Polygalae Radix A Review of Its Traditional Uses

Download as pdf or txt
Download as pdf or txt
You are on page 1of 46

Journal Pre-proof

Polygalae Radix: A review of its traditional uses, phytochemistry,


pharmacology, toxicology, and pharmacokinetics

Xin Zhao, Yueli Cui, Peng Wu, Pan Zhao, Qiao Zhou, Zhihui
Zhang, Yue Wang, Xuelan Zhang

PII: S0367-326X(20)30341-5
DOI: https://doi.org/10.1016/j.fitote.2020.104759
Reference: FITOTE 104759

To appear in: Fitoterapia

Received date: 4 August 2020


Revised date: 7 October 2020
Accepted date: 12 October 2020

Please cite this article as: X. Zhao, Y. Cui, P. Wu, et al., Polygalae Radix: A review
of its traditional uses, phytochemistry, pharmacology, toxicology, and pharmacokinetics,
Fitoterapia (2019), https://doi.org/10.1016/j.fitote.2020.104759

This is a PDF file of an article that has undergone enhancements after acceptance, such
as the addition of a cover page and metadata, and formatting for readability, but it is
not yet the definitive version of record. This version will undergo additional copyediting,
typesetting and review before it is published in its final form, but we are providing this
version to give early visibility of the article. 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.

© 2019 Published by Elsevier.


Journal Pre-proof

Review

Polygalae Radix: A review of its traditional uses,


phytochemistry, pharmacology, toxicology, and
pharmacokinetics
Xin Zhaoa,1 , Yueli Cuia,1, Peng Wua,1 , Pan Zhaoa, Qiao Zhoua, Zhihui Zhanga, Yue Wanga, Xuelan
Zhanga,b,* zhang8832440@sina.com
a School of Chinese Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road, Jinan
250355, Shandong China
b Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole
Industrial Chain of Traditional Chinese Medicine, 4655 Daxue Road, Jinan 250355, Shandong China

of
* Corresponding author at: Shandong University of Traditional Chinese Medicine, 4655 Daxue Road,
Changqing District, Jinan, Shandong Post Code: 250355, China.

ro
Abstract: Polygalae Radix (Polygalaceae), the dried root of Polygala tenuifolia Willd. and Polygala
sibirica L., has been widely used as a medicine for improving cognitive function. In China,
-p
Polygalae Radix has been widely used in the treatment of insomnia, forgetfulness, depression,
cough, palpitation, and other diseases. More than 140 compounds have been isolated from
re
Polygalae Radix, including saponins, xanthones, oligosaccharide esters, and so on. The compounds
and extracts isolated from Polygalae Radix possess wide-ranging pharmacological activities, such
as neuroprotective, antidepressant, hypnotic-sedative, anti-inflammatory, antiviral, antitumor,
lP

antioxidant, antiaging, and antiarrhythmic effects, among others. The clinical practice of traditional
Chinese medicine has proved that raw Polygalae Radix can irritate the throat. Modern studies have
found that raw Polygalae Radix exhibits a certain degree of toxicity to the gastrointestinal tract
na

after long-term use or excessive doses and that its main toxic components are saponins. Thus,
Polygalae Radix is usually processed, and/or combined with other herbs to reduce gastrointestinal
irritation. This review investigated the pharmacokinetics of Polygalae Radix. Future research
perspectives and the existing problems of Polygalae Radix were also discussed. This review can
ur

broaden the understanding regarding Polygalae Radix and provide references for further research.
Jo

Keywords: Polygalae Radix; Traditional uses; Phytochemistry; Pharmacology; Toxicology;


Pharmacokinetics

Abbreviations: PR, Polygalae Radix; AD, Alzheimer’s disease; TCM, traditional Chinese medicine; PD, Parkinson’s disease;

MAP2, microtubule-associated protein 2; Gap-43, growth-associated protein 43; TNF-α, tumor necrosis factor-α; IL-1β,

interleukin-1β; IL-6, interleukin-6; NF-κB, nuclear factor-kappa B; ROCK2, Rhokinase 2; NGF, nerve growth factor; BDNF,

brain-derived neurotrophic factor; tPA, tissue plasminogen activator; cAMP, cyclic adenosine monophosphate; ERK,

extracellular signal-regulated kinase; PI3K, phosphatidylinositol-3-kinase; NOS, nitric oxide synthase; CREB, cyclic

responsive element-binding; CRTC1, CREB-regulated transcription co-activators; mTOR, mammalian target of rapamycin;

CUMS, chronic unpredictable mild stress; NT-3, neurotrophin-3; NLPR3, pyrin domain-containing 3; 5-HT,

5-hydroxytryptamine; IDO, indoleamine 2,3-dioxygenase; AChE, acetylcholinesterase; ChAT, acetyl-transferase; MAPK,

mitogen-activated protein kinase; NA, noradrenaline; HPA, hypothalamus-pituitary-adrenal; MDA, malondialdehyde; GSH,

glutathione; SOD, superoxide dismutase; TrkB, tyrosine kinase B; nAChRα7, nicotinic acetylcholine receptor subunit

1These authors contributed equally to this work and should be considered co-first authors.
Journal Pre-proof

alpha-7; NMDA, N-methyl-D-aspartate; MES, maximal electroshock; PTZ, pentylenetetrazol; ICR, institute of cancer

research; γ-GABA, gamma-aminobutyric acid; Ach, acetylcholine; NO, nitric oxide; iNOS, inducible nitric oxide synthase;

PGE2, prostaglandin E2; COX-2, cyclooxygenase-2; MMPs, matrix metalloproteinase; Nrf2/HO-1, nuclear factor erythroid

2-related factor 2/heme oxygenase; D-GalN, D-galactosamine; GCRV, grass carp reovirus; MyD88, myeloid differentiation

factor 88; Mx1, myxovirus; OVCAR-3, ovarian carcinoma cells-3; EGFR, epidermal growth factor receptor; VEGF, vascular

endothelial growth factor; ROS, reactive oxygen species; RNS, reactive nitrogen free radicals; YZ-OE, YZ oligosaccharide

esters; GSH-PX, glutathione peroxidase; CAT, catalase; LVEDP, left ventricular end-diastolic blood pressure; CPK, creatine

phosphokinase; LDH, lactate dehydrogenase; HMGB1, high-mobility group box protein 1; LVSP, left ventricular systolic

blood pressure; IC50, half maximal inhibitory concentration; LD50, median lethal dose; ICC, interstitial cells of Cajal; NOAEL,

no-observed-adverse-effect level; AUC0–t, the area under the plasma concentration-time curve from zero to last sampling

time; T1/2, elimination half-life; Cmax, peak plasma concentration; LC-MS/MS, liquid chromatography-tandem mass

spectrometry; i.v., intravenous.

of
1. Introduction

ro
Polygalae Radix (PR) (Figure 1) is a Polygala plant, which belongs to the family Polygalaceae
and is mainly distributed in China, South Korea, and the Russian Federation [1]. PR is known as
-p
“Yuanzhi” in Chinese and has been widely used for more than 2000 years as a medicine for
boosting intellectual development. Since 1963, PR has been officially listed in the Chinese
re
Pharmacopoeia. PR is also referred to as “Onji” in Japanese. In Japan, PR is an important medicinal
plant usually included in Kampo prescriptions and is used as an antineurotic, tonic, and sedative
lP

agent [2]. The medicinal use of PR was first described in Shen Nong’s herbal classic, the earliest
pharmaceutical monograph in China [3]. More than 870 Chinese medical formulas contain PR,
among which Kai Xin powder, Di Huang Yin Zi, and Gu Yin Jian are listed in a catalog of ancient
na

classic prescriptions (the first batch) in China. These formulas are commonly used to treat
depression, anxiety, insomnia, Alzheimer’s disease (AD), irregular menstruation, and premature
ur

ovarian aging [4–6]. With the development of methodological approaches, more than 140
compounds have been isolated and identified from PR, including saponins, oligosaccharide esters,
Jo

xanthones, organic acids, and others [7,8]. Pharmacological studies have reported on the
neuroprotective, memory-enhancing, antidepressant, anti-inflammatory, antiviral, and antioxidant
effects of PR [9–12]. The saponins of PR are currently related to gastrointestinal toxicity, resulting in
flatulence, gastrointestinal congestion, and aphasia of animals [13]. After alkaline hydrolysis, PR
can reduce toxicity and maintain cognitive-enhancing efficacy [14]. This toxicity of PR limits its
application and development of new products. Regardless, the quality, efficacy or toxicity of PR can
be altered by processing or compatibility. In addition, the bioavailability of some oligosaccharide
esters and saponins in PR in vivo is low, whereas that of organic acids are high [15,16].
Journal Pre-proof

of
Figure 1. Photograph of PR. (A) Overground part from P. tenuifolia Willd.; (B) The root of P. tenuifolia Willd.;
(C) Commercial herbal pieces of raw PR; (D) Commercial herbal pieces of licorice-boiled PR.

ro
Considerable efforts have been directed toward exploring the phytochemistry and potential
pharmacological activities of PR, and numerous scientific studies have been conducted on the
-p
subject. Meanwhile, the toxicology and pharmacokinetics of PR have rarely been examined. In this
review, we systematically summarize the traditional uses, phytochemistry and pharmacology
re
aspects of PR. On the basis of limited research reports, the toxicity and pharmacokinetics of PR
were summarized and analyzed for the first time. More importantly, we conducted an in-depth
lP

analysis of the existing problems in the current study of PR and discussed the direction of future
research, which was not summarized in the previous review of PR. The review intends to enable
scholars to have a more comprehensive understanding of PR, and to accelerate the exploration and
na

discovery of its pharmacological mechanism, so as to reveal the scientific connotation of its


traditional application better. Meanwhile, it also provides certain scientific basis for the
development and utilization of medicinal value of PR.
ur

2. Traditional uses
Jo

As a traditional medicinal plant, PR has a long history in China because of its widely biological
and pharmacological activities. The initial description of PR is recorded in Shen Nong’s herbal
classic [3], which was written in the Eastern Han Dynasty. Subsequently, in the Compendium of
Materia Medica, which is another outstanding traditional Chinese medicine (TCM) monograph, this
plant was recorded as an effective herbal medicine that can promote intellectual development [17].
In addition, the record about root of P. tenuifolia Willd. and P. sibirica L. as the medicinal part can be
traced back to the Northern and Southern dynasties. Since the Ming dynasty, only the roots have
been used for medicinal purposes. In Chinese clinical practices, raw PR exhibits side effects, such as
gastrointestinal toxicity and throat irritation, and requires processing before being prescribed. The
evolution of the processing methods of PR has changed from simple to complex, and the auxiliary
materials are also diversified. However, now the common processing methods of PR include
discarding the heartwood, licorice boiling, and honey stir-baking. According to the theories of
TCM, processing can reduce the toxicity and enhance the efficacy of PR [18,19]. The main
processing techniques and purposes of PR recorded in some Chinese medicine monographs are
listed in Table 1.
Journal Pre-proof

Table 1
Different processing methods of PR.
Title of books Dynasty Processing method Purpose of processing

Hua Shi Zhong Cang Before the Tang Going to the heart Removing non-medicinal parts and reducing

Jing dynasty side effects to ensure efficacy

Lei Gong Pao Zhi Lun Before the Tang Stir-boiling with licorice Reducing dryness and eliminating throat

dynasty decoction irritation

Pu Ji Ben Shi Fang Song dynasty Stir-baking to yellow Reducing the toxicity and moderating the

effect

Pu Ji Ben Shi Fang Song dynasty Stir-baking with ginger Reducing gastrointestinal toxicity and

enhancing expectorant effect

Tai Ping Hui Min He Ji Ju Song dynasty Stir-frying with wine Tonifying the heart and kidney and treating

of
Fang anxiety

Tai Ping Hui Min He Ji Ju Song dynasty Steaming with wine Tonifying the heart and kidney and calming

ro
Fang the nerves

Pu Ji Fang Ming dynasty Stir-frying with wheat


-p Enhancing the effect of calming the nerves

Jing Yue Quan Shu Ming dynasty Steaming with licorice, Reducing toxicity and enhancing the function

black beans and ginger of relieving cough and eliminating phlegm


re
Zeng Bu Wan Bing Hui Ming dynasty Stir-baking with bile and Reducing gastrointestinal toxicity and

Chun ginger enhancing expectorant effect

Jing Yue Quan Shu Ming dynasty Stir-boiling with black Reducing toxicity and alleviating drug
lP

bean and licorice resistance, and enhancing the function of

calming nerves
na

Lei Zheng Zhi Cai Qing dynasty Carbonizing by Producing hemostatic effect

stir-frying

De Pei Ben Cao Qing dynasty Immersing into rice Strengthening the role of nourishing the
ur

water and licorice spleen and replenishing qi

decoction
Jo

Zhong Yao Pao Zhi Pin Qing dynasty Stir-baking with honey Relieving drug resistance and enhancing

Gu Jin Yan Bian Ping Shu antitussive and expectorant effect

Verification of the medicinal value of PR bears significance. In this review, some representative
classic Chinese herbal prescriptions containing PR are described in Table 2. Among them, 14
prescriptions using processed products of PR. Moreover, PR is often combined with Acori
Tatarinowii Rhizoma, Glycyrrhizae Radix et Rhizoma, Poria, Semen Ziziphi Spinosae, and Ginseng
Radix et Rhizoma to reduce gastrointestinal toxicity and enhance efficacy [20]. These prescriptions
are commonly used to treat spermatorrhea, cough, and sore, as well as numerous neurological
diseases, such as insomnia, forgetfulness, neurasthenia, depression, seizure, and AD [21–23]. In
addition, 79 prescriptions for the treatment of palpitation with insomnia are listed in Zhong Yao
Cheng Fang Zhi Ji, among which more than 50% use PR. Gui Pi soup, Ren Shen Yang Rong soup,
and Di Huang Yin Zi are the representative prescriptions. These prescriptions are based on
tranquilizing drugs, with Qi, blood, and Yin tonics as the core to treat palpitation with insomnia
arising from various symptoms [24].
Journal Pre-proof

Table 2
Examples of traditional and modern Chinese medicine prescriptions containing PR in China.
Preparation name Compositions Traditional and clinical uses Ref.

Ding Xian Pills Polygalae Radix (processed), Castrodiae Rhizoma, Fritillariae Cirrhosae Bulbus, Arisaema Cum Bile, Opening the orifices, calming the nerves, Yi Xue Xin Yu

Pinelliae Rhizoma, Fu Shen, Acori Tatarinowii Rhizoma, Scorpio, Bombyx Batryticatus, Citri and treating epilepsy

f
Reticulatae Pericarpium, Radix Rubra, Ophiopogonis Radix, Glycyrrhizae Radix et Rhizoma,

o
Cinnabaris, Amber

ro
Ding Zhi Pills Polygalae Radix, Acori Tatarinowii Rhizoma, Ginseng Radix et Rhizoma, Poria, Glycyrrhizae Radix Invigorating the spleen and calming the Shen Shi Yao Han

et Rhizoma, Cinnabaris nerves, and treating myopia and

- p forgetfulness

e
Ren Shen Jian Pi Pills Polygalae Radix (processed), Ginseng Radix et Rhizoma, Atractylodis Macrocephalae Rhizoma, Treating dyspepsia, nausea and vomiting, Jing Yue Quan Shu

Astragali Radix, Angelicae Sinensis Radix, Semen Ziziphi Spinosae

P r
Poria, Dioscoreae Rhizoma, Citri Reticulatae Pericarpium, Aucklandiae Radix, Amomi Fructus, abdominal pain, loose stools, weak health

and tiredness caused by weakness of the

Tian Wang Bu Xin Pills

a l
Polygalae Radix (processed), Angelicae Sinensis Radix, Acori Tatarinowii Rhizoma, Codonopsis
spleen and stomach

Treating insomnia, palpitation, wet dream, Xiao Zhu Fu Ren

r n
Radix, Poria, Schisandrae Chinensis Fructus, Ophiopogonis Radix, Asparagi Radix, Rehmanniae forgetfulness Fang

u
Radix, Scrophulariae Radix, Semen Ziziphi Spinosae, Semen Platycladi, Platycodonis Radix,

o
Glycyrrhizae Radix et Rhizoma, Cinnabaris

Gui Pi Soup

J
Polygalae Radix (processed), Codonopsis Radix, Atractylodis Macrocephalae Rhizoma, Astragali

Radix, Glycyrrhizae Radix et Rhizoma, Poria, Semen Ziziphi Spinosae, Longan Arilius, Angelicae

Sinensis Radix, Aucklandiae Radix, Jujubae Fructus


Treating gastric and duodenal ulcer

bleeding, functional uterine bleeding,

aplastic anemia, thrombocytopenic


Zheng Ti Lei Yao

purpura, neurasthenia, heart disease

Ren Shen Yang Rong Soup Polygalae Radix (processed), Ginseng Radix et Rhizoma, Poria, Angelicae Sinensis Radix, Paeoniae Treating tiredness, weakness, palpitation, San Yin Ji Yi Bing

Radix Alba, Citri Reticulatae Pericarpium, Cinnamomi Cortex, Atractylodis Macrocephalae forgetfulness, insomnia, cough and sore Fang Lun

Rhizoma, Glycyrrhizae Radix et Rhizoma, Rehmanniae Radix, Astragali Radix, Schisandrae

Chinensis Fructus

Yang Xin Soup Polygalae Radix (processed), Astragali Radix, Poria, Fu Shen, Angelicae Sinensis Radix, Chuanxiong Nourishing Qi and blood to treat Zheng Zhi Zhun
Journal Pre-proof

Preparation name Compositions Traditional and clinical uses Ref.

Rhizoma, Glycyrrhizae Radix et Rhizoma, Pinellia Ternata, Semen Platycladi, Semen Ziziphi restlessness and palpitation Sheng

Spinosae, Schisandrae Chinensis Fructus, Ginseng Radix et Rhizoma, Cinnamomi Cortex

Kai Xin Powder Polygalae Radix, Ginseng Radix et Rhizoma, Poria, Acori Tatarinowii Rhizoma Curing amnesia Qian Jin

Sang Piao Xiao Powder Polygalae Radix (processed), Mantidis Ootheca, Acori Tatarinowii Rhizoma, Ginseng Radix et Tonifying heart and kidney, astringing Ben Cao Yan Yi

f
Rhizoma, Fu Shen, Angelicae Sinensis Radix, Dragonsbones, Testudinis Carapax et Plastrum spermatorrhea

o
Yuan Zhi Powder Polygalae Radix, Atractylodis Macrocephalae Rhizoma, Cinnamomi Cortex, Ginseng Radix et Curing heart deficiency and strain, Tai Ping Sheng Hui

ro
Rhizoma, Trionycis Carapax, Asparagi Radix, Eucommiae Cortex, Pericarpium Zanthoxyli, weakness of limbs, mind and spirit Fang

Achyranthis Bidentatae Radix, Poria, Dioscoreae Rhizoma, Corni Fructus, Semen Platycladi, lethargy

Rehmanniae Radix, Dendrobii Caulis, Astragali Radix, Glycyrrhizae Radix et Rhizoma

- p
e
Qi Fu Drink Polygalae Radix (processed), Rehmanniae Radix, Angelicae Sinensis Radix, Ginseng Radix et Treating deficiency of Qi and blood Jing Yue Quan Shu

Spinosae

P r
Rhizoma, Atractylodis Macrocephalae Rhizoma, Glycyrrhizae Radix et Rhizoma, Semen Ziziphi

Bu Shen Yi Nao Pills

a l
Polygalae Radix (processed), Cervi Cornu Pantotrichum, Ginseng Radix et Rhizoma Rubra, Poria,

Dioscoreae Rhizoma, Rehmanniae Radix, Angelicae Sinensis Radix, Chuanxiong Rhizoma, Psoraleae
Treating palpitation, shortness of breath,

insomnia, forgetfulness, spermatorrhea,


“Chinese

Pharmacopoeia

r n
Fructus, Achyranthis Bidentatae Radix, Lycii Fructus, Scrophulariae Radix, Ophiopogonis Radix, aching waist and legs, tinnitus and (2015)”

u
Schisandrae Chinensis Fructus, Semen Ziziphi Spinosae, Glycyrrhizae Radix et Rhizoma, Cinnabaris deafness caused by deficiency of kidney

o
and deficiency of Qi and blood

Bai Zi Yang Xin Pills

J
Polygalae Radix (processed), Semen Platycladi, Codonopsis Radix, Astragali Radix, Chuanxiong

Rhizoma, Angelicae Sinensis Radix, Poria, Semen Ziziphi Spinosae, Cinnamomi Cortex, Schisandrae

Chinensis Fructus, Pinellia Ternata, Glycyrrhizae Radix et Rhizoma, Cinnabaris


Treating palpitation, insomnia, dreaminess,

forgetfulness
“Chinese

Pharmacopoeia

(2015)”

Jian Nao Pills Polygalae Radix (processed), Dioscoreae Rhizoma, Amber, Schisandrae Chinensis Fructus, Curing hypomnesia, dizziness, palpitation, “Chinese

Castrodiae Rhizoma, Semen Platycladi, Radix Rubra, Ginseng Radix et Rhizoma, Alpiniae insomnia, waist and knee weakness caused Pharmacopoeia

Oxyphyllae Fructus, Chrysanthemi Flos, Irkutsk Anemone Rhizome, Haematitum, Arisaema Cum by deficiency of heart and kidney (2015)”

Bile, Semen Ziziphi Spinosae, Lycii Fructus

Qiang Yang Bao Shen Pills Polygalae Radix (processed), Epimedii Folium, Cistanches Herba, Psoraleae Fructus, Semen Treating waist acid, mental fatigue, and “Chinese

Astragali Complanati, Rubi Fructus, Semen Euryales, Foeniculi Fructus impotence spermatorrhea caused by yang Pharmacopoeia
Journal Pre-proof

Preparation name Compositions Traditional and clinical uses Ref.

deficiency of kidney (2015)”

Xie Gan An Shen Pills Polygalae Radix (processed), Gentianae Radix et Rhizoma, Scutellariae Radix, Cardeniae Fructus, Curing insomnia, dreaminess and upset “Chinese

Margaritifera Concha, Ostreae Concha, Dragonsbones, Semen Platycladi, Semen Ziziphi Spinosae, caused by hyperactivity of liver fire Pharmacopoeia

Ngelicae Sinensis Radix, Rehmanniae Radix, Ophiopogonis Radix, Tribuli Fructus, Poria, Semen (2015)”

f
Plantaginis, Alismatis Rhizoma, Glycyrrhizae Radix et Rhizoma

o
Fu Fang Chuan Bei Jing Polygalae Radix, Ephedrae Herba, Fritillariae Cirrhosae Bulbus, Citri Reticulatae Pericarpium, Treating cough, asthma, chest distress, “Chinese

ro
Tables Platycodonis Radix, Schisandrae Chinensis Fructus, Glycyrrhizae Radix et Rhizoma, Pinelliae phlegm, acute and chronic bronchi Pharmacopoeia

Rhizoma (2015)”

Fu Ning Kang Tablets

- p
Polygalae Radix, Ginseng Radix et Rhizoma, Lycii Fructus, Angelicae Sinensis Radix, Rehmanniae Treating irregular menstruation, vaginal “Chinese

e
Radix, Paeoniae Radix Rubra, Corni Fructus, Anemarrhenae Rhizoma, Phellodendri Chinensis dryness, emotional depression and Pharmacopoeia

P r
Cortex, Moutan Cortex, Acori Tatarinowii Rhizoma, Poria, Semen Cuscutae, Epimedii Folium,

Morindae Officinalis Radix, Cnidh Fructus, Cibotii Rhizoma, Schisandrae Chinensis Fructus
uneasiness caused by insufficiency of liver

and kidney and disharmony of the


(2015)”

Xin Nao Kang Tablets

a l
Polygalae Radix, Salviae Miltiorrhizae Radix et Rhizoma, Radix Rubra, Puerariae Lobatae Radix,
thoroughfare and conception vessels

Promoting blood circulation, removing “Chinese

r n
Carthami Flos, Achyranthis Bidentatae Radix, Curcumae Radix, Irkutsk Anemone Rhizome, blood stasis, dredging the orifices and Pharmacopoeia

u
Polygoni Multiflori Radix, Lycii Fructus, Chuanxiong Rhizoma, Alismatis Rhizoma, Pheretima, relieving pain, and curing chest (2015)”

o
Semen Ziziphi Spinosae, Deer heart powder, Glycyrrhizae Radix et Rhizoma obstruction, vertigo, coronary heart

Fu Mai Ding Capsules


J
Polygalae Radix, Astragali Radix, Codonopsis Radix, Chuanxiong Rhizoma, Mori Fructus
disease, angina pectoris and cerebral

arteriosclerosis

Treating palpitation and pulse replacement “Chinese

caused by qi deficiency and blood stasis, Pharmacopoeia

and atrial or ventricular premature beat (2015)”

Li Nao Xin Capsules Polygalae Radix, Salviae Miltiorrhizae Radix et Rhizoma, Chuanxiong Rhizoma, Puerariae Treating chest pain and colic caused by qi “Chinese

Thomsonii Radix, Pheretima, Paeoniae Radix Rubra, Carthami Flos, Curcumae Radix, Polygoni stagnation and blood stasis, palpitation, Pharmacopoeia

Multiflori Radix, Alismatis Rhizoma, Lycii Fructus, Semen Ziziphi Spinosae, Irkutsk Anemone dizziness and headache, coronary heart (2015)”

Rhizome, Achyranthis Bidentatae Radix, Glycyrrhizae Radix et Rhizoma disease, myocardial infarction, cerebral
Journal Pre-proof

Preparation name Compositions Traditional and clinical uses Ref.

arteriosclerosis and cerebral thrombosis

Ling Lian Hua Granules Polygalae Radix, Mushroom powder, Cardeniae Fructus, Ligustri LucidiFructus, Ecliptae Herba, Nourishing yin and tranquilizing mind, “Chinese

Lilii Bulbus, Rose Rugosae Flos, Leonuri Herba restoring coordination between heart and Pharmacopoeia

kidney to treat perimenopausal syndrome (2015)”

f
Jie Yu An Shen Granules Polygalae Radix (processed), Bupleuri Radix, Jujubae Fructus, Acori Tatarinowii Rhizoma, Pinelliae Treating insomnia, upset, anxiety and “Chinese

o
Rhizoma, Atractylodis Macrocephalae Rhizoma, Fructus Tritici Ievis, Glycyrrhizae Radix et forgetfulness caused by stagnation of liver Pharmacopoeia

o
Rhizoma, Cardeniae Fructus, Lilii Bulbus, Arisaema Cum Bile, Curcumae Radix, Dens Draconis, qi (2015)”

Semen Ziziphi Spinosae, Poria, Angelicae Sinensis Radix

p r
-
Tian Ma Xing Nao Polygalae Radix, Pheretima, Acori Tatarinowii Rhizoma, Castrodiae Rhizoma, Rehmanniae Radix, Nourishing liver and kidney, and relieving “Chinese

e
Capsules Cistanches Herba pain Pharmacopoeia

P r (2015)”

a l
r n
o u
J
Journal Pre-proof

3. Phytochemistry

More than 140 compounds have thus far been isolated and identified from PR in China, Japan,
and other countries. Saponins, xanthones, and oligosaccharide esters have been identified as the
characteristic constituents of PR. Organic acids, alkaloids, flavonoids, fatty oils, amino acids, and
resins are also found in PR. In this section, the main identified compounds and the corresponding
structures of PR are presented.

3.1. Saponins

Saponins, as the main pharmacological components of PR, have been discovered and
developed for a long time. These saponins exhibit structures typical of oleanolic pentacyclic
triterpenoid saponins, and the types of sugars include glucose, rhamnose, xylose, celery, galactose,

of
and so on [25]. So far, more than 50 saponins have been isolated from PR [26–35] (As shown in
Table 3 and Figure 2). Among them, tenuifolin is one of the index components of PR quality control

ro
in Chinese Pharmacopoeia. Pharmacological studies indicate that saponins exert the functions of
sedation, relieving excitement, expelling phlegm, relieving cough and so on.
-p
Table 3
Saponins isolated from PR.
re
No. Chemical component Chemical formula Source Ref.
1 Tenuifolin C36H56O12 Roots 26
2 Onjisaponin A C80H120O39 Roots 28
3 Onjisaponin B C75H112O35 Roots 28
lP

4 Onjisaponin E C57H92O27 Roots 28


5 Onjisaponin F C75H112O36 Roots 27
6 Onjisaponin G C52H84O22 Roots 27
7 Onjisaponin J C85H126O42 Roots 29
na

8 Onjisaponin L C86H128O43 Roots 29


9 Onjisaponin O C77H116O37 Roots 29
10 Onjisaponin R C76H114O37 Roots 29
11 Onjisaponin S C81H122O40 Roots 29
ur

12 Onjisaponin T C83H124O42 Roots 29


13 Onjisaponin Fg C81H120O40 Roots 29
14 Onjisaponin Gg C76H112O36 Roots 29
Jo

15 Onjisaponin Ng C80H118O38 Roots 29


16 Onjisaponin Pg C76H120O41 Roots 29
17 Onjisaponin Qg C70H110O37 Roots 29
18 Onjisaponin Sg C87H130O44 Roots 29
19 Onjisaponin Tg C90H136O45 Roots 29
20 Onjisaponin Ug C87H130O45 Roots 29
21 (Z)-Polygalasaponin XXXII C79H118O38 Roots 29
22 (Z)-Onjisaponin J C85H126O42 Roots 30
23 (Z)-Onjisaponin L C86H128O43 Roots 30
24 Onjisaponin H C74H110O34 Roots 30
25 (Z)-Onjisaponin H C74H110O34 Roots 30
26 Onjisaponin V C82H122O41 Roots 31
27 Onjisaponin W C81H122O40 Roots 31
28 Onjisaponin X C87H130O45 Roots 31
29 Onjisaponin Y C69H102O30 Roots 31
30 Onjisaponin Z C71H106O32 Roots 31
31 Onjisaponin Vg C82H122O41 Roots 31
32 Polygalasaponin XXXI C75H112O36 Roots 31
33 (E)-Senegasaponin a C74H110O35 Roots 31
34 Onjisaponin Wg C75H112O36 Roots 32
35 Onjisaponin MF C63H92O26 Roots 33
36 Onjisaponin TE C70H110O36 Roots 33
37 Onjisaponin TF C59H94O18 Roots 33
Journal Pre-proof

No. Chemical component Chemical formula Source Ref.


38 Onjisaponin TG C64H100O32 Roots 33
39 Onjisaponin TH C65H96O28 Roots 33
40 Polygalasaponin XLV C78H116O38 Roots 34
41 Polygalasaponin LIII C81H120O39 Roots 34
42 Myrtifolioside A1 C79H118O39 Roots 34
43 Desacylsenegasaponin B C59H94O29 Roots 34
44 Arilloside D C64H102O33 Roots 34
45 Arilloside A C57H72O27 Roots 35
46 Sibiricasaponin A C36H54O12 Roots 35
47 Sibiricasaponin B C35H56O11S Roots 35
48 Sibiricasaponin C C35H56O11S Roots 35
49 Sibiricasaponin D C37H58O12S Roots 35
50 Sibiricasaponin E C66H104O35 Roots 35

of
ro
-p
re
lP
na
ur
Jo
Journal Pre-proof

of
ro
-p
re
lP
na
ur
Jo

Figure 2. Chemical structures of the saponins in PR (1 − 50).

3.2. Xanthones

Xanthones are one of the important components in PR. At present, 27 xanthone (51–77)
Journal Pre-proof

compounds have been isolated and identified from PR (As shown in Table 4 and Figure 3) [36–44].
The mother nucleus of xanthone contains 8 positions that can be replaced, and only 3 types of
substituents are found: the hydroxyl group, the methoxy group, and the methylenedioxy group. A
study showed that the substitution probability of each position of the xanthones are as follows:
R1=97%, R2=68%, R3=86%, R4=20%, R5=3%, R6=30%, R7=70%, R8=14% [45]. Modern
pharmacological studies show that xanthones possess antifungal, analgesic, anti-cancer, and other
effects.
Chinese Pharmacopoeia stipulated that the medicinal part of PR was dry root, and most of the
aboveground parts were discarded. Song et al [46,47] showed that the aerial part of PR also contains
more active components, which provides a new scientific basis for rational development and
utilization of PR resources.
Table 4

of
Xanthones isolated from PR.
No. Chemical component Chemical formula Source Ref.
51 Onjixanthone I C16H14O6 Roots 36

ro
52 Onjixanthone II C15H12O7 Roots 36
53 3-Hydroxy-2,8-dimethoxyxanthone C14H10O6 Roots 37
54 7-Hydroxy-1,2,3-trimethoxyxanthone -p C15H12O7 Roots 37
55 3-Hydroxy-1,2,7-trimethoxyxanthone C15H12O7 Roots 37
56 1,7-Dimethoxy-2,3-methylenedioxyxanthone C15H12O6 Stems 37
57 6,8-Dihydroxy-1,2,4-trimethoxyxanthone C16H14O7 Roots 37
37
re
58 6,8-Dihydroxy-1,2,3-trimethoxyxanthone C16H14O7 Roots
59 Polygalaxanthone III C25H28O15 Roots 38
60 Sibiricaxanthone A C24H26O14 Roots 40
61 Sibiricaxanthone B C24H26O14 Roots 40
lP

62 Polygalaxanthone IV C27H32O15 Roots 41,42


63 Polygalaxanthone V C26H30O15 Roots 41,42
64 Polygalaxanthone VI C23H26O12 Roots 41,42
65 Polygalaxanthone VII C27H32O16 Roots 41,42
na

66 Polygalaxanthone VIII C25H28O15 Roots 41,42


67 Polygalaxanthone IX C25H28O14 Roots 41,42
68 Polygalaxanthone X C29H36O16 Roots 41,42
69 Polygalaxanthone XI C25H28O15 Roots 41,42
ur

70 Lancerin C19H18O10 Roots 41,42


71 6-Hydroxy-1,2,3,7-tetramethoxyxanthone C17H16O7 Stems 43
72 1,3,7-Trihydroxy-2-methoxyxanthone C14H10O6 Stems 43
Jo

73 1,2,3,6,7-Pentamethoxyxanthone C18H18O7 Stems 43


74 7-Hydroxy-1-methoxy-2,3-methylenedioxyxanthone C15H10O6 Roots 44
75 1,3,7-Trihydroxy-2,6-dimethanoxyxanthone C15H12O7 Roots 44
76 7-Hydroxy-1-methoxyxanthone C14H10O4 Roots 44
77 1,7-Dihydroxy-3,4-dimethoxyxanthone C15H12O6 Roots 44
Journal Pre-proof

of
ro
-p
re
lP
na
ur

Figure 3. Chemical structures of the xanthones in PR (51 − 77).

3.3. Oligosaccharide esters


Jo

Oligosaccharide esters represent a sizable class of PR compounds. With a structure that has
sucrose as the common mother nucleus, oligosaccharide esters connect glucose or rhamnose with
various forms of glycosidic bonds to form oligosaccharides and then forms esters with organic acids
[48]. Oligosaccharide esters are widely distributed in plants, but the sugar esters with more than
trisaccharide are only found in Polygalaceae. It is considered that these components are endemic to
Polygalaceae. A total of 36 oligosaccharide esters (78–113) have thus far been found in this plant (As
shown in Table 5 and Figure 4) [48–54]. Oligosaccharide esters have significant biological activity on
anti-dementia, brain protection, and anti-depression.
Table 5
Oligosaccharide esters isolated from PR.
No. Chemical component Chemical formula Source Ref.
78 Tenuifoliside A C31H38O17 Roots [48]
79 Tenuifoliside B C30H36O17 Roots [48]
80 Tenuifoliside C C35H44O19 Roots [48]
81 Tenuifoliside D C18H 24O19 Roots [48]
82 Tenuifoliside E C41H42O22 Roots 49
83 3,6′-Disinapoyl sucrose C34H42O19 Roots 49
Journal Pre-proof

No. Chemical component Chemical formula Source Ref.


84 Tenuifoliose A C62H76O35 Roots 50
85 Tenuifoliose B C60H74O34 Roots 50
86 Tenuifoliose C C58H72O33 Roots 50
87 Tenuifoliose D C60H74O34 Roots 50
88 Tenuifoliose E C58H72O33 Roots 50
89 Tenuifoliose F C68H86O39 Roots 52
90 Tenuifoliose G C66H84O38 Roots 51
91 Tenuifoliose H C61H74O34 Roots 51
92 Tenuifoliose I C59H72O33 Roots 51
93 Tenuifoliose J C59H72O33 Roots 51
94 Tenuifoliose K C57H70O32 Roots 51
95 Tenuifoliose L C67H84O38 Roots 51
96 Tenuifoliose M C65H82O37 Roots 51
97 Tenuifoliose N C63H78O36 Roots 51
98 Tenuifoliose O C61H76O35 Roots 51
99 Tenuifoliose P C59H74O34 Roots 51
100 Tenuifoliose Q C67H82O37 Roots 52
101 Sibiricose A1 C23H32O15 Roots 53

of
102 Sibiricose A2 C24H34O15 Roots 53
103 Sibiricose A3 C19H25O13 Roots 53
104 Sibiricose A4 C34H42O19 Roots 53

ro
105 Sibiricose A5 C22H30O14 Roots 53
106 Sibiricose A6 C23H32O15 Roots 53
107 Polygalatenoside A C19H26O11 Roots 54
108 Polygalatenoside B
-p
C19H26O11 Roots 54
109 Polygalatenoside C C19H26O11 Roots 54
110 Polygalatenoside D C20H28O13 Roots 54
re
111 Polygalatenoside E C22H32O13 Roots 54
112 Sibiriphenone A C20H22O11 Roots 43
113 Sibiricose A7 C17H22O9 Roots 43
lP
na
ur
Jo
Journal Pre-proof

of
ro
-p
re
lP
na
ur
Jo

Figure 4. Chemical structures of the oligosaccharide esters in PR (78 − 113).

3.4. Alkaloids

Alkaloids are widely found in plants and have remarkable and special biological activities. At
present, seven alkaloids were isolated from PR, all of which were β-carboline alkaloids (As shown in
Table 6, figure 5) [55]. Studies have found that β-alkaloids have anti-thrombotic, memory
enhancement, anti-tumor and other effects, and have great development and utilization potential
[56].

3.5. Other components

Organic acids and volatile oils are also important components of PR (As shown in Table 6,
figure 5) [57–60]. Among them, ferulic acid, erucic acid and 3,4,5-trimethoxycinnamic acid were the
Journal Pre-proof

main active components, with anti-seizure, anti-inflammatory, neuroprotective, and anti-amnesia


effects. In addition to the aforementioned chemical components, PR also contains coumarins,
phenylpropanoids, sterides and inorganic metal elements such as Zn, K, Ca, Cu, Fe, Mn, Mg and so
on.
Approximately 140 compounds have been isolated from PR, pharmacological activity studies
mainly focus on the chemical components of saponins and oligosaccharides. However, most other
components have not been further studied. Analysis may be difficult to obtain and purify
compounds, especially saponins and xanthones. Future scholars can use more advanced
instruments and new technologies such as high-speed countercurrent chromatography, molecular
imprinting, and ultrahigh-performance liquid chromatography-tandem mass spectrometry to isolate
and identify more compounds in P. tenuifolia Willd. and P. sibirica L. Through literature review, it is
found that there is little research on the active components of other parts of P. tenuifolia Willd. and P.

of
sibirica L., researchers should study the aboveground parts of P. tenuifolia Willd. and P. sibirica L.,
such as stems and leaves, to contribute to the rational utilization of resources.

ro
In this paper, the chemical constituents of PR were reviewed, which is helpful to further study
the material basis of PR, and provide scientific basis for the development and clinical application of
new drugs.
-p
Table 6
Other components isolated from PR.
re
No. Chemical component Chemical formula Source Ref.
114 N9-formylharman C13H12N2O Roots 55
115 1-Carbobutoxy-β-carboline C16H16N2O2 Roots 55
lP

116 1-Carboethoxy-β-carboline C14H12N2O2 Roots 55


117 1-Carbomethoxy-β-carboline C13H10N2O2 Roots 55
118 Perlolyrine C16H12N2O2 Roots 55
119 Norharman C11H8N2 Roots 55
na

120 Harman C12H10N2 Roots 55


121 3,4,5-Trimethoxycinnamic acid C12H14O5 Roots 57
122 Sinapic acid C11H12O5 Roots 57
123 Ferulic acid C10H10O4 Roots 57
ur

124 p-Hydroxybenzoic acid C7H6O3 Roots 57


125 p-Coumaric acid C9H8O3 Roots 57
126 Benzoic acid C7H6O2 Roots 57
Jo

127 Cinnamic acid C9H8O2 Roots 57


128 p-Methoxy cinnamic acid C10H10O3 Roots 57
129 O-Hydroxybenzoic acid C7H6O3 Roots 58
130 Hexanoic acid C6H12O2 Roots 59
131 Phenethyl alcohol C8H10O Roots 59
132 2,5-Dimethylbenzaldehyde C9H10O Roots 59
133 Stearic acid C18H36O2 Roots 59
134 Oleic acid C18H34O2 Roots 59
135 Palmitic acid C16H12O2 Roots 59
136 Methylsalicylic acid C8H8O3 Roots 59
137 Isorhamnetin-3-O-β-D-glucopyranoside C22H22O12 Roots 60
138 Isorhamnetin-3-O-β-D-galactopyranoside C22H22O12 Roots 60
139 Isorhamnetin C16H12O7 Roots 60
Quercetin-3-O-β-D-glucopyranosyl (1→ Roots
140 C28H32O17 60
2)-β-D-galactopyranoside
141 Linarin C26H28O16 Roots 60
Quercetin-3-O-β-D-glucopyranosyl (1→ Roots
142 C28H32O17 60
2)-β-D-glucopyranoside
143 5,7-Dihydroxy-8-methxoyflavone-7-O-β-D-glucuronoside C22H20O14 Roots 60
144 Kaempferol C15H10O6 Roots 60
145 Quercetin C15H10O7 Roots 60
146 Quercetin-3-O-β-D-glucopyranoside C21H20O12 Roots 60
Journal Pre-proof

of
ro
-p
Figure 5. Chemical structures of other compounds in PR (114 − 146).
re
4. Pharmacology
lP

Previous studies on PR have revealed its wide-spectrum pharmacological activities, including


its effects on the nervous system and the cardiovascular system, as well as its anti-inflammatory,
antiviral, antitumor, and antioxidant properties, among others. The effects on the nervous system
na

are more predominant. The primary pharmacological actions of PR are listed in Table 7 and Figure 6.
ur
Jo
Journal Pre-proof

Table 7
Pharmacological effects of PR.
Pharmacological effects Extracts/compounds Model Dose range tested Mechanism Ref.

Neuroprotective effects Senegenin PC12 cells 1, 5 μg/mL Moderating neurite outgrowth and increasing the 62

expression of MAP2 and Gap-43

63

f
Senegenin Rat hippocampal neural stem 3 μg/mL Not mentioned

o
cells

ro
Onjisaponin B C57BL/6J mice 20, 40 mg/kg/d Inhibiting the RhoA/ROCK2 pathway [64]

Tenuifolin BV2 cells 1, 5, 10 μM Downregulating the NF-κB signaling pathway [65]

1,3,7-Trihydroxyxanthone Rat astrocytes primary 0.1–10 μM

- p Modulating of tPA system and activation of cAMP- 66

e
cultures and ERK- pathway

Tenuifoliside A PC12 cells

r
10 μM

P
Activating the MEK/ERK/CREB and PI3K/Akt

signaling pathways and triggering the


67

3,6′-Disinapoyl sucrose SY5Y cells

a l 75, 150 μM
phosphorylation of CREB

Inhibiting the hyperactivation of NOS, upregulating 68

r n phosphorylation of CREB and increasing the

u
expression of CRTC1 and BDNF

o
Tenuifoliside A SY5Y cells 25, 50 μM Inhibiting the hyperactivation of NOS, upregulating 68

Antidepressant effects Extract of PR


JICR mice and Wistar rats 0.13, 0.25, 0.5, 1 g/kg (p.o., 14 d
phosphorylation of CREB and increasing the

expression of CRTC1 and BDNF

Promoting autophagy and inhibiting 69

for mice), 0.5, 1 g/kg (p.o., 28 d neuroinflammation

for rat)

Senegenin ICR mice 4, 8 mg/kg (p.o., 45 d) Inhibiting of NF-κB and regulating NLRP3 signal 70

Tenuifolin Mice 3, 6, 9 mg/kg (p.o., 28 d) Increasing expression of 5-HT, decreasing expression 71

of IDO, decreasing activity of ache, and enhancing

activity of chat
Journal Pre-proof

Pharmacological effects Extracts/compounds Model Dose range tested Mechanism Ref.

3,4,5-Trimethoxycinnamic acid C57BL/6J mice 25, 50 mg/kg (p.o., 14 d) Enhancing FosB protein and SC1 mRNA expression 69

of in the nucleus accumbens

Sibiricose A5 PC12 cells 0.53, 13.2, 132 μmol/L Inhibiting of Ca2+ release and regulating 72

neurotransmitter release

73

f
3,6′-Disinapoyl sucrose Mice 1, 2, 4 mg/kg (p.o., 3 d) Enhancing the nerve function of 5-HT and NE

74

o
3,6′-Disinapoyl sucrose Chronic unpredictable 5, 10, 20 mg/kg (p.o., 28 d) Modulating the HPA axis

ro
mild stress rats

3,6′-Disinapoyl sucrose Chronic unpredictable 5, 10, 20 mg/kg (p.o., 35 d) Relating to MAO, HPA axis, and oxidative systems 75

mild stress rats

- p 76

e
3,6′-Disinapoyl sucrose Chronic unpredictable 5, 10, 20 mg/kg (p.o., 21 d) Increasing expressions of CAM-L1, laminin, CREB,

Ethanol extracts
mild stress rats

ICR mice and SH-SY5Y cells

P r
In vivo: 200 mg/kg (p.o., 7 d);
and BDNF in the hippocampus

Cytoprotection and neuroproliferating actions 77

3,6′-Disinapoyl sucrose SH-SY5Y cells

a l in vitro: 100, 200 μg/mL

1.5 ×10-5 M Cytoprotection and neuroproliferating actions 77

Tenuifoliside A

r n
SH-SY5Y cells 1.5 ×10-4 M Cytoprotection and neuroproliferating actions 77

u
Cognitive-enhancing Aerial part of P. tenuifolia Male mice 25, 50, 100 mg/kg (p.o., 45 d) Modulating cholinergic activity, inhibiting 78

o
effects neuroinflammation and oxidative stress, and

Water extract of PR
JAD mice and cortical neurons In vivo: 12, 60 mg/kg (p.o., 56

d); in vitro: 0.1, 1, 10, 100


regulating the BDNF and TrkB

Inhibiting endocytosis 79

μg/mL

Onjisaponin B APPswe/PS1ΔE9 In vivo: 10 mg/kg (p.o., 8 d); in Promoting the degradation of amyloid precursor 80

double-transgenic mice and vitro: 10 μg/mL protein

HEK293-APPswe cells

Polygalasaponin XXXII Scopolamine induced mice 0.125, 0.5, 2 mg/kg (p.o., 19 d) Protecting neurons from glutamate and ROS damage, 81

upregulating the phosphorylation of TrkB and


Journal Pre-proof

Pharmacological effects Extracts/compounds Model Dose range tested Mechanism Ref.

maintenance of LTP

Tenuigenin Male mice In vivo: 2, 4 mg/kg (p.o., 20 d) Improving antioxidation, reducing ache activity, and 82

enhancing synaptic plasticity in hippocampus

Tenuigenin Male rats 375 mg/kg (p.o., 35 d) Increasing the expression of nAChRα7 in the 83

f
hippocampus CA1 area

84

o
Hydrolysate of polygalasaponin Senescence accelerate mouse 25, 50 mg/kg (p.o., 10 d) Associating with NMDA receptor (NMDAR)-related

ro
P8 mice pathways

Polygalacicacid male mice 3, 6, 12 mg/kg (p.o., 14 d) Modulating of cholinergic activity and neuro 85

- p inflammation

e
Hypnotic-sedative Ethanol extract of PR ICR mice 1 g/kg (p.o., 7 d) Increasing dopamine content in hippocampus of mice 87

effects

3,4,5-Trimethoxycinnamic acid ICR mice and cerebellar

P r
In vivo: 2, 5, 10 mg/kg (p.o., Modulating GABAergic systems 88

granule cells

a l one administration); in vitro:

10 μM

3,4,5-Trimethoxycinnamic acid Male mice

r n 5, 10, 20 mg/kg (p.o., 21 d) Acting at the GABAA receptor complex 89

u
Tenuifolin ICR mice 20, 40, 80 mg/kg (p.o., one Activating GABAergic systems and/or inhibiting 91

o
administration) noradrenergic systems

Anti-inflammatory
Tenuifolin

Tenuifoliside A
JZebrafish

LPS induced RAW264.7 and


1, 10, 20, 30 μM

5, 10, 20, 40, 80 μM


Mediating by the serotoninergic systems and the

GABAergic systems

Inhibiting JNK/MAPK and NF-κB signaling


92

93

effects murine peritoneal

macrophages

Polygalacic acid SD rats and chondrocytes In vivo: 100 μM (intra-articular Inhibiting MMPs expression and osteoarthritis via 94

injection, 6 weeks); in vitro: 50, Wnt/β-catenin and MAPK signal pathways

100 μM suppression

Tenuigenin LPS induced RAW 264.7 cells 0.98, 1.86, 3.72 μM Inhibiting iNOS and COX-2 expression via 95
Journal Pre-proof

Pharmacological effects Extracts/compounds Model Dose range tested Mechanism Ref.

downregulating MAPK and NF-κB, and upregulating

Nrf2/HO-1 signaling pathways

IL-1β-stimulated osteoarthritis 2, 4, 8, 10 μM Inhibiting PI3K/AKT/NF-κB signaling pathway 96

chondrocytes

97

f
LPS and D-GalN-induced liver 2, 4, 8 mg/kg Inhibiting ASK1, MAPKs, activating NF-κB, and

o
injury mice (intraperitoneally, one upregulating Nrf2 and HO-1 expression

ro
administration)

Male C57BL/6J mice and BV2 in vivo: 25, 50 mg/kg (p.o., 10 Suppressing NLRP3 inflammasome activation in 98

microglia cells

- p
d); in vitro: 2, 4, 8 μM microglia

99

e
Staphylococcus 2, 4, 8 mg/kg (p.o., one Inhibiting NF-kB activation

aureus-induced

pneumonia mice r
administration)

P
a l
LPS-activated rat microglia 10-4, 10-5, 10-6 M Downregulating the release of NO, MMP-9 and

cytokines
100

Antiviral effects 3,4,5-Trimethoxy cinnamic acid

r
Goldfish and

n In vivo: 2.5–20 mg/L (p.o., 2 d); Inducing the expression of MyD88, IL-1β, TNF-α and 101

u
ctenopharyngodon in vitro: 1, 10, 100 μg/L Mx1

o
idella kidney cells

1,5-Anhydro-D-glucitol

JGoldfish and

ctenopharyngodon

idella kidney cells


In vivo: 10–80 mg/L (p.o., 2 d);

in vitro: 1, 10, 100 μg/L


Inducing the expression of MyD88, IL-1β, TNF-α and

Mx1
101

Antitumor effects Polysaccharide OVCAR-3 cells 0.1, 0.2, 0.4, 0.8, 1.6 mg/mL Downregulating bcl-2 and NF-κB expression and 102

upregulating bax, cytochrome c, caspase-3, and

caspase-9 levels through a mitochondrial pathway

Polysaccharide Nude BALB/c mice 10, 20, 40 mg/kg (p.o., 2 Downregulation of mRNA levels of EGFR, VEGF, and 103

weeks) CD34

Polysaccharide OVCAR-3 cells 0.125, 0.25, 0.5, 1, 2 mg/mL Inhibiting Bmi-1 both in protein and transcript levels 104
Journal Pre-proof

Pharmacological effects Extracts/compounds Model Dose range tested Mechanism Ref.

and downregulating telomerase activity

RP02-1 Mice and BxPC-3 and AsPC-1 in vivo: 5, 50 mg/kg (p.o., three Downregulating the expression of Bcl-2, upregulating [105]

cells weeks); in vitro: 4.31, 8.62 μM the expression of Bax, increasing the expression of

Cleaved Caspase 3, and suppressing autophagy

106

f
Antioxidant effects Oligosaccharide ester fractions Senescence-accelerated mice 25, 50 mg/kg (p.o., 90 d) Increasing of the SOD and GSH-Px contents and

o
decreasing of MDA contents

o
Polysaccharide Male mice 0.10, 0.20, 0.40 mg/g (p.o., 30 Not mentioned [107]

d)

p r
-
3,6′-Disinapoyl sucrose Senescence-accelerated mice 25, 50 mg/kg (p.o., 90 d) Increasing of the SOD and GSH-Px contents and 108

e
decreasing of MDA contents

Aqueous extract of PR D-galactose induced aging

mice r
25, 50, 100 mg/kg (p.o., 45 d)

P
Decreasing the MDA contents and increasing the

SOD, CAT and GSH-Px activities and total


108

Cardiovascular effects 3,4,5-Trimethoxycinnamic acid


a l
Rabbit ventricular myocytes 15, 30 μM
antioxidant capacity

Inhibiting calcium channel and suppressing 109

r n intracellular Ca2+ transients

Senegenin SD rats

o u 40, 80 mg/kg (intraperitoneal

injection, 14 d)
Reducing inflammation and inhibiting HMGB1

expression
110

Other pharmacological

effects
Polysaccharide

Extract of PR Jα-Glucosidase enzyme

High-fat diet induced obese

mice and 3T3-L1 adipocytes


0.05–6.40 mg/mL

In vivo: 250 mg/kg (p.o., 5

weeks); in vitro: 125, 250, 500


Inhibiting α-glucosidase enzyme activity

Inhibiting lipid accumulation by inducing the

expression of the master transcription factor PPARα,


111

112

μg/mL attenuating the low-grade chronic inflammation of

obesity, and altering gut microbiota profiles

PR, licorice-boiled PR and Mice 1, 4 g/kg Not mentioned 113,114

honey-stir-baked PR
Journal Pre-proof

of
ro
-p
re
lP
na
ur
Jo

Figure 6. The molecular pharmacological activities mechanisms of PR.

4.1. Effects on the nervous system

4.1.1. Neuroprotective effect

Nervous system diseases, such as cerebrovascular disease, AD, and Parkinson’s disease (PD)
are the major diseases affecting human health [61]. In modern pharmacology, the neuroprotective
effects of PR have been comprehensively investigated. Saponins are abundant in PR and exhibit
good neuroprotective activities. Recent research on PC12 cells incubated with Aβ peptide suggested
that senegenin could increase the viability of PC12 cells by moderating neurite outgrowth and
increasing the expression of microtubule-associated protein 2 (MAP2) and growth-associated
protein 43 (Gap-43) [62]. In 2019, another study on the hippocampal neural stem cells revealed that
senegenin treatment could alleviate damage to neural stem cells induced by oxygen-glucose
Journal Pre-proof

deprivation/reoxygenation [63]. Nevertheless, these two studies on neuroprotective effects of


senegenin lacked a positive control and the analysis of dose-dependent effect. These limitations put
into question the reliability of these in vitro study.
Onjisaponin B at the doses of 20 and 40 mg/kg should be explored as a potential therapeutic
agent for PD because it acts through a mechanism that attenuates microglia over-activation and
secretion of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6),
improves antioxidant enzyme activity, inhibits the expression of nuclear factor-kappa B (NF-κB)
p65, RhoA and Rhokinase 2 (ROCK2) proteins by activating the RhoA/ROCK2 signaling pathway.
In addition, onjisaponin B has a high molecular weight and cannot pass the blood-brain barrier.
One hour after oral administration of onjisaponin B, the metabolite tenuifolin could be detected in
the brain of mice, and entered the brain to play a therapeutic effect [64]. Furthermore, tenuifolin at
the concentrations of 5 and 10 µM has also been shown to reduce inflammation and oxidative stress

of
induced by amyloid-β oligomers in BV2 cells by downregulating the NF-κB signaling pathway [65].
Nonetheless, because the factors affecting PD or AD are numerous, the signaling pathways are

ro
complex and diverse. Thus, more studies are needed to investigate the role and mechanism of the
two agents in the pathological process of neurodegenerative diseases, and to search for the factors
and related signal molecules that regulate their neuroprotective effects.
-p
Another PR component, 1,3,7-trihydroxyxanthone, could significantly increase the synthesis of
the nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) at both the
re
transcription and protein expression levels in a dose-dependent manner. The mechanism
underlying this effect might be associated with the modulation of the tissue plasminogen activator
lP

(tPA) system and the activation of the cyclic adenosine monophosphate (cAMP) and extracellular
signal-regulated kinase (ERK) pathways [66]. Thus, 1,3,7-trihydroxyxanthone may potentially be a
novel candidate to treat AD. Anyhow, the establishment of an appropriate experimental animal
na

model is also crucial to further verify the activity of 1,3,7-trihydroxyxanthone in vivo.


Oligosaccharide esters are the main bioactive substances in PR, and they can be considered as
ur

the lead compounds for the development of new anti-aging and brain protection drugs. Previous
studies indicated that tenuifoliside A exhibited an apparent neuroprotective effect via activating the
Jo

MEK/ERK/CREB and PI3K/Akt signaling pathways [67]. Liu et al. confirmed that co-treatment of
3,6′-disinapoyl sucrose and tenuifoliside A could inhibit the hyperactivation of Glu-induced nitric
oxide synthase (NOS) and upregulate the phosphorylation of CREB, as well as increase the
expression of CREB-regulated transcription co-activators (CRTC1) and BDNF [68]. Nonetheless,
only two doses of 3,6′-disinapoyl sucrose and tenuifoliside A were used throughout the experiment,
limiting any data on dose-dependency. In addition, more experiments should attempt to verify
their additive or synergistic effects on animal models of mental disorders and to explore whether
they can pass the blood-brain barrier.

4.1.2. Antidepressant effect

Depression is a common psychosomatic disease with significant and lasting mood depression
as the main clinical characteristics, which seriously endangers human health. At present, there are
many adverse reactions and slow onset of antidepressant drugs in the process of treating
depression. Therefore, it is urgent to seek an antidepressant Chinese medicine with rapid effect and
small side effects in clinical practice. The antidepressant activities of saponins, organic acids, and
Journal Pre-proof

oligosaccharide esters in PR have been reported in recent years. Animal and cell models of
depression and their mechanisms are discussed in this review. In 2020, Zhou et al. evaluated the
antidepressant effects of PR extracts in behavioral despair mice and chronic restraint stress-induced
rats by behavioral testing and brain tissue biochemical examination. The results showed that RP
(0.5 and 1 g/kg) could decrease the immobility time of mice in forced swimming test (P < 0.01), the
decrease of latency to feeding and the immobility time (P < 0.05), the increase of locomotor activity
(P < 0.05). It could also inhibit the expression of p62, increase the expression of LC3-II and beclin1,
regulate the dysfunction of AMPK-mammalian target of rapamycin (mTOR) pathway in prefrontal
cortex of rats, downregulate the protein levels of NLRP3, ASC and cleaved caspase-1, and inhibit
the overexpression of proinflammatory cytokines. The antidepressant effect of PR extracts was
achieved by promoting autophagy and inhibiting neuroinflammation [69]. However, this study did
not systematically screen natural small molecule compounds with antidepressant effect from PR. In

of
future study, scholars can consider using a variety of chromatographic separation methods and the
modern spectrum methods to isolate and identify the structure of the compounds obtained from PR

ro
extracts, and then use appropriate in vitro experimental model to screen antidepressant active
compounds, so as to lay a pharmacological foundation for better development of antidepressant
natural products with definite clinical efficacy and controllable quality.
-p
In 2017, the antidepressant activity of senegenin in mice with chronic unpredictable mild stress
(CUMS) was investigated. The effect of senegenin was primarily related to the increase in the BDNF
re
and neurotrophin-3 (NT-3) protein levels and inhibition of IL-1β secretion via the downregulation
of the NOD-like receptor family, pyrin domain-containing 3 (NLPR3) inflammasome pathway [70].
lP

Yet this investigation was conducted without the analysis of a dose-response. Wang et al. took the
mice model of depression induced by CUMS and 28-day isolation as the research object to
investigate the antidepressant effect of tenuifolin. The results showed that tenuifolin (6 and 9
na

mg/kg) could reduce the fixed time of forced swimming test and tail suspension test (P < 0.01) in 4
weeks, increase the 5-hydroxytryptamine (5-HT) content in the cerebral cortex of mice, inhibit the
ur

activity of indoleamine 2,3-dioxygenase (IDO) and acetylcholinesterase (AChE), and enhance the
activity of acetyl-transferase (ChAT) (P < 0.05) [71]. However, there was no in-depth study on the
Jo

antidepressant mechanisms of tenuifolin, such as the effect of changes in cAMP, mitogen-activated


protein kinase (MAPK), BDNF, mTOR and other key proteins in the antidepressant pathways.
In mice with stress-induced anxiety and depression-like behavior, 3,4,5-trimethoxycinnamic
acid exhibited antistress and antidepressant activities by enhancing FosB protein and SC1 mRNA
expression in the nucleus accumbens of mice [72]. This study had the same shortage of lacking
dose-dependent analysis and positive control. A series of mechanisms are involved in the
antidepressant effect of 3,6′-disinapoyl sucrose, including regulation of nerve function of 5-HT and
noradrenaline (NA) [73], the hypothalamus-pituitary-adrenal (HPA) axis [74], and the biological
oxidation system [75], improvement of synaptic plasticity of neurons, and promotion of
proliferation of neurons [76]. As these studies of mechanisms were only carried out in animals,
additional studies using clinical trials with appropriate doses are required. Liu et al. found that 50%
of the EtOH fraction in PR exhibited apparent anti-immobility effects in mice. In
corticosterone-induced injury in SH-SY5Y cells, the 50% EtOH fraction of PR and its major
compounds 3,6′-disinapoyl sucrose and tenuifoliside A showed protective properties by increasing
the neuronal survival of cells [77]. However, the antidepressant effects of other components in the
50% EtOH fraction of PR were lacking in this study.
Journal Pre-proof

4.1.3. Cognitive-enhancing effect

A large number of experiments have proved that PR is an effective drug to improve learning
and memory in animals. In 2020, the aerial part of P. tenuifolia was found to exert
memory-enhancing effects against D-galactose/NaNO2-induced learning and memory impairment
in mice by regulating modulating cholinergic activity, inhibiting the formation of IL-1β and
malondialdehyde (MDA), increasing the glutathione (GSH) level and superoxide dismutase (SOD)
activity, as well as increasing the expression of BDNF and tropomyosin receptor kinase B (TrkB)
[78]. Although different doses of extracts from the aerial parts of P. tenuifolia were studied, the safe
range of effective doses was not clear. In another study, the preventive effects against memory
impairment of water extracts from PR were evaluated using 5XFAD mouse models of AD. The
results revealed that the mechanism was potentially related to the prevention of axonal
degeneration and memory impairment via the endocytosis inhibition [79].

of
The effects of saponins in PR on learning and memory have been studied extensively. In 2016,
Li et al. isolated active compounds from PR extracts, among which onjisaponin B possessed exerted

ro
apparent improvement in learning and memory by promoting the degradation of amyloid
precursor protein [80]. Cognitive enhancement attributed to polygalasaponin XXXII was
-p
investigated in mouse models of scopolamine-induced cognitive impairment. Polygalasaponin
XXXII was found to protect neurons from glutamate and reactive oxygen species as well as to
re
upregulate the phosphorylation of TrkB and maintenance of long-term potentiation [81].
Tenuigenin was reported to enhance learning and memory in APP/PS1 transgenic AD mice, and its
lP

mechanism could be related to reversing neuronal apoptosis by inhibiting mitochondrial


membrane-potential loss [82]. Meanwhile, tenuigenin could markedly increase the expression of the
nicotinic acetylcholine receptor subunit alpha-7 (nAChRα7) in the CA1 area of the hippocampus in
na

rat models of epilepsy, which may be part of the mechanism for improving cognitive impairment
[83]. However, the lack of a positive control and single dose is the limitation of this study. In
addition, the biochemical indicators of this study were single, and there was no in-depth study on
ur

the mechanism of tenuigenin improving cognitive impairment. It has been reported that the
hydrolysate of polygalasaponin exerted cognitive-enhancing effects on the senescence-accelerated
Jo

mouse P8 via the N-methyl-D-aspartate (NMDA) receptor-related pathways [84]. Another


pharmacological study revealed the neuroprotective activity of polygalacic acid against
scopolamine-induced cognitive dysfunction [85]. Park et al. recently reported that PSM-04 isolated
from the root of P. tenuifolia Willd. could alleviate cognitive impairment and decrease amyloid
plaque deposition in 5xFAD mice [9]. Though various components in PR have been investigated for
their cognitive-enhancing effect, further research is needed to verify the effectiveness of the
components, the mechanisms involved and effective doses.
PR is widely used in TCM to treat amnesia. The above pharmacological studies show that PR
can significantly improve the learning and memory abilities of various dementia model animals,
which fully indicates that PR can be used as a good source of drugs to improve intelligence.
However, the pharmacodynamic material basis and specific mechanism of PR are not yet very clear.
In the future, scholars need to make joint efforts to conduct in-depth research on it.

4.1.4. Hypnotic-sedative effect

The pathogenesis of depression is relatively complex, and there is no consistent theoretical


Journal Pre-proof

concept in clinical practice. Therefore, it is urgent to seek effective drugs for the treatment of
depression in clinical practice. Modern drug epidemiological investigations have shown that PR is a
common individual Chinese medicine for insomnia [86]. Chen et al. evaluated the hypnotic effect of
PR and found that the ethanol extract of PR could prolong the sleep time and increase the sleep rate
of mice, and the effective component was the ethyl acetate fraction of the ethanol extract. Further
research found that the elution fraction of 70% ethanol in MCI column had the strongest hypnotic
effect, and the DA content in the hippocampus of mice was the highest (P < 0.05) [87]. However, this
study did not explore the mechanism of the hypnotic effect of PR, and lacked the comparative
study of positive drugs.
In 2013, an organic acid isolated from PR, 3,4,5-trimethoxycinnamic acid improved sleep and
altered sleep architecture by GABAAergic systems in mice [88]. In addition,
3,4,5-trimethoxycinnamic acid showed significant antiseizure activity in maximal electroshock

of
(MES) and pentylenetetrazol (PTZ)-induced seizure models, which markedly decreased the
incidence of MES-induced tonic hindlimb extension, severity, and mortality of seizures [89]. In

ro
2019, Zhao et al. examined the anticonvulsant activity of 35 3,4,5-trimethoxycinnamic acid ester
derivatives both in vivo and in silico and found that the anticonvulsant effects of derivatives 28 and
32 were stronger than those of other derivatives, indicating their potential for further development
-p
into anticonvulsant drugs [90]. However, these studies lacked comparative studies of positive drugs
and clinical verification. Future scholars should further analyze the effects of
re
3,4,5-trimethoxycinnamic acid on other hypnotic-sedative targets to improve its mechanism.
A report in 2016 revealed the hypnotic effect of tenuifolin in male institute of cancer research
lP

(ICR) mice and that its ability to prolong total sleep time, reduce NA and gamma-aminobutyric acid
(γ-GABA), as well as increase acetylcholine (Ach). These results demonstrated that tenuifolin could
exhibit sleep-enhancing effects, which might activate GABAAergic systems and/or inhibit
na

noradrenergic systems [91]. A study in 2020 reported that tenuifolin exhibited strong
sleep-promoting activity in zebrafish mediated via the serotoninergic and GABAAergic systems
ur

[92]. However, this study did not explore the effects of tenuifolin on the sleep behaviors of zebrafish
under dark or bright conditions, and lack of research on downstream gene products of biological
Jo

signalling pathways, which is the limitation of this study.


PR, as a classic hypnotic-sedative Chinese medicine, is commonly used in Chinese herbal
compound to treat insomnia and neurasthenia. Although PR or its active components have
hypnotic-sedative effects, the research on the underlying mechanism of PR is not sufficiently. The
current research cannot explain the interaction between various targets and pathways from a
holistic perspective, and cannot connect them into a complete mechanism network. Future scholars
can use transcriptomics, genomics, metabolomics and other methods to conduct in-depth research
on PR, and discover more potential targets and signal pathways.

4.2. Anti-inflammatory effect

Inflammation is caused by the invasion of bacteria or fungi into the human body. Currently,
antibiotics are mainly used to treat inflammatory diseases caused by bacteria. Long-term use of
antibiotics can lead to liver and kidney function damage, intestinal flora disorders, and even drug
resistance. Therefore, the research and development of efficient and low toxicity TCM or
compounds to prevent and treat diseases caused by inflammation has a good application prospect.
Journal Pre-proof

Kim et al. explored the anti-inflammatory effect of tenuifoliside A and found that
tenuifoliside A inhibited the NF-κB pathway in macrophages via inhibiting the production of
pro-inflammatory factors such as TNF-α, and IL-1. It also could reduce the production of nitric
oxide (NO), inducible nitric oxide synthase (iNOS), prostaglandin E2 (PGE2) and cyclooxygenase-2
(COX-2), and suppress the phosphorylation of JNK in a dose-dependent manner to play an
anti-inflammatory role [93]. To our best knowledge, this is the first study to demonstrate that
tenuifoliside A in PR has anti-inflammatory effect. However, this study lacked validation at the
animal and clinical trial levels, which limited the development and application of tenuifoliside A
in the treatment of inflammatory diseases.
In 2018, Xu et al. evaluated the anti-inflammatory activity of polygalacic acid both in vivo and
in vitro. The results indicated that polygalacic acid inhibited the expression of matrix
metalloproteinases (MMPs) and chondrocyte inflammation via the suppression of the

of
Wnt/β-catenin and MAPK signal pathways in rat chondrocytes [94]. Although detailed research
was carried out on anti-inflammatory mechanisms in this study, a major limitation was lack of a

ro
positive control.
Tenuigenin has six anti-inflammatory mechanisms: 1) inhibition of the expression of iNOS and
COX-2 by downregulating MAPK and NF-κB and upregulating the nuclear factor erythroid
-p
2-related factor 2/heme oxygenase (Nrf2/HO-1) signaling pathways in RAW 264.7 [95]; 2) inhibition
of the PI3K/AKT/NF-κB signaling pathway in IL-1β-induced human osteoarthritis cartilage cells
re
[96]; 3) inhibition of ASK1, MAPK, and NF-κB activation, as well as upregulation of Nrf2 and HO-1
expression in lipopolysaccharide and D-galactosamine (D-GalN)-induced liver injury in mice [97];
lP

4) inhibition of the activation of NLPR3 inflammatory bodies in microglia [98]; 5) inhibition of the
NF-κB signaling pathway [99]; and 6) downregulation of the release of NO, MMP-9, and cytokines
[100]. However, the safety or side effects of tenuigenin were not explored in these studies, aspects
na

which should be addressed by a toxicity study prior to use in clinical trials.


Multiple components in PR show anti-inflammatory activity through a plethora of
ur

mechanisms. These results are of high research value, complement the traditional use of PR in the
treatment of sores, swelling and pain of breast, and strengthen the research and development of this
Jo

plant as an anti-inflammatory agent.

4.3. Antiviral effect

Virus is a non cellular parasitic organism composed of a nucleic acid molecule and a protein.
The prevention and treatment of viral diseases in modern medicine has always been a thorny
problem. Many years of practice and research have proved that there are a lot of TCM has a better
effect on viral infection diseases. PR is believed to have the antiviral effect. Yu et al. subsequently
evaluated the antiviral ability of two compounds isolated from PR in vitro and in vivo. The purified
compounds were identified as 3,4,5-trimethoxycinnamic acid and 1,5-anhydro-D-glucitol. In vitro
anti grass carp reovirus (GCRV) experiments showed that the two compounds upregulated the
levels of immune-related genes such as myeloid differentiation factor 88 (MyD88), TNF-α,
myxovirus (Mx1), and IL-1β to varying degrees. According to in vivo insecticidal assays, the efficacy
of 3,4,5-trimethoxycinnamic acid was higher than that of 1,5-anhydro-D-glucitol [101]. However,
this study lacked positive control and the support of animal in vivo experiment and clinical
experiment date.
Journal Pre-proof

4.4. Antitumor effect

Ovarian cancer is one of the most common cancers in women. It has the characteristics of
strong invasion, high mortality and high recurrence rate. Ovarian cancer is still the most lethal
gynecological malignant tumor. Polysaccharide, one polysaccharide extracted from the roots of P.
tenuifolia Willd., has anti-ovarian cancer effects. Zhang et al. revealed that polysaccharide could
significantly induce the apoptosis of human ovarian carcinoma cells-3 (OVCAR-3) in 2015. They
confirmed that the molecular mechanism of the antitumor effect of polysaccharide was associated
with the cell circle arrest at the G0/G1 phase, inhibition of bcl-2 and NF-κB expression, and
upregulation of bax, cytochrome c, caspase-3, and caspase-9 expression [102]. However, there were
no positive control studies in this study, and the effects on downstream targets of the NF-KB
signaling pathway were not discussed in this study. This study was conducted only at the cell level
in vitro, and the mechanism and protein targets of polysaccharides on breast cancer cells in vivo

of
remain to be further explored.
Yao found that polysaccharide could obviously decreased transcription and protein levels of

ro
the epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF) and CD34,
leading to induction of apoptosis in tumor-bearing mice [103]. Zhang et al. studied the antitumor
-p
effect of polysaccharide in OVCAR-3 cells and found that polysaccharide could inhibit cell growth
and colony formation (P < 0.05) of OVCAR-3 cells in a dose-dependent manner. Polysaccharide also
re
effectively inhibited the expression of Bmi-1 at the gene and protein levels and the activity of
telomerase in OVCAR-3 cells [104]. However, this study lacked detailed study on the mechanism of
lP

action of polysaccharide in various growth stages of OVCAR-3 cells, as well as the comparison
study of positive drugs. In the future, flow cytometry can be used to further study the influence of
polysaccharide on each growth stage of OVCAR-3 cells.
na

Pancreatic ductal adenocarcinoma is a highly malignant tumor of the digestive system and one
of the most aggressive tumors. It ranks the fourth in cancer-related deaths in western countries.
Compared with the high toxicity and side effects of chemotherapy drugs, TCM has mild and little
ur

side effects. Bian et al. showed RP02-1, a polysaccharide purified from P. tenuifolia Willd., could
effectively inhibit the growth of pancreatic cancer cell in vitro and in vivo. Mechanism study
Jo

suggested that RP02-1 induced apoptosis of pancreatic cancer cells in a dose-dependent manner by
downregulating the expression of Bcl-2, upregulating the expression of Bax and increasing the
expression of Cleaved Caspase 3. In addition, RP02-1 could inhibit BxPC-3 cells autophagy. Cell
experiments showed that RP02-1 had the biological activity of inhibiting pancreatic cancer cells
growth [105]. However, this study did not study the best therapeutic dose of RP02-1, nor did it
conduct toxicity experiments on RP02-1. In the future research, scholars can consider the acute
toxicity experiment and subacute toxicity experiment to study the safe dose of RP02-1.

4.5. Antioxidant effect

Aerobic organisms are always accompanied by oxidative stress in all stages of life. When the
body is stimulated by oxidation, the excessive expression of reactive oxygen species (ROS) and
reactive nitrogen free radicals (RNS) cannot be cleared in time, which will damage the homeostasis
of oxidation system and antioxidant system, induce cell or tissue damage, and further induce the
generation of various acute and chronic diseases. Therefore, the research and development of TCM
with antioxidant effect has become one of the hot spots in medical science research.
Journal Pre-proof

Liu et al. took the senescence-accelerated mice as the research object to evaluate the antioxidant
activities of resin-fractionated YZ oligosaccharide esters (YZ-OE) and its major constituent
3,6′-disinapoyl sucrose. After 90 days of administration, compared with the model group, the
contents of SOD and glutathione peroxidase (GSH-PX) increased remarkably (P < 0.05, P < 0.01),
and the content of MDA decreased significantly in different doses of YZ-OE and 3,6′-disinapoyl
sucrose (25 and 50 mg/kg). These results indicated that YZ-OE had a high in vivo antioxidant
activity [106]. However, this study lacked in-depth study of antioxidant mechanism, only a few
relatively simple cellular antioxidant enzymes were detected, and the relevant signal pathways and
target proteins were not explored.
Feifei Xie studied the antioxidant activity of PR polysaccharide with different concentrations in
vitro and found that the scavenging rate of PR polysaccharide on hydroxyl free radical and DPPH
free radical was positively correlated with its concentration. The maximum scavenging rates of

of
hydroxyl radical and DPPH radical were 61.3% and 79.5%, respectively (4 mg/mL), which indicated
that PR polysaccharide has good oxidation resistance in vitro [107]. Zhang et al. explored the

ro
antioxidant effect of aqueous extract from P. tenuifolia Willd. seedlings and found that the aqueous
extract from P. tenuifolia Willd. seedlings significantly enhanced learning and memory abilities,
reduced MDA content, and improved SOD, catalase (CAT), GSH-Px, and total antioxidant capacity
-p
levels [108]. However, there was no in-depth study on the transformation pathways and regulatory
mechanisms of aqueous extract from P. tenuifolia Willd. seedlings in vivo. In addition, whether this
re
antioxidant effect is related to the abnormal cholinergic system in the brain and inflammatory
response in vivo remains to be further explored.
lP

4.6. Cardiovascular effect

In 2013, 3,4,5-trimethoxycinnamic acid reportedly showed exhibited antiarrhythmic activity in


na

rabbit ventricular myocytes by inhibiting the effect of the calcium channel [109]. Senegenin
treatment could exert apparent protective effects on myocardial ischemia reperfusion injury in rats.
ur

The mechanism involves the reduction of left ventricular end-diastolic blood pressure (LVEDP);
myocardial infarction area; creatine phosphokinase (CPK) and lactate dehydrogenase (LDH)
Jo

activities; TNF-α, IL-6, and iNOS levels; and high-mobility group box protein 1 (HMGB1)
expression; the mechanism also involves increasing the left ventricular systolic blood pressure
(LVSP) [110]. In conclusion, senegenin inhibited the expression of inflammatory cytokines, which
played a protective role in myocardial ischemia reperfusion injury in rats. When myocardial
ischemia reperfusion occurs, oxidative stress will be triggered. However, this study did not
research on the antioxidant effect of senegenin. In the future, we should also explore the
biochemical indexes such as SOD, GSH-PX, MDA to evaluate the protective effect of senegenin on
myocardial ischemia reperfusion injury in rats.

4.7. Other pharmacological effects

In 2017, an investigation reported that PTP-1 isolated from the PR polysaccharide exhibited
apparent hypoglycemic activity on the α-glucosidase enzyme with the half maximal inhibitory
concentration (IC50) of 0.52 mg/mL [111]. In addition, PR extracts treatment could significantly
inhibit lipid accumulation, attenuate obesity inflammation, as well as alter gut microbiota profiles
in 3T3-L1 adipocytes and high-fat diet-induced obese mice [112]. Moreover, PR is often used to treat
cough, and it requires processing before being prescribed in Chinese clinical practices. In the
Journal Pre-proof

ammonia-induced cough mouse model, raw PR, licorice-boiled PR, and honey-stir-baked PR
relieved cough and resolved phlegm; honey-stir-baked PR significantly reduced the frequency of
coughing in mice and promoted the secretion of phenol red in the respiratory tract [113,114].
At present, the pharmacological effects of PR are mainly focused on neuroprotective,
anti-depressant, cognitive-enhancing, hypnotic-sedative, anti-inflammatory and anti-tumor activity.
Studies showed that oligosaccharide esters and saponins were the main active components of PR.
However, the pharmacological research of PR still mainly focuses on the exploration of single or
multiple molecular mechanisms, and most of them only stay at the phenomenological observation
level. Due to insufficient understanding of the structure-activity relationship of active components
in PR, and the lack of in-depth exploration of the internal relationship between known drug targets,
a more satisfactory explanation for the indications and a more complete explanation of the
pharmacological mechanism have not been obtained. In future research, scholars should focus on
the research on the biomolecular networks of the neurological, immune, and endocrine systems
related to the main indications of PR. At the same time, the pharmacological mechanism of PR
should be studied in multi-level, multi-pathway and multi-target to reveal the potential links, so as

of
to provide a reference for the further development and utilization of the PR.

ro
5. Toxicology

PR exhibits an extensive range of biological activities; however, the gastrointestinal side effects
-p
of raw PR limit its application and development of new products. The potential toxicities and side
effects of PR are summarized in Table 8.
re
Pharmacological research has indicated that long-term and excessive doses of PR can
significantly inhibit gastrointestinal movement and cause gastrointestinal flatulence and intestinal
lP

wall thinning, as well as damage the gastric wall cell structure and intestinal structure [115]. An
acute toxicity study demonstrated that the median lethal dose (LD 50) of raw PR and total saponins
of PR in mice were 15.31 g/kg and 212 mg/kg, respectively [116]. In 2012, Guan et al. compared the
na

acute toxicity test of 60% ethanol and water extracts in different processed products of PR and
found that the toxicity of PR was directly proportional to the total saponin content [13]. One report
in 2011 indicated that raw PR and its total saponins could significantly reduce the interstitial cells of
ur

Cajal (ICC) in the myenteric plexus in the stomach and the small intestine, whereas
honey-stir-baked PR exerted no apparent effect. Therefore, the mechanism underlying the
Jo

gastrointestinal motility caused by raw PR and its total saponins could be related to the reduction in
gastrointestinal ICC, which could be protected by honey processing to a certain extent [117].
In the last several years, the toxicity of active components isolated from PR has been reported.
Wen et al. [14] noted that onjisaponin B (80 mg/L) could lead to the irregular and intense systole of
the isolated gut. Onjisaponin B, tenuifolin, or senegenin could markedly decrease the gastric PGE2
level at a dosage of 200 mg/kg, indicating that these saponins could lead to the loss of gastric
mucosal protection and ultimately result in gastric injury. The “dose-toxicity” relationship of
Yuanzhi-1 in mice (98.0, 89.1, 81.0, and 73.6 mg/kg via intraperitoneal injection for 2 weeks)
indicated that LD50 was 86.5 mg/kg, and the mortality rate of mice was dose-dependent. The side
effects of Yuanzhi-1, such as tremors, convulsion, salivation, and hyper responsiveness, were also
observed [117]. To determine the no-observed-adverse-effect level (NOAEL) and evaluate the
preclinical safety of PR, rats and beagle dogs were given oral extracts for 15 d in a study on acute
toxicity; rats received repeated oral administration of extracts for 90 d in a study on subchronic
toxicity. No treatment-related mortalities and significant changes in clinical parameters were
observed during the toxicity study. The PR root extract had a NOAEL of 1000 mg/kg/day for female
Journal Pre-proof

and male beagles [118]. Combining the results, we concluded that the saponins, alkaloids, and other
chemical components in PR could irritate the body. In clinical applications, PR is typically used in
processed products or combined with other compatible medicinal plants to reduce toxicity and
enhance efficacy. However, the mechanisms by which PR saponins caused toxicity could be
complex and need to be elucidated with further research.
Table 8
Toxicities and side effects of PR.
Extracts/ Animal/
Concentration/dose Toxic reactions Ref.
compounds cell line

Water extract of PR Mice p.o. LD50 = 19.3 g/kg Inflammatory infiltration in 115

submucosa and muscularis of

stomach and small intestine, blood

of
vessel dilatation and endothelial

swelling

ro
Total saponins Mice p.o. LD50 = 212 mg/kg Death 116

Water extract of PR Mice p.o. LD50 = 14.02 g/kg Death 13

13
Water extract of Mice
-p
p.o. LD50 = 13.94 g/kg Death

licorice-boiled PR

Water extract of Mice p.o. LD50 = 14.98 g/kg Death 13


re
honey-stir-baked PR

Ethanol extract of PR Mice p.o. LD50 = 3.80 g/kg Death 13


lP

Ethanol extract of Mice p.o. LD50 = 3.12 g/kg Death 13

licorice-boiled PR

Ethanol extract of Mice p.o. LD50 = 4.81 g/kg Death 13


na

honey-stir-baked PR

Water extract of PR SD rats 10 g/kg (p.o., 14 d) Decreasing interstitial cells of Cajal 117
ur

Total saponins SD rats 5 g/kg (p.o., 14 d) Decreasing interstitial cells of Cajal 117

Onjisaponin B Rabbit 20–40 mg/mL (in vitro, one Irregularing and intensing systole 14
Jo

ileum administration) of isolated gut

Onjisaponin B Male mice 100, 200 mg/kg (p.o., 30 min) Reducing the gastric 14

PGE2 level

Tenuifolin Male mice 100, 200 mg/kg (p.o., 30 min) Reducing the gastric 14

PGE2 level

Senegenin Male mice 100, 200 mg/kg (p.o., 30 min) Reducing the gastric 14

PGE2 level

Yuan zhi-1 Mice p.o. LD50 = 86.5 mg/kg Tremor, convulsion, salivation and 117

hyper

responsiveness.

Ethanol extract of PR Female 1000, 2000 mg/kg (p.o., 15 d) Vomiting, discoloration, or 118

beagle hemorrhage

dogs

6. Pharmacokinetics
Journal Pre-proof

Previous pharmacokinetic studies of PR have focused on oligosaccharide esters and saponins,


including its hydrolysates. Researchers analyzed the chemical and intestinal metabolic profiles of
PR have been analyzed, and glycosylation of intestinal flora has been identified as the main
metabolic pathway of 25 PR saponins. The metabolic pathways of 16 oligosaccharide esters were
oxygenation and cleavage of acetylene and HC. Three xanthone C-glycosides were transformed by
the cleavage of the Api group [120]. In addition, 26 and 33 saponins, which are poorly absorbed by
the body, were identified in urine and feces. These compounds excreted in feces interact with
intestinal microflora and play an important role in the treatment of neurodegenerative diseases
[121]. After oral administration of PR extracts (1.9 g/kg), ultrahigh-performance liquid
chromatography-tandem mass spectrometry was conducted to simultaneously determine the
pharmacokinetics of 7 components in rat plasma. The double-peak absorption of sibiricose A5,
sibiricose A6, tenuifoliside A, and tenuifoliside B was revealed in the mean plasma

of
concentration-time profiles. In addition, the area under the plasma concentration-time curve from
zero to last sampling time (AUC0–t) and the elimination half-life (T1/2) of tenuifoliside B (448.53 ±

ro
121.3 ng h/mL and 8.0 ± 2.1 h) were greater than those of tenuifoliside A (299.22 ± 134.2 ng h/mL
and 5.3 ± 2.2 h) and tenuifoliside C (47.24 ± 19.4 ng h/mL and 3.0 ± 2.1 h). The AUC0–t and peak
plasma concentration (Cmax) of 3,6′-disinapoyl sucrose were lower than those of sibiricose A5,
-p
sibiricose A6, tenuifoliside A, and tenuifoliside B. Notably, 3,4,5-trimethoxycinnamic acid rapidly
reached its peak plasma concentration approximately 0.1 ± 0.05 h after oral administration and
re
maintained a relatively slow elimination rate with T1/2 equal to 7.0 ± 2.9 h [122]. However, this study
did not explore in depth whether the double-peak absorption of sibiricose A5, sibiricose A6,
lP

tenuifoliside A, and tenuifoliside B was caused by the hepatoenteric circulation, presystematic


metabolism, or the mutual transformation between components. In future study, the
pharmacokinetic differences between the PR extracts and oral monomer compounds could be
na

investigated, and pharmacokinetic parameters of the protoglycosides and secondary glycosides or


aglycones in the polygraph could be compared.
ur

The bioavailability of sibiricaxanthone F was detected by liquid chromatography-tandem mass


spectrometry (LC-MS/MS). Sibiricaxanthone F administered once at 150 mg/kg had an AUC0–t of
Jo

49.86 ± 7.63 ng h/mL, a considerably low oral bioavailability with only 0.22%, and multiple peaks in
the mean plasma concentration-time curves [123]. In a similar pharmacokinetic study,
3,6′-disinapoyl sucrose was less absorbed in vivo, and its bioavailability was only 0.5% after an oral
dose of 50 mg/kg and an intravenous (i.v.) dose of 5 mg/kg [15]. 3,6′-disinapoyl sucrose is one of the
main components in PR and has various pharmacological properties. However, the bioavailability
of 3,6′-disinapoyl sucrose in vivo was very low. The fate of 3,6′-disinapoyl sucrose in the
gastrointestinal tract and the existence of its metabolites in plasma should be further study.
Sun et al. developed an online multiple reaction monitoring method to study the differences in
pharmacokinetic parameters of 9 components (polygalaxanthone III, sibiricose A5, sibiricose A6,
3,6′-disinapoyl sucrose, tenuifoliside A, tenuifoliside B, tenuifoliside C, sibiricaxanthone A, and
tenuifolin) in rat plasma after intragastric administration of single herbs PR and PR-Ginseng
combination. It was found that Tmax and T1/2 of other components except for the tenuifolin were
increased in PR-Ginseng group than in PR group, while Cmax was decreased. It was speculated that
some components in Ginseng and PR have competitive inhibition, which might be related to the
reduced toxicity of PR [123]. Future scholars should conduct in-depth research on it.
A sensitive LC-MS/MS method was developed to study the bioavailability of three PR saponin
Journal Pre-proof

hydrolysates, including tenuifolin, 3,4,5-trimethoxycinnamic acid, and p-methoxy cinnamic acid.


Subsequently, PR saponin hydrolysates were administered orally at 100 mg/kg and intravenously at
20 mg/kg. The three active components were quickly absorbed into the circulation, and all their T max
values were less than 25 min. Tenuifolin remained in vivo for a considerably longer period (T1/2, 4.8
± 1.6 h), compared with 3,4,5-trimethoxycinnamic acid (0.6 ± 0.2 h) and p-methoxy cinnamic acid
(0.9 ± 0.4 h). However, the AUC0–t and Cmax values of 3,4,5-trimethoxycinnamic acid and
p-methoxycinnamic acid were much larger than those of tenuifolin after oral administration, with
higher absolute bioavailability levels of 90.1% and 96.5%, respectively [16]. Tenuifolin remained in
the body for a long time, which could display biological activity longer. However, due to the low
oral bioavailability of tenuifolin, it will be a major concern to select appropriate drug dosage forms
and administration methods, such as intravenous injection and sublingual administration, to
improve its bioavailability.

of
Numerous studies have shown in recent years that in the pathology of AD, pharmacokinetic
processes are altered. In 2017, Wang et al. revealed the improved absorption of polygalaxanthone

ro
III, tenuifolin, and sibiricose A6, with 107%–126% increases in Cmax and 108%–218% increases in
AUC0–t in rats with AD after oral administration of the Kai Xin powder formula at 10 g/kg [124].
Reversed-phase HPLC was used to compare the pharmacokinetics of 3,6′-disinapoyl sucrosein the
-p
plasma of the rat model of AD that was orally administered with pure 3,6′-disinapoyl sucrose (48
mg/kg), PR water extracts (3.37 g/kg), and the Kai Xin powder formula (7.65 g/kg). The
re
pharmacokinetic results showed a higher absorption rate (larger AUC0–t and Cmax) and a lower
elimination rate (longer T1/2) in the group administered with the Kai Xin powder formula than any
lP

other groups; significant differences in pharmacokinetics were indicated. Simultaneously, the


AUC0–t values of 3,6′-disinapoyl sucrose in the pure crystal group and the PR water extracts group
were determined to be 52.42 ± 5.13, and 38.90 ± 1.73 mg min/L, respectively. Therefore, some
na

components of PR and its compatibility exerted significant effects on the pharmacokinetic


parameters of 3,6′-disinapoyl sucrose; moreover, PR could improve bioavailability by using its
ur

compatibility with other herbs [125].


Pharmacokinetic research involving PR is highly limited, particularly the study of xanthones
Jo

and saponins. Thus, further research on the distribution, absorption, metabolism, and excretion of
PR and its active components is necessary. In addition, the patients’ weight, age, renal function and
other physiological factors and disease conditions affect the absorption, distribution and
metabolism of PR in the body. The above factors should be taken into account in the design of the
drug administration scheme, so as to rationally use the drug and improve its safety and
effectiveness.

7. Conclusions and future perspectives

PR has been used widely used in Asian countries for a long time, and the past decades have
witnessed its successful application. This review summarizes current research development
regarding the traditional uses, phytochemistry, pharmacology, toxicity, and pharmacokinetics of
PR. More than 140 compounds have been isolated and identified from PR. Meanwhile, modern
pharmacological research has revealed significant pharmacological properties of PR, including its
protective effects on the nervous and cardiovascular systems, as well as its anti-inflammatory,
antioxidant, and antitumor activities. The improper use of PR can cause toxic reactions, such as sore
Journal Pre-proof

throat, vomiting, abdominal distension, and edema. Saponins are the main components in PR that
cause toxicity and side effects. In addition, processing or compatibility with other herbs can not
only reduce the toxicity and enhance the efficacy of PR but also promote the absorption of some
active components of PR in vivo. Regardless, several critical issues have yet to be addressed
regarding the need to further develop PR and to improve its clinical applications.
First, approximately 140 compounds have been isolated from P. tenuifolia Willd. and P. sibirica
L., and various biological activities have been found. Technical difficulties in obtaining and
purifying sufficient compounds, particularly saponins and xanthones, still present a challenge.
Their mechanisms of action and biological activities have yet to be elucidated. To address this
concern, biological chromatography, cell membrane chromatography, affinity ultrafiltration mass
spectrometry, and other strategies have to be combined to screen and identify more active
components in P. tenuifolia Willd. and P. sibirica L.

of
Second, saponins are both effective and toxic components of PR. Evaluating the effective
quantity, as opposed to the quantity that results in toxicity, and achieving a balance between the

ro
effectiveness and possible toxicity of saponins have to be further explored in future research.
Third, saponins comprise the main toxic components of PR; however, the associated toxic
compounds and the mechanism underlying the toxicity have not been clearly determined.
-p
Moreover, the mechanism by which its processing or compatibility reduces toxicity and increases
its efficiency remains poorly understood and thus requires further research.
re
Fourth, processing in TCM directly affects the quality of Chinese medicines, as well as the
safety and effectiveness of its clinical use. However, the current techniques of processing PR remain
lP

traditional with respect to control and lack objective technical parameters. The efficacy of PR
changes during processing, but the quality control of the processed items still depends on the
content determination index of the original medicinal materials. Consequently, the quality standard
na

index of processed products of PR lacks specificity. Therefore, the processing techniques require
further standardization, and the quality standard of PR has to be improved to realize its
ur

standardized production and establish quality guidelines for safety, stability, reliability, and
specificity.
Jo

Finally, the pharmacokinetics of PR was summarized and analyzed for the first time in this
study. It was found that the active components in the pharmacokinetics of PR are still limited, and
there are many medicinal components in PR, including the active components and their
metabolites, which are still unclear. Future scholars can consider analyzing the form of PR in the
body from the perspective of serum pharmacochemistry and in vivo metabolism, so as to
understand its pharmacokinetic process in vivo better. In addition, a
pharmacokinetics-pharmacodynamics model that conforms to the characteristics of TCM was
established to study the relationship between “administration time-drug concentration-drug effect”
of PR and its active components, which could elucidate the pharmacodynamic material basis of PR
and guide the rational use of drugs in clinical settings.

Funding: This research was funded by the National Key Research and Development Program of China (Grant
No. 2018YFC1707204) and the National Natural Science Foundation of China (Grant Nos. 81573606, 81973486,
and 81373968).

Conflicts of Interest: The authors declare no conflict of interest.


Journal Pre-proof

References

[1] H.J. Jiang, T. Liu, L. Li, Y. Zhao, L. Pei, J.C. Zhao, Predicting the potential distribution of Polygala tenuifolia
Willd. under climate change in China, PLoS One 11 (2016) e0163718,
https://dx.doi.org/10.1371/j.pone.0163718.
[2] T. Uchikura, H. Sugiwaki, M. Yoshimura, H. Mitsuhanshi, H. Fuchino, N. Kawahara, T. Hakamatsuka, H.
Amakura, Characterization of UV-sensitive marker constituents of Polygala root for TLC: Applications in
quality control of single crude drug extract preparations, Chem. Pharm. Bull. 66 (2018) 1174–1180,
https://dx.doi.org/10.1248/cpb.c18-00616.
[3] X.Y. Sun, F.Y. Sun, Shen Nong’s herbal classic, Commercial Press, Beijing, China, 1995.
[4] T. Gu, R.H. Wang, T. Wu, Z.H. Ke, H. Yang, D. Wang, Therapeutic effect on mild perimenopausal
depression treated with acupuncture at the “Thirteen Ghost Points” and Kai Xin powder, Chin. Acupunct.
Moxib. 40 (2020) 267–271, https://dx.doi.org/10.13703/j.0255-2930.20190308-0005.

of
[5] L. Zhang, Y.Y. Wang, J.B. Zhou, B. Ke, Y.B. Yang, J. Qin, Y.J. Huang, Effects and mechanisms of Dihuang
Yinzi decoction on the treatment of Alzheimer’s disease patients, China J. Tradit. Chin. Med. Pharm. 33

ro
(2018) 4948–4952.
[6] D. Lan, Y.H. Yan, X. Zhao, X.L. Wang, Making the past prescriptions “Guyin Decoction” to serve present
-p
disease, Clin. J. Tradit. Chin. Med. 27 (2015) 1409–1410, https://dx.doi.org/10.16448/j.cjtcm.2015.0527.
[7] H.L. Wang, J. Gao, D.N. Zhu, B.Y. Yu, Quality evaluation of Polygala japonica through simultaneous
re
determination of six bioactive triterpenoid saponins by HPLC-ELSD, J. Pharm. Biomed. Anal. 43 (2007)
1552–1556, https://dx.doi.org/10.1016/j.jpba.2006. 11.012.
lP

[8] S. Chen, L. Lin, S. Chen, D. Yang, J. Yang, Quantitative determination of nine xanthones in Polygala
Caudata and fingerprinting of Polygala L. by HPLC, J. Liq. Chromatogr. Relat. Technol. 28 (2005) 2953–2963,
https://dx.doi.org/10.1080/10826070500274588.
na

[9] H. Park, S. Kang, E. Nam, Y.H. Suh, K. Chang, The protective effects of PSM-04 against beta
amyloid-induced neurotoxicity in primary cortical neurons and an animal model of Alzheimer’s disease,
Front. Pharmacol. 10 (2019) 1–13, https://dx.doi.org/ 10.3389/fphar.2019.00002.
ur

[10] M.S. Miao, J.J. Shi, L. Guo, T. Wang, M. Bai, Effect of topical application of two Polygala tenuifolia species
decoction on guinea pig skin, Trop. J. Pharm. Res. 16 (2017) 75–81, https://dx.doi.org/10.4314/tjpr.v16i1.10.
Jo

[11] X.B. Yu, G.L. Liu, B. Zhu, K. Hao, F. Ling, G.X. Wang, In vitro immunocompetence of two compounds
isolated from Polygala tenuifolia and development of resistance against grass carp reovirus (GCRV) and
dactylogyrus intermedius in respective host, Fish Shellfish Immunol. 41 (2014) 1–8,
https://dx.doi.org/10.1016/j.fsi.2014.10.004.
[12] H.W. Li, S.Y. Lin, T.T. Qin, H.Y. Li, Z.Q. Ma, S.P. Ma, Senegenin exerts anti-depression effect in mice
induced by chronic un-predictable mild stress via inhibition of NF-κB regulating NLRP3 signal pathway,
Int. Immunopharmacol. 53 (2017) 24–32, https://dx.doi.org/10.1016/j.intimp. 2017.10.001.
[13] S.J. Guan, X.P. Yan, J.K. Lin, Study on acute toxicity test of different processed products of Radix Polygalae,
Chin. J. Integr. Tradit. Chin. West. Med. 32 (2012) 398–401, https://dx.doi.org/10.7661/CJIM.2012.3.398.
[14] L. Wen, N. Xia, P.P. Tang, Y. Hong, Z.Z. Wang, Y.J. Liu, Y.J. Liu, Y.J. Liu, X.Q. Li, The gastrointestinal
irritation of polygala saponins and its potential mechanism in vitro and in vivo, Biomed. Res. Int. 2015 (2015)
918048, https://dx.doi.org/10.1155/2015/918048.
[15] Y. Chen, X.M. Liu, R.L. Pan, G.N. Wang, Y. Fu, Q. Chang, An LC-MS/MS method for determination of
3,6′-disinapoyl sucrose in rat plasma and its application to a pharmacokinetic study, Biomed. Chromatogr.
23 (2009) 1326–1332, https://dx.doi.org/10.1002/bmc.1257.
Journal Pre-proof

[16] Q. Wang, B.X. Xiao, R.L. Pan, X.M. Liu, Y.H. Liao, L. Feng, F.R. Cao, An LC-MS/MS method for
simultaneous determination of three Polygala saponin hydrolysates in rat plasma and its application to a
pharmacokinetic study, J. Ethnopharmacol. 169 (2015) 401–406, https://dx.doi.org/10.1016/j.jep.2015.04.033.
[17] S.Z. Li, Compendium of Materia Medica, People’s Medical Publishing House, Beijing, 1982.
[18] G.Z. Wang, L. Chen, D.G. Wan, Y.P. Liu, J. Pei, Comparative study of pharmacodynamics on Radix
Polygalae by different processed methods, J. Chengdu Med. Coll. 6 (2011) 280–295,
https://dx.doi.org/1674-2257 (2011) 04-0280-04.
[19] W.F. Yang, J. Wang, L.N. Liu, W. Xiao, Study of Cajal interstitial cells in stomach and small intestine of rats
with crude, honey-stir-baked Radix Polygalae and its saponins, J. Chin. Med. Mate. 34 (2011) 33–16,
https://dx.doi.org/10.1159/000327835.
[20] Z.H. Zhang, L. Wen, H.Y. Chen, X.Y. Yu, X.H. Yang, Y.Y. Li, X.J. Li, Common combinations of Polygalae
Radix, Chin. J. Exp. Tradit. Med. Form. 22 (2016) 224–228,

of
https://dx.doi.org/10.13422/j.cnki.syfjx.2016120224.
[21] I.J. Shin, S.U. Son, H. Park, Y. Kim, S.H. Park, K. Swanberg, J. Shin, S.K. Ha, Y. Cho, S.Y. Bang, J.H. Lew,

ro
S.H. Cho, S. Maeng, Preclinical evidence of rapid-onset antidepressant-like effect in Radix Polygalae
extract, PLoS One 9 (2014) e88617, https://dx.doi.org/10.1371/journal.pone.0088617.
[22] W. Sheng, Y. Wang, J.B. Li, H.S. Xu, Clinical and basic research on Renshen Yangrong Decoction, Front.
-p
Nutr. 6 (2019) 1-5, https://dx.doi.org/10.3389/fnut.2019.00175.
[23] Q.Y. Xiao, X.L. Wang, D.M. Qi, X.R. Cheng, Qifu Yin and its modified prescriptions in treating dementia:
re
research progress, Mil. Med. Sci. 43 (2019) 391–396, https://dx.doi.org/10.7644/j.issn.1674-9960.2019.05.015.
[24] Y.R. Cui, S.H. Tang, X. Liu, D. Shen, H.J. Yang, Analysis on Chinese patent drugs rules of palpitations
lP

accompanied by insomnia based on data mining, Chin. J. Tradit Chin. Med. 30 (2015) 1792–1799.
[25] L.L. Han, Z. Li, R.W. Guan, H.B. Lin, J.Q. Lin, J.Q. Lin, Progress in studies on medicinal crop of Polygala
tenuifolia, Chin. Wild Plant Resour. 29 (2010) 1–4, https://dx.doi.org/1006-9690 (2010) 06-0001-04.
na

[26] S.W. Pelletier, S. Nakamura, A prosapogenin from Polygala senega and Polygala tenuifolia, Tetrahedron Lett.
52 (1967) 5303–5306, https://dx.doi.org/10.1016/s0040-4039 (01) 89666-0.
ur

[27] S. Sakuma, J. Shoji, Studies on the constituents of the root of Polygala tenuifolia Willd. Ⅰ: Isolation of
saponins and the structures of onjisaponins G and F, Chem. Pharm. Bull. 29 (1981) 2431–2433,
Jo

https://dx.doi.org/10.1248/cpb.29.2431.
[28] S. Sakuma, J. Shoji, Studies on the constituents of the root of Polygala tenuifolia Willd. Ⅱ: Isolation of
saponins and the structures of onjisaponins A, B and E, Chem. Pharm. Bull. 30 (1982) 810–821,
https://dx.doi.org/10.1248/cpb.30.810.
[29] J.Y. Liu, X.D. Yang, J.M. He, M. Xia, L.Z. Xu, S.L. Yang, Structure analysis of triterpene saponins in Polygala
tenuifolia by electrospray ionization ion trap multiple-stage mass spectrometry, J. Mass Spectrom. 42 (2007)
861–873, https://dx.doi.org/10.1002/jms.1210.
[30] J. Li, Y. Jiang, P.F. Tu, New acylated triterpene saponins from Polygala tenuifolia Willd., J. Asian. Nat. Prod.
Res. 8 (2006) 499–503, https://dx.doi.org/10.1080/10286020500173358.
[31] C.J. Li, J.Z. Yang, S.S. Yu, N.H. Chen, J.F. Hu, D.M. Zhang, Triterpenoid saponins with neuroprotective
effects from the roots of Polygala tenuifolia, Planta. Med. 74 (2008) 133–141,
https://dx.doi.org/10.1055/s-2008-1034296.
[32] C.J. Li, J.Z. Yan, S.S. Yu, N.H. Chen, J.F. Hu, D.M. Zhang, Triterpenoid saponins and oligosaccharides from
the roots of Polygala tenuifolia, Chin. J. Nat. Med. 9 (2011) 0321–0328,
https://dx.doi.org/10.3724/SP.J.1009.2011.00321.
[33] Y. Ling, Z.X. Li, M.C. Chen, Z.L. Sun, M.S. Fan, C.G. Huang, Analysis of multiple constituents in
Journal Pre-proof

Cong-Ming-Tang, a Chinese herbal formula for the treatment of amnesia, by high-performance liquid
chromatography with quadrupole time-of-flight mass spectrometry, Phytochem. Anal. 24 (2013) 677–688,
https://dx.doi.org/10.1002/pca.2454.
[34] G.F. Feng, S. Liu, Z.F. Pi, F.R. Song, Z.Q. Liu, Comprehensive characterization of in vivo metabolic profile of
Radix Polygalae based on ultra-high-performance liquid chromatography-tandem mass spectrometry, J.
Pharm. Biomed. Anal. 165 (2019) 173–181, https://dx.doi.org/10.1016/j.jpba.2018.12.005.
[35] Y.L. Song, K.W. Zeng, T.X. Shi, Y. Jiang, P.F. Tu, Sibiricasaponins A–E, five new triterpenoid saponins from
the aerial parts of Polygala sibirica L., Fitoterapia 84 (2013) 295–301,
https://dx.doi.org/10.1016/j.fitote.2012.12.017.
[36] Y. Ikeya, K. Sugama, M. Okada, H. Mitsuhashi, Two xanthones from Polygala tenuifolia, Phytochemistry 30
(1991) 2061–2065, https://dx.doi.org/10.1016/0031-9422(91)85067-A.
[37] T. Fujita, D.Y. Liu, S. Ueda, Y. Takeda, Xanthones from Polygala tenuifolia, Phytochemistry 31 (1992) 3997–

of
4000, https://dx.doi.org/10.1016/s0031-9422(00)97571-x.
[38] Y. Ikeya, K. Sugama, M. Maruno, Xanthone C-glycoside and acylated sugar from Polygala tenuifolia, Chem.

ro
Pharm. Bull. 42 (1994) 2305–2308, https://dx.doi.org/10.1248/cpb.42.2305.
[39] Chinese Pharmacopoeia Commission, Pharmacopoeia of the People’s Republic of China Part I, People’s
Medical Publishing House, Beijing, 2015.
-p
[40] T. Miyase, H. Noguchi, X.M. Chen, Sucrose esters and xanthone C-glycosides from the roots of Polygala
sibirica, J. Nat. Prod. 62 (1999) 993–996, https://dx.doi.org/10.1021/np990084t.
re
[41] Y. Jiang, P.F. Tu, Xanthone O-glycosides from Polygala tenuifolia, Phytochemistry 60 (2002) 813-816,
https://dx.doi.org/10.1016/s0031-9422(02)00184-x.
lP

[42] Y. Jiang, P.F. Tu, Xanthone glycosides from Polygala tenuifolia and their conformational analyses, J. Nat.
Prod. 68 (2005) 875–879, https://dx.doi.org/10.1021/np050026+.
[43] Y.H. Zhou, S.Y. Zhang, Q. Guo, X.Y. Chai, Y. Jiang, P.F. Tu, Chemical investigation of the roots of Polygala
na

sibirica L., Chin. J. Nat. Med. 12 (2014) 225–228, https://dx.doi.org/10.1016/S1875-5364(14)60038-8.


[44] L.L. Xu, C.J. Li, J.Z. Yang, Y.M. Luo, D.M. Zhang, Chemical constituents of Polygala tenuifolia root, J. Chin.
ur

Med. Mater. 37 (2014) 1594–1596, https://dx.doi.org/10.13863/j.issn1001-4454.2014.09.022.


[45] Y.S. Zhao, Z.Y. Yan, Z.L. Li, The study on structure features and pharmacological activity of active
Jo

materials of Polygala, Chin. Arch. Tradit. Chin. Med. 8 (2005) 1420-1423.


https://dx.doi.org/10.13193/j.archtcm.2005.08.69.zhaoysh.029.
[46] Y.L. Song, Y. Jiang, S.X. Zhou, P.F. Tu, Study on the chemical composition of the aboveground part of
Polygala sibirica L. Chin. Tradit. Herb Drugs 1 (2010) 27-29.
[47] Y.L. Song, Y. Jiang, S.X. Zhou, D. Bi, P.F. Tu, Study on the ketone components in the aboveground part of
Polygala sibirica L., China J. Chin. Mater. Med. 5 (2009) 574-576.
[48] X.J. Yang, L.J. Zhang, B. Liang, S.L. Yang, Oligosaccharide esters isolated from plants of polygonaceae,
Chin. Tradit. Herb. Med. 33 (2002) 954–958, https://dx.doi.org/CNKI:SUN:ZCYO.0.2002-10-042.
[49] Y. Ikeya, K. Sugama, M. Okada, H. Mitsuhashi, Four new phenolic glycosides from Polygala tenuifolia,
Chem. Pharm. Bull. 39 (1991) 2600–2605, https://dx.doi.org/10.1248/cpb.39.2600.
[50] T. Miyase, A. Ueno, Sucrose derivative from Polygala tenuifolia, Jpn. J. Pharm. 47 (1993) 267–278.
[51] T. Miyase, Y. Iwata, A. Ueno, Tenuifolioses A–F, oligosaccharide multiesters from the roots of Polygala
tenuifoia, Chem. Pham. Bull. 39 (1991) 3082–3084, https://dx.doi.org/10.1248/cpb.39.3082 .
[52] T. Miyase, Y. Iwata, A. Ueno, Tenuifolioses G–P oligosaccharide multiester from the root of Polygala
tenuifolia, Chem. Pham. Bull. 40 (1992) 2741–2748, https://dx.doi.org/10.1248/cpb.40.2741.
[53] Y. Jiang, P.F. Tu, Tenuifoliose Q a new oligosaccharide ester from the root of Polygala tenuifolia, J. Asian.
Journal Pre-proof

Nat. Prod. Res. 5 (2003) 279–283, https://dx.doi.org/10.1080/1028602031000111987.


[54] T. Miyase, H. Noguchi, X.M. Chen, Sucrose esters and xanthone C-glycosides from the roots of Polygala
sibirica, J. Nat. Prod. 62 (1999) 993–996, https://dx.doi.org/10.1021/np990084t.
[55] B.Y. Jin, Z.Y. Piao, Study on the alkaloid constituents of Polygala tenuifolia, China J. Chin. Mater. Med. 11
(1993) 675–677, https://dx.doi.org/CNKI:SUN:ZGZY.0.1993-11-020.
[56] D.J. Moura, C. Rorig, D.L. Vieira, J.A. Henriques, R. Roesler, J. Saffi, J.M. Boeira, Effects of β-carboline
alkaloids on the object recognition task in mice, Life Sci. 79 (2006) 2099-2104,
https://dx.doi.org/10.1016/j.lfs.2006.07.004.
[57] M.H. Song, P. Wu, X.L. Zhang, H.F. Li, J.T. Liu, Y. Meng, J.X. Wang, X. Zhao, HPLC comparison of 8
organic acids in 3 processed products of Polygala tenuifolia, Chin. Tradit. Pat. Med. 38 (2016) 1565–1569,
https://dx.doi.org/10.3969/j.issn.1001-1528.2016.07.027.
[58] Y.Y. Jiang, Y.Y. Duan, Y. Liu, M. Fang, R.B. Shi, Isolation and structure identification of chemical

of
constituent of Polygala tenuifolia, J. Beijing Univ. Tradit. Chin. Med. 34 (2011) 122–125,
https://dx.doi.org/CNKI: SUN: JZYB.0.2011-02-014.

ro
[59] Z.J. Wu, Analysis of the volatile oil components of Polygala tenuifolia Willd. by GC-MS, Med. Plant 38 (2010)
4562–4574, https://dx.doi.org/CNKI:SUN:AHNY. 0.2010-09-057.
[60] T. Shi, Y.G. Li, Y. Jiang, Isolation of flavonoids from the aerial parts of Polygala tenuifolia Willd. and their
-p
antioxidant activities, Chin. Pham. J. 22 (2013) 36–39, https://dx.doi.org/10.5246/jcps.2013.01.004.
[61] GBD 2016 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national
re
burden of neurological disorders, 1990-2016: a systematic analysis for the global burden of disease study
2016, Lancet Neurol. 18 (2019) 459–480, https://dx.doi.org/10.1016/S1474–4422(18)30499-X.
lP

[62] R. Jesky, H. Chen, The neuritogenic and neuroprotective potential of senegenin against Aβ-induced
neurotoxicity in PC12 cells, BMC Complement Altern. Med. 16 (2016) 26,
https://dx.doi.org/10.1186/s12906-016-1006-3.
na

[63] D.Q. Liao, L.C. Zhang, Y. Lan, J.L. Wang, J.P. Zhang, Protective effect of senegenin on the injury of rat
hippocampal neural stem cells induced by oxygen-glucose deprivation/reoxygenation culture in vitro,
ur

China J. Tradit. Chin Med. 34 (2019) 2990–2993.


[64] F. Peng, L. Lu, F. Wei, D. Wu, K. Wang, J. Tang, The onjisaponin B metabolite tenuifolin ameliorates
Jo

dopaminergic neurodegeneration in a mouse model of Parkinson’s disease, Neuroreport. 31 (2020) 456-465.


http://dx.doi.org/10.1097/WNR.0000000000001428.
[65] S. Chen, J. Jia, Tenuifolin attenuates amyloid-β42-induced neuroinflammation in microglia through the
NF-κB signaling pathway, J Alzheimers Dis. 76 (2020) 195-205. http://dx.doi.org/10.3233/JAD-200077.
[66] H.G. Yang, W.J. Xu, W. Zhao, M.L. Gu, W. Wang, 1,3,7-Trihydroxyxanthone, derived from Polygalae
Radix, a herbal medicine, stimulates the expression of neurotrophic factors in rat astrocyte primary
cultures via cAMP- and ERK-dependent pathways, Biome. Pharmacother. 98 (2018) 762–768,
https://dx.doi.org/10.1016/j.biopha.2017.12.085.
[67] X. Liu, X. Wang, J. Lu, Tenuifoliside A promotes neurite outgrowth in PC12 cells via the PI3K/AKT and
MEK/ERK/CREB signaling pathways, Mol. Med. Rep. 12 (2015) 7637–7642, https://dx.doi.org/10.3892/mmr.
2015.4397.
[68] X. Liu, D.X. Wang, R.Q. Zhao, X.Z. Dong, Y. Hu, P. Liu, Synergistic neuroprotective effects of two herbal
ingredients via CREB-dependent pathway, Front. Pharmacol. 7 (2016) 337,
https://dx.doi.org/10.3389/fphar.2016.00337.
[69] Y. Zhou, M. Yan, R. Pan, Z. Wang, X. Tao, C. Li, T. Xia, X. Liu, Q. Chang, Radix Polygalae extract exerts
antidepressant effects in behavioral despair mice and chronic restraint stress-induced rats probably by
Journal Pre-proof

promoting autophagy and inhibiting neuroinflammation, J Ethnopharmacol 265 (2020) 113317,


https://dx.doi.org/10.1016/j.jep.2020.113317.
[70] H.W. Li, S.Y. Lin, T.T. Qin, H.Y. Li, Z.Q. Ma, S.P. Ma, Senegenin exerts anti-depression effect in mice
induced by chronic un-predictable mild stress via inhibition of NF-κB regulating NLRP3 signal pathway,
Int. Immunopharmacol. 53 (2017) 24–32, https://dx.doi.org/10.1016/ j.intimp. 2017.10.001.
[71] L.Q. Wang, S.Z. Zhu, D.M. Li, Effect of tenuifolin on mental behavior in depression model mice, Chin. J.
public health 17 (2019) 3710–3713, https://dx.doi.org/10.11847/zgggws1122400.
[72] Y.H. Leem, S. Oh, 3,4,5-Trimethoxycinnamin acid ameliorates restraint stress-induced anxiety and
depression, Neurosci. Lett. 585 (2015) 54–59, https://dx.doi.org/10.1016/j.neulet. 2014.11.016.
[73] P. Liu, D.X. Wang, D.H. Guo, H.H. Tu, L. Ma, T.T. Xie, L.Y. Kong, Antidepressant effect of 3, 6′-disinapoyl
sucrose from Polygala tenuifolia Willd. in pharmacological depression model, Chin. Pharm. J. 43 (2008)
1391–1394, https://dx.doi.org/1001-2494(2008)18-1391-04.

of
[74] Y. Hu, H.B. Liao, P. Liu, K. Rahman, A bioactive compound from Polygala tenuifolia regulates efficiency of
chronic stress on hypothalamic-pituitary-adrenal axis, Pharmazie 64 (2009) 605–608,

ro
https://dx.doi.org/10.1691/ph.2009.9580.
[75] Y. Hu, M. Liu, P. Liu, D.H. Guo, R.B. Wei, K. Rahman, Possible mechanism of the antidepressant effect of
3,6′-disinapoyl sucrose from Polygala tenuifolia Willd., J. Pharm. Pharmacol. 63 (2011) 869–874,
-p
https://dx.doi.org/10.1111/j.2042-7158.2011.01281.x.
[76] Y. Hu, H.B. Liao, D.H. Guo, P. Liu, Y.Y. Wang, K. Rahman, Antidepressant-like effects of 3,6′-disinapoyl
re
sucrose on hippocampal neuronal plasticity and neurotrophic signal pathway in chronically mild stressed
rats, Neurochem. Int. 56 (2010) 461–465, https://dx.doi.org/10.1016/j.neuint.2009.12.004.
lP

[77] P. Liu, Y. Hu, D.H. Guo, D.X. Wang, H.H. Tu, L. Ma, T.T. Xie, L.Y. Kong, Potential antidepressant
properties of Radix Polygalae (Yuan Zhi), Phytomedicine 17 (2010) 794–799,
https://dx.doi.org/10.1016/j.phymed.2010. 01.004.
na

[78] D. Zhang, X. Wang, R. Li, Lin. Wang, Z. Zhou, Q. Fu, Y.J. Cao, M.F. Fang, Extract of the aerial part of
Polygala tenuifolia attenuates d-Galactose/NaNO2-induced learning and memory impairment in mice,
ur

Planta. Med. (2020), https://dx.doi.org/10.1055/a-1212-3212.


[79] T. Kuboyama, K. Hirotsu, T. Arai, H. Yamasaki, C. Tohda, Polygalae Radix extract prevents axonal
Jo

degeneration and memory deficits in a transgenic mouse model of Alzheimer’s disease, Front. Pharmacol.
8 (2017) 805, https://dx.doi.org/10.3389/fphar.2017.00805.
[80] X.H. Li, J. Cui, Y. Yu, W. Li, Y.J. Hou, X. Wang, D.P. Qin, C. Zhao, X.S. Yao, J. Zhao, G. Pei, Traditional
Chinese nootropic medicine Radix Polygalae and its active constituent onjisaponin B reduce β-amyloid
production and improve cognitive impairments, PLoS One 11 (2016) e0151147,
https://dx.doi.org/10.1371/journal.pone. 0151147.
[81] H. Zhou, W. Xue, S.F. Chu, Z.Z. Wang, C.J. Li, Y.N. Jiang, L.M. Luo, P. Luo, G. Li, D.M. Zhang, N.H. Chen,
Polygalasaponin XXXII, a triterpenoid saponin from Polygalae Radix, attenuates scopolamine-induced
cognitive impairments in mice, Acta Pharmacol. Sin. 37 (2016) 1045–1053,
https://dx.doi.org/10.1038/aps.2016.17.
[82] L. Wang, G.F. Jin, H.H. Yu, X.H. Lu, Z.H. Zou, J.Q. Liang, H. Yang, Protective effects of tenuifolin isolated
from Polygala tenuifolia Willd. roots on neuronal apoptosis and learning and memory deficits in mice with
Alzheimer's disease, Food Funct. 10 (2019) 7453-7460, https://dx.doi.org/10.1039/c9fo00994a.
[83] W. Jing, Y. Li, B. Ma, X.Y. Li, Effect of tenuigenin on the expression of nAChRα7 in epileptic model rats,
Chin. J Integr. Med. Cardio/Cerebrovasc Dis. 15 (2017) 3118–3120,
https://dx.doi.org/10.3969/j.issn.1672-1349.2017.24.009.
Journal Pre-proof

[84] P. Xu, S.P. Xu, K.Z. Wang, C. Lu, H.X. Zhang, R.L. Pan, C. Qi, Y.Y. Yang, Y.H. Li, X.M. Liu,
Cognitive-enhancing effects of hydrolysate of polygala saponin in SAMP8 mice, J. Zhejiang Univ. 17 (2016)
503–514, https://dx.doi.org/10.1631/jzus.B1500321.
[85] C.R. Guo, J.Y. Shen, Z.Q. Meng, X.L. Yang, F. Li, Neuroprotective effects of polygalacic acid on
scopolamine-induced memory deficits in mice, Phytomedicine 23 (2016) 149–155,
https://dx.doi.org/10.1016/j.phymed.2015.12.009.
[86] F.P. Chen, M.S. Jong, Y.C. Chen, Y.Y. Kung, F.J. Chen, S.J. Hwang, Prescriptions of Chinese herbal
medicines for insomnia in Taiwan during 2002, J. Evidence-Based Complement Altern. Med. 2011 (2011) 1–
9, https://dx.doi.org/10.1093/ecam/nep018.
[87] L.H. Chen, D.P. Xu, J.M. Kong, M.H. Yao, D. Zhang, L. Gu, J.Q. Gao, Separation and identification of the
hypnotic active ingredients of Polygala tenuifolia Willd., Food & Machinery. 34 (2018) 119–122,
https://dx.doi.org/10.13652/j.issn.1003-5788.2018.08.024.

of
[88] C.I. Lee, J.Y. Han, J.T. Hong, K.W. Oh, 3,4,5-Trimethoxycinnamic acid (TMCA), one of the constituents of
Polygalae Radix enhances pentobarbital-induced sleeping behaviors via GABAAergic systems in mice,

ro
Arch. Pharm. Res. 36 (2013) 1244–1251, https://dx.doi.org/10.1007/s12272-013-0167-6.
[89] C.Y. Chen, X.D Wei, C.R. Chen, 3,4,5-Trimethoxycinnamic acid, one of the constituents of Polygalae Radix
exerts anti-seizure effects by modulating GABAAergic systems in mice, J. Pharmacol. Sci. 131 (2015) 1–5,
-p
https://dx.doi.org/10.1016/j.jphs.2015.07.021.
[90] Z.F. Zhao, Y.J. Bai, J. Xie, X.F. Chen, X.R. He, Y. Sun, Y.J. Bai, Y.Y. Zhang, S.P. Wu, X.H. Zheng, Excavating
re
precursors from the traditional Chinese herb Polygala tenuifolia and Gastrodia elata: Synthesis,
anticonvulsant activity evaluation of 3,4,5-trimethoxycinnamic acid (TMCA) ester derivatives, Bioorg.
lP

Chem. 88 (2019) 102832, https://dx.doi.org/10.1016/j.bioorg.2019.03.006.


[91] Q. Cao, Y. Jiang, S.Y. Cui, P.F. Tu, Y.M. Chen, X.L. Ma, X.Y. Cui, Y.L. Huang, H. Ding, J.Z. Song, B. Yu, Z.F.
Sheng, Z.J. Wang, Y P. Xu, G. Yang, H. Ye, X. Hu, Y.H. Zhang, Tenuifolin, a saponin derived from Radix
na

Polygalae, exhibits sleep-enhancing effects in mice, Phytomedicine 23 (2016) 1797–1805,


https://dx.doi.org/10.1016/j.phymed.2016.10.015.
ur

[92] Z.W. Chen, C.B. Peng, Z. Pei, M.R. Zhang, T.C. Yun, Z.M. Yang, F.P. Xu, Effects of tenuifolin on rest/wake
behavior in zebrafish, Exp. Ther. Med. 19 (2020) 2326–2334, https://dx.doi.org/10.3892/etm.2020.8476.
Jo

[93] K.S. Kim, D.S. Lee, G.S. Bae, S.J. Park, D.G. Kang, H.S. Lee, H. Oh, Y.C. Kim, The inhibition of JNK MAPK
and NF-κB signaling by tenuifoliside A isolated from Polygala tenuifolia in lipopolysaccharide-induced
macrophages is associated with its anti-inflammatory effect, Eur. J. Pharmacol. 721 (2013) 267–276,
https://dx.doi.org/10.1016/j.ejphar.2013.09.026.
[94] K. Xu, C.Y. Ma, L.H. Xu, J.S. Ran, L.F. Jiang, Y.Z. He, S.A. Moqbel, Z. Wang, L.D. Wu, Polygalacic acid
inhibits MMPs expression and osteoarthritis via Wnt/β-catenin and MAPK signal pathways suppression,
Int. Immunopharmacol. 63 (2018) 246–252, https://dx.doi.org/10.1016/j.intimp.2018.08.013.
[95] H.M. Lv, W.Z. Ren, Y.W. Zheng, L.D. Wang, G.J. Lu, P.F. Yi, X.X. Ci, Tenuigenin exhibits anti-inflammatory
activity via inhibiting MAPK and NF-κB and inducing Nrf2/HO-1 signaling in macrophages, Food Funct.
10 (2016) 355–363, https://dx.doi.org/10.1039/c5fo00807g.
[96] C.L. Wang, L.H. Zeng, T. Zhang, J.K. Liu, W.B. Wang, Tenuigenin prevents IL-1β-induced inflammation in
human osteoarthritis chondrocytes by suppressing PI3K/AKT/NF-κB signaling pathway, Inflammation 39
(2016) 807–812, https://dx.doi.org/10.1007/s10753-016-0309-3.
[97] R. Jia, H.G. Zhang, W.J. Zhang, H. Zhao, C.J. Zha, Y.H. Liu, Protective effects of tenuigenin on
lipopolysaccharide and D-galactosamine-induced acute liver injury, Microb. Pathog. 112 (2017) 83–88,
https://dx.doi.org/10.1016/j.micpath.2017.09.051.
Journal Pre-proof

[98] Z. Fan, Z.G. Liang, H. Yang, Y.T. Pan, Y. Zheng, X.M. Wang, Tenuigenin protects dopaminergic neurons
from inflammation via suppressing NLRP3 inflammasome activation in microglia, J. Neuroinflammation
14 (2017) 256, https://dx.doi.org/10.1186/s12974-017-1036-x.
[99] B. Yu, J.T. Qiao, Y.B. Shen, L.Y. Li, Protective effects of tenuigenin on staphylococcus aureus-induced
pneumonia in mice, Microb. Pathog. 110 (2017) 385–389, https://dx.doi.org/10.1016/j.micpath.2017.07.023.
[100] L. Lu, X.R. Li, P.X. Xu, Y. Zheng, X.M. Wang, Tenuigenin down-regulates the release of nitric oxide, matrix
metalloproteinase-9 and cytokines from lipopolysaccharide-stimulated microglia, Neurosci. Lett. 650
(2017) 82–88, https://dx.doi.org/10.1016/j.neulet.2017.04.001.
[101] X.B. Yu, G.L. Liu, B. Zhu, K. Hao, F. Ling, G.X. Wang, In vitro immunocompetence of two compounds
isolated from Polygala tenuifolia and development of resistance against grass carp reovirus (GCRV) and
Dactylogyrus intermedius in respective host, Fish Shellfish Immunol. 41 (2014) 541–548,
https://dx.doi.org/10.1016/j.fsi.2014.10.004.

of
[102] F.B. Zhang, X.W. Song, L. Li, J.F. Wang, L.Y. Lin, C. Li, H.T. Li, Y.J. Lv, Y.H. Jin, Y. Liu, Y. Hu, T. Xin,
Polygala tenuifolia polysaccharide PTP induced apoptosis in ovarian cancer cells via a mitochondrial

ro
pathway, Tumor. Biol. 36 (2015) 2913–2919, https://dx.doi.org/10.1007/s13277-014-2921-x.
[103] H. Yao, P. Cui, D. Xu, Y.D. Liu, Q.H. Tian, F.B. Zhang, A water-soluble polysaccharide from the roots of
Polygala tenuifolia suppresses ovarian tumor growth and angiogenesis in vivo, Int. J. Biol. Macromol. 107
-p
(2018) 713–718, https://dx.doi.org/10.1016/j.ijbiomac.2017.09.043.
[104] F.B. Zhang, X.W. Song, L. Li, J.F. Wang, L.Y. Lin, C. Li, H.T. Li, Y.J. Lv, Y.H. Jin, Y. Liu, Y. Hu, T. Xin,
re
Polygala tenuifolia polysaccharide (PTP) inhibits cell proliferation by repressing Bmi-1 expression and
downregulating telomerase activity, Tumor. Biol. 36 (2015) 2907–2912,
lP

https://dx.doi.org/10.1007/s13277-014-2920-y.
[105] Y. Bian, H. Zeng, H. Tao, L. Huang, Z. Du, J. Wang, K. Ding, A pectin-like polysaccharide from Polygala
tenuifolia inhibits pancreatic cancer cell growth in vitro and in vivo by inducing apoptosis and suppressing
na

autophagy, Int. J. Biol. Macromol. 162 (2020) 107-115. https://doi.org/10.1016/j.ijbiomac.2020.06.054 .


[106] P. Liu, Y. Hu, D.H. Guo, B.R. Lu, K. Raham, L.H. Mu, D.X. Wang, Antioxidant activity of oligosaccharide
ur

ester extracted from Polygala tenuifolia roots in senescence-accelerated mice, Pharm. Biol. 48 (2010) 828–833,
https://dx.doi.org/10.3109/13880200903283707.
Jo

[107] F.F. Xie, Effects of anti-fatigue in vivo and anti-oxidant in vitro of polysaccharides from Polygala tenuifolia
Willd. on exhaustive exercise mice, Science and Technology of Food Industry 16 (2020) 1-11,
https://doi.org/10.13386/j.issn1002-0306.2020050292.
[108] D. Zhang, Y. Wang, X. Wang, L. Wang, R. Li, M.F. Fang, Antioxidant effect of aqueous extract of Polygala
tenuifolia Willd. seedlings on D-galactose aging mice, China J. Chin. Mater. Med. 34 (2019) 4322–4326.
[109] Z.H. Zhao, M.F. Fang, D.D. Xiao, M. Liu, N. Fefelova, C. Huang, W.J. Zang, L.H. Xie, Potential
antiarrhythmic effect of methyl 3,4,5-trimethoxycinnamate, a bioactive substance from roots of Polygalae
Radix: suppression of triggered activities in rabbit myocytes, Biol. Pharm. Bull. 36 (2013) 238–244,
https://dx.doi.org/10.1248/bpb.b12-00654.
[110] H.Y. Li, Y.F. Chi, Effects of senegenin pretreatment on myocardial ischemia-reperfusion injury and HMGB
1 expression in rats. Chin. Tradit. Pat. Med. 41 (2019) 1165–1168.
https://dx.doi.org/0.3969/j.issn.1001-1528.2019.05.044.
[111] Y.S. Jing, D.S. Zhang, L.F. Wu, Y.G. Zheng, Purification and structural characterization of polysaccharide
from Polygala tenuifolia and its biological activity, Food Science 38 (2017) 126–131,
https://dx.doi.org/10.7506/spkx1002-6630-201717021.
[112] C.C. Wang, J.H. Yen, Y.C. Cheng, C.Y. Lin, C.T. Hsieh, R.J. Gau, S.J. Chiou, H.Y. Chang, Polygala tenuifolia
Journal Pre-proof

extract inhibits lipid accumulation in 3T3-L1 adipocytes and high-fat diet-induced obese mouse model and
affects hepatic transcriptome and gut microbiota profiles, Food Nutr. Res. 61 (2017) 1379861,
https://dx.doi.org/10.1080/16546628.2017.1379861.
[113] X.W. Liu, H.H. Wu, J. Wang, H.P. Zhao, H. Yian, Contrast research on preventing cough and eliminating
phlegm actions between various honey-stir-baked and crude Radix Polygalae, Lishizhen Med. Mater. Med.
Res. 12 (2006) 2379–2380, https://dx.doi.org/1008-0805 (2006) 12-2379-02.
[114] G.Z. Wang, L. Chen, D.G. Wang, Y.P. Liu, J. Pei, Comparative study of pharmacodynamics on Radix
Polygalae by different processed methods, J. Chengdu Med. Coll. 6 (2011) 280–295,
https://dx.doi.org/1674-2257 (2011) 04-0280-04.
[115] R. Wang, T. Wu, Y. Liu, M.X. Yu, X.J. Tao, H.B. Wang, The effect of Radix Polygala and honey stir-baking
Radix Polygala on acute gastrointestinal toxicity of mice, Chin. Med. Mod. Dis. Educ. China 16 (2018) 88–
90. https://dx.doi.org/10.3969/j.issn.1672-2779.2018.08.037.

of
[116] J. Wang, H.H. Wu, Y. Wu, H.Z. Bao, A comparative study on acute toxicity of Radix Polygalae and its total
saponins with honey-stir-baked Radix polygalae, Pharmacolo. Clin. Chin. Mater. Med. 21 (2004) 120–122,

ro
https://dx.doi.org/10.13412/j.cnki.zyyl. 2004.06.013.
[117] Z.L. Jin, N. Gao, J.R. Zhang, X.R. Li, H.X. Chen, J. Xiong, Y.F. Li, Y. Tang, The discovery of Yuanzhi-1, a
triterpenoid saponin derived from the traditional Chinese medicine, has antidepressant-like activity, Prog.
-p
Neuro-Psychopharmacol. Biol. Psychiatry 53 (2014) 9–14, https://dx.doi.org/10.1016/j. pnpbp.2014.02.013.
[118] K.Y. Shin, B.Y. Won, H.J. Ha, Y.S. Yun, H.G. Lee, Preclinical safety of the root extract of Polygala tenuifolia
re
Willdenow in Sprague-Dawley rats and Beagle dogs, J. Evidence-Based Complementary Altern. Med. 2014
(2014) 570134, https://dx.doi.org/10.1155/2014/570134.
lP

[119] G.F. Feng, S. Liu, Z.F. Pi, F.R. Song, Z.Q. Liu, Studies on the chemical and intestinal metabolic profiles of
Polygalae Radix by using UHPLC-IT-MSn and UHPLC-Q-TOF-MS method coupled with intestinal
bacteria incubation model in vitro, J. Pharm. Biomed. Anal. 148 (2018) 298–306,
na

https://dx.doi.org/10.1016/j.jpba.2017.10.017.
[120] G.F. Feng, S. Liu, Z.F. Pi, F.R. Song, Z.Q. Liu, Comprehensive characterization of in vivo metabolic profile
ur

of Polygalae radix based on ultra-high-performance liquid chromatography-tandem mass spectrometry, J.


Pharm. Biomed. Anal. 165 (2019) 173–181, https://dx.doi.org/10.1016/j.jpba.2018.12.005.
Jo

[121] B.X. Xu, C.C. Qu, W.H. Zheng, Y.Y. Xi, X. Zhao, H.F. Li, J.T. Liu, X.L. Zhang, UHPLC-MS/MS method for
simultaneous determination of Radix Polygalae glycolipids and organic acids in rat plasma and application
in a pharmacokinetic study, J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci. 15 (2018) 165–173,
https://dx.doi.org/10.1016/j.jchromb.2018.09.037.
[122] C.J. Yang, X.B. Yin, X.X. Dong, X. Zhang, L.T. You, W.P. Wang, J.H. Wang, Q.H. Chen, J. Ni, Determination
of the phytochemical composition of Jingning fang and the in vivo pharmacokinetics of its metabolites in rat
plasma by UPLC-MS/MS, J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci. 1067 (2017) 71–88,
https://dx.doi.org/10.1016/j.jchromb.2017.09.019.
[123] Y. Sun, G. Feng, Y. Zheng, S. Liu, Y. Zhang, Z. Pi, F. Song, Z. Liu, Putative multiple reaction monitoring
strategy for the comparative pharmacokinetics of postoral administration Renshen-Yuanzhi compatibility
through liquid chromatography-tandem mass spectrometry, J. Ginseng Res. 44 (2020) 105-114,
https://dx.doi.org/10.1016/j.jgr.2018.09.007.
[124]X.T. Wang, Y. Zhang, H.B. Niu, Y.J. Geng, B. Wang, X.M. Yang, P.Y. Yan, Q. Li, K.S. Bi, Ultra-fast liquid
chromatography with tandem mass spectrometry determination of eight bioactive components of
Kai-Xin-San in rat plasma and its application to a comparative pharmacokinetic study in normal and
Alzheimer’s disease rats, J. Sep. Sci. 40 (2017) 2131–2140, https://dx.doi.org/10.1002/jssc.201601343.
Journal Pre-proof

[125]Y.Y. Ba, Y.Y. Jiang, Y. Liu, H. Lv, C.L. Sa, X.T. Li, G.M. She, R.B. Shi, Comparative pharmacokinetics of
3,6′-disinapoyl sucrose after oral administration of pure 3,6′-disinapoyl sucrose, Radix Polygalae extract
and Kai-Xin-San in acquired dysmnesia model rats, Chin J. Exp. Tradit. Med. Form. 18 (2012) 138–142,
https://dx.doi.org/10.3969/j.issn.1005- 9903.2012.14.042.

of
ro
-p
re
lP
na
ur
Jo
Journal Pre-proof

Graphical abstract

of
ro
-p
re
lP
na
ur
Jo

You might also like