japonicus. Chemistry of Natural

Compounds 48, 613–615.

Editorial Board of Flora of China, 1978. Flora of China, Science Publishing House, Beijing, p. 163.

Fan, Y., Ma, X., Zhang, J., Ma, L., Gao, Y., Zhang, W., Song, X., Hou, W., Guo, C., Tong, D., 2015.

Ophiopogon polysaccharide liposome can enhance the non-specific and specific immune response

in chickens. Carbohydrate Polymers 119, 219-227.

Fan, Y., Song, X., Gao, Y., Chen, Y., Ma, L., Zhang, W., Hou, W., Guo, C., Tong, D., 2014.

Preparation and optimization of ophiopogon polysaccharide liposome and its activity on Kupffer

cells. International Journal of Pharmaceutics 477, 421-430.

Guan, J., Li, S.P., 2010. Discrimination of polysaccharides from traditional Chinese medicines

using saccharide mapping-Enzymatic digestion followed by chromatographic analysis. Journal of

Pharmaceutical and Biomedical Analysis 51, 590-598.

Guan, T., Liu, Q., Qian, Y., Yang, H., Kong, J., Kou, J., Yu, B., 2013. Ruscogenin reduces cerebral

ischemic injury via NF-kappaB-mediated inflammatory pathway in the mouse model of

experimental stroke. European Journal of Pharmacology 714, 303-311.

Hu, Y., Song, J., Wang, X., Zhang, M., Wang, X., Li, B., 2009. Genotoxicity study of Radix

Ophiopogonis Decotion. Chinese Journal of Information on TCM 16, 38-40.

Huang, Q., Gao, B., Wang, L., Zhang, H.-Y., Li, X.-J., Shi, J., Wang, Z., Zhang, J.-K., Yang, L.,

Luo, Z.-J., Liu, J., 2015. Ophiopogonin D: A new herbal agent against osteoporosis. Bone 74,

18-28.

34
Hung, T.M., Thu, C.V., Dat, N.T., Ryoo, S.W., Lee, J.H., Kim, J.C., Na, M., Jung, H.J., Bae, K.,

Min, B.S., 2010. Homoisoflavonoid derivatives from the roots of Ophiopogon japonicus and their

in vitro anti-inflammation activity. Bioorganic & Medicinal Chemistry Letters 20, 2412-2416.

Iqbal, Z., Hiradate, S., Araya, H., Fujii, Y., 2004a. Plant growth inhibitory activity of Ophiopogon

japonicus Ker Gawler and role of phenolic acids and their analogues: a comparative study. Plant

Growth Regulation 43, 245-250.

Iqbal, Z., Hiradate, S., Araya, H., Fujii, Y., 2004b. Plant growth inhibitory activity of Ophiopogon

japonicus Ker-Gawler and role of phenolic acids and their analogues: a comparative study. Plant

Growth Regulation 43, 245-250.

Ishuibashi, H., Mochidome, T., Okai, J., Ichiki, H., Shimada, H., Takahama, K., 2001. Activation of

potassium conductance by ophiopogonin-D in acutely dissociated rat paratracheal neurones. British

Journal of Pharmacology 132, 461-466.

Izawa, M.D., 1967. Colored Illustrations of Medicinal Plants (Material Medica) of Japan.

Seibundo-shinkosha Publishing Co Tokyo, Japan, pp, 272-273.

Jiang, H., Jin, H., Jiang, J., Tian, R., Wen, D., Yang, J., 2012. Comparative Study on 1H-NMR

Fingerprints of Root of Ophiopogon japonicus Cultivated in Different Areas. Progress in Modern

Biomedicine 12, 5665-5668.

Kaneda, N., Nakanishi, H., Kuraishi, T., Katori, T., 1983. Studies on the components of

Ophiopogon roots(China). I. Journal of the Pharmaceutical Society of Japan 103, 1133-1139.

Kato, H., Sakuma, S., Tada, A., Kawanishi, S., Shoji, J., 1968. Studies on the constituents of

Ophiopogonis tuber. I. Isolation of steroidal glycosides from tuber of Ophiopogon japonicus

35
Ker-Gawler var. genuinus Maxim. Journal of the Pharmaceutical Society of Japan 88, 710–714.

Kishima, M.O., 1963. Dictionary of Hirokawa Medicinal plants, Tokyo, Japan.

Kou, J., Sun, Y., Lin, Y., Zhihong, C., Zheng, W., Yu, B., Xu, Q., 2005b. Anti-inflammatory

Activities of Aqueous Extract from Radix Ophiopogon japonicus and Its Two Constituents.

Biological and Pharmaceutical Bulletin 28, 1234-1238.

Kou, J., Tian, Y., Tang, Y., Yan, J., Yu, B., 2006. Antithrombotic Activities of Aqueous Extract from

Radix Ophiopogon japonicus and Its Two Constituents. Biological and Pharmaceutical Bulletin 29,

1267-1270.

Kou, J., Yu, B., Xu, Q., 2005. Inhibitory effects of ethanol extract from Radix Ophiopogon

japonicus on venous thrombosis linked with its endothelium-protective and anti-adhesive activities.

Vascular pharmacology 43, 157-163.

Lan, S., Yi, F., Shuang, L., Chenjie, W., Zheng, X.W., 2013. Chemical constituents from the fibrous

root of Ophiopogon japonicus, and their effect on tube formation in human myocardial

microvascular endothelial cells. Fitoterapia 85, 57-63.

Li, J.S., Wang, H.F., Li, S.Y., Yu, X.Q., Wang, Z.W., 2011. Shenmai injection for chronic

pulmonary heart disease: a systematic review and meta-analysis. Journal of Alternative and

Complementary Medicine 17, 579-587.

Li, N., Che, Y., Zhang, L., Zhang, J., Zhou, Y., Jiang, Y., Tu, P., 2013a. Fingerprint analysis of

Ophiopogonis Radix by HPLC-UV-ELSD coupled with chemometrics methods. Journal of Chinese

Pharmaceutical Sciences 22, 50-56.

Li, N., Zhang, J.Y., Zeng, K.W., Zhang, L., Che, Y.Y., Tu, P.F., 2012a. Anti-inflammatory

36
homoisoflavonoids from the tuberous roots of Ophiopogon japonicus. Fitoterapia 83, 1042-1045.

Li, N., Zhang, L., Zeng, K.W., Zhou, Y., Zhang, J.Y., Che, Y.Y., Tu, P.F., 2013b. Cytotoxic steroidal

saponins from Ophiopogon japonicus. Steroids 78, 1-7.

Li, P.B., Lin, W.L., Wang, Y.G., Peng, W., Cai, X.Y., Su, W.W., 2012b. Antidiabetic activities of

oligosaccharides of Ophiopogonis japonicus in experimental type 2 diabetic rats. International

Journal of Biological Macromolecules 51, 749-755.

Li, W.J., Cheng, X.L., Liu, J., Lin, R.C., Wang, G.L., Du, S.S., Liu, Z.L., 2012c. Phenolic

compounds and antioxidant activities of Liriope muscari. Molecules 17, 1797-1808.

Li, Y., Zhu, Y.-Y., Shi, L.-L., Shen, L., Wei, H., Wang, Y., Ruan, K.-F., Feng, Y., 2014.

UPLC-TOF/MS based urinary metabonomic studies reveal mild prevention effects of MDG-1 on

metabolic disorders in diet-induced obese mice. Analytical Methods 6, 4171-4180.

Liang, H., Xing , Y., Chen, J., Zhang, D., Guo, S., Wang, C., 2012. Antimicrobial activities of

endophytic fungi isolated from Ophiopogon japonicus (Liliaceae). BMC Complementary and

Alternative Medicine 238, 1472-1477.

Lin, D., Tsuzuki, E., Sugimoto, Y., Dong, Y., Matsuo, M., Terao, H., 2004. Elementry Identification

of Phenolic Allelochemicals from Dwarf Lilyturf Plant(Ophiopogon japonicus K.) and Their

Growth-Inhibiting Effects for Two Weeds in Paddy Rice Field. Plant Production Science 7,

260-265.

Lin, L.G., Liu, Q.Y., Ye, Y., 2014. Naturally occurring homoisoflavonoids and their

pharmacological activities. Planta medica 80, 1053-1066.

Lin, M., Sun, W., Gong, W., Zhou, Z., Ding, Y., Hou, Q., 2015. Methylophiopogonanone A Protects

37
against Cerebral Ischemia/Reperfusion Injury and Attenuates Blood-Brain Barrier Disruption In

Vitro. Plos One 10, e0124558.

Lin, Y., Zhu, D., Qi, J., Qin, M., Yu, B., 2010. Characterization of homoisoflavonoids in different

cultivation regions of Ophiopogon japonicus and related antioxidant activity. Journal of

Pharmaceutical and Biomedical Analysis 52, 757-762.

Liu, B., Peng, H., Yao, Q., Li, J., Van Damme, E., Balzarini, J., Bao, J.K., 2009. Bioinformatics

analyses of the mannose-binding lectins from Polygonatum cyrtonema, Ophiopogon japonicus and

Liparis noversa with antiproliferative and apoptosis-inducing activities. Phytomedicine 16,

601-608.

Liu, J., Chen, X., Yang, W., Zhang, S., Wang, F., Tang, Z., 2010. Chemical Fingerprinting of Wild

Germplasm Resource of Ophiopogon Japonicus from Sichuan Basin, China by RP-HPLC Coupled

with Hierarchical Cluster Analysis. Analytical Letters 43, 2411-2423.

Liu, L., Lu, Y., Shao, Q., Cheng, Y., Qu, H., 2007. Binary chromatographic fingerpringting for

quality evaluation of Radix Ophiogogonis by high performance liquid chromatography coupled

with ultraviolet and evaporative light-scattering detectors. Journal of Separation Science 30,

2628-2637.

Liu, N., Wen, X.B., Liu, J.H., Liang, M., Zeng, H.J., Lin, Y., 2006. Determination of ruscogenin in

crude Chinese medicines and biological samples by immunoassay. Analytical and Bioanalytical

Chemistry 386, 1727-1733.

Liu, Y., Meng, L.Z., Xie, S.X., Xu, T.H., Sun, L.K., Liu, T.H., Xu, Y.J., Xu, D.M., 2014. Studies on

chemical constituents of Ophiopogon japonicus. Journal of Asian Natural Products Research 16,

38
982-990.

Liu, Y.X., 2013. Study on steroidal saponins from Ophiopogon japonicus. Master thesis of Nanhua

University.

Lu, H.-J., Tzeng, T.-F., Liou, S.-S., Da Lin, S., Wu, M.-C., Liu, I.M., 2014. Ruscogenin ameliorates

diabetic nephropathy by its anti-inflammatory and anti-fibrotic effects in streptozotocin-induced

diabetic rat. Bmc Complementary and Alternative Medicine 14, 1.

Ma, S., Kou, J., Yu, B., 2011. Safety evaluation of steroidal saponin DT-13 isolated from the tuber

of Liriope muscari (Decne.) Baily. Food and Chemical Toxicology 49, 2243-2251.

Nakanishi, H., Kameda, N., 1987. Studies on the component of Ophiopogon tuber (China) II.

Journal of the Pharmaceutical Society of Japan 107, 780–784.

Nguyen, T.H., Van Sung, T., Porzel, A., Franke, K., Wessjohann, L.A., 2003. Homoisoflavonoids

from Ophiopogon japonicus Ker-Gawler. Phytochemistry 62, 1153-1158.

Niu, H.Z., Li, X.M., Chen, X.Z., Zhang, X.H., Wang, X.F., 2000. Influence of Shengmai Capsule

on patients with coronary syndrome. Chinese Journal of Integrative Medicine 20, 290-291.

Okanishi, T., Akahori, A., Yasuda, F., Takeuchi, Y., Iwat, T., 1975. Steroidal Sapogenins of Sixteen

Liliaceae Plants. Chemical and Pharmaceutical Bulletin 23, 575-579.

Pharmacopoeia Commission of PRC, 2015. Pharmacopoeia of the People’s Republic of China,

2015ed. China Medical Science Press, Beijing, China.

Qi, J., Hu, Z.-f., Zhou, Y.-f., Hu, Y.-j., Yu, B.-y., 2015a. Steroidal Sapogenins and Glycosides from

the Fibrous Roots of Ophiopogon japonicus and Liriope spicata var. prolifera with

Anti-inflammatory Activity. Chemical & Pharmaceutical Bulletin 63, 187-194.

39
Qi, J., Hu, Z.F., Zhou, Y.F., Hu, Y.J., Yu, B.Y., 2015b. Steroidal Sapogenins and Glycosides from

the Fibrous Roots of Ophiopogon japonicus and liriope spicata var.prolifera with

Anti-inflammatory Activity. Chemical and Pharmaceutical Bulletin 63, 187-194.

Qi, J., Xu, D., Zhou, Y.F., Qin, M.J., Yu, B.Y., 2010. New features on the fragmentation patterns of

homoisoflavonoids in Ophiopogon japonicus by high-performance liquid

chromatography/diode-array detection/electrospray ionization with multi-stage tandem mass

spectrometry. Rapid Communications in Mass Spectrometry 24, 2193-2206.

Qian, J., Jiang, F., Wang, B., Yu, Y., Zhang, X., Yin, Z., Liu, C., 2010. Ophiopogonin D prevents

H2O2-induced injury in primary human umbilical vein endothelial cells. Journal of

Ethnopharmacology 128, 438-445.

She, G., Shi, J., 2003. Structural features of two neutral polysaccharides Md-1, Md-2 from

Ophiopogon japonocus. Zhong Yao Cai 26, 100-101.

Shen, H.L., Xiang, N.J., Xu, Y., Gao, Q., Miao, M.M., Li, J.M., 2008. Analysis of fat-soluble

components of Ophiopogon japonicus by GC-MS. Chinese Journal of Spectroscopy Laboratory 4,

669–672.

Shi, L.L., Li, Y., Wang, Y., Feng, Y., 2015. MDG-1, an Ophiopogon polysaccharide, regulate gut

microbiota in high-fat diet-induced obese C57BL/6 mice. International Journal of Biological

Macromolecules 81, 576-583.

Song, J., Kou, J., Huang, Y., Yu, B., 2010. Ruscogenin Mainly Inhibits Nuclear Factor-late gut

microbiota in high-fat diet-induced obese C57BL/6 mice. International journal of biological

macromolecules urnal of Pharmacological Sciences 113, 409-413.

40
Sun, K., Cao, S., Pei, L., Matsuura, A., Xiang, L., Qi, J.H., 2013. A Steroidal Saponin from

Ophiopogon japonicus Extends the Lifespan of Yeast via the Pathway Involved in SOD and UTH1.

International Journal of Molecular Sciences 14, 4461-4475.

Sun, L., Lin, S., Zhao, R., Yu, B., Yuan, S., Zhang, L., 2010. The Saponin Monomer of Dwarf

Lilyturf Tuber, DT-13, Reduces Human Breast Cancer Cell Adhesion and Migration during

Hypoxia via Regulation of Tissue Factor. Biological and Pharmaceutical Bulletin 33, 1192-1198.

Sun, Q., Chen, L., Gao, M., Jiang, W., Shao, F., Li, J., Wang, J., Kou, J., Yu, B., 2012. Ruscogenin

inhibits lipopolysaccharide-induced acute lung injury in mice: involvement of tissue factor,

inducible NO synthase and nuclear factor (NF)-kappaB. International Immunopharmacology 12,

88-93.

State Administration of Traditional Chinese Medicine of PRC, 1999. Chinese Materia Medica.

Shanghai Science and Technology Publishers, Shanghai, China.

Tada, A., Kasai, R., Saitoh, T., Saitoh, J., 1980b. Studies on the constituents of ophiopogonis tuber.

VI. Structures of homoisoflavonoids. (2) Chemical and Pharmaceutical Bulletin 28, 2039-2044.

Tada, A., Kasai, R., Saitoh, T., Shoji, J., 1980a. Studies on the constituents of Ophiopogonis tuber.

V. Isolation of a novel class of homoisoflavonoids and determination of their structures (1).

Chemical and Pharmaceutical Bulletin 28, 1477-1484.

Tada, A., Kobayashi, M., Shoji, J., 1973. Studies on the constituents of Ophiopogonis tuber III. On

the structure of ophiopogonin D. Chemical and Pharmaceutical Bulletin 21, 308-311.

Tada, A., Shoji, J., 1972. Studies on the constituents of Ophiopogonis tuber. II. On the structure of

ophiopogonin B. Chemical and Pharmaceutical Bulletin 20, 1729-1734.

41
Tao, J., Wang, H., Chen, J., Xu, H., Li, S., 2005. Effects of Saponin Monomer 13 of Dwarf Lilyturf

Tuber on L-Type Calcium Currents in Adult Rat Ventricular Myocytes. The American Journal of

Chinese Medicine 33, 797-806.

Tian, Q., Wang, W., Miao, C., Peng, H., Liu, B., Leng, F., Dai, L., Chen, F., Bao, J., 2008.

Purification, characterization and molecular cloning of a novel mannose-binding lectin from

rhizomes of Ophiopogon japonicus with antiviral and antifungal activities. Plant Science 175,

877-884.

Tian, Y., Kou, J., Li, L., Yu, B., 2011. Anti-inflammatory Effects of Aqueous Extract from Radix

Liriope muscari and Its Major Active Fraction and Component. Chinese Journal of Natural

Medicines 9, 222-226.

Wang, H., 2013. Preventive effects of ophiopogon-polysaccharide on apiponectin in gestational

diabetes mellitus rat. Asian Pacific Journal of Tropical Medicine 6, 296-299.

Wang, J., Liu, H.Q., Liu, L., Tang, H.T., Zhang, P., Tang, Y., 2013a. Development of full-quantified

HPLC fingerprint for quality evaluation of ophiopogonis radix of Sichuan. Zhong Yao Cai 36,

721-725.

Wang, J.Z., Ye, L.M., Chen, X.B., 2008. A new C27-steroidal glycoside from Ophiopogon

japonicus. Chinese Chemical Letters 19, 82-84.

Wang, L.Y., Wang, Y., Xu, D.S., Ruan, K.F., Feng, Y., Wang, S., 2012a. MDG-1, a polysaccharide

from Ophiopogon japonicus exerts hypoglycemic effects through the PI3K/Akt pathway in a

diabetic KKAy mouse model. Journal of Ethnopharmacology 143, 347-354.

Wang, S., Zhang, Z., Lin, X., Xu, D.S., Feng, Y., Ding, K., 2010. A polysaccharide, MDG-1,

42
induces S1P1 and bFGF expression and augments survival and angiogenesis in the ischemic heart.

Glycobiology 20, 473-484.

Wang, X.M., Sun, R.G., Zhang, J., Chen, Y.Y., Liu, N.N., 2012b. Structure and antioxidant activity

of polysaccharide POJ-U1a extracted by ultrasound from Ophiopogon japonicus. Fitoterapia 83,

1576-1584.

Wang, Y., Xu, J., Qu, H., 2011a. Structure characterization and identification steroidal saponins

from Ophiopogon japonicus Ker-Gawler (Liliaceae) by high-performance liquid chromatography

with ion trap mass spectrometry. Phytochemical Analysis 22, 166-171.

Wang, Y., Xu, J., Qu, H., 2013b. Determination of three steroidal saponins from Ophiopogon

japonicus (Liliaceae) via high-performance liquid chromatography with mass spectrometry. Natural

Product Research 27, 72-75.

Wang, Y., Xu, J., Zhang, L., Qu, H., 2011b. A new steroidal glycoside from the Ophiopogon

japonicus Ker-Gawler (Liliaceae). Natural Product Research 25, 31-35.

Wang, Y., Yan, T., Shen, J., Guo, H., Xiang, X., 2007. Preventive effect of Ophiopogon japonicus

polysaccharides on an autoallergic mouse model for Sjogren's syndrome by regulating the Th1/Th2

cytokine imbalance. Journal of Ethnopharmacology 114, 246-253.

Wang, Y., Zhu, Y.Y., Ruan, K.F., Wei, H., Feng, Y., 2014. MDG-1, a polysaccharide from

Ophiopogon japonicus, prevents high fat diet-induced obesity and increases energy expenditure in

mice. Carbohydrate Polymers 114, 183-189.

Watanabe, Y., Sanada, S., Ida, Y., Shoji, J., 1985. Comparative studies on the constituents of

Ophiopogonis tuber and its congeners. IV. Studies on the homoisoflavonoids of the Subterranean

43
part of Ophiopogon ohwii Okuyama and Ophiopogon jaburan (Kunth) Lodd. Chemical and

Pharmaceutical Bulletin 33, 5358-5363.

Watanabe, Y., Sanada, S., Tada, A., Shoji, J., 1977. Studies on the constituents of Ophiopogonis

tuber. IV. On the structures of ophiopogonin A, B', C, C', and D'. Chemical and Pharmaceutical

Bulletin 25, 3049-3055.

Wu, F., Cao, J., Jiang, J., Yu, B., Xu, Q., 2000. Ruscogenin glycoside (Lm-3) isolated from Liriope

muscari improves liver injury by dysfunctioning liver-infiltrating lymphocytes. Journal of

Pharmacy and Pharmacology 53, 681-688.

Wu, X., Dai, H., Huang, L., Gao, X., Karl, W.T., Tu, P., 2006a. A Fructan, from Radix

Ophiopogonis, Stimulates the Proliferation of Cultured Lymphocytes: Structural and Functional

Analyses. Journal Natural Products 69, 1257-1260.

Wu, X., Xu, D., Feng, Y., Gu, F., 2006b. Quantitative determination of ophiopogonin D and

ophiopogonin D' in Fibre Ophiopogon enriched extract by macroporous resin. Chinese Traditional

Patent Medicine 28, 1638-1640.

Wu, Y., Dong, Z., Wu, H., Ding, W., Zhao, M., Shi, Q., Wang, Q., 2014. Comparative studies on

Ophiopogonis and Liriopes based on the determination of 11 bioactive components using LC–

MS/MS and Hierarchical clustering analysis. Food Research International 57, 15-25.

Xia, C.H., Sun, J.G., Wang, G.J., Shang, L.L., Zhang, X.X., Zhang, R., Wang, X.J., Hao, H.P., Xie,

L., 2009. Differential effect of Shenmai injection, a herbal preparation, on the cytochrome P450

3A-mediated 1'-hydroxylation and 4-hydroxylation of midazolam. Chemico-Biological Interactions

180, 440-448.

44
Xiao, P.G., 2002. Modern Chinese Materia Medica. Chemical Industry Press Beijing, pp, 77-81.

Xie, T., Liang, Y., Hao, H., A, J., Xie, L., Gong, P., Dai, C., Liu, L., Kang, A., Zheng, X., Wang, G.,

2012. Rapid identification of ophiopogonins and ophiopogonones in Ophiopogon japonicus extract

with a practical technique of mass defect filtering based on high resolution mass spectrometry.

Journal of Chromatography A 1227, 234-244.

Xiong, S.-L., Hou, D., Huang, N., Li, A., 2012. Preparation and biological activity of saponin from

Ophiopogon japonicus. African Journal of Pharmacy and Pharmacology 6, 1964-1970.

Xiong, S.l., Li, A., Huang, N., Lu, F., Hou, D., 2011. Antioxidant and immunoregulatory activity of

different polysaccharide fractions from tuber of Ophiopogon japonicus. Carbohydrate Polymers 86,

1273-1280.

Xu, J., Wang, Y., Xu, D.S., Ruan, K.F., Feng, Y., Wang, S., 2011. Hypoglycemic effects of MDG-1,

a polysaccharide derived from Ophiopogon japonicus, in the ob/ob mouse model of type 2 diabetes

mellitus. International Journal of Biological Macromolecules 49, 657-662.

Xu, T.H., Xu, Y.J., Chen, P., Han, D., Zhao, H.F., Si, Y.S., Xu, D.M., Li, Y., Niu, J.Z., 2007. A New

Furospirostanol Saponin, Ophiofurospiside B from Ophiopogon japonicus (Thunb.) Ker-Gawl.

Chemical Research in Chinese Universities 23, 742-744.

Xu, T.H., Xu, Y.J., Xie, S.X., Zhao, H.F., Han, D., Li, Y., Niu, J.Z., Xu, D.M., 2008a. A novel

steroidal glycoside, ophiofurospiside A from Ophiopogon japonicus (Thunb.) Ker-Gawl. Journal of

Asian Natural Products Research 10, 415-418.

Xu, Y.J., Xu, T.H., Hao, L.Z., Zhao, H.F., Xie, S.X., Si, Y.S., Han, D., Xu, D.M., 2008. Two new

steroidal glucosides from Ophiopogon japonicus (L.f.) Ker-Gawl. Chinese Chemical Letters 19,

45
825-828.

Yao, H.T., Chang, Y.W., Chen, C.T., Chiang, M.T., Chang, L., Yeh, T.K., 2008. Shengmai San

reduces hepatic lipids and lipid peroxidation in rats fed on a high-cholesterol diet. Journal of

Ethnopharmacology 116, 49-57.

Ye, G., Ye, M., Guo, D., Huang, C., 2005a. Determination of Homoisoflavonoids in Ophiopogon

japonicus by RP-HPLC. Chromatographia 61, 121-125.

Ye, M., Guo, D., Ye, G., Huang, C., 2005b. Analysis of homoisoflavonoids in Ophiopogon

japonicus by HPLC-DAD-ESI-MSn. Journal of the American Society for Mass Spectrometry 16,

234-243.

Yu, B.Y., Hirai, Y., Shoji, J., Xu, G.J., 1990. Comparative Studies on the Constituents of

Ophiopogonis Tuber and Its Congeners. VI. : Studies on the Constituents of the Subterranean Part

of Liriope spicata var. prolifera and L. muscari. (1). Chemical and Pharmaceutical Bulletin 38,

1931-1935.

Zhang, J., Wang, Y., 2012. Determination of total flavonoids and total saponins in Ophiopogon

japonicus(Thunb.) Ker. Gawl by NIR spectroscopy. Chinese Journal of Spectroscopy laboratory 29,

551-555.

Zhang, L., Hu, J., Xiao, L., Zhang, Y., Zhao, W., Zheng, W., Shang, H., 2010a. Adverse drug

reactions of Shenmai injection: a systematic review. Journal of Evidence-based Medicine 3,

177-182.

Zhang, M., Liu, X., Song, J., Hu, Y., Wang, X., Li, B., 2010b. Effects of Radix Ophiopogonis is

decoction on embryo-fetal development in rats. China Journal of Chinese Materia Medica 35,

46
2334-2337.

Zhang, T., Kang, L.P., Yu, H.S., Liu, Y.X., Zhao, Y., Xiong, C.Q., Zhang, J., Zou, P., Song, X.B.,

Liu, C., Ma, B.P., 2012a. Steroidal saponins from the tuber of Ophiopogon japonicus. Steroids 77,

1298-1305.

Zhang, T., Zou, P., Kang, L.P., Yu, H.S., Liu, Y.X., Song, X.B., Ma, B.P., 2009. Two novel

furostanol saponins from Ophiopogon japonicus. Journal of Asian Natural Products Research 11,

824-831.

Zhang, Y., Liu, J., Kou, J., Yu, J., Yu, B., 2012b. DT-13 suppresses MDA-MB-435 cell adhesion

and invasion by inhibiting MMP-2/9 via the p38 MAPK pathway. Molecular Medicine Reports 6,

1121-1125.

Zhang, Y.-Y., Meng, C., Zhang, X.-M., Yuan, C.-H., Wen, M.-D., Chen, Z., Dong, D.-C., Gao, Y.-H.,

Liu, C., Zhang, Z., 2015. Ophiopogonin D Attenuates Doxorubicin-Induced Autophagic Cell Death

by Relieving Mitochondrial Damage In Vitro and In Vivo. Journal of Pharmacology and

Experimental Therapeutics 352, 166-174.

Zhao, R., Sun, L., Lin, S., Bai, X., Yu, B., Yuan, S., Zhang, L., 2013. The saponin monomer of

dwarf lilyturf tuber, DT-13, inhibits angiogenesis under hypoxia and normoxia via multi-targeting

activity. Oncology Reports 29, 1379-1386.

Zhao, R.-P., Sen-Sen, L.I.N., Yuan, S.-T., Yu, B.-Y., Bai, X.-S., Sun, L., Zhang, L.-Y., 2014. DT-13,

a saponin of dwarf lilyturf tuber, exhibits anti-cancer activity by down-regulating C-C chemokine

receptor type 5 and vascular endothelial growth factor in MDA-MB-435 cells. Chinese Journal of

Natural Medicines 12, 24-29.

47
Zheng, Q., Feng, Y., Xu, D.S., Lin, X., Chen, Y.Z., 2009. Influence of sulfation on anti-myocardial

ischemic activity of Ophiopogon japonicus polysaccharide. Journal of Asian Natural Products

Research 11, 306-321.

Zhou, C.X., Zou, L., Mo, J.X., Wang, X.Y., Yang, B., He, Q.J., Gan, L.S., 2013a.

Homoisoflavonoids from Ophiopogon japonicus. Helvetica Chimica Acta 96, 1397-1405.

Zhou, Q., Qin, W.Z., Liu, S.B., Kwong, J.S.W., Zhou, J., Chen, J., 2014. Shengmai (a traditional

Chinese herbal medicine) for heart failure (Review). The Cocbrane Library 4, 1-95.

Zhou, Y., Jin, Q., Zhu, D.N., Yu, B., 2008a. Homoisoflavonoids from Ophiopogon japonicus and Its

Oxygen Free Radicals (OFRs) Scavenging Effects. Chinese Journal of Natural Medicines 6,

201-204.

Zhou, Y.F., Hu, Y.Y., Mao, J.W., Yu, B.Y., 2013b. Steroidal aglycones from acid-hydrolyzed

products of Ophiopogon japonicus. Biotechnology 8, 1674-1677.

Zhou, Y.F., Jin, Q., Zhu, D., Yu, B., 2008b. Homoisoflavonoids from Ophiopogon japonicus and its

oxygen free radicals (OFRs) scavenging effects. Chinese Journal of Natural Medicines 6, 201-204.

Zhu, Y., Yan, K., Tu, G., 1987. Two homoisoflavones from Ophiopogon japonicus. Phytochemistry

26, 2873-2874.

Zhu, Y.Y., Cong, W.J., Shen, L., Wei, H., Wang, Y., Wang, L.Y., Ruan, K.F., Wu, F., Feng, Y., 2014.

Fecal metabonomic study of a polysaccharide, MDG-1 from Ophiopogon japonicus on diabetic

mice based on gas chromatography/time-of-flight mass spectrometry (GC TOF/MS). Molecular

bioSystems 10, 304-312.

48
Figure captions

Fig. 1 The whole plant (A) and roots (B) of Ophiopogon japonicus.

Fig. 2 Chemical structures of the other saponins from Ophiopogon japonicus.

Fig. 3 Pharmacological activities of different types of components from Ophiopogon

japonicus.

49
Table 1. Spirostanol saponins isolated from Ophiopojon japonicus.
No Compounds Structures Ref.
R1 R2 R3 R4 R5 R6 R7 25-C

O
F
R3
E R6
O R7
R1
C D R5

A B R4

R 2O

1 glycoside C O-rha(1→2)fuc H H H H H H S (Asano et al., 1993b)


2 LS-10 O-rha(1→2)xyl(1→3)fuc H H H H H H S (Asano et al., 1993b)
3 nolinospiroside F O-fuc rha H H H H H S (Sun et al., 2013)
4 25(S)-ruscogenin OH H H H H H H S (Liu et al., 2006)
5 25(R)-ruscogenin OH H H H H H H R (Liu et al., 2006)
6 ophiopogonin B O-rha(1→2)fuc H H H H H H R (Tada and Shoji, 1972)
7 ophiopogonin D O-rha(1→2)xyl(1→3)fuc H H H H H H R (Tada et al., 1973)
8 ophiopogonin A O-[Ac-rha(1→2)]fuc H H H H H H R (Watanabe et al., 1977)
9 ophiopogonin C O-[rha(1→2)]xyl(1→3)fuc Ac H H H H H R (Watanabe et al., 1977)
10 ruscogenin 1-O-sulfate O-SO3M H H H H H H R (Asano et al., 1993b)
11 (25R)-ruscogenin-3-yl α- L-rhamno O-rha(1→2)xyl(1→4)glc H H H H H H R (Duan et al., 2010a)
pyranosyl-(1→2)-[β-D-xylopyranosy
l-(1→4)]-β-D-glucopyranoside
12 (25R)-3β-hydroxyspirost-5-en-1-β-yl O-rha(1→2)xyl(1→3)ara H H H H H H R (Wang et al., 2011b)
-3-O-α-L-rhamnopyranosyl-(1→2)-
O-β-D-xylopyranosyl-(1→3)-α-L-ar
abinopyranoside
13 (23S,24S,25S)-23,24-dihydroxyrusco O-rha(1→2)xyl(1→3)ara H H H H OH fuc R (Asano et al., 1993b)
genin1-O-[α-L-rhamnopyranosyl(1
→2)][β-D-xylopyranosyl(1→3)]-α-L

50
-arabinopyranoside
24-O-β-D-fucopyranoside
14 (23S,24S,25S)-23,24-dihydroxyrusco O-[Ac-rha(1→2)]xyl(1→3)ara H H H H OH fuc R (Asano et al., 1993b)
geninI-O-[α-L-2,3,4-tri-O-acetylrha
mnopyranosyl(1→2)][β-D-xylopyran
osyl(1→3)]-α-L-arabinopyranoside
24-O-β-D-fucopyranoside
15 (23S,24S,25S)-23,24-dihydroxyrusco O-[Ac-rha(1→2)] xyl(1→3)ara H H H H OH fuc R (Asano et al., 1993b)
genin1-O-[α-L-2,3,4-triO-acetylrham
nopyranosyl(1→2)][β-D-xylopyrano
syl(1→3)]-α-L-arabinopyranoside
24-O-β-D-fucopyranoside
16 diosgenin H H H H H H H Unknown (Okanishi et al., 1975)
17 ophiopogoninB’ H [Ac-rha(1→2)] xyl(1→3)glc H H H H H Unknown (Watanabe et al., 1977)
18 ophiopogonin C’ H rha(1→2) glc H H H H H Unknown (Watanabe et al., 1977)
19 ophiopogonin D’ H rha(1→2)xyl(1→3)glc H H H H H Unknown (Watanabe et al., 1977)
20 diosgenin-3-O-[2-O-acetyl-α-L-rham H [Ac-rha(1→2)] xyl(1→4)glc H H H H H Unknown (Duan et al., 2010a)
nopyranosyl-(1→2)][β-D-xylopyran
osyl-(1→4)]-β-D-glucopyranoside
21 ophiopogonin P H [Ac-rha(1→2)] xyl(1→4)glc H H H H H Unknown (Li et al., 2013b)
22 ophiopogonin Q H rha(1→2)[ Ac-xyl(1→4)]glc H H H H H Unknown (Li et al., 2013b)
23 sprengerinin A H xyl(1→4)glc H H H H H Unknown (Li et al., 2013b)
24 sprengerin C H rha(1→2) xyl(1→4)glc H H H H H Unknown (Li et al., 2013b)
25 ophiogenin H H H OH OH H H R (Nakanishi and
Kameda, 1987)
26 ophiogenin H rha(1→2)glc H OH OH H H R (Adinolfi et al., 1990)
3-O-α-L-rhamnopyranosyl-(1→2)-β-
D-glucopyranoside
27 Bornyl H ara(1→6)glc H OH OH H H R (Adinolfi et al., 1990)
7-O-α-L-arabinofuranosyl(1→6)-β-
D-glucopyranoside
28 cixi-ophiopogon A H rha(1→2)xyl(1→3)glc H OH OH H H R (Chen et al., 2000)
51
29 cixi-ophiopogon B H rha(1→2)xyl(1→3)glc(1→4)glc H OH OH H H R (Chen et al., 2000)
30 ophiopojaponin C H rha(1→2) xyl(1→4)glc H OH OH H H R (Dai et al., 2005)
31 (25R)-14α,17α-hydroxyspirost-5-en- H rha(1→2) glc(1→3)glc H OH OH H H R (Wang et al., 2011b)
3β-yl3-O-α-L-rhamnpyranosyl-(1→2
)-β-D-glucopyranosyl-(1→3)-β-D-gl
ucopyranoside
32 ophiopogonin O H rha(1→2) xyl(1→4)glc H OH OH H glc R (Zhang et al., 2012a)
33 ophiopogonin R H rha(1→2) glc OH OH OH H H R (Li et al., 2013b)
34 ophiopojaponin A H [Ac-rha(1→2)] xyl(1→3)glc H H OH H H R (Dai et al., 2000)
35 ophiopogonin E H xyl(1→4)glc H H OH H H R (Cheng et al., 2006)
36 pennogenin-3-O-α-L-rhamnopyranos H rha(1→2)xyl(1→4)glc H H OH H H R (Wang et al., 2008)
yl-(1→2)-β-D-xylopyranosyl-(1→4)
-β-D-glucopyranosid
37 floribundasaponin B H rha(1→4)glc H H OH H H R (Wang et al., 2008)
38 pennogenin-3-O-[2-O-acetyl-α-L-rha H [Ac-rha(1→2)] xyl(1→4)glc H H OH H H R (Duan et al., 2010a)
mnopyranosyl-(1→2)][β-D-xylopyra
nosyl-(1→4)]-β-D-glucopyranoside
39 (25R)spirost-5-ene-3β,14α-diol-3-β- H rha(1→2)xyl (1→4)glc H OH H H H R (Xu et al., 2008)
O-β-L-rhamnopyranosyl(1α-dioβ-D-
xylopyranosyl(1(1α-dβ-D-glucopyra
noside
40 ophiopogonin S H xyl (1→4)glc H OH H H H R (Li et al., 2013b)
41 14-hydroxydiosgenin3-O-α-L-rhamn H rha(1→2) glc H OH H H H R (Li et al., 2013b)
opyranosyl-(1→2)-β-D-
glucopyranoside
42 14-hydroxydiosgenin H rha(1→2)xyl(1→4)glc H O H H H R (Li et al., 2013b)
3-O-α-L-rhamnopyranosyl-(1→2)-β-
D-xylopyranosyl-(1→4)-β-D-glucop
yranoside
43 prazerigenin A H H H OH H H H R (Zhou et al., 2013b)

52
44 ruscogenin1-O-α-L-rhamnopyranosy O-rha(1ė2)-4-O-sulfo-ara glc H H H H H R (Qi et al., 2015b)
l-(1→2)-4-O-sulfo-α-L-arabinopyran
oside-3-O-β-D-glucopyranoside
45 ruscogenin1-O-[2-O-acetyl-α-L-rha O-[Ac-rha(1→2)]xyl(1→3)fuc H H H H H H R (Dai et al., 2005)
mnopyranosyl(1→2)]-β-D-xylopyran
osyl(1→3)-β-D-fucopyranoside
46 ruscogenin1-O-[β-D-glucopyranosyl- glc(1→2)xyl(1→3)fuc H H H H H H R (Ma et al., 2011)
(1→2)]-[β-D-xylopyranosyl-(1→3)]-
β-D-fucopyranoside
O
OR 2
F
E
O

C D

A B

R 1O

47 ophiofurospiside A rha(1→2)xyl(1→4)glc glc ü ü ü ü ü ü (Xu et al., 2008a)


48 ophiofurospiside B man(1→2)ara(1→4)glc glc ü ü ü ü ü ü (Xu et al., 2007)

53
Table 2. Furostanol saponins isolated from Ophiopojon japonicus
No Compounds Structures Ref.
R1 R2 R3 R4 R5 R6
OH
OR6
R3
E
O
R1
C D R5

A B R4

R 2O

49 ophiopojaponin B H rha(1→2)glc H OH H glc (Dai et al., 2000)


50 ophiopogonin J H rha(1→2)xyl(1→4)glc H OH H glc (1→2)glc (Zhang et al., 2012a)
51 ophiopogonin L H rha(1→2)xyl(1→4) glc OH OH H glc (Zhang et al., 2012a)
52 ophiopogonin N H rha(1→2)xyl(1→4)glc H OH H glc (1→6)glc (Zhang et al., 2012a)
53 ophiopogonin K H rha(1→2)glc H OH OH glc (1→2)glc (Zhang et al., 2012a)
54 ophiopogonin M H xyl(1→4)rha(1→2)glc OH OH OH glc (Zhang et al., 2012a)
55 ophiopogonin F H xyl(1→4)rha(1→2)glc H H H glc (1→2)glc (Zhang et al., 2009)
56 ophiopogonin G H xyl(1→4)rha(1→2)glc H H H glc (1→6)glc (Zhang et al., 2009)
57 ophiopogonin H H rha(1→2)glc H H H glc (1→2)glc (Duan et al., 2010b)
58 Ophiofurospiside C H rha(1→2)xyl(1→4)glc H H OH glc(1→6)glc (Liu, 2013)
59 Ophiofurospiside D H rha(1→2)glc OH OH OH glc (Liu, 2013)
60 Ophiofurospiside E H rha(1→2)xyl(1→4)glc H OH OH glc (Liu, 2013)
61 Ophiofurospiside F H rha(1→2)glc H OH OH glc (Liu, 2013)
62 Ophiofurospiside G H xyl(1→4)glc H H H glc(1→6)glc (Liu, 2013)
63 Ophiofurospiside H H xyl(1→4)glc H H H glc (1→2)glc (Liu, 2013)
64 Ophiofurospiside I H rha(1→2)glc H OH H glc (1→2)glc (Liu, 2013)
65 Ophiofurospiside J H glc H H H glc (1→2)glc (Liu, 2013)
66 Ophiofurospiside K H rha(1→2)glc H H H glc(1→6)glc (Liu, 2013)
67 Ophiofurospiside L OH rha(1→2)xyl(1→4) glc H H H glc (Liu, 2013)
68 Ophiofurospiside M OH rha(1→2)glc H H H glc (Liu, 2013)
69 Ophiofurospiside N O-rha(1→2)xyl(1→3)fuc H H H H glc (Liu, 2013)

54
70 26-O-β-D-glucopyranosyl(25S)-furost-5-ene-1β,3β,22α,2 O-xyl(1→3)rha(1→2)fuc H H H H glc (Xu et al., 2008)
6- tetraol 1-O-aol -glucopyranosyl(25S)[α-L-
rhamnopyranosyl-(1nosyl(25S)[α-L-st-5-ene

71 26-O-β-D-glucopyranosyl-(25R)- furost-5-en-3β,14αˈ H rha(1→2)glc H OH OH glc (Liu et al., 2014)


17α,22α,26-pentaol-3-O-α-L-rhamnopyranosyl-(1→2)-
β-D-glucopyranoside

HO OR2

E
O

C D

A B

R 1O

72 ophiopogonin I glc(1ė2)glc rha(1ė2)glc ü ü ü ü (Duan et al., 2010b)


73 (25R)-26-[(O-β-D-glucopyranosy-(1→2)-β-D-glucopyran rha(1ė2)xyl(1ė4)glc glc(1ė2)glc ü ü ü ü (Duan et al., 2012)
osyl)]-20α-hydroxyfurost-5,22-diene-3-O-α-L-rhamnopyr
anosyl-(1→2)-[β-D-xylopyranosyl(1→4)]-β-D-
glucopyranoside

55
Table 3. Homoisoflavonoids isolated from Ophiopojon japonicus.

No Compounds Structures Ref.


R1 R2 R3 R4 R5 R6 R7 R8 R9 R10

76 methylophiopogonone B OH CH3 OH CH3 H H OCH3 ü ü ü (Guan and Li, 2010;


Tada et al., 1980a)
77 ophiopogonone B OH CH3 OH H H H OCH3 ü ü ü (Tada et al., 1980b)
78 6-formylisoophiopogonone B OH CHO OH CH3 H H OCH3 ü ü ü (Zhu et al., 1987)
79 5,7-dihydroxy-3-(4′-hydroxybenzyl)-6-methylchromo OH CH3 OH H H H OH ü ü ü (Asano et al., 1993a)
ne
80 desmethylisoophiopogonone B OH H OH CH3 H H OH ü ü ü (Asano et al., 1993a)
81 ophiopogonone D OH CH3 OH H OH H OH ü ü ü (Duan et al., 2009)
82 8-formyl-7-hydroxy-5,4′-dimethoxy-6-methylhomois OCH3 CH3 OH CHO H H OCH3 ü ü ü (Lan et al., 2013)
oflavone
83 8-formylophiopogonone B OH CH3 OH CHO H H OCH3 ü ü ü (Wang et al., 2013a)
84 methylophiopogonone A OH CH3 OH CH3 H -O-CH2-O- ü ü ü (Tada et al., 1980a)
85 isoophiopogonone A OH H OH CH3 H -O-CH2-O- ü ü ü (Tada et al., 1980b)
86 6-aldehydo-isoophiopogonone A OH CHO OH CH3 H -O-CH2-O- ü ü ü (Zhu et al., 1987)
87 ophiopogonone A OH CH3 OH H H -O-CH2-O- ü ü ü (Asano et al., 1993a)
88 ophiopogonone C OH OH OH CH3 CHO -O-CH2-O ü ü ü (Chang et al., 2002)
89 2′-hydroxy-methylophiopogonone A OH CH3 OH CH3 OH -O-CH2-O- ü ü ü (Watanabe et al., 1985)
90 5,7,2′-trihydroxy-6-methyl-3-(3′,4′-methylenedioxybe OH CH3 OH H OH -O-CH2-O- ü ü ü (Asano et al., 1993a)
nzyl)chromone
91 5,7,2′-trihydroxy-8-methyl-3-(3′,4′-methylenedioxybe OH H OH CH3 OH -O-CH2-O- ü ü ü (Asano et al., 1993a)
nzyl)chromone

56
No Compounds Structures Ref.
R1 R2 R3 R4 R5 R6 R7 R8 R9 R10
92 5,7,2′-trihydroxy-6,8-dimethyl-3-(3′,4′-methylenediox OH CH3 OH CH3 OH -O-CH2-O- ü ü ü (Zhou et al., 2008b)
ybenzyl)chromone

93 5-hydroxy-7,8-dimethoxy-6-methyl-3-(3′,4′-dihydrox OH CH3 OCH3 OCH3 H OH OH H H H (Asano et al., 1993a)


ybenzyl)chroman-4-one
94 ophiopogonanone E OH CH3 OH OCH3 OH H OCH3 H H H (Chang et al., 2002)
95 5,8,4′-trimethoxy-6-methyl-7,2′-dihydroxy-3-benzylc OCH3 CH3 OH OCH3 OH OCH3 H H H H (Chang et al., 2002)
hroman-4-one
96 5,7-dihydroxy-6,8-dimethyl-3-(4′-hydroxy-3′-methox OH CH3 OH CH3 H H OH OCH3 H H (Nguyen et al., 2003)
ybenzyl)chroman-4-one
97 5-hydroxy-7,8-dimethoxy-6-methyl-3-(3′,4′-dihydrox OH CH3 OCH3 OCH3 H H OH OH H H (Nguyen et al., 2003)
ybenzyl)chroman-4-one
98 5,7-dihydroxy-6,8-dimethyl-3-(4′-hydroxy-3′5′-dimet OH CH3 OH CH3 H OCH3 OH OCH3 H H (Nguyen et al., 2003)
hoxybenzyl)chroman-4-one
99 5,7-dihydroxy-6-methyl-3(R)-(2,4-dihydroxybenzyl) OH CH3 OH H OH H OH H H H (Duan et al., 2009)
chroman-4-one
100 8-Formyl-5-O-methyl ophiopogonanone B OCH3 CH3 OH CHO H H OCH3 H H H (Zhou et al., 2013)
101 5,7,2′,3′-tetrahydroxy-6-methyl-8-methoxy-3-(4′-meth OH CH3 OH OCH3 OH OH OCH3 H H H (Hung et al., 2010)
oxybenzyl)chroman-4-one
102 methylophiopogonanone B OH CH3 OH CH3 H H OCH3 H H H (Tada et al., 1980a)
103 2,5,7-trihydroxy-6,8-dimethyl-3-(4′-methoxybenzyl)c OH CH3 OH CH3 H H OCH3 H OH H (Nguyen et al., 2003)
hroman-4-one
104 8-formylophiopogonanone B OH CH3 OH CHO H H OCH3 H H H (Zhou et al., 2013)
105 methylophiopogonanone A OH CH3 OH CH3 H -O-CH2-O- H H H (Tada et al., 1980a)
106 ophiopogonanone A OH CH3 OH H H -O-CH2-O- H H H (Kaneda et al., 1983)

57
No Compounds Structures Ref.
R1 R2 R3 R4 R5 R6 R7 R8 R9 R10
107 ophiopogonanone C OH CH3 OH CHO H -O-CH2-O- H H H (Chang et al., 2002)
108 ophiopogonanone H OH CH3 OH CH3 H -O-CH2-O- H H OH (Li et al., 2012a)
109 5-methoxy-6-methyl-7-hydroxy-8-aldehydo-3-(3′,4′- OCH3 CH3 OH CHO H -O-CH2-O- H H H (Chang et al., 2002)
methylenedioxybenzyl)chroman-4-one
110 2,5,7-trihydroxy-6,8-dimethyl-3-(3′,4′-methylenediox OH CH3 OH CH3 H -O-CH2-O- H OH H (Nguyen et al., 2003)
ybenzyl)-chroman-4-one
111 5,7-trihydroxy-6,8-dimethyl-3-(2′-hydroxy-3′,4′-meth OH CH3 OH CH3 OH -O-CH2-O- H H H (Nguyen et al., 2003)
ylenedioxybenzyl)chromone

58
Table 4. Polysaccharides isolated and purified from Ophiopojon japonicus.
Name MW(kDa) Molar Ratio Structural characteristics(backbone and branch) Ref.
Md-1 2.7064 --- Backbone: D-glucose and α-(1→4)glc (She and Shi, 2003)
Md-2 4.8651 --- Backbone: D-glucose and α-(1→4)glc (She and Shi, 2003)
MDG-1 3.4 --- Backbone: β-(2→1)-Fruf and β-(2→6)-Fruf (Wang et al., 2010)
Branch: Fruf (2→6) Fruf (2→ per average 2.8 of main chain residues and trace of α-D-Glc) Fruf
FOJ-5 5 --- Backbone: β-(2→1)-Fruf and β-(2→6)-Fruf (Zheng et al., 2009)
Branch: Fruf (2→6) Fruf (2→ per average 2.8 of main chain residues and trace of α-D-Glc) Fruf
Opaw-2 14 fru: glc=30:1 Backbone: β-(1→2) - Fruf and β-(2→6) -Fruf (Wu et al., 2006a)
Branch: (2→1) D- fructosyl
OJP-1 2.74 --- --- (Xiong et al., 2011)
OJP-2 124.30 --- --- (Xiong et al., 2011)
OJP-3 324.65 --- --- (Xiong et al., 2011)
OJP-4 6.75 --- --- (Xiong et al., 2011)
POJ-U1a 4.02 glcf:glcp=3:11 Backbone:1,6-α-D-glucopyranose and 1,3,6-α-D-glucofuranose (Wang et al., 2012b)
Branch: 1,3-α-D-glucopyranose and 1-α-D-glucopyranose
OJP1 35.2 ara:glc:gal=1:16:8 Backbone:1,6-linked glc; 1,4-linked glc; and 1,4,6-linked glc (Chen et al., 2011)

59
Table 5. Selected applications of HPLC for qualitative and quantitative analysis of Ophiopogon japonicus.

Analytes Sample Sample preparation Column Mobile phase Detection Ref.


3 homoisoflavonoids O. 1. Ultrasonication with Zorbax Extend-C18 Acetonitrile - 0.3% UV 296 nm / (Ye et al.,
(methylophiopogonanone A, japonicus chloroform-methanol(1:1) (4.6 mm i.d. × 250 mm, 5 μm) acetic acid with 274 nm 2005a)
methylophiopogonanone B, 2. Evaporation to dryness gradient elution
6-aldehydo-isoophiopogonanone A) 3. Dissolving with acetonitrile
18 homoisoflavonoids O. 1. Ultrasonication with Zorbax Extend-C18 Acetonitrile - 0.3% UV 285 nm (Ye et al.,
japonicus chloroform-methanol (1:1) (4.6 mm i.d. × 250 mm, 5 μm) acetic acid with ESI-MS 2005b)
2. Evaporation to dryness gradient elution
3. Dissolving with acetonitrile
28 homoisoflavonoids O. 1. Reflux with ethanol Zorbax SB-C18 Acetonitrile - 0.1% UV 296 nm (Qi et al.,
japonicus 2. Evaporation to dryness and dissolving (4.6 mm i.d. × 50 mm, 1.8 μm) formic acid with ESI/MS 2010)
with water gradient elution
3. Fraction on SPE column (C18)
4. Evaporation of the 95% ethanol eluent to
dryness and dissolving with methanol
3 steroidal saponins O. 1. Ultrasonication with 90% ethanol Tigerkin C18 Acetonitrile - 0.02% ESI-MS (Wang et
(cixi-ophiopogon A, japonicus (4.6 mm i.d.× 250 mm, 5 μm) formic acid with al.,
cixi-ophiopogon B, gradient elution 2013b)
cixi-ophiopogon C)
8 steroidal saponins O. 1. Reflux with 90% ethanol Tigerkin C18 Acetonitrile - 0.05% UV 203 nm (Wang et
japonicus 2. Evaporation to dryness and dissolving (4.6 mm i.d.× 250 mm, 5 μm) formic acid with ESI-MS al.,
with water gradient elution 2011a)
3. Fraction on a D101 resin column
4. Concentration of the 70% ethanol eluent
and dissolving in methanol

60
Analytes Sample Sample preparation Column Mobile phase Detection Ref.
6 phenolic acids (salicylic acid, O. 1. Soak with methanol Zorbax ODS Methanol - 2% acetic UV 254 nm (Lin et al.,
syringic acid, syringaldehyde, japonicus 2. Evaporation to dryness and dissolving (4.6 mm i.d. × 250 mm, 5 μm) acid with gradient 2004)
vanillic acid, p-hydroxybenzoic with methylene chloride elution
acid, sinapic acid) 3. Evaporation to dryness and redissolving
with methanol
6 steroidal saponins (ophiopogon O. 1. Ultrasonication with 50% ethanol Luna-C18 Acetonitrile - 0.3% ESI-MS (Wu et
Ra, ophiopojaponin C, japonicus, 2. Concentration to dryness (2.00 mm i.d. × 150 mm, 5 μm) acetic acid with al., 2014)
ophiopogonin D, ophiopogonin D’, Liriope 3. Dissolving with 50% ethanol gradient elution
liriope muscari baily saponins C, spicata var.
liriopesides B) and 5 flavonoids prolifera
(ophiopogonanone E, and Liriope
methylophiopogonanone A, muscari
methylophiopogonanone B,
ophiopogonanone C,
methylophiopogonone A)
50 ophiopogonins and 27 O. 1. Ultrasonication with 75% ethanol Luna-C18 Acetonitrile - 0.02% Q-TOF-MS (Xie et
ophiopogonones japonicus 2. Evaporation to dryness and dissolving (2.1 mm i.d. × 150 mm, 5 μm,) acetic acid with al., 2012)
with water gradient elution
3. Fraction on SPE cartridge (C18)
4. Evaporation of methanol eluent to
dryness
5. Reconstitution with acetonitrile
Fingerprint O. 1. Reflux with methanol Lichrospher C18 0.05% formic acid in UV 280 nm (Liu et al.,
japonicus, 2. Evaporation to dryness and dissolving (4.6 mm i.d. × 250 mm, 5 μm) acetonitrile – 0.05% ELSD 2007)
Liriope spp. with water formic acid in water
3. Extraction with n-butanol with gradient elution
4. Evaporation to dryness and dissolving
with methanol

61
Analytes Sample Sample preparation Column Mobile phase Detection Ref.
Fingerprint O. 1. Ultrasonication with ethanol Symmetry-C18 Methanol - 0.2% UV 280 nm (Liu et al.,
japonicus 2. Evaporation to dryness and dissolving (4.6 mm i.d. × 250 mm, 5 μm) acetic acid with 2010)
with water gradient elution
3. Extraction with ether
4. Evaporation to dryness and dissolving
with methanol
Fingerprint O. 1. Ultrasonication with methanol Zorbax SB-C18 Acetonitrile/methanol UV 296 nm (Li et al.,
japonicus 2. Concentration to dryness and dissolving (4.6 mm i.d. × 250 mm, 5 μm) (3:1) - 0.1% formic ELSD 2013a)
with water acid with gradient ESI-MS
3. Fraction on SPE column (C18) elution
4. Evaporation of methanol eluent to
dryness
5. Dissolving with methanol

62
Table 6. The traditional and clinical uses of Ophiopogon japonicus in China.

Preparation Name Compositions Traditional uses Ref.


Shashen Maidong Glehnia littoralis F.Schmidt ex Miq., Ophiopogon japonicus (Thunb.) Ker Clearing heat of lung and stomach, promoting fluid Wenbing Tiaobian(Qing Dynasty,
Decoction Gawl., Polygonatum odoratum (Mill.) Druce, Glycyrrhiza uralensis Fisch., production and moisturizing dryness AD 1798)
Morus alba L., Lablab purpureus (L.) Sweet, Trichosanthes kirilowii Maxim
Zengye Decoction Scrophularia ningpoensis Hemsl., Ophiopogon japonicus (Thunb.) Ker Promoting body fluid to moisturize dryness, curing Wenbing Tiaobian(Qing Dynasty,
Gawl., Rehmannia glutinosa (Gaertn.) DC. constipation AD 1798)
Maidong Maren Ophiopogon japonicus (Thunb.) Ker Gawl., Cannabis sativa L., Paeonia Nourishing yin of stomach, promoting fluid production Wenbing Tiaobian(Qing Dynasty,
Decoction lactiflora Pall., Reynoutria multiflora (Thunb.) Moldenke, Prunus and moisturizing dryness, curing constipation due to AD 1798)
mume (Siebold) Siebold & Zucc., Anemarrhena asphodeloides Bunge impairment of the body fluid
Maimendong Curing epistaxis and hematemesis due to deficiency of Sheng Hui Fang (Song Dynasty,
Ophiopogon japonicus (Thunb.) Ker Gawl., Cirsium japonicum (Thunb.)
Yinzi both qi and yin AD 992)
Fisch. ex DC., Rehmannia glutinosa (Gaertn.) DC.
Maimendong Ophiopogon japonicus (Thunb.) Ker Gawl., Platycodon grandiflorus (Jacq.) Curing pulmonary abscess Shengji Zonglu(Song Dynasty,
Decoction A.DC., Glycyrrhiza uralensis Fisch. AD 1117)
Maimendong Ophiopogon japonicus (Thunb.) Ker Gawl., Pinellia ternata (Thunb.) Curing asthmatic cough due to fluids deficiency in the Jingui Yaolue(Donghan Dynasty,
Decoction Makino, Panax ginseng C.A.Mey., Glycyrrhiza uralensis Fisch., Oryza lung and stomach with the flaring up of deficiency fire AD 300)
sativa L., Ziziphus jujuba Mill.
Maimendong Pill Ophiopogon japonicus (Thunb.) Ker Gawl., Coptis chinensis Franch. Curing sore throat, pulmonary heat symptoms Pu Ji Fang (Ming Dynasty, AD
1390)
Shengmai San Panax ginseng C.A.Mey., Ophiopogon japonicus (Thunb.) Ker Gawl., Curing promordial qi due to lung-heat, increasing the Qian Jin Fang (Tang Dynasty,
Schisandra chinensis (Turcz.) Baill. body fluid and quenching thirst AD 652)
Shendong Yin Panax ginseng C.A.Mey., Ophiopogon japonicus (Thunb.) Ker Gawl. Curing pant due to qi deficiency and clearing heat Zheng Yin Mai Zhi (Qing
Dynasty, AD 1706)
Erdong Gao Asparagus cochinchinensis (Lour.) Merr., Ophiopogon japonicus (Thunb.) Nourishing yin and moisturizing the lung, curing “Chinese Pharmacopoeia”, vol. 1.
Ker Gawl. cough caused by dryness and pharyngalgia
Kouyanqing Asparagus cochinchinensis (Lour.) Merr., Ophiopogon japonicus (Thunb.) Nourishing yin and clearing heat, detumescence by “Chinese Pharmacopoeia”, vol. 1.
granule Ker Gawl., Scrophularia ningpoensis Hemsl., Lonicera japonica Thunb., detoxification, curing inflammation in oral cavity
Glycyrrhiza uralensis Fisch.

63
Preparation Name Compositions Traditional uses Ref.
Xiao’er Zhisou Scrophularia ningpoensis Hemsl, Ophiopogon japonicus (Thunb.) Ker Clearing heat by nourishing lung, relieving cough and “Chinese Pharmacopoeia”, vol. 1.
syrup Gawl., Arisaema heterophyllum Blume, Prunus armeniaca L., reducing sputum
Areca catechu L., Platycodon grandiflorus (Jacq.) A.DC., Bambusa
tuldoides Munro, Morus alba L., Trichosanthes kirilowii Maxim.,
Fritillaria cirrhosa D.Don, Trichosanthes kirilowii Maxim.,
Glycyrrhiza uralensis Fisch., Perilla frutescens (L.) Britton, Anemarrhena
asphodeloides Bunge
Shengmai capsule Panax ginseng C.A.Mey., Ophiopogon japonicus (Thunb.) Ker Gawl., Nourishing yin and promoting fluid production, curing “Chinese Pharmacopoeia”, vol. 1.
Schisandra chinensis (Turcz.) Baill. palpitate and short of breath, making one feel at ease
and energetic
Xuanmai Ganju Scrophularia ningpoensis Hemsl., Ophiopogon japonicus (Thunb.) Ker Nourishing yin and clearing heat, curing sore throat “Chinese Pharmacopoeia”, vol. 1.
tablets Gawl., Glycyrrhiza uralensis Fisch., Platycodon grandiflorus (Jacq.) A.DC. and reducing pharyngeal swelling
Maiwei Dihuang Ophiopogon japonicus (Thunb.) Ker Gawl., Schisandra chinensis (Turcz.) Curing syndrome of yin deficiency in lung and kidney, “Chinese Pharmacopoeia”, vol. 1.
pill Baill., Rehmannia glutinosa (Gaertn.) DC., Cornus officinalis Siebold & reducing weak in waist and knee
Zucc., Paeonia × suffruticosa Andrews, Dioscorea oppositifolia L., Poria
cocos(Schw.)Wolf, Alisma plantago-aquatica L.
Yangxin Dingji Rehmannia glutinosa (Gaertn.) DC., Ophiopogon japonicus (Thunb.) Ker Nourishing the blood and qi, calming the mind and “Chinese Pharmacopoeia”, vol. 1.
oral liquid Gawl., Panax ginseng C.A.Mey., Ziziphus jujuba Mill., Equus asinus L., relieving palpitation, curing arrhythmia, insomnia and
Sesamum indicum L., Cinnamomum cassia (L.) J.Presl, night sweat
Zingiber officinale Roscoe, Glycyrrhiza uralensis Fisch.
Yangyin Qingfei Rehmannia glutinosa (Gaertn.) DC., Ophiopogon japonicus (Thunb.) Ker Nourishing yin and moisturizing the dryness, curing “Chinese Pharmacopoeia”, vol. 1.
pill Gawl., Scrophularia ningpoensis Hemsl., Fritillaria cirrhosa D.Don, dry cough with little sputum
Paeonia lactiflora Pall., Paeonia suffruticosa Andrews,
Mentha canadensis L., Glycyrrhiza uralensis Fisch.
Guanxin Panax ginseng C.A.Mey., Ophiopogon japonicus (Thunb.) Ker Gawl., Tonifying qi, promoting fluid production and coronary “Chinese Pharmacopoeia”, vol. 1.
Shengmai oral Schisandra chinensis (Turcz.) Baill., Salvia miltiorrhiza Bunge, circulation, curing palpitate and short of breath
liquid Paeonia lactiflora Pall., Curcuma aromatica Salisb.,
Panax notoginseng (Burkill) F.H.Chen

64
Preparation Name Compositions Traditional uses Ref.
Tiedi oral liquid Ophiopogon japonicus (Thunb.) Ker Gawl., Scrophularia Promoting fluid production and quenching thirst, “Chinese Pharmacopoeia”, vol. 1.
ningpoensis Hemsl., Trichosanthes kirilowii Maxim., curing sore throat caused by yin deficiency and lung
Terminalia chebula Retz., Canarium album (Lour.) DC., Gallus gallus heat
domesticus Brisson, Platycodon grandiflorus (Jacq.) A.DC.,
Fritillaria thunbergii Miq., Poria cocos(Schw.)Wolf,
Glycyrrhiza uralensis Fisch.
Yixinshu capsule Panax ginseng C.A.Mey., Ophiopogon japonicus (Thunb.) Ker Gawl., Nourishing yin, promoting blood circulation to dispel “Chinese Pharmacopoeia”, vol. 1
Schisandra chinensis (Turcz.) Baill., Astragalus mongholicus Bunge, blood stasis, curing thoracic obstruction
Salvia miltiorrhiza Bunge, Ligusticum striatum DC.,
Crataegus pinnatifida Bunge
Zixinyin oral Ophiopogon japonicus (Thunb.) Ker Gawl., Paeonia lactiflora Pall., Nourishing heart yin, promoting blood circulation and “Chinese Pharmacopoeia”, vol. 1.
liquid Glehnia littoralis F.Schmidt ex Miq., Panax notoginseng (Burkill) F.H.Chen relieving pain in thorax
Qinghuo Zhimai Andrographis paniculata (Burm.f.) Nees, Gardenia jasminoides J.Ellis, Clearing away heat and toxic materials, cooling the “Chinese Pharmacopoeia”, vol. 1.
tablet Ophiopogon japonicus (Thunb.) Ker Gawl. blood and apocatastasis, clearing away heat in lung
and stomach, curing toothache, redeyes and sore throat

65
Table 7. Summary of pharmacological activities of the extracts/compounds from Ophiopogon japonicus
Activity Extract/Compoun Model Effect Dosage Reference
d
cardioprotection ROJ-ext Rats ↓ the dried weight of thrombus (36.0% and 70.6%) and 12.5 and 25 mg/kg (Kou et al., 2005)
endothelium injury
HL-60 and ECV304 cells ↓ cells adhesion 0.1, 1.0 and 10 μg/ml
ROJ-ext Male ICR mice ↓ tail thrombosis in mice 12.5 and 25 mg/kg (Kou et al., 2006)
Male Sprague–Dawley ↓ arterio-venous shunt-induced thrombus in rats 6.25 and 12.5 mg/kg
rats ↓ platelet aggregation in rats 12.5 and 0.7 mg/kg
saponins Rats ↓ arrhythmias 10 mg/kg (Chen et al., 1990)
Dogs ↓ ventricular arrhythmia
DT-13 Adult rat ventricular ↓ L-type Ca2+ currents (15.4±2.1%, 26.7±4.9%, 0.001, 0.01, 0.1 and 1 μM (Tao et al., 2005)
myocytes 38.3±8.2% and 61.7±7.4%);
↑ current voltage curve
ophiopogonin D HMMEC-1 cells ↑ tube formation, tube length and tube branching points 10 and100 μg/ml (Lan et al., 2013)
and compound 45
(saponin)
ophiopogonin J FAS inhibition assay ↓ animal FAS with IC50 of 76 ±2 μM (Duan et al., 2012)
RUS Male C57BL/6 mice ↓ infarct size (37.4% and 63% ); brain water content; 5 and 10 mg/kg (Guan et al., 2013)
p65 expression (46.1%) and phosphorylation by (26.6%)
(10 mg/kg); NF-κB activation/translocation via inhibiting
p65 nuclear translocation and NF-κB p65 subnit DNA
binding capacity; mRNA levels of IL-1β, TNF-α, ICAM-1,
COX-2 and IL-6; protein levels of iNOS (by 34.0% and
49.4%), ICAM-1 (by 43.3% and 52.2%), COX-2 (by
43.4% and 58.7%); ↑ neurological performances
ophiopogonin D H9C2 cells ↓ LC-3-II/LC3-I ratio; p62 expression; DOX-induced 1 μM (Zhang et al., 2015)
autophagic damage; activation of JNK and ERK
C57BL/6J mice ↓ DOX-induced cardiac dysfunction in mice 10 mg/kg
MDG-1 HMEC-1 cells ↓ cell death; ↑ cell migration and tube formation 0.4, 1.2, 4 and 10 mM (Wang et al., 2010)
↑ SPHK1 expression, S1P1 expression, Akt, ERK, eNOS

66
phosphorylation, NO production and bFGF expression
rats ↓ average infarct size and cells damage induced by acute 3, 9 and 30 mg/kg
myocardial ischemia
↑ neovascularization in chemic myocardium
FOJ-5 Langendorff model ↑ heart contraction and coronary blood flux 1-100 μg/ml (Zheng et al., 2009)
rats ↓ heart rate caused by ischemia 20, 40 mg/kg
↓ LDH
Anti-inflammation ROJ-ext Mice and rats ↓ ear swelling, paw edema, pleural leukocyte migrations, 25 and 50 mg/kg (Kou et al., 2005b)
peritoneal total leukocyte and neutrophil migration;
HL-60 and ECV304 cells ↓ adhesion of HL-60 cells to ECV304 cells, with IC50 of
42.85μg/ml.
RUS LPS-induced mice ↓ lung wet to dry weight ratio (72.65%), MPO activity 0.3, 1.0 and 3.0 mg/kg (Sun et al., 2012)
(43.12%, 69.10% and 92.48%) and nitrate/nitrite content
(44.47%, 58.97% and 64.57%)
↓ TF, iNOS and NF-κB p-p65 expression in the lung
tissue.
RUS Human umbilical vein ↓ICAM-1 expression (53.7% ), p65 phosphorylation (40% 1 μM (Song et al., 2010)
endothelial cells and 70% at 5,15 min), IKKα phosphorylation (48.0% and
58.9% at 5,15 min), IKKβ phosphorylation ( 67.8% and
91.1% at 5,15 min)
RUS Diabetic rats ↓ macrophage influx (43.4 ± 3.9%); 3.0 mg/kg/day for 8 weeks (Lu et al., 2014)
↓ TNF-α, IL-6 and IL-1β expression
RUS Pulmonary arterial ↓ RVSP, mPAP values; pulmonary arterial wall thickness; 0.1, 0.4, and 0.7 mg/kg (Bi et al., 2013)
hypertension rats expression of eNOS, CD31, and caveolin-1 in the lungs;
leukocyte invasion into lung tissue and endothelial cell
apoptosis
↑ NF-κB phosphorylation;
Ophiopogonin C ECV304 cells ↓ adhesion of HL-60 to ECV304 cells stimulated by 0.01. 0.1 and 1μM (Tian et al., 2011)
TNF-α (IC50 of 19.9 nM) or PMA (IC50 of 4.35 nM)
Ophiopogonanone bronchial epithelial ↓ IL-4-induced eotaxin production and eotaxin expression 25.0 μM (Hung et al., 2010)
G BEAS-2B cells

67
ophiopogoside A
ophiopogoside B
ophiopogonanone murine microglial BV-2 ↓ NO production with IC50 of 20.1, 7.8, 19.2 and 14.4 μM, -- (Li et al., 2012a)
H; cells respectively
methylophiopogona
none B;
methylophiopogono
ne A;
ophiopogonanone E
Anti-oxidation Saponins DPPH scavenging model; ↓ DPPH radical with IC50 of 1.46 mg/ml and hydroxyl 2 and 5 mg/ml (Xiong et al., 2012)
Hydroxyl radical radical
scavenging model; ↑ macrophage activity 100, 200, 400 μg/ml
cells ↑ NO production and IL-1 production
Ophiopogonin D HUVECs ↓ H2O2-induced oxidative stress, apoptosis and ERK1/2 0.6 to 60.0 μM (Qian et al., 2010)
activation
Ophiopogonin D MC3T3-E1 cells and ↓H 2O2-induced MC3T3-E1 dysfunction 1, 10, 100 μM (Huang et al., 2015)
RAW264.7 cells; ↓induced MC3T3-E
BALB/c female mice ↓induced MC3T3-E1 dysfunctionellsnRAW264.7 cells
↓Aosteoclastic markers in mice 5 and 25 mg/kg
Nolinospiroside F K6001 yeast cells ↑ the yeast replicative lifespan 1, 3 and 10 μM (Sun et al., 2013)

compound 109 Free radical scavenging ↓ superoxide anion radical with IC50 of 4.52 μM, H2O2 -- (Zhou et al., 2008a)
(homoisoflavonoid) model radical IC50 of 0.05μM and hydroxyl radical 0.58 μM.
OJP-1, OJP-2, Macrophages from male ↓ DPPH radical with IC50 of 145.26, 34.21, 11.12, 7.52 100-400 μg/ml (Xiong et al., 2011)
OJP-3 and OJP-4 Kunming strain mice mg/ml and hydroxyl radical with IC50 of 0.04, 8.65, 5.89,
2.07 mg/ml
↑ macrophage activation
POJ-U1a Free radical scavenging ↓ DPPH radical by 35.08%, hydroxyl radical by 32.50% 8 mg/ml (Wang et al., 2012b)
model and superoxide anion by 45.01%
Phenolic Free radical scavenging ↓ DPPH radical and ABTS radical 12.5–100 μg/mL (Li et al., 2012c)
compounds model

68
Anticancer DT-13 Breast cancer ↓ tumor cell adhesion (43.5%, 60.8% ) and migration 1 and 10 μM (Sun et al., 2010)
MDA-MB-435 cells (20%, 30% )
↓ expression of tumor αvβ3 integrin, TF (40.8%, 87.9%)
and early growth response gene-1 (50%, 62%)
decreased excretion of MMP-9
DT-13 Breast cancer ↓ tumor cell adhesion(54%, 72%) and invasion(57%, 10 and 30 μM (Zhang et al., 2012b)
MDA-MB-435 cells 64%); expression of MMP-2/9 and p38 kinase
DT-13 Breast cancer ↓expression of VEGF, CCR5 and factor 1α 0.01 to 1 μM (Zhao et al., 2014)
MDA-MB-435 cells
Ophiopogonin B Human non-small cell; ↓ p-Akt both at Ser308 and Thr473 ,induced autophagy, (Chen et al., 2013b)
lung cancer cells with IC50 of 2.86 μM for H157 cells and IC50 of 4.61 μM
for H460 cells
↓ P13K/Akt/mTOR/p70S6K pathway in NCI-H460 cells
Ophiopogon human breast cancer ↓ MCF-7 cell growth and induce MCF-7 cell apoptosis via -- (Liu et al., 2009)
japonicus lectin MCF-7 cells caspase pathway, with IC50 of 22 μg/ml
DT-13 HUVECs ↓ VEGF secretion under normoxia (11, 15% and 40%) and 0.01, 0.1, 1μM (Zhao et al., 2013)
under hypoxia (37, 42 and 38%).
↓ VEGF-induced tube migration (61.5,40.1 and 38.9%)
and tube formation (63.4, 49.3 and 35%)
↓ angiogenesis by inhibiting the VEGF pathway,
↓ p-VEGFR2 and p-Akt level under hypoxia
Anti-diabetes polysaccharides NIT-1 cells; ↓ the glucose uptake into BBMV by 50% and the activity 0.06-240 mg/ml (Ding et al., 2012)
Rat hepatoma H4IIE cell; of α-glucosidase by 70%;
3T3-L1 mouse ↑ NIT-1 cells damaged by STZ;
adipocytes;
BBMV assay model
polysaccharides Gestational diabetes ↓ blood glucose,serum insulin 125, 250, 500 mg/kg (Wang, 2013)
mellitus rat ↑ adiponectin level
↑ APN mRNA level
OJP1 Sprague–Dawley rats ↓ blood glucose level (32.9%, 150 mg/kg) and insulin 150 and 300mg/kg (Chen et al., 2011)
level compared with the diabetic control (P<0.05)
↑ the insulin antigenesity of diabetic islet of β-cells.
69
OJP1 Diabetic rats ↓ blood glucose, MDA, serum TG, TC and LDL-C 150 or 300 mg/kg (Chen et al., 2013c)
Increased SOD and GPx activity in diabetic rats
↓ liver index, Scrs, serum UA and UREA levels, serum
ALT and AST activities, SCr levels
↓ expression of mRNA for TGF-β1 and CTGF
MDG-1 ob/ob mice ↓ fed blood glucose levels (13.5%, 23.7%) and the fasting 150 and 300 mg/kg (Xu et al., 2011)
blood glucose levels (8.8%, 16.3%)
↓ the liver triglyceride content of mice, the body weight
gain (15.1%) and subcutaneous fat
MDG-1 Female KKAy and ↓ the blood glucose, insulin levels and plasma lipids (total 75 and 300 mg/kg (Wang et al., 2012a)
C57BL/6J mice cholesterol, triglyceride, LDL-C and HDL-C)
↑thymus and spleens indices; IRS-1/PI3-K association;
Akt phosphorylation and Glut 4 protein
↓ the hepatocyte hypertrophy and lipid droplet
accumulation
MDG-1 Obese C57BL/6 mice ↑ weight loss and reduce adipose tissue mass (50%) 300 mg/kg (Wang et al., 2014)
↑ oxygen consumption and energy expenditure
ameliorated plasma lipid profiles,
↓ leptin secretion, attenuate hepatic lipid accumulation
increased the expressions of genes related to lipid and
energy metabolism
MDG-1 Female KKay mice ↓ intestinal glucose absorption, enhanced liver 300 mg/kg (Zhu et al., 2014)
glycogenesis,
↓ glycogenolysis,
↑ GLP-1 secretion
MDG-1 Obesity (DIO) mouse ↓ the body weight gain, fed blood glucose, oral glucose 300 mg/kg (Li et al., 2014)
tolerance test and insulin
MDG-1 Obese C57BL/6 mice Regulated gut microbiota to normal state 300 mg/kg (Shi et al., 2015)
↑ the proliferation of intestinal probiotics
↓ the level of D-galactosamine
increased the level of taurine, SCFAs

70
Oligosaccharides Type 2 diabetic rats ↑ body weight and decreased organ related weights of liver 225 and 450 mg/kg (Li et al., 2012b)
and kidney;
↓ fasting blood glucose level and improved oral glucose
tolerance;
↓ urinary protein levels;
↑ hepatic glycogen content;
↑ GCK activity and decreased PEPCK activity;
↑ serum active GLP-1 level and reduced glucagon level
Antitussive ophiopogonin-D paratracheal neurones hyperpolarized the paratracheal neurones from a resting 10 μM (Ishuibashi et al., 2001)
membrane potential of -65.7 to -73.5 mV;
↑ K+ channels
Antimicrobial OJL Vero cells ↓ herpes simplex virus type II(40.9%, 52.3% and 85.4%) 2, 4 and 8 μg/mL (Tian et al., 2008)
with EC50 of 3.93 μg/mL
↓ the growth of fungi Gibberella saubinetii and 0.06 and 0.05 mg/ml
Rhizoctonia solani
Immunomodulation Saponins DPPH and hydroxyl ↓ DPPH radical by 99.64% (IC50 of 1.46 mg/ml) and 5 mg/ml (Xiong et al., 2012)
radical scavenging model hydroxyl radical
peritoneal macrophage ↑ phagocytic capacity, macrophage viability, NO 2 mg/ml
cells production(200.71%,335.81% and 538.07%) and IL-1 100, 200, 400 μg/mL
release
DT-13 ICR mice ↓ ALT level, hepatocelluar necrosis and adipose 10 or 20 mg/kg (Wu et al., 2000)
RUS degeneration
hepatocytes and spleen ↓ the release of ALT in nonparenchymal cells with IC50 of
cells 6.3× 10-10 M and 3.9 × 10-7 M
↓lympho proliferation in spleen cells 10-5-10-4 M
polysaccharides C57BL/6 mice ↑ body weight, water intake and salivary flow 0.05 and 0.1 g/kg (p.o.) (Wang et al., 2007)
↓ SMG, spleen index, lymphocytic infiltration, the level of
IFN-γ the IFN-γ/IL-4 ratio

71
OJP-1, OJP-2, DPPH radical scavenging ↓ DPPH radical with IC50 of 145.26, 34.21, 11.12, 7.52 (Xiong et al., 2011)
OJP-3 and OJP-4 model; mg/ml
hydroxyl radical ↓ hydroxyl radical with IC50 of 8.65, 5.89. 2.07, 0.04
scavenging model; mg/ml
macrophages cells ↑ macrophage and its energy metabolism rate 100-400 μg/ml
↑ NO release (17.78%, 84.92, 141.93% and 190.43%) and
IL-1 production
OPL kupffer cells ↑ A490 values 7.813-125 μg/ml (Fan et al., 2014)
↑ NO, iNOS production, IL-6, IL-12 contents
↑ CD80 and CD86 expression
OPL One-day-old White ↑ A490 values 7.813-125 μg/ml (Fan et al., 2015)
Roman chickens (male) ↑ IL-2, IL-6 secretion and phagocytic index
↑ lymphocytes proliferation
↑CD4+ T and CD8+ T cells and enhanced the humoral and
cellular immune function

72
73
74
75
*Graphical Abstract

Graphical abstract

Phytochemistry Pharmacology

Steroidal saponins Ophiopogon japonicus Cardiovascular protection


Homoisoflavonoids Anti-inflammation
Polysaccharides Anticancer
Anti-oxidation
Immunomodulation
Antitussive activity
Antimicrobial
Anti-diabetes

You might also like