Sei sulla pagina 1di 20

Journal of Ethnopharmacology 172 (2015) 10–29

Contents lists available at ScienceDirect

Journal of Ethnopharmacology
journal homepage: www.elsevier.com/locate/jep

Review

Sophora flavescens Ait.: Traditional usage, phytochemistry


and pharmacology of an important traditional Chinese medicine
Xirui He a,b, Jiacheng Fang b, Linhong Huang a,n, Jinhui Wang c, Xiaoqiang Huang a,n
a
Hong-Hui Hospital, Xi’an Jiaotong University College of Medicine, Xi’an 710054, PR China
b
The College of Life Sciences, Northwestern University, Xi’an 710069, PR China
c
Department of Pharmacy, University Hospital of Gansu Traditional Medicine, Lanzhou 730020, PR China

art ic l e i nf o a b s t r a c t

Article history: Ethnopharmacological relevance: Sophora flavescens (Fabaceae), also known as Kushen (Chinese: 苦参),
Received 2 January 2015 has been an important species in Chinese medicine since the Qin and Han dynasties. The root of Sophora
Received in revised form flavescens has a long history in the traditional medicine of many countries, including China, Japan, Korea,
3 June 2015
India and some countries in Europe. In traditional Chinese medicine (TCM), Sophora flavescens has been
Accepted 5 June 2015
used extensively, mainly in combination with other medicinal plants in prescriptions to treat fever,
Available online 16 June 2015
dysentery, hematochezia, jaundice, oliguria, vulvar swelling, asthma, eczema, inflammatory disorders,
Keywords: ulcers and diseases associated with skin burns. The aim of this review is to provide updated and
Sophora flavescens comprehensive information regarding the botany, ethnopharmacology, phytochemistry, biological
Ethnopharmacology activities and toxicology of Sophora flavescens and to discuss possible trends and opportunities for
Flavonoids
further research on Sophora flavescens.
Alkaloids
Materials and methods: We systematically searched major scientific databases (PubMed, Elsevier,
Antitumor activity
Antimicrobial activity
SpringerLink, Google Scholar, Medline Plus, ACS, “Da Yi Yi Xue Sou Suo (http://www.dayi100.com/login.
jsp)”, China Knowledge Resource Integrated (CNKI) and Web of Science) for information published
between 1958 and 2015 on Sophora flavescens. Information was also acquired from local classic herbal
literature, conference papers, government reports, and PhD and MSc dissertations.
Results: The broad spectrum of biological activities associated with Sophora flavescens has been
considered a valuable resource in both traditional and modern medicine. Extracts are taken either orally
or by injection. More than 200 compounds have been isolated from Sophora flavescens, and the major
components have been identified as flavonoids and alkaloids. Recent in vitro and in vivo studies indicate
that at least 50 pure compounds and crude extracts from Sophora flavescens possess wide-ranging
antitumor, antimicrobial, antipyretic, antinociceptive, and anti-inflammatory pharmacological abilities.
The anticancer and anti-infection abilities of these components are especially attractive areas for
research.
Conclusions: Sophora flavescens is a promising traditional medicine, but there is a need for more precise
studies to test the safety and clinical value of its main active crude extracts and pure compounds and to
clarify their mechanisms of action. Moreover, some existing studies have lacked systematic methods and

Abbreviations: 5-HT, 5-hydroxytryptamine; 5R-DHT, 5R-dihydrotestosterone; Ach, Acetylcholine; AGE, Advanced glycation end product; AHR, Airway hyperreactivity; ALP,
Alkaline phosphatase; ANP, Atrial natriuretic peptide; ASM, Airway smooth muscle; cGMP-PDE5, Cyclic guanosine monophosphate (cGMP)-phosphodiesterase type 5; CK,
Casein kinase; COX-2, Cyclooxygenase-2; DGAT, Diacylglycerol acyltransferase; DHT, Dihydrotestosterone; DNFB, 1-Fluoro-2,4-dinitrofluorobenzene; EC50, Concentration for
50% of maximal effect; ECA, Ehrlich ascites carcinoma cell lines; Eca-109, Human esophageal squamous carcinoma Eca-109 cell line; ER, Estrogen receptor; ERK, Extracellular
regulated protein kinases; EtOAc, Ethyl acetate; H460, Non-small cell lung cancer H460; HBeAg, Hepatitis B e antigen; HBsAg, Hepatitis B surface antigen; HBV, Viral hepatitis
type B; HDL-C, High-density lipoprotein-cholesterol; HMC-1, Human mast cell-1; HPLC, High performance liquid chromatography; HPLC-DAD-ESI/MS, High performance
liquid chromatography with diode-array detector and electrospray ionization-tandem mass spectrometry; HRAR, Human recombinant aldose reductase; HSV, Herpes
simplex virus; HT29, Colon cancer cell line; IC50, 50% inhibition concentration; IGF-1, Insulin and insulin-like growth factor-1; IL, Interleukin; IFN-γ, Interferon-gamma; iNOS,
Inducible nitric oxide synthase; KGF, Kinetics gain factor; LD50, Median lethal dose; LDL-C, Low-density lipoprotein-cholesterol; LH, Luteinizing hormone; LPS,
Lipopolysaccharide; LVSP, Left ventricular systolic pressure; LVEDP, Left ventricular end-diastolic pressure; MAO, Monoamine oxidase; MC3T3-E1, Murine calvaria-derived
osteoblastic cell line; MCF-7, Human breast adenocarcinoma cell lines; MCP-1, Monocyte chemoattractant protein-1; MDA, Malondialdehyde; MIC, Minimal inhibitory
concentration; MTT, Methylthiazolyldiphenyl-tetrazolium bromide; NF-κB, Nuclear factor kappa B; NO, Nitric oxide; PDE, Phosphodiesterase; PFS, Prenylated flavonoid-
enriched fraction; PGE2, Prostaglandin E2; PMA, Phorbol 12-myristate 13-acetate; PPARs, Peroxisome proliferator-activated receptors; RANKL, Receptor activator for nuclear
factor-κ B ligand; RLAR, Rat lens aldose reductase; ROS, Reactive oxygen species; S180, Sarcoma 180; SFE, Sophora flavescens extract; SFL, Lectin from Sophora flavescens; sGC-
cGMP, Soluble guanylyl cyclase-cyclic guanosine monophosphate; SGLT, Na þ -glucose cotransporter; SOD, Superoxide dismutase; SR, Root of Sophora flavescens; TC, Total
cholesterol; TCM, Traditional Chinese Medicine; TG, Triglycerides; TLC, Thin layer chromatography; TNF-α, Tumor necrosis factor-alpha
n
Corresponding authors. Tel.: þ 86 29 87800002.
E-mail addresses: xiruihe6105194@163.com (X. He), hxrhist@163.com (L. Huang).

http://dx.doi.org/10.1016/j.jep.2015.06.010
0378-8741/& 2015 Published by Elsevier Ireland Ltd.
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 11

integration with the existing literature, and some of the experiments were isolated, used small sample
sizes and were unreliable. More validated data are therefore required.
& 2015 Published by Elsevier Ireland Ltd.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2. Botany and ethnopharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2.1. Botany . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2.2. Ethnopharmacology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
3. Phytochemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
3.1. Flavonoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
3.2. Alkaloids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
3.3. Triterpenoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.4. Other compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4. Methods of quality control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5. Effects of crude extract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
5.1. Antitumor activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
5.2. Anti-inflammatory and antinociceptive activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
5.3. Antianaphylaxis and antiasthma activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.4. Antimicrobial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.4.1. Antibacterial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.4.2. Antiviral activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.4.3. Insecticidal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.4.4. Antifungal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.5. Effect on the cardiovascular system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.5.1. Antiarrhythmic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
5.5.2. Antimyocardial fibrosis activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
5.5.3. Effect on myocardial contractility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
5.5.4. Vascular relaxing and antimyocardial hypoxic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
5.6. Immunoregulatory activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
5.7. Other activities. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
6. Toxicity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
7. Future perspectives and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

Fig. 1. Sophora flavescens Ait. (A) the aerial parts, (B) roots, and (C) traditional Chinese medicine decoction pieces.
12 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

1. Introduction flavescens was listed in the first Chinese medical classic,


“Shen-Nung’s Pen-Ts’ao”, in 200 A. D., where it was ranked as a
Sophora is a widespread genus in the Fabaceae family that “Middle grade” medicine. According to the “Compendium of Materia
consists of approximately 52 species, nineteen varieties, and seven Medica”, another authoritative monograph of TCM, the root of
forms that grow throughout Asia, Oceania, and the Pacific Islands. Sophora flavescens is bitter in taste, cold in nature and described as
Approximately fifteen species in this genus have been the focus of having activities related to such processes as clearing away the heat-
the most attention since antiquity (Panthati et al., 2012). The root evil, expelling superficial evils, dispelling wind, relieving rheuma-
of Sophora flavescens, also known as Kushen (Chinese: 苦参), has a tism, improving eyesight, and nourishing the liver and gall. In
strong, bitter taste and cold properties and has been widely used ancient times, more than fifteen classical pharmaceutical books
in traditional and popular medicine as a functional food ingredient created by distinguished medical experts in China contained records
for the treatment of conditions including fevers, dysentery, jaun- of this medicinal plant. It was described as the main component in
dice, vaginal itching with leukorrhagia, abscesses, carbuncles, some noted prescriptions and recipes. Briefly, according to “Tang
enteritis, leukorrhea, pyogenic infections of the skin, scabies, materia medica”, the dried root of Sophora flavescens was used to
swelling, and pain (State Administration of Traditional Chinese improve leg numbness and as an insecticide in the Tang dynasty. In
Medicine, 1999; Chinese Pharmacopoeia Commission, 2010). In the Ming dynasty, according to two famous classical books from the
China, Sophora flavescens is documented in editions of the Phar- Chinese materia medica called “Ben Cao Jing Bai Zhong Lu” and “Dian
macopoeia of the People’s Republic of China from 1977 to 2010. Nan Ben Cao”, the root of Sophora flavescens was considered to be
Extracts of Sophora flavescens were, in the past, mainly used in cold in nature, was thought to act on the heart meridian, and was
compound formulas or as decoctions for other herbal drug used as a treatment for itching skin, hematochezia, solid tumors,
products and was always taken orally. More recently, with the purulent phlegm, inflammation and other diseases. In the Qing
emergence and development of new dosage forms (e.g., tablets, dynasty, it was described in the Chinese book “Ben Cao Cun Xin” as
capsules and injections) and the characterization of a pharmaco- being used to eliminate dampness, to clear away the heat-evil and to
logical profile for Sophora flavescens, injection administration has treat spermatorrhea. Some of the abovementioned uses have been
become gradually more accepted and used in clinics for the supported by evidence from an increasing number of studies and
treatment of cancer and infectious diseases (Chen et al., 2012; clinical reports.
Yanju et al., 2014). In China, Sophora flavescens is currently distributed mainly in
The chemical profile of Sophora flavescens is extensive and is the Shanxi, Hubei, Henan and Hebei provinces. The powder from
known to contain molecules with therapeutic importance. Exten- the dried root of Sophora flavescens has been listed in the
sive studies have led to a rapid increase in the amount of Pharmacopoeia of the People0 s Republic of China for the treatment
information available regarding the pharmacological activities of of dysentery, hematochezia, jaundice, oliguria, vulvar swelling,
Sophora flavescens components. Several authors have published eczema, ulcers, scabies, and leprosy (Chiang Su New Medical
reviews about the chemistry and biological activity of Sophora College, 1977; Zhu, 1998; Chinese Pharmacopoeia Commission,
flavescens components. However, no comprehensive review has 2010). The 2010 edition of the Chinese Pharmacopoeia officially
yet been published. Here, we systematically review the ethno- listed nine Chinese patented medicines that contained mainly
pharmacology, phytochemistry, pharmacological activity and toxi- Sophora flavescens, including Kushen tablets, Danggui Kushen
city of Sophora flavescens based on a survey of the scientific tablets, Kushen capsules, and compound Kushen injections.
literature between 1958 and 2015. This current and comprehen- Kushen tablets, for example, are a common traditional Chinese
sive information is provided to support the development of better medicine that has been used for the treatment of infectious
therapeutic agents and good clinical practices related to Sophora diseases in China for over 40 years. Reported effects of Kushen
flavescens. tablets include clearing heat, eliminating dampness and killing
insects, and it is very effective for treating dysentery, intestinal
catarrh and eczema in clinics. A multitude of classic prescriptions
2. Botany and ethnopharmacology (e.g., Kushen decoction, Kushen powder, Kushen Shierwei pills)
created by ancient doctors and pharmacists continue to be
2.1. Botany frequently used in clinics (State Administration of Traditional
Chinese Medicine, 1999).
Sophora flavescens Ait. (Fabaceae) is a deciduous shrub that is The root of this herb is also used as an effective agent and is
0.5 1.5 m tall and is distributed throughout East Asia (especially available in other countries. In Korea, Sophora flavescens is com-
China, Korea and Japan) and some European countries. The root of mercially available as the generic “Kosam”, and it has long been
Sophora flavescens is cylindrical in shape, 10–30 cm in length, used in popular medicine as an antipyretic, analgesic, antihelmin-
1–6.5 cm in diameter, and externally gray-brown. The leaves are tic and stomachic, as well as for the treatment of fever, inflam-
pinnately compound (odd) and up to 12–25 cm long. The raceme matory disorders, ulcers and skin burns (Hur, 1994; Bae, 2000). In
is 10–20 cm long and has numerous mitriform calyces and a Japan, the dried root of Sophora flavescens, known as “Kujinn”, was
yellowish white corolla that is slightly longer than the other documented in the 16th edition of the Japanese Pharmacopoeia for
petals. The linear pod is 5–12 cm long, has an apex with a long its use as a stomachic, antifebrile, analgesic and anthelmintic. (Wu
beak, is slightly beaded, and does not crack after maturity. Sophora et al., 1985a). In Hawaiian popular medicine, “Kushen” (Sophora
flavescens flowers between May and July and its fruit ripens flavescens Ait.) has been used for treating asthma by practitioners
during August and September into a brown, spherical pod with of Hawaiian herbal medicine.
1–5 seeds (Fig. 1).

2.2. Ethnopharmacology 3. Phytochemistry

Current available resources show that Sophora flavescens plants Sophora flavescens produces a wide range of secondary metabo-
have been pharmacologically used by ethnic groups as a popular lites, some of which have received considerable attention due to
herbal medicine in various regions of the world, especially in Asian their wide-reaching biological activities and novel structures. Prior
countries. Two thousand years ago, the dried root of Sophora to early 2015, more than 200 compounds had been isolated and
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 13

Table 1
The compounds and activities of some flavonoids isolated from the roots of Sophora flavescens.

No. Compounds Biological activity Results References

1 Quercetin Li et al. (2004)


2 Rutin Li et al. (2004)
3 Kushenol C Antibacterial activity Inhibited the growth of Staphylococcus aureus and Streptococcus mutans Wu et al. (1985a); Yamaki
with MIC of 12.5 and 12.5 μg/mL, respectively. et al. (1990)
Aldose reductase Showed marked inhibitory activity on HRAR with IC50 values of Jung et al. (2008)
inhibitory activity 0.85 μM, and also possessed good inhibitory activity toward AGE
formation with IC50 values of 84.6 μg/mL.
4 5-methylkushenol C Yagi et al. (1989)
5 Kushenol G Wu et al. (1985b)
6 Noranhydroicaritin Inhibited the growth of S. aureus and S. mutans with MIC of 25 and Komatsu et al. (1970);
50 mg/mL, respectively. Yamaki et al.(1990)
7 Isoanhydroicaritin Komatsu et al. (1970)
8 Sophoflavescenol PDEs inhibitory Showed the most potent inhibitory activity (IC50 ¼ 13 nM) against Shin et al. (2002)
activity cGMP-PDE5 with 31.5- and 196.2-fold selectivity over PDE3 and PDE4.
9 Resokaempferol Ding and Chen, 2005
10 8-lavandulylkaempferol Aldose reductase Showed inhibitory activity on RLAR and HRAR with IC50 values of Shen et al. (2006); Jung
inhibitory activity 3.8 and 0.79 μM, respectively. It also possessed good inhibitory activity et al. (2008)
toward AGE formation with IC50 values of 132.1 μg/mL.
11 8-prenylkaempferol Estrogenic activity Exhibited estrogenic activity by binding to rat uterine ER with EC50 of Hillerns and Wink (2005)
(Desmethylanhydroicaritin) 3.71 70.62 mg/mL.
Aldose reductase Showed marked inhibitory activity on RLAR and HRAR with IC50 values Jung et al. (2008)
inhibitory activity of 0.95 and 0.45 μM, respectively. And it also possessed good inhibitory
activity toward AGE formation with IC50 values of 104.3 μg/mL.
Osteogenesis Significantly promoted ALP activity, up-regulated mRNA expressions of Chiou et al. (2011)
promising osteocalcin and osteopontin in MC3T3-E1 cells.
0
12 8-lavandulvl-5,7,4 -trihydroxy- Cao (2007)
flavonol
13 Citrusinol Lin (2006)
14 Flavenochromane B Antitumor activity Active against A549 (human lung cancer line), 1A9 (ovarian carcinoma), Ding et al. (2004)
MCF-7 (breast adenocarcinoma), KB (epidermoid carcinoma of the
nasopharynx), and KB-Vin (drug-resistant variant KB) with IC50 values
1.0, 1.2, 3.6, 1.7 and 1.3 mM, respectively.
15 Flavenochromane C Antitumor activity Against A549, 1A9, KB, and KB-Vin with IC50 values r 1.7 mM, and Ding et al. (2004)
against the MCF-7 with IC50 value of 3.6 mM.
Flavanones
16 Kushenol A Antibacterial activity Against the bacteria S. aureus, B. subtilis, S. epidermidis and P. acnes with Wu et al. (1985a);
the MIC of 5.0, 2.5, 5.0 and 10.0 mg/mL, respectively. Kuroyanagi et al. (1999)
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 51.4 % and 89.3 %, respectively.
Neuraminidase Dose-dependently inhibited the neuraminidase activity with IC50 of Ryu et al. (2008)
inhibitory activity 14.8 mM.
Glycosidase inhibitory Showed strong α-glucosidase inhibitory activity with IC50 values of Kim et al. (2006)
activity 45 μM, and was shown to be noncompetitive inhibitor.
17 Kushenol B Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Wu et al. (1985a); Ryu
HCT-15 with the ED50 of 3.8, 4.3, 3.4, 2.7 and 3.0 mg/mL. et al. (1997)
PLCγ1 inhibitory Showed potent PLCγ1 inhibitory activity with IC50 values of 7.5 mM. Lee et al. (1997)
activity
18 Kushenol E Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Wu et al. (1985b); Ryu
HCT-15 with the ED50 of 6.4, 6.4, 5.3, 5.0 and 4.6 mg/mL. et al. (1997)
PLCγ1 inhibitory Showed PLCγ1 inhibitory activity with IC50 values of 11.8 mM. Lee et al. (1997)
activity
19 Kushenol F Monoamine oxidase Dose-dependenly inhibited the activity of MAO-A and MAO-B with IC50 Wu et al. (1985b); Hwang
inhibitory activity values of 103.7 and 63.1 mM, respectively. et al. (2005)
20 Kushenol P Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 76.1 % and 77.0 %, respectively.
21 Kushenol Q Antibacterial activity Against the bacteria S. aureus and Bacillus subtilis with the MIC of Kuroyanagi et al. (1999)
5.0 and 5.0 mg/mL.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 34.8 % and 82.4 %,
respectively.
22 Kushenol R Antibacterial activity Against the bacteria S. aureus, B. subtilis, S. epidermidis and Kuroyanagi et al. (1999)
Propionibacterium acnes with the MIC of 5.0, 2.5, 5.0 and 10.0 mg/mL.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 23.6 % and 67.5 %, respectively.
23 Kushenol S Antibacterial activity Against the bacteria S. aureus and B. subtilis with the MIC of 5.0 and Kuroyanagi et al. (1999)
5.0 mg/mL.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 22.9 % and 47.7 %, respectively.
24 Kushenol T PPARs Significantly activated the transcription of PPARα and PPARγ with EC50 Kuroyanagi et al. (1999);
transactivational values of 16.0 73.4 and 25.5 7 3.1 mM, respectively. Quang et al. (2013)
properties
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 54.8 % and 84.9 %,
respectively.
25 Kushenol U Antibacterial activity Kuroyanagi et al. (1999)
14 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

Table 1 (continued )

No. Compounds Biological activity Results References

Against the bacteria S. aureus and B. subtilis with the MIC of 10.0 and
10.0 mg/mL.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 50.3 % and 85.1 %,
respectively.
26 Kushenol V Antibacterial activity Against the bacteria S. aureus and B. subtilis with the MIC of 10.0 and Kuroyanagi et al. (1999)
10.0 mg/mL.
27 Kushenol W Antibacterial activity Against the bacteria S. aureus and B. subtilis with the MIC of 10.0 and Kuroyanagi et al. (1999)
10.0 mg/mL.
28 Kurarinone Antitumor activity Inhibited proliferation of HL-60 and HepG2 cells with an IC50 of 18.5 Hatayama and Komatsu
and 36.2 mM, respectively; inhibited the proliferation of A549, SK-OV-3, (1971); Ko et al. (2000);
SK-MEL-2, XF498 and HCT-15 with the ED50 of 9.0, 9.4, 6.4, 5.9 and Ryu et al. (1997)
8.6 mg/mL.
Anti-inflammatory Dose-dependently inhibited TNFα-induced NF-κB transcriptional Quang et al. (2013); Kim
activity activity in HepG2 cells with IC50 values of 4.0 μM. and Kim, 2013
Antibacterial activity Against the bacteria S. aureus and B. subtilis with the MIC of 2.5 and Kuroyanagi et al. (1999);
2.5 mg/mL, respectively. Besides, it also exhibited potent antibacterial Ryu et al. (2008)
activity with 10.0 mg/disk against Gram-positive bacteria (B. cereus and
Escherichia coli).
Antifungal activity Against P. vanterpooli and P. Graminicola at a concentration of 12.5 mg/ Yagi et al. (1989)
mL, and weakly inhibited the growth of P. aphanidermatum
(MIC ¼ 25.0 mg/mL).
Insecticidal activity 50 and 25 mg/mL showed insecticidal activity on Mosquito larvae. Zheng et al. (1999)
PLCγ1 inhibitory Showed inhibitory activity against PLCγ1 with IC50 values of 14.1 mM. Lee et al. (1997)
activity
β-secretase inhibitory It was shown to be a noncompetitive inhibitor toward β-secretase with Hwang et al.(2008)
activity IC50 of 3.3 mM.
Neuraminidase Dose-dependently inhibited the neuraminidase activity with IC50 of Ryu et al. (2008)
inhibitory activity 15.1 mM.
Tyrosinase inhibitory Exhibited extremely tyrosinase inhibitory activity (noncompetitive Ryu et al. (2008); Sasaki
activity inhibitors, IC50 of 2.2 mM), and also demonstrated to be noncompetitive et al. (2014)
inhibitors of protein tyrosine phosphatase 1B based on a kinetic
analysis with IC50 o30.0 mM.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 40.5 % and 89.2 %,
respectively.
Glycosidase inhibitory Showed strong α-glucosidase and β-amylase inhibitory activities with Kim et al. (2006)
activity IC50 values of 68 and 85 μM, respectively, and were shown to be
noncompetitive inhibitor.
Aldose reductase Showed marked inhibitory activity on RLAR with IC50 values of 2.99 μM. Jung et al. (2008)
inhibitory activity
DGAT inhibitory Dose-dependently inhibited DGAT activity with IC50 values of 10.9 μM. Chung et al. (2004)
activity
SGLT inhibitory Showed strong inhibitory activity against SGLT1 and SGLT2 at 50 μM Sato et al. (2007)
activity with inhibition rate of 98.8 % and 99.7 %, respectively.
MCP-1 inhibitors Inhibited the migration of THP-1 cells induced by MCP-1 with IC50 Lee et al. (2005)
value of 19.2 μg/mL.
0
29 2 -methoxykurarinone Antitumor activity Inhibited proliferation of HL-60 and HepG2 cells with an IC50 of 13.7 Kang et al. (2000a); Ko
and 21.1 mM, respectively. et al. (2000)
PPARs Dose-dependently activated the transcription of PPARα and PPARβ(δ) Quang et al. (2013)
transactivational with EC50 values of 11.17 2.2 and 21.4 72.5 mM, respectively.
properties
β-secretase inhibitory It was shown to be a noncompetitive inhibitor toward β-secretase with Hwang et al. (2008)
activity IC50 of 6.7 mM.
Tyrosinase inhibitory Demonstrated to be noncompetitive inhibitors of protein tyrosine Sasaki et al. (2014)
activity phosphatase 1B based on a kinetic analysis with IC50 o 30 mM.
Glycosidase inhibitory Showed strong α-glucosidase and β-amylase inhibitory activities with Kim et al. (2006)
activity IC50 values of 155 and 45 μM, respectively.
Aldose reductase Showed inhibitory activity on RLAR with IC50 values of 3.77 μM. Jung et al. (2008)
inhibitory activity
Osteoclast Dose-dependently inhibited osteoclast differentiation in bone marrow Kim et al. (2014)
differentiation cell-osteoblast cocultures and RANKL-induced osteoclast
inhibition differentiation from bone marrow macrophages.
30 Isokurarinone Antibacterial activity Against the bacteria S. aureus, B. subtilis, S. epidermidis and P. acnes with Kyogoku et al. (1973);
the MIC of 2.5, 2.5, 5.0 and 10.0 mg/mL, respectively. Kuroyanagi et al. (1999)
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 87.1 % and 99.8 %, respectively.
31 (þ ) -norkurarinone PPARs Activated the transcription of PPARα, PPARβ(δ) and PPARγ with EC50 Hatayama and Komatsu
transactivational values of 5.6 7 0.2, 20.7 71.5 and 6.6 7 1.1 mM, respectively. (1971); Quang et al. (2013)
properties
Antibacterial activity Against the bacteria S. aureus, B. subtilis, S. epidermidis and P. acnes with Kuroyanagi et al. (1999)
the MIC of 2.5, 2.5, 10.0 and 10.0 mg/mL, respectively.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 86.4 % and 85.8 %,
respectively.
Estrogenic activity Hillerns and Wink (2005)
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 15

Table 1 (continued )

No. Compounds Biological activity Results References

Exhibited estrogenic activity by binding to rat uterine ER with EC50 of


8.22 7 4.14 mg/mL.
32 Kurarinol Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Kyogoku et al. (1973); Ryu
HCT-15 with the ED50 of 30.3,25.8, 21.8, 25.9 and 28.7 mg/mL. et al. (1997)
Antiviral activity Against Herpes simplex virus types I and II with the EC50 of 60 73.4 Woo et al. (1998)
and 447 2.9 mg/mL.
Neuraminidase Dose-dependently inhibited the neuraminidase activity with IC50 of Ryu et al. (2008)
inhibitory activity 12.1 mM.
Tyrosinase inhibitory It was shown to be a noncompetitive inhibitor toward tyrosinase with Ryu et al. (2008)
activity IC50 of 1.0 mM.
Antiandrogen activity 200 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 11.9 % and 65.8 %, respectively.
Aldose reductase Showed marked inhibitory activity on RLAR with IC50 values of 2.13 μM. Jung et al. (2008)
inhibitory activity
DGAT inhibitory Dose-dependently inhibited DGAT activity with IC50 values of 8.6 μM. Chung et al. (2004)
activity
cGMP-PDE5 Showed inhibitory activity against cGMP-PDE5 with an IC50 values of Shin et al. (2002)
inhibitory activity 6.1 μM.
33 Neokurarinol Antibacterial activity Against the bacteria S. aureus, B. subtilis, S. epidermidis and P. acnes with Kyogoku et al. (1973);
the MIC of 2.5, 2.5, 10.0 and 25.0 mg/mL, respectively. Kuroyanagi et al. (1999)
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 79.3 % and 58.9 %,
respectively.
34 Norkurarinol Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Kyogoku et al. (1973); Ryu
HCT-15 with the ED50 of 15.4, 14.8, 12.9, 10.7 and 16.5 mg/mL. et al. (1997)
Tyrosinase inhibitory Showed inhibitory activity on mushroom tyrosinase with IC50 values of Son et al. (2003)
activity 2.1 mM.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 41.3 % and 75.4 %, respectively.
35 Isoxanthohumol Glycosidase inhibitory Showed α-glucosidase inhibitory activity. Komatsu et al. (1970);
activity Quang et al. (2012b)
36 Leachianone A Antitumor activity Inhibited proliferation of HL-60 and HepG2 cells with an IC50 of 11.3 Kang et al. (2000a)
and 13.3 mM, respectively.
β-secretase inhibitory It was shown to be a noncompetitive inhibitor toward β-secretase with Hwang et al.(2008)
activity IC50 of 2.6 mM.
Estrogenic activity Exhibited estrogenic activity by binding to rat uterine ER with EC50 of Hillerns and Wink (2005)
8.95 7 3.41 mg/mL.
37 Leachianone G Antibacterial activity Against the bacteria S. aureus and Bacillus subtilis with the MIC of Kuroyanagi et al. (1999)
5.0 and 2.5 mg/mL.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 80.5 % and 84.8 %,
respectively.
38 Sophoraflavanone B Zhao et al. (1993)
39 5-methylsophoraflavanone B Antibacterial activity Against Candida albicans, E. coli, Salmonella typhimurium, S. epidermis Kang et al. (2000b); Sohn
and S. aureus. with MIC of 25, 20, 20, 20, 20 mg/mL. et al. (2004)
40 (  ) -sophoraflavanone G Antitumor activity Inhibited proliferation of HL-60 and HepG2 cells with an IC50 of 12.5 Wang and Han (1996); Ko
and 13.3 mM, respectively; inhibited the proliferation of A549, SK-OV-3 et al. (2000); Ryu et al.
(ovary), SK-MEL-2 (skin), XF498 (central nerve system) and HCT-15 (1997)
(colon) with the ED50 of 6.4, 7.9, 3.9, 5.8 and 5.7 mg/mL.
Anti-Inflammatory 2.5–20 μM inhibited the levels of NO and PGE2 and decreased the Wun et al. (2013)
activity production of IL-1b, IL-6 and TNFα.
Antibacterial activity 10 mg/disk against Gram-positive bacteria (B. subtilis, B. cereus and E. Ryu et al. (2008); Kim
coli). Besides, it showed strong antibacterial activity on E.coli, S. et al. (2013); Sohn et al.
epidermis and S. aureus with MIC of 20, 20 and 20 mg/mL. (2004)
Insecticidal activity 50 and 25 mg/mL showed insecticidal activity on Mosquito larvae. Zheng et al. (1999)
PLCγ1 inhibitory Showed potent PLCγ1 inhibitory activity with IC50 values of 8.2 mM. Lee et al. (1997)
activity
β-secretase inhibitory It was shown to be a noncompetitive inhibitor toward β-secretase with Hwang et al. (2008)
activity IC50 of 5.2 mM.
Neuraminidase Dose-dependently inhibited the neuraminidase activity with IC50 of Ryu et al. (2008)
inhibitory activity 13.5 mM.
Tyrosinase inhibitory It was shown to be a noncompetitive inhibitor toward tyrosinase with Ryu et al. (2008)
activity IC50 of 4.7 mM.
Glycosidase inhibitory Showed strong α-glucosidase and β-amylase inhibitory activities with Kim et al. (2006)
activity IC50 values of 37 and 55 μM, respectively.
Naþ -Glucose Showed strong inhibitory activity against SGLT1 and SGLT2 at 50 μM Sato et al. (2007)
cotransporter (SGLT) with inhibition rate of 99.2 % and 100 %, respectively.
inhibitory activity
41 Sophoraflavanone K Shen et al. (2006)
42 Sophoraflavanone L Shen et al. (2006)
43 Naringenin Yagi et al. (1989)
44 Naringenin-7-O-β-D-glucosyl-40 -O-β- Shen and Zhang(2012)
D-glucose
45 5-methoxy-7,20 ,40 -trihydroxy-8- Li et al. (2008)
prenylflavanone
46 Cao (2007)
16 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

Table 1 (continued )

No. Compounds Biological activity Results References

(2S)-6-lavandulyl-isopentenyl-5-
methoxy-7,20 ,40 ,- trihydroxy-
flavonone
47 (2S)-8-isopentenyl-7,20 ,40 ,-5- Cao (2007)
methoxyflavonone
48 (2S)-6[2(3-hydroxyisopropyl)-5- Ma et al. (2008)
methyl-4-hexenyl]-5-methoxy-
7,20 ,40 -trihydroxyflavanone
49 (2S)-5, 40 -dimethoxy-8- lavandulyl-7, Ma et al. (2008)
20 -dihydroxy flavanone
50 (2S)-8-(5-hydroxy-2-isopropenyl-5- Ma et al. (2008)
methylhexyl)-7-methoxy-5,20 , 40 -
trihydroxy-flavanone
51 (2S)-7, 40 -dihydroxy-5-methoxy- 8- Aldose reductase Showed marked inhibitory activity on HRAR with IC50 values of Kang et al. (2000b); Jung
(γ, γ- dimethylallyl) -flavanone inhibitory activity 0.37 μM. et al. (2008)
52 (2R)-3α,7,40 -trihydroxy-5-methoxy-8-PPARs Activated the transcription of PPARα, PPARβ(δ) and PPARγ in a dose- Quang et al. (2013)
(γ, γ-dimethylallyl)-flavanone transactivational dependent manner, with EC50 values of 5.5 7 1.8, 11.4 7 1.0 and
properties 10.5 7 2.0 mM, respectively.
0
53 (2S)-3β,7,4 -trihydroxy-5-methoxy-8- Aldose reductase Showed inhibitory activity on RLAR with IC50 values of 3.63 μM, and Quang et al. (2013); Jung
(γ, γ-dimethylally)-flavanone inhibitory activity also possessed good inhibitory activity toward AGE formation with IC50 et al. (2008)
values of 261.0 μg/mL.
54 (2S)-7,20 ,40 -triihydroxy-5-methoxy-8- Anti-angiogenesis Blocked cell cycles in the G0/G1 phase without inducing apoptosis, and Quang et al. (2013); Zhang
dimethylallylflavanone activity down regulates vascular endothelial growth factor (VEGF) expression. et al. (2013)
55 3β, 7, 40 - trihydroxy-5-methoxy 8- Li et al. (2008)
[3, 3- dimethylallyl] - flavanone
56 40 -hydroxyisolonchocarpin Lin (2006)
Flavanonols
57 Kushenol J Wu et al. (1985c)
58 Kushenol H Antiviral activity Against Herpes simplex virus types II with an EC50 of 147 78.8 mg/mL Wu et al. (1985b); Woo
et al. (1998)
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 50.9 % and 81.4 %,
respectively.
DGAT inhibitory Dose-dependently inhibited DGAT activity with IC50 values of 142.0 μM. Chung et al. (2004)
activity
cGMP-PDE5 Showed potent cGMP-PDE5 inhibitory activity with IC50 values of Shin et al. (2002)
inhibitory activity 4.75 μM.
59 Kushenol I Wu et al. (1985b)
60 Kushenol K Antiviral activity Against Herpes simplex virus types II with an EC50 of 162 76.5 mg/mL. Wu et al. (1985c); Woo
et al. (1998)
DGAT inhibitory Dose-dependently inhibited DGAT activity with IC50 values of 250.0 μM. Chung et al. (2004)
activity
PLCγ1 inhibitory Showed potent PLCγ1 inhibitory activity with IC50 values of 34.9 mM. Lee et al. (1997)
activity
cGMP-PDE5 Showed potent cGMP-PDE5 inhibitory activity with IC50 values of Shin et al. (2002)
inhibitory activity 10.6 μM.
61 Kushenol L Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Wu et al. (1985c); Ryu
HCT-15 with the ED50 of 11.4, 11.0, 12.4, 12.0 and 8.4 mg/mL. et al. (1997)
PLCγ1 inhibitory Showed potent PLCγ1 inhibitory activity with IC50 values of 11.6 mM. Lee et al. (1997)
activity
62 Kushenol M Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Wu et al. (1985c); Ryu
HCT-15 with the ED50 of 5.5, 6.4, 5.5, 5.7 and 5.0 mg/mL. et al. (1997)
PLCγ1 inhibitory Showed potent PLCγ1 inhibitory activity with IC50 values of 12.2 mM. Lee et al. (1997)
activity
63 Kushenol N Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Wu et al. (1986); Ryu et al.
HCT-15 with the ED50 of 13.1, 13.8, 6.7, 9.6 and 14.5 mg/mL. (1997)
PLCγ1 inhibitory Showed potent PLCγ1 inhibitory activity with IC50 values of 31.2 mM. Lee et al. (1997)
activity
SGLT inhibitory Showed strong inhibitory activity against SGLT1 and SGLT2 at 50 μM Sato et al. (2007)
activity with inhibition rate of 58.4 % and 87.3 %, respectively.
64 Kushenol X Antibacterial activity Against the bacteria S. aureus and B. subtilis with the MIC of 10.0 and Kuroyanagi et al. (1999)
10.0 mg/mL.
Estrogenic activity Exhibited estrogenic activity by binding to rat uterine ER with EC50 of Hillerns and Wink (2005)
4.39 7 1.26 mg/mL.
Antiandrogen activity 100 mg/mL showed antiandrogen activity on 5α- reductase and 5α-DHT- Kuroyanagi et al. (1999)
receptor binding with the inhibitory rate 84.5 % and 90.5 %,
respectively.
65 Kosamol A PLCγ1 inhibitory Showed potent PLCγ1 inhibitory activity with IC50 values of 10.2 mM. Ryu et al. (1996); Lee et al.
activity (1997)
66 (2R, 3R) -8-isopentenyl-7,40 - Cao (2007)
dihydroxy -5-methoxy-flavanonol
67 (2R, 3R)-8-lavanduly-5,7,40 - Cao (2007)
trihydroxy-20 -methoxy-flavanonol
68 (2R, 3R)-8-isopentenyl-7,20 ,40 - Cao (2007)
trihydroxy-5-methoxy-flavanonol
69 Flavenochromanes Ding et al. (2004)
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 17

Table 1 (continued )

No. Compounds Biological activity Results References

Isoflavones
70 Formononetin PPARs Activated the transcription of PPARα, PPARβ(δ) and PPARγ with EC50 Kyogoku et al. (1973);
transactivational values of 0.9 7 0.2, 1.5 7 0.3 and 1.6 7 0.5 mM, respectively. Quang et al. (2013)
properties
Monoamine oxidase Dose-dependenly inhibited the activity of MAO-A and MAO-B with IC50 Hwang et al. (2005)
inhibitory activity values of 21.2 and 11 mM.
71 Daidzein Ding and Chen (2005)
73 Biochanin A Ding and Chen (2005)
74 7-methoxy -40 - hydroxyisoflavone Chen et al. (2011)
75 7,40 -dihydroxy-30 -methoxyisoflavone Ding and Chen (2005)
76 Calycosin Ding and Chen (2005)
77 Kushenol O Wu et al. (1986)
78 3’-hydroxykushenol O Shih (1999)
79 Ononin Shen and Zhang (2012)
80 Pseudobatigenin Lin (2006)
81 Pseudobatigenin-7-O-β-D-xylose- Shen and Zhang(2012)
(1-6)-β-D- glucopyranoside
82 Daidzein-7-O-β-D-xylose-(1-6)-β-D- Shen and Zhang(2012)
glucopyranoside
83 30 -methoxy-40 -hydroxyisoflavone-7- Shen and Zhang(2012)
O-β-D-xylose -(1-6) -β-D-
glucopyranoside
84 30 -hydroxy-40 -methoxyisoflavone-7- Hepatoprotective Exhibited weak inhibition of the cytotoxic effect of D-galactosamine on Shen et al. (2013)
O-β-D-xylopyranosyl-(1-6)-β-D- activity the human hepatic cell line HL-7702 with an inhibition rate of 36.6 %.
glucopyranoside
85 30 -hydroxy-40 -methoxyisoflavone-7- Hepatoprotective Exhibited weak inhibition of the cytotoxic effect of D-galactosamine on Shen and Zhang (2012),
O-β-D-apiofuranosyl-(1-6)-β-D- activity the human hepatic cell line HL-7702 with an inhibition rate of 46.3 %. Shen et al. (2013)
glucopyranoside
86 30 -methoxy-40 -hydroxy-isoflavone-7- Shen and Zhang(2012)
O-β-D-apiose -(1-6) -β-D-
glucopyranoside
87 40 -Hydroxy-30 -methoxyisoflavone-7- Hepatoprotective Exhibited weak inhibition of the cytotoxic effect of D-galactosamine on Shen et al. (2013)
O-β-D-xylopyranosyl-(1-6)-β-D- activity the human hepatic cell line HL-7702 with an inhibition rate of 45.8 %.
glucopyranoside
88 40 -hydroxy-30 -methoxyisoflavone-7- Shen et al. (2013)
O-β-D-apiofuranosyl-(l-6)-β-D-
glucopyranoside
89 40 -hydroxyisoflavone -7-O-β-D- Shen and Zhang(2012)
apiose -(1-6) -β-D- glucopyranoside
90 40 -methoxy-isoflavone-7-O-β-D- Shen and Zhang(2012)
apiose-(1-6) - β-D- glucopyranoside
91 30 ,40 -methylenedioxyisoflavone-7-O- Shen et al. (2013)
β-D-apiofuranosyl-(l-6)-β-D-
glucopyranoside
92 30 ,40 -dihydroxy-isoflavone-7-O-β-D- Shen and Zhang(2012)
glucopyranoside
93 5-hydroxy-40 -methoxy-isoflavone-7- Shen and Zhang(2012)
O-β-D- xylose -(1-6) -β-D-
glucopyranoside
94 5-hydroxy-40 -methoxy-isoflavone-7- Shen and Zhang(2012)
O-β-D- apiose-(1-6)-β-D-
glucopyranoside
95 50 -hydroxy-40 -methoxyisoflavone-20 - Shen et al. (2013)
β-D-glucopyranoside
96 5,40 -dihydroxy-isoflavone -7-O- β-D- Shen and Zhang(2012)
xylose -(1-6) -β-D- glucopyranoside
Isoflavanones
97 7-hydroxy-40 -hydroxy-isoflavonone- Shen and Zhang(2012)
30 -O-β-D-glucopyranoside
98 2,3-dihydroxy-40 -methoxy- Shen and Zhang(2012)
isoflavonone -7-O-β- D-xylose-(1-
6)-β- D- glucopyranoside
99 2,3-dihydroxy-40 -methoxy- Shen and Zhang(2012)
isoflavonone 7-O-β-D-apiose -(1-6)-
β- D- glucopyranoside
Homoisoflavones
100 2,3,40 - trihydroxy-homoisoflavone 7- Shen and Zhang(2012)
O- β-D- glucopyranoside
101 2,3-dihydroxy-40 -methoxy- Shen and Zhang(2012)
homoisoflavone -7-O -xyloside
Chalcones
102 20 , 4- dihydroxy -40 , 60 - Liu and Shi (2007)
dimethoxychalcone
103 Kuraridin Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Hatayama and Komatsu
HCT-15 with the ED50 of 7.1, 5.8, 5.4, 5.8 and 5.0 mg/mL. (1971); Ryu et al. (1997)
Quang et al. (2013)
18 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

Table 1 (continued )

No. Compounds Biological activity Results References

PPARs Dose-dependently inhibited TNFα-induced NF-κB transcriptional


transactivational activity in HepG2 cells with IC50 values of 4.4 μM, and activated the
properties transcription of PPARα, PPARβ(δ) and PPARγ with EC50 values of
4.8 7 0.9, 10.3 7 1.2 and 15.7 7 1.0 mM.
Antibacterial activity Inhibited the growth of S. aureus and S. mutans. Yamaki et al. (1990)
PLCγ1 inhibitory Showed inhibitory activity toward PLCγ1 with IC50 values of 7.5 μM. Lee et al. (1997)
activity
Tyrosinase inhibitory Possessed potent inhibitory activity on mushroom tyrosinase with IC50 Son et al. (2003); Sasaki
activity values of 0.6 mM. Besides, it was demonstrated to be noncompetitive et al. (2014)
inhibitors of protein tyrosine phosphatase 1B based on a kinetic
analysis with IC50 o30 mM.
Glycosidase inhibitory Showed strong β-amylase inhibitory activity with IC50 values of 57 μM, Kim et al. (2006)
activity and was shown to be noncompetitive inhibitor.
Aldose reductase Showed marked HRAR inhibitory activity with IC50 values of 0.27 μM. Jung et al. (2008)
inhibitory activity
DGAT inhibitory Dose-dependently inhibited DGAT activity with IC50 values of 9.8 μM. Chung et al. (2004)
activity
cGMP-PDE5 Showed potent cGMP-PDE5 inhibitory activity with IC50 values of Shin et al. (2002)
inhibitory activity 0.64 μM.
SGLT inhibitory Showed strong inhibitory activity against SGLT1 and SGLT2 at 50 μM Sato et al. (2007)
activity with inhibition rate of 71.1 % and 100 %, respectively.
104 Kuraridinol Kyogoku et al. (1973)
105 Kushenol D Antibacterial activity Inhibited the growth of S. aureus and S. mutans. Wu et al. (1985a); Yamaki
et al. (1990)
106 Xanthohumol Komatsu et al. (1970)
107 Cyclokuraridin Shen et al. (2006)
108 (Z)-4, 20 ,40 - trihydroxy chalcone Cao (2007)
Biflavonoids
109 Sophflavone A Cao (2007)
110 Sophflavone B Cao (2007)
Pterocarpans
111 Kushecarpin A Kuroyanagi et al. (1999)
112 Kushecarpin B Kuroyanagi et al. (1999)
113 Kushecarpin C Kuroyanagi et al. (1999)
114 Kushecarpin D Anti-angiogenesis Showed antiangiogenic activity via inhibitory effects on proliferation, Pu et al. (2013)
activity migration, adhesion and tube formation of ECV340 cell line.
Meanwhile, it down-regulated ROS levels and up-regulated catalase
activity.
115 Maackiain Neuraminidase Dose-dependently inhibited the activity of neuraminidase with IC50 of Zhao et al. (1993); Ryu
inhibitory activity 3.2 mM. et al. (2008)
116 l-maackiain Yagi et al. (1989)
117 4-methoxy-maackiain Zhao et al. (1993)
118 Maackiain -7-O-β-D- apiose -(1-6) Shen and Zhang(2012)
-β-D-glucopyranoside
119 Pterocarpin Neuraminidase Dose-dependently inhibited the activity of neuraminidase with IC50 of Ryu et al. (2008)
inhibitory activity 1.4 mM.
120 3-hydroxy-4-methoxy-8,9- Lin (2006)
methylenedioxypterocarpan
121 Trifolirhizin Antitumor activity Inhibited the proliferation of A549, SK-OV-3, SK-MEL-2, XF498 and Yagi et al. (1989); Ryu
HCT-15 with the ED50 of 16.3, 24.9, 15.0, 37.8 and 21.0 mg/mL. et al. (1997)
ASM relaxation effect Inhibited acetylcholine mediated ASM contraction or directly relaxes Yang et al. (2013)
pre-contracted ASM independent of β2-adrenoceptors.
122 Trifolirhizin-60 -monoacetate Li et al. (2004)
123 Kushenin Wu et al. (1985a)
124 Medicarpin-3-O-β-D- apiose -(1-6) Shen and Zhang(2012)
-β-D- glucopyranoside
Alkaloids
125 (þ ) -matrine Bohlmann et al.(1958)
126 (þ ) -isomatrine Ueno et al. (1975)
127 (þ ) -allomatrine Sekine et al. (1993)
128 cis-neomatrine Ueno et al. (1975)
129 trans-neomatrine Zhang et al. (2003)
130 (  )  9α-hydroxy-7, 11- Liu et al. (2010)
dehydromatrine
131 (þ ) -oxymatrine Bohlmann et al.(1958)
132 (þ ) -9α-hydroxymatrine Ding et al. (2006)
133 (1) -14β-hydroxymatrine Liu et al. (2010)
134 (þ ) -Sophoranol Bohlmann et al.(1958)
135 (þ ) -Sophoranol N-oxide Sekine et al. (1993)
136 (þ ) -oxysophoranol N-oxide Zhao et al. (1994)
137 13, 14-dehydroxysophoridine Ding et al. (2006)
138 (  )-sophocarpine Okuda et al. (1965)
139 (þ ) -oxysophocarpine Wang et al. (1996)
140 (  ) -sophocarpine N-oxide Sekine et al. (1993)
141 (  ) -9α-hydroxysophocarpine Ding et al. (2006);
142 9α-hydroxysophocarpine N-oxide Wang et al. (1996)
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 19

Table 1 (continued )

No. Compounds Biological activity Results References

143 ( þ) -12α-hydroxysophocarpine Ding et al. (2006)


144 ( þ) -sophoramine Sekine et al. (1993)
145 (  ) -13-ethylsophoramine Okuda et al. (1965)
146 Tetrahydroneosophoramine Ibragimov et al. (1981)
147 (  ) -cytisine Wang et al. (1981)
148 (  ) -methylcytisine Okuda et al. (1965)
149 (  ) -baptifoline Bohlmann et al. (1958)
150 (  ) -anagyrine Bohlmann et al.(1958)
151 Flavascensine Liu et al. (2010)
Triterpenoids
152 Lupeol Zhao (2008)
153 Lupenone Cao (2007)
154 Monogynol B Cao (2007)
155 β-amyrenol Cao (2007)
156 Soyasaponin I Yoshikawa et al.(1985)
157 Sophoraflavoside I Yoshikawa et al.(1985)
158 Sophoraflavoside II Ding et al. (1992)
159 Sophoraflavoside III Ding et al. (1992)
160 Sophoraflavoside IV Ding et al. (1992)
Others
161 Umbelliferone Zhang et al. (2000)
162 Citrusin A Shen and Zhang(2012)
163 Citrusin B Shen and Zhang(2012)
164 Alaschanioside A Shen and Zhang(2012)
165 Sophodibenzoside A Shen et al. (2013)
166 Sophodibenzoside B Shen et al. (2013)
167 Sophodibenzoside C Shen et al. (2013)
168 Sophodibenzoside D Shen et al. (2013)
169 Sophodibenzoside E Shen et al. (2013)
170 Sophodibenzoside F Shen et al. (2013)
171 Sophodibenzoside G Shen et al. (2013)
172 Sophodibenzoside H Shen et al. (2013)
173 Sophodibenzoside I Shen et al. (2013)
174 Sophodibenzoside J Shen et al. (2013)
175 Sophodibenzoside K Shen et al. (2013)
176 Sophodibenzoside L Shen et al. (2013)
177 Kusheearpin D Cao (2007)
178 Kushequinone Wu et al. (1986)
179 β-sitosterol Zhang et al. (2000)
180 2, 4-dihydroxy benzoic acid Li et al. (2004)
181 Tetracosanoic acid Zhang et al. (2000)

identified from Sophora flavescens. The constituents are classified 3.2. Alkaloids
into four groups: flavonoids (15 flavonols, 41 flavanones, 13 flavano-
nols, 27 isoflavones, 3 isoflavanones, 2 homoisoflavones, 7 chalcones, There are two other major groups of active constituents in
2 biflavonoids, and 14 pterocarpans), 47 alkaloids, 9 terpenoids and 26 Sophora flavescens. One of these groups of metabolites is alka-
other components (Tables 1 and 2). Among these compounds, loids, which have received a large amount of attention in recent
alkaloids and flavonoids are considered to be the major active decades in China. Currently, 27 alkaloids (125–151) have been
components of this Chinese medicine (Table 1, Figs. 2 and 3). isolated and identified from the roots of Sophora flavescens
(Table 1), and another 20 alkaloids have been reported from the
aerial parts, flowers and seeds of Sophora flavescens (Table 2).
Alkaloids are characteristic compounds of this plant and they can
3.1. Flavonoids be classified by structure into groups including matrine-type
alkaloids, cytisine-type alkaloids, anagyrine-type alkaloids, and
Flavonoids, which are important secondary metabolites, are wide- lupinine-type alkaloids. It is worth noting that the alkaloids, and
spread throughout the plant kingdom. Flavonoids and their derivatives especially the matrine-type alkaloids, have been identified as the
are the main bioactive components of Sophora flavescens, and they are bioactive component that contributes to a variety of pharmaco-
therefore receiving increased attention. A series of characteristic logical effects on cancers, hepatitis B and C, and cardiac diseases.
isoprenylated or lavandulylated flavonoids have been isolated from Modern pharmacological studies have shown that the most
this plant. To date, 124 flavonoids have been isolated from the roots of active of these components, matrine, has multi-pharmacological
Sophora flavescens. Among these compounds, kushenol A, B, C, E, F, H, effects, including significant antitumor, antiviral, anti-inflamma-
K, L, M, N, P, Q, R, S, T, U, V, W and X, kurarinone, isokurarinone, tory, hepatoprotective, antiarrhythmic, analgesic, antipyretic,
norkurarinone, (2S)-20 -methoxykurarinone, kurarinol, neokurarinol, anti- anaphylactic and antiasthma activities (Zhang et al., 2012;
norkurarinol, kosamol A, leachianone A, leachianone G, sophoraflava- Chang et al., 2013; Shao et al., 2013; Shi et al., 2013a; Tan et al.,
none G, 8-lavandulylkaempferol, formononetin, and isoxanthohumol 2013; Deng et al., 2014; Huang et al., 2014; Liu et al., 2014; Shao
have been confirmed to possess various pharmacological activities et al., 2014; Zhou et al., 2014; Gong et al., 2015; Wang et al.,
in vivo and in vitro (Table 1 and Fig. 2). 2015). Oxymatrine has, in particular, been demonstrated to be
20 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

Table 2
The compounds isolated from the aerial parts, flowers and seeds of Sophora flavescens.

No. Compounds Resource References

Alkaloids
1 Matrine-N-oxide Seeds Sekine et al. (1993)
2 (  ) -7, 11-dehydromatrine Flowers Murakoshi et al. (1982)
3 ( þ ) -5α,9 α-hydroxymatrine Flowers, Seeds Murakoshi et al. (1982); Sekine et al. (1993)
4 (  )-Leontalbinine N-oxide Seeds Sekine et al. (1993)
5 (  ) -sophoridine Aerial parts Morinaga et al. (1978)
6 ( þ ) -isosophoridine Aerial parts Morinaga et al. (1978)
7 Isosophocarpine Aerial parts Morinaga et al. (1978)
8 5epi-sophocarpine Aerial parts Sekine et al. (1993)
9 (  ) -5α-hydroxysophocarpine Seeds Saito et al. (1990)
10 ( þ ) -sophoramine-N-oxide Aerial parts Ueno et al. (1978)
11 (  ) -9α- hydroxysophoramine Flowers, Aerial parts Murakoshi et al. (1982); Sekine et al. (1993)
12 (  )-△7-dehydrosophoramine Aerial parts Ueno et al. (1978)
13 (  ) -N-methylcytisine Aerial parts Ueno et al. (1978)
14 (  ) -rhombifoline Flowers Murakoshi et al.(1982)
15 ( þ ) -manmanine Flowers Ibragimov et al. (1981)
16 ( þ ) -kuraramine Flowers Ibragimov et al. (1981)
17 Isokuraramine Flowers Murakoshi et al.(1982)
18 Lupanine Flowers Sekine et al. (1993)
19 (-) -5, 6-dehydrolupanine Flowers Sekine et al. (1993)
20 ( þ )-lehmannine Aerial parts Sekine et al. (1993)
Others
21 Syringin Stems, Leaves Zhang et al. (2013)
22 Corchionoside C Stems, Leaves Zhang et al. (2013)
23 Coniferin Stems, Leaves Zhang et al. (2013)
24 Benzyl O-β-D-glucopyranoside Stems, Leaves Zhang et al. (2013)
25 Piscidic acid Stems, Leaves Zhang et al. (2013)

effective as an anti-inflammatory, antitumor, antihypertensive, flavescens (Shen and Zhang, 2012). In a study by Shen et al. (2013),
antioxidant, antiapoptotic, and antiviral, and at protecting hepa- twelve new dibenzoyl derivatives (sophodibenzoside A-L) were been
tocytes, inhibiting the immune reaction and reducing hepatic purified from the n-butanol fraction of Sophora flavescens for the first
fibrosis (Yamazaki et al., 1984; Dong et al., 2013; Shi et al., 2013b; time. Several other compounds, including syringin, corchionoside C,
Wang et al., 2013; Liu et al., 2014; Zhang et al., 2014a, 2014b; Wei coniferin, piscidic acid and benzyl O-β-D-glucopyranoside have also
et al., 2014; Guo et al., 2015; Wu et al., 2015). The derivatives been isolated from the stems and leaves of this plant (Zhang et al.,
( þ)-oxysophocarpine, ( )-sophocarpine, and ( þ)-lehmannine, 2013).
at a dose of 0.2 mmol/mL, have shown significant anti-HBV
activity with inhibitory potency against HBsAg secretion at
48.3%, 57.2%, 52.6%, respectively, and against HBeAg secretion at 4. Methods of quality control
24.6%, 34.6%, and 25.4%, respectively (Ding et al., 2006). Thus, of
these alkaloids, matrine and oxymatrine have most frequently Quinolizidine alkaloids and flavonoids are considered to be the
been used as a quality control marker for Sophora flavescens and main active components of Sophora flavescens. In the 2010 edition
its medicinal extracts and preparations (Chinese Pharmacopoeia of the Chinese Pharmacopoeia, only the main alkaloids matrine
Commission, 2010). Moreover, the China State Food and Drug and oxymatrine are included as standards for the evaluation of
Administration database of drug manufacturing certificates Kushen quality in the pharmaceutical market. According to this
currently lists 20 clinical uses for matrine and oxymatrine source, the matrine and oxymatrine content, analyzed by HPLC in
preparations (http://www.sda.gov.cn/WS01/CL0001/). These the roots of Sophora flavescens, should both be no less than 1.2%
pharmaceutical products, including suppositories, tablets, gran- (Chinese Pharmacopoeia Commission, 2010). Over the past few
ules, capsules, and injectable dosage forms, have been used in years, various different methods, including TLC, HPLC (Li and
China for the treatment of cancer and infectious and cardiac Wang, 2004), and capillary electrophoresis (Wu et al., 2006a,
diseases (Liu et al., 2001; Long et al., 2004; Lao, 2005). 2006b; Wang et al., 2011), have been used to describe the chemical
constituents and to control the quality of derivatives isolated from
3.3. Triterpenoids the plant. However, interactions between multiple chemical com-
pounds may contribute to the therapeutic effects observed in
So far, only nine triterpenoid compounds have been purified herbal medicine. Recently, therefore, the analysis of multiple
and characterized from the roots of Sophora flavescens. These are components (mainly matrine-type alkaloids and flavonoids) is
lupeol, lupenone, monogynol B, β-amyrenol, soyasaponin І, and increasingly used to validate the quality of this herb. In 2004,
sophoraflavoside І, II, III and IV (Yoshikawa et al., 1985; Ding et al., the use of reversed-phase HPLC was developed and validated for
1992; Cao, 2007; Zhao, 2008). the analysis of three bioactive alkaloids, including matrine,
sophoridine and oxymatrine, derived from Sophora flavescens.
3.4. Other compounds The analysis was performed using a Kromasil C18 column
(250 mm  4.6 mm i.d.; particle size 5 mm) with a gradient elution
A range of other compounds, including some lignans, phenylpro- of 0.01 mol/L KH2PO4 buffer-methanol-triethylamine in the ratio
panoids, coumarins, and phenolic acids, have also been isolated from 94:6:0.01 (v/v) and detected by ultraviolet absorbance at 208 nm.
the roots of Sophora flavescens (Wu et al., 1986; Zhang et al., 2000; Li The analysis time was 24 min per injection. The calibration for
et al., 2004). Three lignans, including citrusin A, citrusin B and matrine, sophoridine and oxymatrine was linear and ranged from
alaschanioside A, have been identified in the roots of Sophora 0.2–120.0 μg/mL, 0.2–115.2 μg/mL and 0.2–110.4 μg/mL,
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 21

Fig. 2. The chemical structure of some active flavonoids from Sophora flavescens.

respectively (Li and Wang, 2004). Wu et al. (2006a, 2006b) flavanones, 4 chalcones, 4 pterocarpans, 3 isoflavones, 1 isoflava-
established a simple and accurate capillary electrophoresis none, 1 flavanonol and 1 flavonol were identified or tentatively
method for the characterization of four quinolizidine alkaloids characterized by an Agilent 1100 HPLC system and Agilent 1100
(matrine, oxymatrine, sophoridine and sophocarpine) in Sophora series SL ion trap mass spectrometer (Zhang et al., 2007).
flavescens, as follows. Electrophoretic separation was performed on
a 75 μm i.d. uncoated fused-silica capillary (65 cm length with
56 cm effective length) with a running buffer of aqueous solution 5. Effects of crude extract
composed of 60 mmol/L sodium borate, and the limits of detection
for the four alkaloids were determined to range from 8.8 to 5.1. Antitumor activity
48.0 μg/mL. We suggest that flavonoids should also be added into
the quality control system of Sophora flavescens due to their Sophora flavescens extract has demonstrated obvious antitumor
favorable and crucial pharmacological activities. Fortunately, a activity in vitro and in vivo and has been commonly used in
powerful and new method that couples HPLC-DAD-ESI with MS Chinese herbal medicines to treat cancer in clinical practice. The
has been established, providing a chemical mechanism for the antitumor activity of Sophora flavescens has been confirmed in SK-
separation and characterization of flavonoids in Sophora flavescens. OV-3 (ovary), A549, SK-MEL-2 (skin), HT29, NCI-H460 (non-small
Using this method, a total of 24 flavonoids, including 10 cell lung), XF498 (central nerve system), HCT-15 (colon), HL-60
22 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

Fig. 2. (continued)

(myeloid leukemia), and SPC-A-1 (lung) cell lines, in mouse et al., 1966; Li et al., 2002; Sun et al., 2007, 2008). Experimental
models, including S180, H22 and Lewis lung tumors, and in nude studies have explored the systematic activities of Sophora flaves-
mice with human H460 and Eca-109 xenografted tumors (Abbott cens and its antitumor mechanisms, including inducing apoptosis,
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 23

O O
O O O
H H
H N H N
N N N
H H
H H H
H H H H
H H N H H N H H
N N N
O O
Matrine Oxymatrine (-)-sophocarpine (+) -oxysophocarpine (+) -lehmannine
Fig. 3. The chemical structure of main active alkaloids from Sophora flavescens.

inducing cancer cells to differentiate to normal cells, inhibiting weights after 14 days treatment with 55 mg/kg decreased to
certain enzyme activities, inhibiting cancer cell DNA synthesis, 0.3170.09 g and 0.5370.31 g, respectively, a reduction of 49.18%
affecting cell cycle arrest and tumor metastasis, controlling the and 38.37%, respectively, whereas in the positive control group
expression levels of tumor-related factors, and affecting telomer- (administered cisplatin), the inhibitory ratio was 88.52% and
ase activity. However, it is important to note that most of the 79.07%, respectively. It was not determined whether NF-κB, ERK,
research performed to study antitumor activity has employed p53, and p21 proteins were also involved in SFL-induced apoptosis in
in vitro-based methods and that further in vivo tests should nude mice (Shi et al., 2014).
therefore be encouraged.
In vitro, the aqueous extract of Sophora flavescens, at concentra-
tions of 100 and 200 μg/mL, inhibited growth in Ehrlich ascites 5.2. Anti-inflammatory and antinociceptive activity
carcinoma (EAC) cell lines with an inhibition rate of 50% and 66%,
respectively (Li et al., 2002). An ethanol extract of Radix Sophora In vivo and in vitro studies have shown that Sophora flavescens
flavescens (95% ethanol, solid:liquid ratio of 1:9, 80 min heat inhibits inflammatory reactions and suppresses inflammatory
reflux) demonstrated significantly inhibited proliferation of colon factors, including the inhibition of production of COX-2, iNOS,
cancer HT29 cells with relatively high inhibition rates, as analyzed NO, IL-8, IL-6 and TNF-α. Administration of 200 mg/kg of Sophora
by MTT assay (Xiao et al., 2013). The total extract, which had a cell flavescens dried root extract inhibited the mast cell-mediated
inhibition rate of 52%, must contain the matrines and flavonoids passive cutaneous anaphylaxis reaction in vivo and reduced
that have been described in the literature. Additionally, a compound 48/80-induced histamine release from rat peritoneal
mannose-binding lectin isolated from Sophora flavescens (SFL) mast cells. In addition, this crude extract directly decreased the
markedly inhibited the growth of MCF-7 cells in a dose- and expression of phorbol 12-myristate 13-acetate (PMA) and also
time-dependent manner and with an IC50 value of 20 μg/mL. decreased stimulation of TNF-a, IL-6, and IL-8 by the calcium
Concentrations of 12.5, 25, 50, and 100 μg/mL SFL induced an ionophore A23187, indicating that Sophora flavescens may be a
apoptotic morphology, dose-dependently increased the sub-G1 source for new drugs that can treat mast cell-derived allergic
proportion of cells and triggered G2/M phase cell cycle arrest in inflammatory diseases (Hong et al., 2009). In a 1-fluoro-2,4-
the MCF-7 cell line. SFL also dose-dependently increased the dinitrofluorobenzene (DNFB)-induced mouse model of contact
activity of both caspase-3 and caspase-9 and the release of dermatitis, 10 mg/mL of dried root of Sophora flavescens (SR)
cytochrome C from mitochondria into the cytoplasm, up- effectively inhibited the enlargement of ear thickness and weight
regulated Bax and Bid, and down-regulated Bcl-2 and Bcl-XL in that is normally induced by repeated application of DNFB. Topical
MCF-7 cells. Western blot data demonstrated that treatment of application of SR also inhibited hyperplasia, edema, spongiosis and
MCF-7 cells with SFL resulted in down-regulation of protein levels infiltration of mononuclear cells in ear tissue. In addition, SR
of NF-κB and ERK and up-regulation of protein levels of p53 and treatment (10 mg/mL) effectively reduced production levels of
p21. Finally, SFL showed a strong cytotoxic effect in HeLa cells by IFN-γ and TNF-α in vivo (Kim et al., 2012). SR at concentra-
inducing apoptosis in a time- and dose-dependent manner, and tions of 50, 100 and 200 mg/mL also inhibited histamine and
SFL from 0.01 to 5 mg/mL exerted a potent inhibitory effect on the β-hexosaminidase release and migration in an in vitro model using
growth of HeLa cells (Liu et al., 2008). RBL-2H3 cells. Finally, SR was also effective at reducing the
In vivo, the total flavonoids isolated from Sophora flavescens Pseudomonas aeruginosa-induced expression of IL-1β in lung
(200 mg/kg, i.v.) inhibited growth in five cell lines, including H22 epithelial cells and macrophages (Lee et al., 2014).
liver cancer, sarcoma 180 (S180), Lewis lung cancer, human esopha- Oral administration of 10–100 mg/kg/day of the alkaloid-free
geal squamous carcinoma Eca-109 and non-small cell lung cancer prenylated flavonoid-enriched fraction (PFS) of rhizomes of
H460 cell lines, with inhibitory rates of 40%, 82.14%, 59.74%, 42.71% Sophora flavescens significantly inhibited arthritic inflammation
and 46.8%, respectively, and with an oral or intravenous maximum in Wistar rats in a dose-dependent manner. When paws were
tolerated dose of the extract of more than 2.8 g/kg or 0.75 g/kg, treated with heat-killed Mycobacterium butyricum, PFS (100 mg/
respectively (Sun et al., 2008). The Sophora flavescens polysaccharide kg/day) inhibited paw volume by 32.8% and 31.8% at 14 and 18
promoted splenocyte proliferation and significantly strengthened days, respectively, while in non-treated paws, PFS inhibited paw
peritoneal macrophages, which digest H22 tumor cells. The anti- volume by 92.8% and 43.9% at 14 and 18 days, respectively. It was
tumor effect of this polysaccharide may therefore be associated with suggested that this inhibition was due to multiple actions by
its potent immunostimulatory effect (Bai et al., 2012). Sophora proinflammatory enzymes and cytokines, including COX, iNOS and
flavescens polysaccharide was administered once per day (p.o.) for IL-6 (Jin et al., 2010). PFS also inhibited cyclooxygenase-2 (COX-2)-
14 consecutive days at 25, 50 and 100 mg/kg to H22 tumor-bearing catalyzed PGE2 and inducible nitric oxide synthase (iNOS)-cata-
mice. This treatment effectively inhibited tumor growth with an lyzed NO production in lipopolysaccharide (LPS)-treated RAW
inhibitory rate of 29.02%, 35.96%, and 46.06%, respectively, and the 264.7 cells at doses ranging from 10 to 50 mg/mL. These effects
average survival time was prolonged at least 1.9, 2.9 and 3.6 times, occurred primarily through COX-2 inhibition and iNOS down-
respectively, compared to the control group. Intraperitoneal admin- regulation, respectively. Finally, PFS also inhibited production of
istration of 55 mg/kg of SFL significantly decreased subcutaneous IL-6 and TNF-α (Jin et al., 2010). We suggest that these antiarthritic
tumor mass volume and weight in MCF-7 bearing mice. Tumor properties of Sophora flavescens might explain its use in local and
24 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

traditional treatments for rheumatism, chronic bronchitis and 5.4.2. Antiviral activity
chronic hepatitis. Oral administration of Sophora flavescens has been used in
PFS has also displayed a significant antinociceptive effect in an remedies against infectious viral diseases in China, but existing
acetic acid-induced writhing model. PFS, administered orally at 50, studies have been performed using mixtures instead of pure
100 and 400 mg/kg, potently inhibited acetic acid-induced wri- compounds. An ethyl acetate extract demonstrated weak antiviral
thing in mice, yielding an inhibition rate of 46%, 63% and 68.8%, activity against Herpes simplex virus types I and II (HSV-1 and 2),
respectively. The inhibition rate of PFS at 400 mg/kg was stronger with EC50 values of 4125 mg/mL and 93.3 712.5 mg/mL, respec-
than that of aspirin at 100 mg/kg (Jin et al., 2010). Advanced tively (Woo et al., 1998). Ma et al. (2002) selected forty-four
studies should be performed to investigate these biochemical medicinal herbs that are used for the treatment of respiratory
pathways and to develop better antinociceptive therapeutic tract infectious diseases in China, and tested the antiviral activities
agents. of these herbs against respiratory syncytial virus (RSV) using the
cytopathogenic effect assay. The aqueous extract of Sophora
flavescens roots showed potent antiviral activity against RSV, with
an IC50 of 50.0 mg/mL and a selectivity index of 16 (Ma et al., 2002).
5.3. Antianaphylaxis and antiasthma activity
In 2014, Alfajaro et al. used a porcine rotavirus G5[P7] strain to
examine the antirotaviral activities of Sophora flavescens extracts
One of the most common uses of Sophora flavescens root in
(SFE) and stevioside, either singly or in various combinations,
local medicine is the treatment of allergic diseases, including
in vitro and in vivo. The results suggested that drug-mixtures of
atopic dermatitis and asthma. Several reports have been carried
SFE and stevioside inhibited in vitro virus replication more
out to support these ethnobotanical applications. In a mouse
efficiently than each single treatment. Moreover, in a piglet model,
model of atopic dermatitis, methanol extract of Sophora flavescens,
SFE improved rotavirus enteritis but did not completely cure it.
administered perorally (p.o.) at a dose of 50–200 mg/kg, dose-
Interestingly, piglets given a combination therapy of 1.6 g SFE and
dependently and significantly inhibited a serotonin (5-HT)-
12 g stevioside displayed a rapid decrease in viral copy number, a
induced itch-related response (scratching) and spontaneous
marked improvement in small intestinal lesion scores and fecal
scratching in NC mice (Yamaguchi-Miyamoto et al., 2003). In a
virus shedding, and a reduction in diarrhea (Alfajaro et al., 2014).
separate study, Sophora flavescens was administered intragastri-
cally at a dose equivalent to a standard human adult dose twice
daily for 17 days in a murine model of allergic asthma. This 5.4.3. Insecticidal activity
treatment significantly reduced the development of airway hyper- A number of pharmacological tests and clinical observations
reactivity (AHR), reduced airway inflammation, and decreased have shown that different extract and/or compound prescriptions
serum IgE, IL-4 and IL-5 levels in splenocyte cultures of derived from Sophora flavescens have significant insecticidal effects
conalbumin-treated mice (Wen et al., 2004). The antiasthma against diseases and organisms including trichomoniasis, schisto-
activity of water extracted from Sophora flavescens was then somiasis, and Toxoplasma gondii. The methanolic extract of Sophora
investigated. This crude extract significantly reduced allergic flavescens had high anti-T. gondii activity (EC50 ¼0.2 mg/mL) and
asthma airway hyperreactivity in mice and inhibited acetylcholine high selective toxicity (Choi et al., 2008). The alcoholic extract of
(ACh) induced airway smooth muscle (ASM) contractions in Sophora flavescens from South Korea was effectively used in vitro to
tracheal rings in allergic asthmatic mice (Yang et al., 2013). reduce the replication of Toxoplasma gondii and Neospora caninum
(Youn et al., 2003). Sophora flavescens inhibited T. gondii prolifera-
tion by 98.7%, 83.0% and 27.2%, in a range from 0.156 to 0.039 mg/
mL. Sophora flavescens inhibited N. caninum proliferation by 98.6%,
5.4. Antimicrobial activity 97.0%, 69.5% and 14.0% in the range from 0.16 to 0.02 mg/mL. The
active antineosporal constituents in Sophora flavescens extracts
5.4.1. Antibacterial activity were sophoridane, furosardonin A, and tetraisopropylidene-
The roots of Sophora flavescens have been used as traditional cyclobutane (Seo et al., 2013). The aqueous extract of Sophora
medicines for microbial infections for thousands of years, and they flavescens roots demonstrated inhibitory effects against Eimeria
have outstanding potential as antimicrobial agents. Di et al. (2006) tenella (Youn and Noh, 2001) and Giardia lamblia (Zhu et al., 2005).
showed that an aqueous extract of Sophora flavescens roots
exhibited significant antibacterial activity against Escherichia coli,
Staphylococcus aureus, Streptococcus, Bacillus dysenteriae, Salmo- 5.4.4. Antifungal activity
nella and Bacillus proteus. This crude Sophora flavescens extract also In 2012, Liu et al. found that acetone extract of Sophora
exhibited remarkable inhibitory activity against the methicillin- flavescens at a concentration of 0.2 g/mL had in vitro antifungal
resistant Staphylococcus aureus and methicillin-sensitive Staphylo- activity against Candida albicans and Saccharomyces cerevisiae
coccus epidermidis, with MIC50 values of 286 and 143 mg/mL, growth, with inhibition rates of 33% and 23%, respectively. These
respectively, and MIC90 values of 1185 and 286 mg/mL, respectively, results provided more scientific evidence supporting the clinical
displaying particularly strong antibacterial activity against Staphy- use of these plants in Chinese popular medicine (Liu et al., 2012).
lococcus epidermis (Yang et al., 2007). Moreover, using double Moreover, the flavonoids of Sophora flavescens were demonstrated
dilution methods, total alkaloids, total flavonoids and mixtures of to have antifungal activity against Aspergillus niger, Penicillium
these compounds from Sophora flavescens potently inhibited chrysogenum and Saccharomyces cerevisiae, with MIC values of
Pseudomonas aeruginosa, Escherichia coli, Staphylococcus aureus, 0.28, 0.56 and 0.56 g/L, respectively (Zheng et al., 2008).
Shigella, Shigella flexneri, Staphylococcus citreus, Staphylococcus
albus and Bacillus paratyphosus B in vitro with MIC values of 9.0, 5.5. Effect on the cardiovascular system
3.38, 8.25, 9.0, 4.50, 13.50, 6.75, 7.13 mg/mL, 3.0, 2.63, 3 56, 3.38,
1.50, 2.44, 3.56, 2.35 mg/mL and 2.25, 2.07, 3.28, 3.94, 1.22, 2.44, 5.5.1. Antiarrhythmic activity
2.07, 1.97 mg/mL, respectively (Du et al., 2010). These findings A total alkaloid solution derived from Sophora flavescens dis-
suggest that antibacterial activity is an important property of played preventive effects against chloroform- and isoproterenol-
Sophora flavescens and that this plant is a potential source of induced arrhythmia in anesthetized cats. It also significantly
preservatives for the food and pharmaceutical industries. improved arrhythmia caused by ouabain, barium chloride and
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 25

strophanthin in neonatal rats (Zha et al., 1981; Zhang et al., 1985). consecutive 14 days significantly strengthened peritoneal macro-
In addition, the total flavonoids solution derived from Sophora phages that ingest H22 tumor cells and stimulated macrophages to
flavescens had a significantly preventive effect against chloroform- produce nitric oxide (NO) by up-regulating inducible NO synthase
induced atrial fibrillation in mice, and it showed the same (iNOS) activity, suggesting that the polysaccharide is a strong
preventive effect in a rabbit model of arrhythmia caused by natural immunomodulator (Bai et al., 2012). Advanced studies
chloroform and/or isoproterenol. In vitro cultured myocardial cell should be performed to explore the composition and structure of
experiments showed that the total flavonoids (125–250 mg/mL) polysaccharide derived from this plant with a goal of developing a
derived from Sophora flavescens significantly slowed down spon- potent new immunomodulator.
taneous pulsation frequency in suckling mouse myocardial cells
and improved strophanthin-induced arrhythmia (Wang et al., 5.7. Other activities
1981, 1983).
A number of extracts from Sophora flavescens have reported
5.5.2. Antimyocardial fibrosis activity roles in the regulation of hair growth, skin whitening, and the
Total flavonoids derived from Sophora flavescens inhibited lowering of cholesterol, in addition to documented neuroprotec-
myocardial fibrosis and protected the myocardium in rats. In an tive effects. The methanol extract of this herb displayed a strong
isoprenaline (5 mg/kg)-induced myocardial fibrosis rat model, 200 hair growth promoting effect. It also induced an earlier conversion
and 400 mg/kg flavonoids from Sophora flavescens markedly of telogen-to-anagen in female C57BL/6 mice and affected mRNA
decreased the activity of CK in blood serum and increased SOD levels of growth factors such as IGF-1 and KGF in human hair
activity in the left ventricle. It also markedly reduced hydroxypro- dermal papilla cells. Moreover, the extract displayed a potent
line and angiotensin II levels in the left ventricle and increased NO inhibitory effect on type II 5α-reductase activity and thus could
content and decreased MDA content in blood serum (Fan et al., potentially be used in hair growth products (Roh et al., 2002). The
2013). ethanol extract showed a whitening effect that acted by strongly
inhibiting tyrosinase activity, with an IC50 value of 10.4 μg/mL.
5.5.3. Effect on myocardial contractility DNA microarray, immunohistochemistry and immunofluorescence
The ethanol extract of Sophora flavescens decreased atrial ANP analyses showed that the extract dramatically down-regulated
secretion concomitant with an increase in atrial dynamics, includ- melanosomes at both the mRNA and protein level in keratinocytes.
ing stroke volume and pulse pressure, in euthyroid atria of sham- In a clinical trial of a formula containing 0.05% Sophora flavescens
extract used for 8 weeks on human skin, the formula had a
treated rabbits. However, in rabbits with hyperthyroid atria
induced by daily administration of sodium L-thyroxine (0.5 mg/ significant effect on skin whitening, as evaluated both mechani-
cally and visually (Shin et al., 2013). The ethanol extract also
kg) for 12 days, the SF-induced decrease in atrial ANP secretion
was attenuated, and the SF-induced increase in atrial dynamics reduced the short circuit current in forskolin-activated rat ileal
epithelia, measured using an Ussing chamber and voltage  cur-
was heightened (Wen et al., 2009). Moreover, the ethanol extract
of Sophora flavescens decreased cardiac parameters including LVSP, rent technique, implying that this extract may affect ion transport
in the rat ileum epithelia and that this may be critical to the
LVEDP, þ dp/dtmax,  dp/dtmax and heart rate in both euthyroid and
hyperthyroid groups (Wen et al., 2011). therapeutic effects of antidiarrheal agents (Tsai et al., 2004).
The ethyl acetate (EtOAc) extract from Sophora flavescens
displayed antihyperlipidemic effects. When orally administered
5.5.4. Vascular relaxing and antimyocardial hypoxic activity at doses of 250 and 100 mg/kg, once daily for 3 days, to poloxamer
The ethanol extract of the roots of Sophora flavescens 407-induced hyperlipidemic rats, the extract significantly
(0.1–100 mg/mL) induced relaxation of the phenylephrine-precon- increased HDL-C levels and decreased total cholesterol (TC),
tracted aortic rings in isolated rat aorta in a dose-related manner. triglycerides (TG), and low-density lipoprotein-cholesterol (LDL-C)
These vasorelaxation effects were mediated by the release of NO levels in rat serum (Kim et al., 2008). It could therefore be used as
from endothelium and activation of sGC-cGMP signaling (Jin et al., an effective cholesterol- lowering agent and could also be useful
2011). Intragastric administration of the aqueous extract of for preventing hypercholesterolemic atherosclerosis. Furthermore,
Sophora flavescens at a dose of 10 and 20 g/kg significantly an EtOAc extract enriched in flavonoids from the root of Sophora
prolonged survival time (by 26.08% and 45.85%, respectively) and flavescens had neuroprotective effects in a model of focal cerebral
reduced oxygen consumption in isoprenaline-induced hypoxic ischemia. The flavonoids in this formula might therefore be
cardiomyocyte mice (Xu et al., 2008). The aqueous extract of responsible for its neuroprotective activity in rats, either alone or
Sophora flavescens exhibited a positive antimyocardial hypoxic in part (Park et al., 2009).
effect, but the dosage used in this study was excessive, equaling
a human equivalent dosage of 813 mg/kg, which would be
equivalent to a dose of approximately 48 g aqueous extract/day 6. Toxicity
for a 60 kg person. Therefore, the protective effect of this extract
against myocardial ischemia remains to be investigated. Despite the long use of Sophora flavescens root in traditional
medicine to treat numerous diseases, it has also known been known
5.6. Immunoregulatory activity to be harmful when used at an excessive dosage. The alkaloids from
Sophora flavescens, in particular, cause spasms and palsies of the
Studies have shown that root extracts of Sophora flavescens may respiratory center in both cold- and warm-blooded animals. The
have immunoregulatory effects and that they could be recom- intravenous administration of the sophocarping of Sophora flavescens
mended for use as a potential supplements to enhance other at doses of 150 mg/kg in rabbits caused venous congestion, lowered
treatments for immunity and prevention of various diseases. A diet blood pressure, breathing difficulties and death. The injection of
supplement containing 0.025%, 0.05%, 0.1%, 0.2%, or 0.4% Sophora matrine into rabbits was found to induce central nervous paralysis
flavescens significantly increased serum lysozyme, antiprotease and spasms, with a minimum lethal dose of 0.4 g/kg. An intramuscular
activity, natural hemolytic complements and cellular myeloperox- injection of oxymatrine in mice was found to be lethal at 256.747
idase activity (Wu et al., 2013). Administration (p.o.) of 25, 50 and 573.6 mg/kg, while its intravenous LD50 value was 144.2722.8 mg/kg.
100 mg/kg of Sophora flavescens polysaccharide once per day for An injection of matrine to a frog could induce slow and irregular
26 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

breathing, and sometimes respiratory arrest resulting in death. The the various biological properties and traditional uses of the plant
LD50 values of matrine administered orally or by subcutaneous in herbal medicine. However, there are also a number of points
injection in mice were found to be 1.18 g/kg70.1 and 571.27 and aspects that need to be improved and researched further: b
48.8 mg/kg, respectively (State Administration of Traditional Chinese Limited work has been performed on the aerial parts of the
Medicine, 1999). In addition, administration of larger doses of matrine Sophora flavescens plant, and further extensive phytochemical
in mice caused irritability, restlessness and spasmodic tics. Matrine investigation on these are necessary. (ii) The flavonoids, and
may also inhibit sperm activity and lead to sperm malformation in especially lavanduly and prenyl flavonoids, have received heigh-
mice after long-term oral administration (Su et al., 2007). However, no tened public attention due to their hypotoxicity and high medic-
significant poisoning symptoms were found in dogs treated with an inal value, and further research on these compounds should be a
intramuscular injection of matrine (200 mg/kg) and Sophora flavescens priority. More specifically, the mechanisms responsible for the
extract (100 mg/kg, 13 days). An intramuscular injection of matrine anti-tumor, anti-inflammatory, antibacterial and PLCγ1 inhibitory
was also non-toxic to pigeons at a dose of 100 mg/kg (Xia et al., 1979; effects of these flavonoids need to be fully defined and studied. We
State Administration of Traditional Chinese Medicine, 1999). also suggest that the active flavonoids should be used as controls
Compared to alkaloids, the crude extract and flavonoids derived in studies of Sophora flavescens compounds. (iii) Although Sophora
from Sophora flavescens have less pronounced side effects, and related flavescens root is officially listed in the Chinese Pharmacopoeia and
systematic toxicity and safety evaluations have been rare. The oral and is used widely and successfully in clinics across China, the
intramuscular injection LD50 values of the extract of Sophora flavescens experimental animal studies and randomized clinical trials invol-
roots in mice were found to be 14.5 g/kg and 14.4 g/kg, respectively. ving this plant and its derivatives are still in the exploration stage,
The intravenous LD50 value of the total flavonoids of Sophora flavescens and the exact mechanisms for their therapeutic effects are
in mice was found to be 103.177.66 g/kg. Furthermore, treatment unknown. (iv) Although matrine and oxymatrine compounds
with the ethanol extract of Sophora flavescens at 1.25 g/kg or 2.5 g/kg derived from Sophora flavescens have been used as effective
twice daily for 3 days significantly elevated alanine aminotransferase human drug products for the treatment of various diseases, there
and aspartate aminotransferase levels in the serum. The changes in the are more than 50 other main constituents with prominent
levels of transaminases were supported by the remarkable fatty biological activity, including other alkaloids, lavanduly and pre-
degeneration observed in liver histopathology. Histopathological nylated flavonoids, flavonols, and flavanones, that have not been
photomicrographs of the liver showed remarkable fatty degeneration, developed for use in clinical practice. Further research should be
with numerous micro and macro vesicles in the portal area of the encouraged to study these components or their analogues. We
2.5 g/kg ESF treated rats compared to the control group. The 1.25 g/kg expected that more safe, effective and quality-controlled compo-
ESF-treated group exhibited moderate fatty infiltration. Further inves- nents will be widely used in the pharmaceutical, food, and
tigations using bioassay-guided isolation and analysis indicated that cosmetics industries in the future. (v) Because the Sophora
prenylated flavanones accounted for the positive hepatotoxic results flavescens plant contains potentially toxic compounds, reliable
(Yu et al., 2013). analytical methods are required for proper quality control of
The above analysis shows that studies of the side effects and product development to ensure that potentially toxic components
toxicity associated with the use of Sophora flavescens are not are kept below tolerance levels in Sophora flavescens products.
systematic or sufficiently comprehensive. To guard against toxic Future studies should include the identification of any side effects
effects, the 2010 edition of the Chinese Pharmacopoeia describes and/or toxicity.
an exact dosage of the root as 4.5–9 g and includes this precau-
tion: incompatible with the root and rhizome of Veratrum nigrum
L. (Chinese Pharmacopoeia Commission, 2010). Although daily References
clinical administration of this extract usually does not result in
any significant adverse effects in humans, systemic toxicity and Abbott, B.J., Hartwell, J.L., Leiter, J., Perdue Jr., R.E., Schepartz, S.A., 1966. Screening
data from the cancer chemotherapy national service center screening labora-
safety issues are in urgent need of further attention and study. tories. Cancer Res. 26, 1461–1656.
Large scale clinical trials are also required to assess the potential of Alfajaro, M.M., Rho, M.C., Kim, H.J., Park, J.G., Kim, D.S., Hosmillo, M., Son, K.Y., Lee, J.H.,
this extract or its derivates as an alternative to current treatments Park, S.I., Kang, M.I., Ryu, Y.B., Park, K.H., Oh, H.M., Lee, S.W., Park, S.J., Lee, W.S., Cho,
K.O., 2014. Anti-rotavirus effects by combination therapy of stevioside and Sophora
of tumors and infectious diseases.
flavescens extract. Res. Vet. Sci. 96, 567–575.
Bae, K., 2000. Bae, K. (Ed.), Medicinal Plants of Korea. Kyo-Hak Pub, Seoul, p. 261.
Bai, L., Zhu, L.Y., Yang, B.S., Shi, L.J., Liu, Y., Jiang, A.M., Zhao, L.L., Song, G., Liu, T.F.,
7. Future perspectives and conclusions 2012. Antitumor and immunomodulating activity of a polysaccharide from
Sophora flavescens Ait. Int. J. Biol. Macro-mol. 51, 705–709.
Bohlmann, F., Rahtz, D., Arndt, C., 1958. Lupinen-alkaloide, XI. Die Alkaloide aus
The present review summarizes the multifaceted uses and recent Sophora flavescens. Chem. Ber. 91, 2189–2193.
findings regarding studies of the phytochemistry, traditional use, Cao, M.A., 2007. Studies on the chemical constituents and bioactivities of Sophora
flavescens Ait. [D]. Dissertation of Master Degree of Lanzhou University, pp. 1–3.
pharmacology and toxicity of different extracts and compounds of Chang, C., Liu, S.P., Fang, C.H., He, R.S., Wang, Z., Zhu, Y.Q., Jiang, S.W., 2013. Effects of
Sophora flavescens. Sophora flavescens has been traditionally and matrine on the proliferation of HT29 human colon cancer cells and its
popularly used in various indigenous populations to treat disease. antitumor mechanism. Oncol. Lett. 6, 699–704.
Chen, L., Liu, Y., Liang, S.W., 2011. Study on the chemical constituents of Sophora
Recent reports have focused primarily on evaluating the anticancer, flavescens. J. Guangdong Pharm. Univ. 27, 471.
antibacterial, anti-inflammatory, and antiasthmatic activities of the Chen, Y.Z., Li, Y.Q., Wang, W., Zhang, Y., Li, P.P., 2012. Fufang Kushen injection in
herbal medicines derived from this plant. The alkaloids and flavonoids treatment of mild or moderate cancer pain. J. Beijing Univ. Tradit. Chin. Med. 35,
61–69.
are the two major groups of plant derivatives, and they have been Chiang Su New Medical College, 1977. Dictionary of Chinese Crude Drugs. Shanghai
demonstrated to be the main active ingredients in this plant. In Scientific Technologic Publisher, Shanghai p. 1283.
particular, the lavandula and prenyl flavonoids (e.g., sophoraflavanone Chiou, W.F., Lee, C.H., Liao, J.F., Chen, C.C., 2011. 8-Prenylkaempferol accelerates
osteoblast maturation through bone morphogenetic protein-2/p38 pathway to
G and kurarinone), have been confirmed to be hypotoxic and to have
activate Runx2 transcription. Life Sci. 88, 335–342.
high medicinal value through various pharmacological activities in vivo Choi, K.M., Gang, J.G., Yun, J.S., 2008. Anti-toxoplasma gondii RH strain activity of
and in vitro. herbal extracts used in traditional medicine. Int. J. Antimicrob. Agents 32,
Much of the work on Sophora flavescens during the last five 360–362.
Chung, M.Y., Rho, M.C., Ko, J.S., Ryu, S.Y., Jeune, K.H., Kim, K., Lee, H.S., Kim, Y.K.,
decades has focused on obtaining more extracts and active 2004. In vitro inhibition of diacylglycerol acylt ransferase by prenylflavonoids
compounds from the root of this plant, in addition to studying from Sophora flavescens. Planta Med. 70, 258–260.
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 27

Chinese Pharmacopoeia Commission, 2010. The Pharmacopoeia of the People’s 407-induced hyperlipidemic and cholesterol-fed rats. Biol. Pharm. Bull. 31,
Republic of China, 2010 ed. Part I. China Medical Science Press, Beijing, China, 73–78.
pp. 188–189. Kim, J.H., Ryu, Y.B., Kang, N.S., Lee, B.W., Heo, J.S., Jeong, I.Y., Park, K.H., 2006.
Deng, X., Shen, C., Meng, Q., 2014. Screening of herbal components for attenuating Glycosidase inhibitory flavonoids from Sophora flavescens. Biol. Pharm. Bull. 29,
amiodarone-induced hepatotoxicity on gel-entrapped rat hepatocytes. Drug 302–305.
Chem. Toxicol. 37, 100–106. Kim, J.Y., Kim, J.Y., Kim, J.J., Oh, J., Kim, Y.C., Lee, M.S., 2014. (2S)-20 -Methoxykur-
Di, D.L., Li, F.Q., Chen, L., Liu, G.J., 2006. Study on growth inhibition effect of Radix arinone inhibits osteoclastogenesis and bone resorption through down-
Sophora flavescens on bacteria in vitro. Lishizhen Med. Mater. Med. Res. 17, 1974. regulation of RANKL signaling. Biol. Pharm. Bull. 37, 255–261.
Ding, P.L., Chen, D.F., Bastow, K.F., Nyarko, A.K., Wang, X.H., Lee, K.H., 2004. Ko, W.G., Kang, T.H., Kim, N.Y., Lee, S.J., Kim, Y.C., Ko, G.I., Ryu, S.Y., Lee, B.H., 2000.
Cytotoxic isoprenylated flavonoids from the roots of Sophora flavescens. Helv. Lavandulylflavonoids: a new class of in vitro apoptogenic agents from Sophora
Chim. Acta 87, 2574–2580. flavescens. Toxicol. in vitr. 14, 429–433.
Ding, P.L., Liao, Z.X., Huang, H., Zhou, P., Chen, D.F., 2006. ( þ )-12a- Hydroxysopho- Komatsu, M., Tomimori, T., Hatayama, K., Mikuriya, N., 1970. Studies on the
carpine, a new quinolizidine alkaloid and related anti-HBV alkaloids from constituents of Sophora species. IV. Constituents of the root of Sophora
Sophora flavescens. Bioorganic Med. Chem. Lett. 16, 1231–1235. angustifolia siebet zucc. Yakugaku Zasshi 90, 463–468.
Ding, P.L., Chen, D.F., 2005. Studies on the phenolic constituents of Sophora Kuroyanagi, M., Arakawa, T., Hirayama, Y., Hayashi, T., 1999. Antibacterial and
flavescens. Asia-Pac. Tradit. Med. 3, 79–83. antiandrogen flavonoids from Sophora flavescens. J. Nat. Prod. 62, 1595–1599.
Ding, Y., Tian, R.H., Kinjo, J., Nohara, T., Kitagawa, I., 1992. Three new oleanene Kyogoku, K., Hatayama, K., Komastu, M., 1973. Constituents of a Chinese crude drug
glycosides from Sophora flavescens. Chem. Pharm. Bull. 40, 2990–2994. Kushen (the root of Sophora flavescens). Isolation of five new flavonoids and
Dong, X.Q., Du, Q., Yu, W.H., Zhang, Z.Y., Zhu, Q., Che, Z.H., Chen, F., Wang, H., Chen, J., formononetin. Chem. Pharm. Bull. 21, 2733–2738.
2013. Anti-inflammatory effects of oxymatrine through inhibition of nuclear factor- Lao, Y., 2005. Clinical study of matrine injection on preventing liver function
kappa B and mitogen-activated protein kinase activation in lipopolysaccharide- damage of anti-tumor drugs during chemotherapy of breast cancer. J. Chin.
induced BV2 microglia cells. Iran. J. Pharm. Res. 12, 165–174. Med. Mater. 28, 735–737.
Du, S.M., Ma, L.Q., Sun, J.J., Yang, Z.X., 2010. Study on in vitro antibacterial activity of Lee, H.S., Ko, H.R., Ryu, S.Y., Oh, W.K., Kim, B.Y., Ahn, S.C., Mheen, T.I., Ahn, J.S., 1997.
extracts from Sophora flavescens Ait. Acta Chin. Med. Pharmacol. 38, 74–76. Inhibition of phospholipase Cγ1 by the prenylated flavonoids from Sophora
Fan, H.Y., Wang, Y.H., Ren, K., Shen, N., Gu, R.S., Zhao, L.J., Chang, Y., 2013. Effect of flavescens. Planta Med. 63, 266–268.
flavonoids from Sophora fiavescens on the myocardial fibrosis induced by Lee, J.H., Shin, H., Kim, Y.J., Paek, S.H., Jin, S., Ha, U.H., 2014. Pseudomonas
isoprenaline in rats. Pharmacol. Clin. Chin. Mater. Med. 29, 76–78. aeruginosa-induced IL-1β production is inhibited by Sophora flavescens via
Gong, X.B., Gao, Y., Guo, G.Q., Vondran, F.W.R., Schwartlander, R., Efimova, E., Pless, G., the NF-κB/inflammasome pathways. J. Microbiol. 52, 1044–1049.
Sauera, I.M., Neuhaus, P., 2015. Effect of matrine on primary human hepatocytes Lee, S.W., Lee, H.S., Nam, J.Y., Kwon, O.E., Baek, J.A., Chang, J.S., Rho, M.C., Kim, Y.K.,
in vitro. Cytotechnology 67, 255–265. 2005. Kurarinone isolated from Sophora flavescens Ait. inhibited MCP-1-
Guo, B., Zhang, T., Su, J., Wang, K., Li, X., 2015. Oxymatrine targets EGFR (p-Tyr845) induced chemotaxis. J. Ethnopharmacol. 97, 515–519.
and inhibits EGFR-related signaling pathways to suppress the proliferation and Li, D., Zuo, H.J., Gao, H.Y., Wu, L.J., 2004. Study on the chemical constituents of
invasion of gastric cancer cells. Cancer Chemother. Pharmacol. 75, 353–363. Sophora flavescens Ait. J. Shenyang Pharm. Univ. 21, 346–348.
Hatayama, K., Komatsu, M., 1971. Studies on the constituents of Sophora species. Li, J.L., Cheng, A.M., Jin, Y.H., Cai, F.Q., 2002. Experiment study on the antitumor
effects of Sophora flavescens Ait. Chin. J. Clin. Oncol. Rehabil. 9, 28–29.
V. constituents of the root of Sophora flavescens Ait. var. angustifolia Siebet Zucc.
Li, K., Wang, H.J., 2004. Simultaneous determination of matrine, sophoridine and
Chem. Pharm. Bull. 19, 2126–2131.
oxymatrine in Sophora flavescens Ait. by high performance liquid chromato-
Hillerns, P.I., Wink, M., 2005. Binding of flavonoids from Sophora flavescens to the
graphy. Biomed. Chromatogr. 18, 178–182.
rat uterine estrogen receptor. Planta Med. 71, 1065–1068.
Li, W., Liang, H., Yin, T., Wang, B., Zhao, Y.Y., 2008. Main flavonoids from Sophora
Hong, M.H., Lee, J.Y., Jung, H., Jin, D.H., Go, H.Y., Kim, J.H., Jang, B.H., Shin, Y.C., Ko, S.G.,
flavescens. Acta Pharm. Sin. 43, 833–837.
2009. Sophora flavescens Ait. inhibits the production of pro-inflammatory cytoki-
Lin, D.W., 2006. Analysis of the chemical constituents from Sophora flavescens root
nesthrough inhibition of the NF jB/IjB signal pathway in human mast cell line
[D]. Chung Yuan Christian University, pp. 1–10.
(HMC-1). Toxicol. in vitr. 23, 251–258.
Liu, B., Shi, R.B., 2007. Chemical principles in flavonoid fraction of alcohol extract
〈http://www.sda.gov.cn/WS01/CL0001/〉 (in Chinese).
from Kushen decoction. J. Beijing Univ. Tradit. Chin. Med. 30, 263–270.
〈http://www.dayi100.com/login.jsp〉 (in Chinese).
Liu, X.J., Cao, M.A., Li, W.H., Shen, C.S., Yan, S.Q., Yuan, C.S., 2010. Alkaloids from
Huang, W.C., Chan, C.C., Wu, S.J., Chen, L.C., Shen, J.J., Kuo, M.L., Chen, M.C., Liou, C.J.,
Sophora flavescens Ait. Fitoterapia 81, 524–527.
2014. Matrine attenuates allergic airway inflammation and eosinophil infiltra-
Liu, Y., Xu, Y., Ji, W., Li, X., Sun, B., Gao, Q., Su, C., 2014. Anti-tumor activities of
tion by suppressing eotaxin and Th2 cytokine production in asthmatic mice. J.
matrine and oxymatrine: literature review. Tumour Biol. 35, 5111–5119.
Ethnopharmacol. 151, 470–477.
Liu, Y.H., Liu, Y.F., Guo, X.X., 2001. Current studies on anti-endotoxic chemical
Hur, J., 1994. Tonguibokam “Medical Dictionary of Oriental Crude Drugs”. Yeokang
components of traditional Chinese medicine in China. Acta Pharmacol. Sin. 22,
Publising Co., Korea p. 1645.
1071–1077.
Hwang, J.S., Lee, S.A., Hong, S.S., Lee, K.S., Lee, M.K., Hwang, B.Y., 2005. Monoamine
Liu, Z., Liu, B., Zhang, Z.T., Zhou, T.T., Bian, H.J., Min, M.W., Liu, Y.H., Chen, J., Bao, J.K.,
oxidase inhibitory components from the roots of Sophora flavescens. Arch.
2008. A mannose-binding lectin from Sophora flavescens induces apoptosis in
Pharm. Res. 28, 190–194. HeLa cells. Phytomedicine 15, 867–875.
Hwang, E.M., Ryu, Y.B., Kim, H.Y., Kim, D.G., Hong, S.G., Lee, J.H., Curtis-Long, M.J., Liu, Q., Luyten, W., Pellens, K., Wang, Y., Wang, W., Thevissen, K., Liang, Q., Cammue,
Jeong, S.H., Park, J.Y., Park, K.H., 2008. BACE1 inhibitory effects of lavandulyl B.P., Schoofs, L., Luo, G., 2012. Antifungal activity in plants from Chinese
flavanones from Sophora flavescens. Bioorganic Med. Chem. 16, 6669–6674. traditional and folk medicine. J. Ethnopharmacol. 143, 772–778.
Ibragimov, B.T., Talipov, S.A., Tishchenko, G.N., Kushmuradov, Y.K., Aripov, T.F., Long, Y., Lin, X.T., Zeng, K.L., Zhang, L., 2004. Efficacy of intramuscular matrine in the
Kuchkarov, S., 1981. Molecular and crystal structure of tetrahydroneo- treatment of chronic hepatitis B. Hepatobiliary Pancreat. Dis. Int. 3, 69–72.
sophoramine. Chem. Nat. Compd. 17, 546–551. Ma, S.C., Du, J., But, P.P.H., Deng, X.L., Zhang, Y.W., Ooi, V.E.C., Xu, H.X., Lee, S.H.S.,
Jin, J.H., Kim, J.S., Kang, S.S., Son, K.H., Chang, H.W., Kim, H.P., 2010. Anti- Lee, S.F., 2002. Antiviral Chinese medicinal herbs against respiratory syncytial
inflammatory and anti-arthritic activity of total flavonoids of the roots of virus. J. Ethnopharmacol. 79, 205–211.
Sophora flavescens. J. Ethnopharmacol. 127, 589–595. Ma, X.C., Xin, X.L., Zhang, B.J., Li, F.Y., Liu, K.X., Guo, D.A., 2008. Structural
Jin, S.N., Wen, J.F., Li, X., Kang, D.G., Lee, H.S., Cho, K.W., 2011. The mechanism of determination of flavonoids from Sorphora flavescens. Magn. Reson. Chem. 46,
vasorelaxation induced by ethanol extract of Sophora flavescens in rat aorta. 903–906.
J. Ethnopharmacol. 137, 547–552. Morinaga, K., Ueno, A., Fukushimae, S., Namikoshi, M., Iitaka, Y., Okuda, S., 1978.
Jung, H.A., Yoon, N.Y., Kang, S.S., Kim, Y.S., Choi, J.S., 2008. Inhibitory activities of Studies on the lupin alkaloids VIII. a new stereoisomer of sophocarpine. Chem.
prenylated flavonoids from Sophora flavescens against aldose reductase and Pharm. Bull. 26, 2483–2488.
generation of advanced glycation end products. J. Pharm. Pharmacol. 60, Murakoshi, L., Kidoguchi, E., Hagniwa, J., Ohmiy, S, Higashiyama, K., Otomasu, H.,
1227–1236. 1982. Isokuraramine and (-)-7, 11-dehydromatrine lupin alkaloids from flowers
Kang, T.H., Seong, S.J., Ko, W.Q., Kim, N.Y., Lee, B.H., Inagaki, M., Miyamoto, T., of Sophora flavescens. Phytochemistry 21, 2379–2384.
Higuchi, R., Kim, Y.C., 2000a. Cyotoxic lanvandulyl flavanones from Sophora Okuda, S., Murakoshi, I., Kamata, H., Kashida, Y., Haginiwa, J., Tsuda, K., 1965.
flavescens. J. Nat. Prod. 63, 680–681. Studies on the lupine alkaloids. I. the minor alkaloids of Japanese Sophora
Kang, S.S., Kim, J.S., Son, K.H., Chang, H.W., Kim, H.P., 2000b. A new prenylated flavescens. Chem. Pharm. Bull. 13, 482–487.
flavanone from the roots of Sophora flavescens. Fitoterapia 71, 511–515. Panthati, M.K., Rao, K.N.V., Sandhya, S., David, B., 2012. A review on phytochemical,
Kim, B.H., Kim, T.Y., 2013. Kurarinone, isolated from the roots of Sophora flavescens ethnomedical and pharmacological studies on genus Sophora, Fabaceae. Rev.
Aiton, regulates skin inflammation by regulation of the immune responses. Bras. Farmacogn.-Braz. J. Pharmacogn. 22, 1145–1154.
J. Dermatol. Sci. 69, e16. Park, S.J., Nam, K.W., Lee, H.J., Cho, E.Y., Koo, U., Mar, W., 2009. Neuroprotective
Kim, C.S., Park, S.N., Ahn, S.J., Seo, Y.W., Lee, Y.J., Lim, Y.K., Freire, M.O., Cho, E., Kook, effects of an alkaloid-free ethyl acetate extract from the root of Sophora
J.K., 2013. Antimicrobial effect of sophoraflavanone G isolated from Sophora flavescens Ait. against focal cerebral ischemia in rats. Phytomedicine 16,
flavescens against Mutans streptococci. Anaerobe 19, 17–21. 1042–1051.
Kim, H.W., Lee, M.R., Lee, G.S., An, W.G., Cho, S.I., 2012. Effect of Sophora flavescens Pu, L.P., Chen, H.P., Cao, M.A., Zhang, X.L., Gao, Q.X., Yuan, C.S., Wang, C.M., 2013. The
Ait. extract on degranulation of mast cells and contact dermatitis induced by antiangiogenic activity of Kushecarpin D, a novel flavonoid isolated from
dinitrofluorobenzene in mice. J. Ethnopharmacol. 142, 253–258. Sophora flavescens Ait. Life Sci. 93, 791–797.
Kim, H.Y., Jeong-da, M., Jung, H.J., Jung, Y.J., Yokozawa, T., Choi, J.S., 2008. Quang, T.H., Ngan, N.T., Minh, C.V., 2012b. alpha-Glucosidase inhibitors from the
Hypolipidemic effects of Sophora flavescens and its constituents in Poloxamer roots of Sophora flavescens. Bull. Korean Chem. Soc. 33, 1791–1793.
28 X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29

Quang, T.H., Ngan, N.T., Minh, C.V., Kiem, P.V., Tai, B.H., Nhiem, N.X., Thao, N.P., Tan, C., Qian, X., Jia, R., Wu, M., Liang, Z., 2013. Matrine induction of reactive oxygen
Luyen, B.T., Yang, S.Y., Kim, Y.H., 2013. Anti-inflammatory and PPAR transactiva- species activates p38 leading to caspase-dependent cell apoptosis in non-small
tional properties of flavonoids from the roots of Sophora flavescens. Phytother. cell lung cancer cells. Oncol. Rep. 30, 2529–2535.
Res. 27, 1300–1307. Tsai, J.C., Tsai, S., Chang, W.C., 2004. Effect of ethanol extracts of three Chinese
Roh, S.S., Kim, C.D., Lee, M.H., Hwang, S.L., Rang, M.J., Yoon, Y.K., 2002. The hair medicinal plants with anti-diarrheal properties on ion transport of the rat
growth promoting effect of Sophora flavescens extract and its molecular intestinal epithelia. J. Pharmacol. Sci. 94, 60–66.
regulation. J. Dermatol. Sci. 30, 43–49. Ueno, A., Morinaga, K., Fukushima, S., Iitaka, Y., Koiso, Y., Okuda, S., 1975. Studies on
Ryu, S.Y., Choi, S.U., Kim, S.H., 1997. In vitro antitumor activity of flavoniod from the lupin alkaloids VI. isolation and structure of ( þ ) -isomatrine. Chem. Pharm.
Sophora flavascens. Phytother. Res. 11, 51–53. Bull. 23, 2560–2566.
Ryu, S.Y., Kim, S.K., No, Z., Ahn, J.W., 1996. A novel flavonoid from Sophora Ueno, A., Morinaga, K., Fukushima, S., Okuda, S., 1978. Studies on the lupin alkaloids
flavescens. Planta Med. 62, 361–363. VII. isolation and structure of (-)-Δ7-dehydrosophoramine. Chem. Pharm. Bull.
Ryu, Y.B., Curtis-Long, M.J., Kim, J.H., Jeong, S.H., Yang, M.S., Lee, K.W., Lee, W.S., 26, 1832–1836.
Park, K.H., 2008. Pterocarpans and flavanones from Sophora flavescens display- Wang, H., Li, Y., Dun, L., Xu, Y., Jin, S., Du, J., Ma, L., Li, J., Zhou, R., He, X., Sun, T., Yu, J.,
ing potent neuraminidase inhibition. Bioorganic Med. Chem. Lett. 18, 2013. Antinociceptive effects of oxymatrine from Sophora flavescens, through
6046–6049. regulation of NR2B-containing NMDA receptor-ERK/CREB signaling in a mice
Saito, K., Arai, N., Sekine, T., Ohmiya, S., Kubo, H., Otomasu, H., Murakoshi, I., 1990. model of neuropathic pain. Phytomedicine 20, 1039–1045.
(-)-5α-Hydroxysophocarpine, a new lupine alkaloid from the seeds of Sophora Wang, H.Q., Jin, J.J., Wang, J., 2015. Matrine induces mitochondrial apoptosis in
flavescens var. angustifolia. Planta Med. 56, 487–488. cisplatin-resistant non-small cell lung cancer cells via suppression of β-catenin/
Sasaki, T., Li, W., Higai, K., Quang, T.H., Kim, Y.H., Koike, K., 2014. Protein tyrosine survivin signaling. Oncol. Rep. 33, 2561–2566.
phosphatase 1B inhibitory activity of lavandulyl flavonoids from roots of Wang, H.Y., Lu, Y.C., Chen, J., Li, J.C., Liu, S.H., 2011. Subcritical water extraction of
Sophora flavescens. Planta Med. 80, 557–560. alkaloids in Sophora flavescens Ait. and determination by capillary electrophor-
Sato, S., Takeo, J., Aoyama, C., Kawahara, H., 2007. Na þ -glucose cotransporter (SGLT) esis with field amplified sample stacking. J. Pharm. Biomed. Anal. 58, 146–151.
inhibitory flavonoids from the roots of Sophora flavescens. Bioorganic Med. Wang, J.Z., Sun, F.L., Han, H.W., Zhang, B.H., Yuan, S.H., Ma, Li., 1981. Experimental
Chem. 15, 3445–3449. study of total flavonoids from Sophora flavescens on cultured myocardial cell
Sekine, T., Saito, K., Minami, R., Arai, N., Suzuki, H., Koike, Y., Murakoshi, I., 1993. A with arrhythmia. Chin. Pharm. J. 16, 53.
new lupin alkaloid, (-)-leontalbinine N-oxide, in Sophora flavescens var. angu- Wang, J.Z., Sun, F.L., Han, H.W., Zhang, B.H., Yuan, S.H., Ma, Li., 1983. Experimental
stifolia seeds and its synthesis by biomimetic transformation from ( þ) study of total flavonoids from Sophora flavescens on cultured myocardial cell
-matrine N-oxide. Yakugaku Zasshi 113, 53–62. with arrhythmia. Acta Pharmacol. Sin. 4, 32–35.
Seo, H.S., Kim, K.H., Kim, D.Y., Park, B.K., Shin, N.S., Kim, J.H., Youn, H., 2013. GC/MS Wang, X.H., Han, G.Q., 1996. Study on the flavonoids of Sophora flavescens. Nat.
analysis of high-performance liquid chromatography fractions from Sophora Prod. Res. Dev. 8, 7–9.
flavescens and Torilis japonica extracts and their in vitro anti-neosporal effects Wang, X.K., Li, J.S., Yan, Y.N., 1996. Research situation of lupine alkaloids in sophora
on Neospora caninum. J. Vet. Sci. 14, 241–248. and its pharmacological action. World Phytomed.: Plant Med. Fasc. 11, 9–14.
Wei, J., Zhu, Y., Xu, G., Yang, F., Guan, Z., Wang, M., Fang, Y., 2014. Oxymatrine
Shao, Q., Zhao, X., Yao, L., 2014. Matrine inhibits the growth of retinoblastoma cells
extracted from Sophora flavescens inhibited cell growth and induced apoptosis
(SO-Rb50) by decreasing proliferation and inducing apoptosis in a mitochon-
in human osteosarcoma MG-63 cells in vitro. Cell Biochem. Biophys. 70,
drial pathway. Mol. Biol. Rep. 41, 3475–3480.
1439–1444.
Shao, H., Yang, B., Hu, R., Wang, Y., 2013. Matrine effectively inhibits the prolifera-
Wen, J.F., Jin, S.N., Zhou, G.H., Wang, Y.L., Zhang, F., Wang, X.N., Cho, K.W., 2009.
tion of breast cancer cells through a mechanism related to the NF-κB signaling
Effects of Sophora flavescens on the cardiac contractile and secretory functions
pathway. Oncol. Lett. 6, 517–520.
and its alterations in the hyperthyroidism. Int. J. Cardiol. 137, S48.
Shen, C.C., Lin, T.W., Huang, Y.L., Wan, S.T., Shien, B.J., Chen, C.C., 2006. Phenolic
Wen, J.F., Jin, S.N., Zhou, G.H., Wang, Y.L., Zhang, F., Wang, X.N., Cho, K.W., 2011.
constituents of the roots of Sophora flavescens. J. Nat. Prod. 69, 1237–1240.
Effects of Sophora flavescens ethanol extract on cardiac dynamics in hyperthyr-
Shen, Y., Feng, Z.M., Jiang, J.S., Yang, Y.N., Zhang, P.C., 2013. Dibenzoyl and
oid rabbits. Int. J. Cardiol. 152, S53.
isoflavonoid glycosides from Sophora flavescens: inhibition of the cytotoxic
Wen, M.C., Huang, C.K., Srivastava, K.D., Zhang, T.F., Schofield, B., Sampson, H.A., Li,
effect of D-galactosamine on human hepatocyte HL-7702. J. Nat. Prod. 76,
X.M., 2004. Kushen (Sophora flavescens Ait.), a single Chinese herb, abrogates
2337–2345.
airway hyperreactivity in a murine model of asthma. J. Allergy Clin. Immunol.
Shen, Y., Zhang, P.C., 2012. Studies on the chemical constituents and pharmacolo-
113, S218.
gical activities of Chinese medicine Sophora flavescens [C]. In: Proceedings of
Woo, E.R., Kwak, J.H., Kim, H.J., Park, H., 1998. A new prenylated flavonol from the
the Eleventh National Youth Pharmaceutical Workers of the Latest Scientific
roots of Sophora flavescens. J. Nat. Prod. 61, 1552–1554.
Research Exchange, Hangzhou, China, pp. 663-669.
Wu, C., Huang, W., Guo, Y., Xia, P., Sun, X., Pan, X., Hu, W., 2015. Oxymatrine inhibits
Shi, D., Zhang, J., Qiu, L., Li, J., Hu, Z., Zhang, J., 2013a. Matrine inhibits infiltration of
the proliferation of prostate cancer cells in vitro and in vivo. Mol. Med. Rep. 11,
the inflammatory Gr1(hi) monocyte subset in injured mouse liver through
4129–4134.
inhibition of monocyte chemoattractant protein-1. Evid.-based Complement. Wu, L.J., Miyase, T., Ueno, A., Kuroyanagi, M., Noro, T., Fukushima, S., 1985a. Studies
Altern. Med. 2013, 580673. on the constituents of Sophora flavescens Ait. II. Chem. Pharm. Bull. 33,
Shi, L.J., Shi, L., Song, G.Y., Zhang, H.F., Hu, Z.J., Wang, C., Zhang, D.H., 2013b. 3231–3236.
Oxymatrine attenuates hepatic steatosis in non-disease rats fed with high Wu, L.J., Miyase, T., Ueno, A., Kuroyanagi, M., Noro, T., Fukushima, S., 1985b. Studies
fructose diet through inhibition of sterol regulatory element binding transcrip- on the constituents of Sophora flavescens Ait. III. Yakugaku Zasshi 105, 736–741.
tion factor1 and activation of peroxisome proliferator activated receptor alpha. Wu, L.J., Miyase, T., Ueno, A., Kuroyanagi, M., Noro, T., Fukushima, S., 1985c. Studies
Eur. J. Pharmacol. 714, 89–95. on the constituents of Sophora flavescens Ait. IV. Yakugaku Zasshi 105,
Shi, Z., Chen, J., Li, C.Y., An, N., Wang, Z.J., Yang, S.L., Huang, K.F., Bao, J.K., 2014. 1034–1039.
Antitumor effects of concanavalin A and Sophora flavescens lectin in vitro and Wu, L.J., Miyase, T., Ueno, A., Kuroyanagi, M., Noro, T., Fukushima, S., 1986. Studies
in vivo. Acta Pharmacol. Sin. 35, 248–256. on the constituents of Sophora flavescens Ait. V. Yakugaku Zasshi 106, 22–26.
Shih, H.Z., 1999. Isoflavone and isoflavone glycosides antidiabetics. Japan, 11- 116487 [P]. Wu X.L.,Hang T.J, Zang Z.X., Shen J.P., Zhang Y.D., 2006a. Studies on pharmacoki-
Shin, D.H., Cha, Y.J., Joe, G.J., Yang, K.E., Jang, I.S., Kim, B.H., Kim, J.M., 2013. netics of oxymatrine and its metabolites in healthy volunteers [C]. Report of the
Whitening effect of Sophora flavescens extract. Pharm. Biol. 51, 1467–1476. 6th Chinese Pharmaceutical Association annual meeting. Guangzhou, China,
Shin, H.J., Kim, H.J., Kwak, J.H., Chun, H.O., Kim, J.H., Park, H., Kim, D.H., Lee, Y.S., November, pp. 3527  3537.
2002. A prenylated flavonol, sophoflavescenol: a potent and selectiveinhibitor Wu, Y.J., Shao, Q., Zhen, Z.Q., Cheng, Y.Y., 2006b. Determination of quinolizidine
of cGMP phosphodiesterase 5. Bioorganic Med. Chem. Lett. 12, 2313–2316. alkaloids in Sophora flavescens and its preparation using capillary electrophor-
Sohn, H.Y., Son, K.H., Kwon, C.S., Kwon, G.S., Kang, S.S., 2004. Antimicrobial and esis. Biomed. Chromatogr. 20, 446–450.
cytotoxic activity of 18 prenylated flavonoids isolated from medicinal plants: Wu, Y.R., Gong, Q.F., Fang, H., Liang, W.W., Chen, M., He, R.J., 2013. Effect of Sophora
Morus alba L., Morus mongolica Schneider, Broussnetia papyrifera (L.) Vent, flavescens on non-specific immune response of tilapia (GIFT Oreochromis
Sophora flavescens Ait. and Echinosophora koreensis Nakai. Phytomedicine 11, niloticus) and disease resistance against Streptococcus agalactiae. Fish Shellfish
666–672. Immunol. 34, 220–227.
Son, J.K., Park, J.S., Kim, J.A., Kim, Y., Chung, S.R., Lee, S.H., 2003. Arenylated Wun, Z.Y., Lin, C.F., Huang, W.C., Huang, Y.L., Xu, P.Y., Chang, W.T., Wu, S.J., Liou, C.J.,
flavonoids from the roots of Sophora flavescens with tyrosinase inhibitory 2013. Anti-inflammatory effect of sophoraflavanone G isolated from Sophora
activity. Plana Med. 69, 559–561. flavescens in lipopolysaccharide-stimulated mouse macrophages. Food Chem.
State Administration of Traditional Chinese Medicine, 1999. Chinese Materia Toxicol. 62, 255–261.
Medica, 11. Shanghai Science and Technology Publishing Edition, China p. 634. Xia, B.N., Bao, S.J., Zhou, W.Z., Zeng, X.D., 1979. Pharmacological studies of Radix
Su, J.L., Lu, Y., Rong, X.L., Qu, J.F., Han, J., 2007. Effects of matrine on mice sperm Sophora flavescens. China Pharm. J. 9, 402–403.
activity. Strait Pharm. J. 19, 16–19. Xiao, Z.M., Wang, A.M., Wang, X.Y., Shen, S.R., 2013. Effects of ethanol extract of
Sun, M.Y., Zuo, J., Duan, J.F., Han, J., Fan, S.M., Zhang, W., Zhu, L.F., Yao, M.H., 2008. Radix Sophora flavescens on activity of colon cancer HT29 cells. Afr. J. Tradit.,
Antitumor activities of Kushen flavonoids in vivo and in vitro. J. Chin. Integr. Complement. Altern. Med. 10, 352–355.
Med. 6, 51–59. Xu, X.Y., Wang, X.D., Xin, X.M., Sun, Q.H., Gao, Y.S., Zhu, Y.Y., 2008. Anti-myocardial
Sun, M., Han, J., Duan, J., Cui, Y., Wang, T., Zhang, W., Liu, W., Hong, J., Yao, M., Xiong, S., hypoxic effects of extract from Sophora flavescens Ait. Herald Med. 27, 520–521.
Yan, X., 2007. Novel antitumor activities of Kushen flavonoids in vitro and in vivo. Yang, N., Liang, B., Srivastava, K., Zeng, J., Zhan, J., Brown, L., Sampson, H., Goldfarb,
Phytother. Res. 21, 269–277. J., Emala, C., Li, X.M., 2013. The Sophora flavescens flavonoid compound
X. He et al. / Journal of Ethnopharmacology 172 (2015) 10–29 29

trifolirhizin inhibits acetylcholine induced airway smooth muscle contraction. Zhang, J.H., Zhao, Y.Y., Liu, Q.C., Ye, X.J., 2000. Studies on the chemical constituents
Phytochemistry 95, 259–267. from Sophora flavescens Ait. China J. Chin. Mater. Med. 25, 37–38.
Yagi, A., Fukunaga, M., Okuzako, N., Mifuchi, I., Kawamoto, F., 1989. Antifungal Zhang, L., Xu, L., Xiao, S.S., Liao, Q.F., Li, Q., Liang, J., Chen, X.H., Bi, K.S., 2007.
substance from Sophora flavescens. Jpn. J. Pharmacogn. 43, 343–347. Characterization of flavonoids in the extract of Sophora flavescens Ait. by high-
Yamaguchi-Miyamoto, T., Kawasuji, T., Kuraishi, Y., Suzuki, H., 2003. Antipruritic performance liquid chromatography coupled with diode-array detector and
effects of Sophora flavescens on acute and chronic itch-related responses in electrospray ionization mass spectrometry. J. Pharm. Biomed. Anal. 44,
mice. Biol. Pharm. Bull. 26, 722–724. 1019–1028.
Yamaki, M., Kashihara, M., Takagi, S., 1990. Activity of Kushen compounds against Zhang, S., Zhang, Y., Zhuang, Y., Wang, J., Ye, J., Zhang, S., Wu, J., Yu, K., Han, Y., 2012.
Staphylococcus aureus and Streptococcus mutans. Phytother. Res. 4, 235–236. Matrine induces apoptosis in human acute myeloid leukemia cells via the
Yamazaki, M., Arai, A., Suzuki, S., Takeuchi, T., 1984. Protective effects of matrine mitochondrial pathway and Akt inactivation. PLoS ONE 7, e46853.
and oxymatrine on stress ulcer in relation to their effects on the central Zhang, X.L., Cao, M.A., Pu, L.P., Huang, S.S., Gao, Q.X., Yuan, C.S., Wang, C.M., 2013. A
nervous system. Yakugaku Zasshi 104, 293–301. novel flavonoid isolated from Sophora flavescens exhibited anti- angiogenesis
Yang, J., Liu, P., Wu, X.Y., 2007. In vitro antibacterial activity of extractum Sophora activity, decreased VEGF expression and caused G0/G1 cell cycle arrest in vitro.
flavescens against Staphylococcus epidermidis. Chin. J. Nosocomiol. 17, Pharmazie 68, 369–375.
1357–1358. Zhang, Y., Sun, S., Chen, J., Ren, P., Hu, Y., Cao, Z., Sun, H., Ding, Y., 2014a. Oxymatrine
Yanju, B., Yang, L., Hua, B., Hou, W., Shi, Z., Li, W., Li, C., Chen, C., Liu, R., Qin, Y., Lv,
induces mitochondria dependent apoptosis in human osteosarcoma MNNG/
W., 2014. A systematic review and meta-analysis on the use of traditional
HOS cells through inhibition of PI3K/Akt pathway. Tumor Biol. 35, 1619–1625.
Chinese medicine compound Kushen injection for bone cancer pain. Support
Zhang, Z.T., Yang, B.L., Liu, Q.G., Yu, K.B., 2003. Crystal structure of stereoisomers of
Care Cancer 22, 825–836.
matrine from Sophora flavescens in the Qin Mountain. Acta Chim. Sin. 61,
Yoshikawa, M., Wang, H.K., Kayakiri, H., Taniyama, T., Kitagawa, I., 1985. Saponin
1058–1064.
and sapogenol. XL. Structure of sophoraflavoside I, a bisdesmoside of soyasa-
Zhao, Y.P., Pang, Q.Y., Liu, J.B., Chen, Y.Y., Lou, Z.C., 1994. Study on the alkaloids of
pogenol B, from Sophorae Radix, the root of Sophora flavescens Ait. Chem.
Sophora flavescens Ait. Nat. Prod. Res. Dev. 6, 10–13.
Pharm. Bull. 33, 4267–4274.
Zhao, Y.Y., Wang, B., Lei, L.M., Guo, M.J., Zhang, R.Y., Huang, L.R., Yi, Y.Y., Lou, Z.C.,
Youn, H.J., Noh, J.W., 2001. Screening of the anticoccidial effects of herb extracts
1993. Study on the flavonoids of Sophora flavescens. J. Integr. Plant Biol. 35,
against Eimeria tenella. Vet. Parasitol. 96, 257–263.
Youn, H.J., Lakritz, J., Kim, D.Y., Rottinghaus, G.E., Marsh, A.E., 2003. Anti- protozoal 304–306.
efficacy of medicinal herb extracts against Toxoplasma gondii and Neospora Zhao, Y.Z., 2008. Studies on the Chemical Constituents and Quality Control of
caninum. Vet. Parasitol. 116, 7–14. Sophora flavescens Ait.. Master Degree Thesis of Central South University, pp.
Yu, Q., Cheng, N., Ni, X., 2013. Identifying 2 prenylflavanones as potential 2–19.
hepatotoxic compounds in the ethanol extract of Sophora flavescens. J. Food Zheng, J.H., Wang, W., Huang, H., 2008. Antimicrobial activity of the flavonoids in
Sci. 78, T1830–1834. Sophora flavescens extraction. J. Wuhan Univ. (Nat. Sci. Ed.) 54, 439–442.
Zha, L., Qian, J.Q., Lv, F.H., 1981. Antiarrhythmic effect of Angelica sinensis, Zheng, Y.Q., Yao, J.R., Shao, X.D., 1999. Bio-active compounds from Sophora
tetrandrine and Radix Sophora flavescens. Chin. Pharm. J. 16, 53–54. flavescens Ait. Chin. J. Pestic. Sci. 1, 91–93.
Zhang, B.F., Song, L.Y., Zhu, L.G., Liu, Y.L., Li, X.Q., 1985. Anti-arrhythmic effect of Zhou, Y., Wu, Y., Deng, L., Chen, L., Zhao, D., Lv, L., Chen, X., Man, J., Wang, Y., Shan,
total alkaloids from Sophora flavescens. China J. Chin. Mater. Med. 10, 229–230. H., Lu, Y., 2014. The alkaloid matrine of the root of Sophora flavescens prevents
Zhang, B., Niu, W., Xu, D., Li, Y., Liu, M., Wang, Y., Luo, Y., Zhao, P., Liu, Y., Dong, M., arrhythmogenic effect of ouabain. Phytomedicine 21, 931–935.
Sun, R., Dong, H., Li, Z., 2014b. Oxymatrine prevents hypoxia- and Zhu, Y.H., Niu, A.O., Lu, S.Q., 2005. Preliminary studies on the effects of a Chinese
monocrotaline-induced pulmonary hypertension in rats. Free Rad. Biol. Med. herb-Radix Sophora flavescens on trophozoites of Giardia lamblia in vitro. Chin.
69, 198–207. J. Parasit. Dis. Control 18, 19–21.
Zhang, C., Ma, Y., Gao, H.M., Liu, X.Q., Chen, L.M, Zhang, Q.W., Wang, Z.M., Li, A.P., Zhu, Y.P., 1998. Chinese materia medica: chemistry, pharmacology, and applica-
2013. Non-alkaloid chemical constituents from Sophora flavescens. China J. Chin. tions. Harwood Academic Publishers, Amsterdam, The Netherlands, pp.
Mater. Med. 38, 3520–3524. 149–155.

Potrebbero piacerti anche