Sei sulla pagina 1di 18

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/266377108

Cuscuta chinensis lam.: A systematic review on Ethnopharmacology,


phytochemistry and pharmacology of an important traditional herbal
medicine

Article  in  Journal of Ethnopharmacology · October 2014


DOI: 10.1016/j.jep.2014.09.032

CITATIONS READS

27 322

7 authors, including:

Jin Li Paul Owusu Donkor


59 PUBLICATIONS   372 CITATIONS    University of Health and Allied Sciences
23 PUBLICATIONS   67 CITATIONS   
SEE PROFILE
SEE PROFILE

yan-xu Chang
Tianjin University of Traditional Chinese Medicine
79 PUBLICATIONS   692 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Review of locally and traditionally used Ligusticum species View project

TCM pk-pd View project

All content following this page was uploaded by yan-xu Chang on 10 January 2018.

The user has requested enhancement of the downloaded file.


Journal of Ethnopharmacology 157 (2014) 292–308

Contents lists available at ScienceDirect

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

Review

Cuscuta chinensis Lam.: A systematic review on ethnopharmacology,


phytochemistry and pharmacology of an important traditional
herbal medicine
Sineeporn Donnapee a,b, Jin Li a, Xi Yang a,b, Ai-hua Ge a,b, Paul Owusu Donkor a,
Xiu-mei Gao a,b, Yan-xu Chang a,b,n
a
Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
b
Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China

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

Article history: Ethnopharmacological relevance: Cuscuta chinensis Lam. has found its use as a traditional medicine in
Received 18 April 2014 China, Korea, Pakistan, Vietnam, India and Thailand. It is commonly used as an anti-aging agent, anti-
Received in revised form inflammatory agent, pain reliever and aphrodisiac. To provide an overview of the ethnopharmacology,
18 September 2014
phytochemistry, pharmacokinetics, pharmacology and clinical applications of Cuscuta chinensis, as well
Accepted 19 September 2014
Available online 2 October 2014
as being an evidence base for further research works of the plant.
Materials and methods: The present review covers the literature available from 1985 to 2014. The
Keywords: information was collected from journals, books, theses and electronic search (Google Scholar, PubMed,
Cuscuta chinensis Lam ScienceDirect, ESBCO, Springerlink and CNKI). Literature abstracts and full-text articles were analyzed
Tu-Si-Zi and included in the review.
Ethnopharmacology
Results: Many phytochemicals have been isolated, identified and published to date, including: at least
Phytochemistry
18 flavonoids; 13 phenolic acids; 2 steroids; 1 hydroquinone; 10 volatile oils; 22 lignans; 9 polysacchar-
Pharmacokinetics
Clinical applications
ides; 2 resin glycosides; 16 fatty acids. These phytochemicals and plant extracts exhibit a range of
pharmacological activities that include hepatoprotective, renoprotective, antiosteoporotic, antioxidant,
Chemical compounds studied in this article:
anti-aging, antimutagenic, antidepressant, improve sexual function, abortifacient effects, etc.
Quercetin (PubChem CID: 5280343)
Conclusion: This present review offers primary information for further studies of Cuscuta chinensis. The
Kaempferol (PubChem CID: 5280863)
Isorhamnetin (PubChem CID: 5281654)
in vitro studies and in vivo models have provided a bioscientific explanation for its various ethnophar-
Astragalin (PubChem CID: 5282102) macological uses and pharmacological activities (most notably antioxidant effects) especially in the
Hyperoside (PubChem CID: 5281643) prevention of hepatic disease and renal failure. It is necessary and important to do more pharmacokinetic
Rutin (PubChem CID: 5280805) and toxicological research works on human subjects in order to inform the possible active compounds in
Calycopteretin (PubChem CID: 10429470) the body and validate its safety in clinical uses.
Cinnamic acid (PubChem CID: 444539) & 2014 Elsevier Ireland Ltd. All rights reserved.
Caffeic acid (PubChem CID: 689043)
p-coumaric acid (PubChem CID: 637542)

Abbreviations: ALP, alkaline phosphatase; AMMC, amelanotic elanocytes; APAP, acetaminophen; ARF, acute renal failure; AST, aspartate aminotransferase; CCP, Cuscuta
chinensis polysaccharides; CHO, cholesterol; CK, creatine kinase; CK-MB, creatine kinase muscle and brain (subunits); CMSD, Cuscuta chinensis with other 7 medicinal herbs;
CNS, central nervous system; DC, dendritic cell; DMBA, 7,12-dimethylbenz(α)anthracene; DPPH, 2,2-Diphenyl-1-picrylhydrazyl; EC, 50% of effective concentration; ED90, 90%
of effective concentration; EESC, ethanol extract of the dry seed of Cuscuta chinensis; ERα, estrogen receptor alpha; ERβ, estrogen receptor beta; FSC, Flavonoids from Cuscuta
chinensis seeds; FST, force swimming test; H2O2, hydrogen peroxide; IC50, 50% of inhibition concentration; I.P., intraperitoneal; I.V., intravenous; LD50, 50% of lethal dose;
LDH, lactate dehydrogenase; LF, Lipofuscin; LH, Luteininzing hormone; LPS, lipopolysaccharides; MDA, malondialdehyde; MI/RI, myocardial ischemia /reperfusion injury;
P.O., per oral; OVA, ovalbumin; PCC, penile corpus cavernosum; PK, pharmacokinetics; ROS, reactive oxygen species; S.C., subcutaneous; SD rats, Sprague-Dawley rats; SOD,
superoxide diamutase; TBHP, tertiary butyl hydroperoxide; TG, triglyceride; TST, tail suspension test
n
Corresponding author at: Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
Tel.: þ 86 22 59596163; fax: þ 86 25 59596163.
E-mail address: Tcmcyx@126.com (Y.-x. Chang).

http://dx.doi.org/10.1016/j.jep.2014.09.032
0378-8741/& 2014 Elsevier Ireland Ltd. All rights reserved.
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 293

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
2. Phytochemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294
3. Ethnopharmacology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
4. Pharmacokinetics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
5. Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 297
5.1. Effect on skin protection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 297
5.2. Hepatoprotective activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 298
5.3. Anti-osteoporotic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 298
5.4. Immunological effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
5.5. Effect on neuronal system. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
5.6. Antioxidant activities. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
5.7. Anti-aging activities. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
5.8. Effects on tumor cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
5.9. Renoprotective effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
5.10. Effect on reproductive system. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
5.11. Prevent abortion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
5.12. Anti-mutagenic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
5.13. Anti-diabetic activities. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5.14. Cardiovascular activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5.15. Anti-depressant activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5.16. CNS depressant activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5.17. Effect on melanin production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5.18. Anthelmintic activities. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5.19. Anti-nociceptive and anti-inflammatory activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5.20. Toxicological reports . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6. Clinical applications. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.1. Female infertility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.2. Prevent abortion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.3. Male reproductive system disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.4. Chyluria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.5. Chloasma faciei . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.6. Glomerulonephritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.7. Nocturia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.8. Treatment and prevent diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.9. Herpes zoster . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
6.10. Acne . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
6.11. Vitiligo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306

1. Introduction minute scales. The flowers are hermaphrodite. Inflorescence lateral,


compact cymose glomelurous, few to many flowered clusters or
Cuscuta chinensis Lam. (Cuscuta chinensis; Convolvulaceae, (The racemes, white; bracteoles scale-like and bracts. Pedical ca. 1 mm.
Plant List., 2013)) is a parasitic plant which is also known as Calyx lobe 4–5, capular, sepals triangular ovate ca. 1.5 mm, ridged
Chinese Dodder (Mavlonov et al., 2008), or Tu-Si-Zi in Chinese on outer surface, apex obtuse, partly thickened. Corolla 3–3.5 mm
(Flora of China, 2006). It is commonly used in traditional medicine long, white, urceolate, 4 or 5 lopes, lopes detoid-ovate, apex acute
as a tonic and aphrodisiac in China and other Asian countries. It is or obtuse, spreading horizontally, fimbriate, reflexed, infrastaminal
often used as a functional food by adding to alcoholic beverages or scales shorter than tube. Stamens anthers ovoid, exserted. Ovary
porridge to improve sexual potency and vision and prevent subglobose, locues 2, ovules 4, styles 2, slender. Stigmas globose,
abortion (Zheng et al., 1998). capitates. Capsules globose, ca. 3 mm wide, enclosed by persistent
Cuscuta chinensis has two synonyms (Cuscuta. carinata R. Br. and corolla; pericarp thin, circumscissile. The seeds 2–4, broadly
Cuscuta. chinensis var. carinata (R. Br.) Engelm) (The Plant List., ovalate, 1–2 mm long, pale brown, not smooth. Flowering in
2013). No articles were found after searching by using these two June–October; December–March; February–May and fruits in
names. However, it was found that Cuscuta chinensis Lam. was used August–October. It can grow in semi-shade (light woodland) or
in 80 reports (both in English and Chinese articles including 35 no shade and requires moist soil. It is often on the plants of
plant-authorized articles) and was not used in 17 Chinese articles in Fabaceae, Asteraceae, and Zygophyllaceae. Cuscuta chinensis is
which the plant was just defined as Tu-Si-Zi. Cuscuta chinensis Lam. distributed in Africa: Ethiopia; Middle Asia: Kazakhstan, Kyrgyz-
should be used instead of Tu-Si-Zi in all publications in the future. stan, Tajikistan, Turkmenistan, Uzbekistan; Mongolia; Russia;
Cuscuta chinensis (Fig. 1) is a parasitic plant that wraps around China; Western Asia: Iran, Iraq, Afghanistan; Tropical Asia: India,
other plants and uses them for its nourishment. The plant grows Sri Lanka; Indonesia; Eastern Asia: Korea, Japan, Taiwan, Thailand;
near seaside. Stems thin, twining, filiform, glabrous, yellowish or Australasia. It is distributed in most parts of China mainly in
pale yellowish, ca. 1 mm diam. The plant has no leaf or reduced to Henan, Jiangsu, Shandong, Hebei, Jilin, Liaoning provinces and
294 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

Fig. 1. Cuscuta chinensis Lam. (A) Habitus, (B) stem and flowers, (C) fruits, and (D) dried seeds.

Taiwan region. Its aerial parts are excavated in autumn when the hyperoside, quercetin, astragalin, kaempferol-3-O-galactoside
fruits are ripe and dried by natural sunlight and thrashed for and quercetin-3-O-glucoside) with various quantities, and Cuscuta
seeds. It is used in its unprocessed form or boiled after removal of chinensis having extremely high content of kaempferol-3-O-
impurities. It is also pounded into cake for use after being steamed glucoside (29–34% of the total phenolics) among all species. The
(Liao et al., 2000; Lei, 2000; Teng, 2007 ; Costea et al., 2011) . water-soluble constituents of Cuscuta chinensis include a trypto-
Although many researchers have attempted to find out the phan derivative alkaloid (cuscutamine), and 2 new lignans (cus-
chemical constituents including the pharmacological activities and cutosides A and B), along with 4 known flavonoids (astragalin,
clinical applications of Cuscuta chinensis which are evidenced by an hyperoside, quercetin, quercetin-3-O-apiosyl(1-2)-galactoside),
increasing number of published scientific literatures in this area, there 3 known lignans (pinoresinol-4-O-glucoside, pinoresinol, epipi-
is no review literature available. This current review therefore aims to noresinol), p-coumaric acid, caffeic acid, chlorogenic acid, and
provide an overview of the ethnomedicinal use, phytochemical arbutin (Yahara et al., 1994). The microelements of this plant such
properties, pharmacokinetics, pharmacological activities and clinical as calcium, magnesium, iron, manganes, and copper have so far
applications of Cuscuta chinensis, as well as being an evidence base for been identified using atomic absorption spectrometry method
further research works of this plant. (Zhao et al., 1990). Moreover, five other compounds were isolated
from petroleum ether and chloroform fractions from the stem of
Cuscuta chinensis for the first time. Their structures were identified
2. Phytochemistry as β-sitosterol, d-sesamin, 9(R)-hydroxy-d-sesamin, D-pinoresinol
and daucosterol by chemical and spectroscopical methods (Ye
Cuscuta chinensis is a parasitic plant so that its phytochemical et al., 2001). They also isolated 5 principal flavonoids, quercetin
constituents depend on the type of host (Lin et al., 2007). The 3-O-β-D-galactoside-7-O-β-D-glucoside, quercetin 3-O- β-D-apio-
chemical constituents of Cuscuta chinensis are flavonoids, poly- furanosyl-(1etin 3D-galactoside, hyperoside, quercetin and kaemp-
saccharides, alkaloids, steroids, volatile oils, lignans and others ferol by RP-HPLC (Ye et al., 2002). Furthermore, other 2 new lignan
(Fig. 2) (Miyahara et al., 1996; Du et al., 1998; Wang et al., 2000, glycosides (cuscutoside C and D) have been isolated in 2010 (He
2001; Bao et al., 2002; Hou et al., 2003). Flavonoids account for et al., 2010). In addition, Cheng et al. (2013) studied about the
about 3.0% of the total chemical constituents. The main flavonoids content of fatty acid in the Cuscuta chinensis seed oil n-hexane
including kaempferol, quercetin, hyperoside, astragalin and lig- extract by using capillary gas chromatography and they found 16
nans play an important role in the pharmacological activity kinds of fatty acid, primarily including palmitic acid, linoleic acid,
(Cornwell et al., 2004; Williamson et al., 2005). Loffler et al. oleic acid, and linolenic acid.
(1997) studied the plants in the genus Cuscuta and found that all Cuscuta chinensis is an important traditional medicine, which
species had the same soluble phenolic compounds (Chlorogenic has many isolated phytochemical compounds including at least
acid, 3,5-dicaffeoylquinic acid, 4,5-dicaffeoylquinic acid, 18 flavonoids; 13 phenolic acids; 2 steroids; 1 hydroquinone; 10
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 295

Kaempferol Quercetin Isorhamnetin Rutin Hyperoside

Astragalin d - sesamin Chlorogenic acid Cinnamic acid Caffeic acid

p-coumaric acid β-sitosterol daucosterol Arbutin caryophyllene


Fig. 2. Structure of some chemical constituents isolated from Cuscuta chinenesis.

volatile oils; 22 lignans; 9 polysaccharides; 2 resin glycosides and reproductive organs, spermatorrhea, frequent urination, thirst, mouth's
16 fatty acids (more details about the occurrence, the chemical bitter taste feeling or aggregation of the blood due to cold (Li, 2009).
group and pure compounds isolated from Cuscuta chinensis are Moreover, it has been listed in the Pharmacopoeia of the People's
summarized in Table 1). The main chemical constituents of Republic of China (Committee for the Pharmacopoeia of PR China,
Cuscuta chinensis seed are flavonoids (principally hyperoside, 1995) since 1995, some of its prescriptions have been used to improve
quercetin, rutin, isorhametin) which were selected as markers to sexual function, prevent and treat cardiovascular diseases, treat osteo-
evaluate the quality of the plant (Shekarchi et al., 2014). Never- porosis and prevent senescence in clinic. Among them, Gu Jing Bu Shen
theless, the studies on phytochemistry of the plant are mainly pills have been employed for the treatment of premature ejaculation
targeting on the seed while the studies on other parts of the plant and spleen and kidney deficiency. In addition, many classical medicine
are minority. Therefore, more phytochemistry studies on other books in China, like Ben Cao Hui Yan, Ben Cao Yuan Shi, Ben Jing Feng
parts of the plant are needed. Meanwhile, the influence of Yuan and Ben Cao Zheng Yi, have recorded that the plant and its
different hosts on the contents of phytochemical constituents also famous prescriptions are used to treat various diseases such as
needed to clarify in the future. impotence, infertility, wet dreams, urinary retention, urinary incon-
tinence, lower abdominal and back pain etc. (Ni, 2005; Li, 2013; Zhang,
2011; Zhang, 2013).
3. Ethnopharmacology Furthermore, the paste of the plant is applied on chronic ulcers and
wounds. Poultice is applied on painful inflammations in Pakistan
Around 2000 years ago, Cuscuta chinensis was recorded in Shen (Qureshi et al., 2010). In India, the stems from this plant are used in the
Nong Ben Cao Jing (Shen Nong's Herbal, the ancient Chinese herbal treatment of sore heads, inflamed eyes (Petel and Petel, 2010),
classic) for the first time in Chinese history. It was listed as a “top jaundice (Patil and Patil, 2012) and for removing hair dandruff
grade” drug for the kidney-tonifying and liver-strengthening functions (Shubhangi and Patil, 2012). The leaves and the stems of this plant
(Mou, 2002). The plant was used as a tonic medicine by traditional are used to increase lactation (Patil and Birada, 2011 and Patil et al.,
Chinese medicine practitioners for the treatment of kidney and liver 2010). In Vietnam, the whole plant of Cuscuta chinensis is used for back
deficiency (He et al., 2010). Cuscuta chinensis has an ability to tonify pain and constipation (Sam et al., 2008). In Korea, the seeds with other
kidney and nourish liver, it can replenish essence, improve vision, stop herbal medicines are used to improve sexual function and health
diarrhea and prevent abortion. It has been used not only in kidney (Sohn et al., 2008). In Thailand, the whole plant is boiled in water and
insufficiency with lumbago, impotence, spermatorrhea, frequent urina- used as an anthelminthic and as a body wash for treatment of jaundice
tion, sterility due to cold uterus, threatened abortion or habitual (Smittinan, 2011; Cruz-Garcia and Price, 2011) as shown in Table 2.
abortion, but also used in treatment of the liver insufficiency with
dim and blurred vision (Teng, 2007). In the famous classic book of
Chinese materia medica, Ben Cao Gang Mu (Compendium of Materia 4. Pharmacokinetics
Medica), Cuscuta chinensis improves eyesight and prevents aging. It can
tonify the muscles, enhance the activity of bone and tendon, principally The studies about pharmacokinetics (PK) of Cuscuta chinensis
used to treat or improve excessive cold in male and female are very limited. There are 2 reports: the PK study of quercetin in
296 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

Table 1
Phytochemistry of Cuscuta chinensis.

Chemical constituents Part of plant Reference

Flavonoids
Quercetin Seed; fruit; whole plant; Ye et al., (2002, 2005); Yahara et al., (1994); Loffler et al.,
stem (1997); Kwon et al., (2000)
Pan et al., (2014)
Quercetin-3-O-glucoside Whole plant Loffler et al., (1997)
Quercetin-3-O-apiosyl(1-2)-galactoside Fruit; seed; stem Yahara et al., (1994); Ye et al., (2005)
Quercetin-3-O-β-D-galactoside-7-O-β-glucoside Seed Ye et al., (2002, 2005)
Quercetin-3-O-β-D-apiofuranosyl(1-2)-β-D-galactoside Seed Ye et al., (2002); Pan et al., (2014)
Quercetin-3-O-rhamnosylgalactoside Stem Ye et al., (2005)
Quercetin-3-O-acetylgalactoside Seed; stem Ye et al., (2005)
Kaempferol Seed; stem Ye et al., (2002, 2005); Hajimehdipoor et al., (2012);
Kwon et al., (2000); Pan et al., (2014)
Kaempferol 3,7-di-O-β-D-glucopyranoside Seed He et al., (2010)
Keampferol-3-O-galactoside Whole plant; seed; stem Loffler et al., (1997); Ye et al., (2005)
Keampferol-3-O- β-D-glucoside Seed Pan et al., (2014)
Isorhamnetin Seed; stem Hajimehdipoor et al., (2012); Ye et al., (2005)
Isorhamnetin-3-O-glucoside Seed; stem Ye et al., (2005)
Astragalin Seed;whole plant; fruit; Yang et al., (2009a, 2009b); Loffler et al., (1997);
stem Yahara et al., (1994); Ye et al., (2005)
Hyperoside Seed; Whole plant; stem Hajimehdipoor et al., (2012); Loffler et al., (1997);
Yahara et al., (1994); Pan et al., (2014);
Ye et al., (2005)
Rutin Seed Ye et al., (2001); Hajimehdipoor et al., (2012)
Calycopteretin Seed Kwon et al., (2000)
Apigenin Seed; stem Ye et al., (2005)

Phenolic acid
Chlorogenic acid Seed; stem Pan et al., (2014); Ye et al., (2005)
Caffeic acid Fruit Yahara et al., (1994)
4-Caffeoyl quinic acid Seed He et al., (2010)
5-Caffeoyl quinic acid Seed He et al., (2010)
4-Caffoyl-5-coumaroylquinic acid Seed; stem Ye et al., (2005)
Methyl 4-hydroxy-3,5-dimethoxycinnamate Seed Kwon et al., (2000)
4-Feruoyl-5-caffeoylquinic acid Stem Ye et al., (2005)
3,5-Dicaffeoylquinic acid Whole plant; seed; stem Loffler et al., (1997); Ye et al., (2005)
3,5-Dicaffeoyl-4-feruoylquinic acid or 3-feruoyl-4,5-dicaffeoylquinic acid Seed; stem Ye et al., (2005)
4,5-Dicaffeoylquinic acid Whole plant; seed; stem Loffler et al., (1997); Ye et al., (2005)
3,4,5-Tricaffeoylquinic acid Seed Ye et al., (2005)
p-Coumaric acid Fruit Yahara et al., (1994)
Cinnamic acid Seed; whole plant; fruit He et al., (2010); Loffler et al., (1997); Yahara et al., (1994)

Steroids
β-Sitosterol Seed Ye et al., (2001)
Daucosterol Seed Ye et al., (2001)

Hydroquinone
Arbutin Fruit Yahara et al., (1994)

Volatile oil
2 – Pentyl furan Seed Hou et al., (2003)
Dodecane Seed Hou et al., (2003)
3- Butene-2 –alcohol Seed Hou et al., (2003)
Furfural Seed Hou et al., (2003)
Furan-2-ylmethanol Seed Hou et al., (2003)
Heptanal Seed Hou et al., (2003)
3, 7 – Dimethyl-1, 6 – octadiene, 3 – alcohol Seed Hou et al., (2003)
Borneol Seed Hou et al., (2003)
α-Terpineol Seed Hou et al., (2003)
Caryophyllene Seed Hou et al., (2003)
α-Caryophyllene Seed Hou et al., (2003)

Lignans
20 -Hydroxyl asarinin 20 -O-β-D-apiofuranosyl-(1-2)- β –D-glucopyranoside Seed He et al., (2010); Du et al., (1998)
(Cuscutoside A)
20 -Hydroxy asarinin 20 -0-j-xylopyranosyl-(1-6)-β-glucopyranoside Seed He et al., (2010); Du et al., (1998)
(Cuscutoside B)
20 -Hydroxyl asarinin 20 -O-β-D-glucopyranoside (Cuscutoside C) Seed He et al., (2010); Du et al., (1998)
20 -Hydroxyl asarinin 20 -O-β-D-apiofuranosyl-[(1-2)- β –D-glucopyranosyl- Seed He et al., (2010); Du et al., (1998)
(1-6)-β-D-glucopyraniside (Cuscutoside D)
( þ )-Pinoresinol Fruit Yahara et al., (1994)
D-Pinoresinol Seed Ye et al., (2001)
( þ )-Epipinoresinol Fruit Yahara et al., (1994)
( þ )-Pinoresinol-4-O-glucoside Fruit Yahara et al., (1994)
d-Pinoresinol-4-O-glucoside Seed; stem Ye et al., (2005)
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 297

Table 1 (continued )

Chemical constituents Part of plant Reference

Feruoylcaffeoyl-d-pinoresinol Seed; stem Ye et al., (2005)


Dicaffeoyl-d-piniresinol Stem Ye et al., (2005)
d-Sesamin Seed; stem Ye et al., (2005)
9(R)-Hydroxy-d-sesamin Seed Ye et al., (2001)
9α-Hydroxy-d-sesamin-9-O-glucoside Seed; stem Ye et al., (2005)
9α-Hydroxy-d-sesamin-O-glucoside Seed Ye et al., (2005)
5,50 -Dihydroxy-d-sesamin-5-O-glucoside Stem Ye et al., (2005)
4-Methyl-3-methoxy-9α-hydroxyligballinol-O-glucoside Seed; stem Ye et al., (2005)
3,4-Demethyleudesmin-4-O-glucoside Seed Ye et al., (2005)
Piperitol-4-O-glucoside Seed; stem Ye et al., (2005)
20 -Hydroxyasarinin-20 O-rhamnosylglucoside Seed Ye et al., (2005)
Lignin-O-coumaroylglucoside Stem Ye et al., (2005)
Lignin-4-O-coumaroylglucoside Stem Ye et al., (2005)

Polysaccharides
Neutral heteropolysaccharide: arabinose, rhamnose, xylose and galactose Seed Wang et al., (2001)
composition
Acidic heteropolysaccharide mainly branched: CHC-1, H3, CS-A, CS-B, CS-C Seed Wang et al., (2001)

Resin glycosides
Acylated trisaccharide: α-L-rhamnopyranosyl-(1- 3) -[2-O-(11S)-11- Seed Miyahara et al., (1996)
hydroxytetradecanoyl]-[4-O-(2R,3R) -3-hydroxy-2-methylbutyryl]-α-L-
rhamnopyranose- (1-20-[6-O-acetyl]-D-glucopyranoside
α-L-rhamnopyranosyl-(1- 3) – [2-O-(11S) -11-hydroxypamitoyl]-[4-O-(2R, Seed Miyahara et al., (1996)
3R)-3- hydroxy-2-methylbutyryl]-α-L-rhamnopyranose-(1-2-0-[6-O-
acetyl]-D-glucopyranoside

Fatty acid
Methylmyristate Seed Cheng et al., (2013)
Methyl pentadecanoate Seed Cheng et al., (2013)
Methyl hexadecanoate Seed Cheng et al., (2013)
Methyl palmitoleate Seed Cheng et al., (2013)
Methyl heptadecanoate Seed Cheng et al., (2013)
Methyl stearate Seed Cheng et al., (2013)
Methyl oleate Seed Cheng et al., (2013)
Methyl linoleate Seed Cheng et al., (2013)
Methyl linolenate Seed Cheng et al., (2013)
Methyl Arachidate Seed Cheng et al., (2013)
Methyl cis-11-eicosenoate Seed Cheng et al., (2013)
Methyl 11c,14c- eicosadienoate Seed Cheng et al., (2013)
Methyl behenate Seed Cheng et al., (2013)
Sinapic acid methylester Seed Cheng et al., (2013)
Methyl tricosanoate Seed Cheng et al., (2013)
Methyl lignocerate Seed Cheng et al., (2013)

Microelements
Calcium Seed Zhao et al., (1990)
Magnesium Seed Zhao et al., (1990)
Iron Seed Zhao et al., (1990)
Manganese Seed Zhao et al., (1990)
Copper Seed Zhao et al., (1990)

rat and hyperoside in human after oral administration of Cuscuta PK and pharmacodynamics especially in human subjects should be
chinensis. Zheng et al. studied about the PK of Cuscuta chinensis in conducted.
rat's serum by using HPLC-UV to determine the concentrations of
quercetin in rats plasma. The rats were given 0.85 g/kg 95%
ethanol extract of the Cuscuta chinensis seeds by gavage. They 5. Pharmacology
found that the highest concentration of quercetin was 0.401 ng/mL
after 0.333 h (Zheng et al., 2004). The PK of hyperoside was The long history of medicinal applications of Cuscuta chinensis
studied after oral administration of Cuscuta chinensis ethanol has inspired many pharmacological research works. The study
extract in 5 healthy female volunteers by HPLC-UV. At 3.259 h, indicates that Cuscuta chinensis shows a broad range of biological
the highest concentration of hyperoside was 3.902 ng/mL (Peng, activities, which are briefly shown in Table 4.
2010). All the parameters of PK are shown in Table 3. This
information showed that the main phytochemical constituents 5.1. Effect on skin protection
that were found in the body after oral administration were
flavonoids(quercetin and hyperoside). Moreover, there is a report that Nisa et al. reported the inhibition of whole plant extract of
quercetin can inhibit CYP450 enzymes (Gaudineau et al., 2004). Due to Cuscuta chinensis on 7,12-dimethylbenz(α)anthracene(DMBA)-
the limited information about the PK of Cuscuta chinensis, further induced skin papillomas and carcinomas in mice. The water
research works such as the study of the PK of Cuscuta chinensis and the extract 1 g/kg was orally administrated thrice a week to 22 mice
individual chemical constituents, the bioavailability, the relationship of and started on the 10th day after the first application of DMBA to
298 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

Table 2
Ethnopharmacology of Cuscuta chinensis.

No. Ethnomedicinal uses Parts of the plant Locality References

1 Treatment of male infertility, importance, spermatorrhea Seed China Teng, (2007), Li, (2009); Committee for
Treatment of female infertility due to cold uterus the Pharmacopoeia of PR China, (1995);
Treatment of frequent urination Ni, (2005); Li, 2013; Zhang, (2011);
Treatment of diarrhea Zhang, (2013)
Prevent abortion
Improve vision
Prevent aging
Improve activity of bone and tendon
Treatment of thirsty, bitter taste feeling or dry mouth
Treatment of blood aggregation due to the cold
Treatment of urinary retention
Treatment of urinary incontinence
Treatment of lower abdominal and back pain
2 Treatment of chronic ulcers, wounds, painful inflammation Whole pant Pakistan Qureshi et al., (2010)
3 Ttreatment of sore heads and inflamed eyes Stem India Petel and Petel, 2010
Treatment of jaundice Stem Patil et al., (2012)
Removes hair dandruff Stem Shubhangi and Patil, 2012
Facilitates lactation Leaf Patil et al., (2011); Patil et al., (2010)
4 Treatment of back pain and constipation Whole plant Vietnam Sam et al., (2008)
5 Improves sexual function and health Seed Korea Sohn et al., (2008)
6 As an anthelminthic, and to alleviate jaundice Whole plant Thailand Smittinan, (2011)

Table 3
Pharmacokinetic information of Cuscuta chinensis.

Species/ bio-matrix Analytes Administration route/ PK parameters (or important information) References
dosage

Rat/serum Quecrcetin Gavage 0.85g/kg of Cuscuta Kα(h-1): 5.214, α(h-1): 3.614, β(h-1): 0.568, T1/2(α) (h): 0.19, T1/2(β) (h): Zheng et al., (2004)
chinensis 95% ethanol 1.22, T1/2(AB) (h): 0.13, Tmax (h): 0.333, Cmax (μg/ ml): 0.401
extract

Human/plasma Hyperoside Oral administration 360 g A(μg/ml): 13.45, Ke(h-1): 0.199, Ka(h-1): 0.448, Lag time(h): 0.444, T1/2ka(h): Peng, (2010)
of Cuscuta chinensis ethanol 1.55, T1/2ke(h): 3.48, Tmax(h): 3.259, Cmax(μg/ml): 3.902, AUC((μg/ml)nh):
extract 37.423, CL/F(s)(μg/h/(μg/ml)): 0.027, V/F(c)(μg/(μg/ml): 0.134

the 252nd day. They observed the decreases in the number of enzymes may be the major mechanism of Cuscuta chinensis
papillomas per mouse in 190th day and the volume of papillomas ethanol extract to prevent the development of liver damage
per mouse on 206th day. The results show that Cuscuta chinensis induced by APAP (Yen et al., 2007). Moreover, it will be more
significantly inhibited the appearance and development of skin effective when the seed of Cuscuta chinensis is prepared in
papillomas and also reduced the incidence of carcinoma (Nisa nanoparticles (Yen et al., 2008b). These two studies indicate that
et al., 1986). Cuscuta chinensis seeds have an exact effect on prevention of
hepatotoxicity.
5.2. Hepatoprotective activity
5.3. Anti-osteoporotic activity
There are two ethnomedicinal references for the use of Cuscuta
chinensis in liver disease. The study of hepatoprotective activity Cuscuta chinensis has been used for treatment of osteoporosis in
was performed to prove the claims that Cuscuta chinensis has China and in some Asian countries. Yao et al. (2005) demonstrated
hepatoprotective effect. Yen et al. studied about hepatoprotective that addition of an aqueous extract of Cuscuta chinensis in rat bone
effect of the aqueous and ethanol extracts of Cuscuta chinensis cells significantly promoted the differentiation and proliferation of
against acetaminophen (APAP)-induced hepatotoxicity in rats. osteoblasts, but the activities of osteoclasts were inhibited. The
They found that the seed of Cuscuta chinensis ethanol extract at water extract of dry Cuscuta chinensis seed can promote
oral dose of both 125 and 250 mg/kg significantly reduced the osteoblast-like MG-63 cells differentiation in a dose-dependent
evaluation of glutamate oxaloacetate transaminase, glutamate manner. The extract enhances ALP activity, collagen synthesis,
pyruvate transaminase, and alkaline phosphatase (ALP). In addi- bone morphogenetic protein (BMP-2) expression, and mineraliza-
tion, the ethanol extract can prevent liver histopathological tion in MG-63 cells (Yang et al., 2009a). These observations
changes such as centribular hepatic necrosis, kupffer cell infiltrat- indicate that Cuscuta chinensis has osteogenic effects. Moreover,
ing lymphocytes, ballooning degradation, and APAP-induced hepa- Yang et al. studied about antiosteoporotic compounds from seeds
totoxicity in rats. They also found that high doses of APAP could of Cuscuta chinensis. They isolated 5 flavonoids from Cuscuta
lead to decreased levels of antioxidant enzymes (glutathione chinensis and found that kaempferol and hyperoside significantly
peroxidase superoxide diamutase (SOD), and catalase) and indi- increased the ALP activity in osteoblast-like UMR-106 cells, astra-
cate a significant level of hepatotoxcity, and the ethanol extract of galin promoted the proliferation of UMR-160 cells. 5 flavonoids
Cuscuta chinensis could raise the levels of SOD, catalase and also promoted Hela cell, a human cervical cancer cell via estro-
glutathione peroxidase. In contrast, the aqueous extract did not genic receptor ERα and ERβ. They suggested that kaempferol and
show any effect. However, the enhanced levels of antioxidant hyperoside are the active compounds in this plant for osteogenic
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 299

Table 4
Pharmacology of Cuscuta chinensis.

No. Activities Part of Model Formulation /dosage/ Control Result Reference


plant extract

1 Skin protective Whole Mice (in vivo) P.O. 1 g/kg body wt of Positive control: 150 μg Cuscuta chiensis inhibit both the appearance Nisa et al.,
activity plant water extract 3 times/ DMBA appy on skin for and development of skin papillomas and also (1986)
week 7 days; negative control: reduced the incidence of carcinoma
P.O. distilled water transformation.

2 Hepatoprotective Seed Rats (in vivo) P.O. 125 and 250 Positive control: P.O. 125 and 250 mg/kg ethanol extract can Yen et al.,
activity mg/kg of water and APAP 835 mg/kg single significantly inhibit liver damage caused by (2007)
ethanol extract dose; negative control: APAP-induced hepatotoxicity
I.P. normal saline
Seed Rats (in vivo) P.O. 125 and 250 Positive control: P.O. 25 and 50 mg/kg seeds ethanol extract Yen et al.,
mg/kg ethanol extract, APAP 835 mg/kg single nanoparticles formulation show significantly (2008b)
and 25 and 50 mg/kg dose; negative control: hepatoprotective effects the same as the
nanoparticles of I.P. normal saline normal 125, 250 mg/kg seeds ethanol
ethanol extract extract.

3 Musculoskeletal Seed cultured Aqueous extract Aqueous extract of Cuscuta chinensis in rat Yao et al.,
(osteoporosis neonatal rat bone cells clearly promoted the proliferation (2005)
treatment) calvarias organ and differentiation of the osteoblasts, but
(in vitro) inhibited the activities of osteoclasts
Seed MG-63 cells 100, 500 and 1000 Negative control: culture 500 and 1000 μg/ml dry seed water extract Yang et al.,
(in vitro) μg/ml aqueous extract medium mildly promoted the proliferation of MG-63 (2009a)
cells. Dose-dependent increases in ALP activity
and collagen synthesis, the release of BMP-2
but not osteocalcin in the MG-63 cells was
induced (100–1000 μg/ml). In addition, the
extract markedly increased mRNA expression
of ALP, collagen, and BMP-2 in the MG-63 cells
in a dose-dependent manner. Mineralization in
the culture of MG-63 cells was significantly
induced at 500 and 1000 μg/ml
Seed (UMR-106 1, 5, 10, 50, 100 μg/ml Positive control: 0.1 μM Isolated flavonoids, show osteoporotic Yang et al.,
cells) in vitro of its organic fraction 17-β-estradiol; negative activities, keampferol and hyperoside (2011)
significantly increased the ALP activity in
and 1, 5, 10, 50 μM of control: DMSO 0.1 v/v
oteoblast-like UMR-106 cells and astragalin
its isolated flavonoids
promoted the proliferation of UMR-160 cells.

4 Immune Seed Mice (in vivo) P.O. 100, 200 or Positive control: S.C. The ethanol extract can significantly enhance Pan et al.,
regulation 400 μg of ethanol OVA 100 μg negative the mitogen- and ovalbumin (OVA)-stimulated (2005)
extract control: S.C. normal splenocyte proliferation in OVA-immunized
saline mice, and enhance a specific antibody and
cellular response against OVA in mice.
Seed Mouse bone 50 μg/ml methanol Positive control: LPS Keampferol can reduce cytokines and Lin et al.,
marrow- ectract and its organic 100 ng/ml negative chemokines produced by LPS-stimulated (2011)
derived DCs control: DMSO 0.1% v/v dendritic cells (DCS), and abrogate the ability
fraction and 20 μg/ml
(in vitro) of LPS-stimulated DCs to promote Ag-specific
kaempferol
T cell activation, both in vitro and in vivo.
Seed Mice (in vivo) P.O. 20 ml/kg of the Negative control: P.O. Cuscuta chinensis has the immune Lin et al.,
ethanol extract 200 g/l 20 mg/kg normal saline enhancement, anti-fatigue and anoxia (2003)
tolerance effects.

5 Neuroprotection Seed PC12 cells Cuscuta chinensis Positive control: 7S Cuscuta chinensis glycoside can induce Liu et al.,
activity (in vitro) glycoside up to nerve growth factor; neuronal differentiation with resulting (2003)
200 mg/l negative control: outgrowth of neutrites and increase of
distilled water acetylcholinesterase activity in rat
pheochromocytoma PC12 cells in a dose-
dependent manner
Seed Mice (in vivo) P.O. decoction of 20 g Positive control: Cuscuta chinensis with other Invigorating-Qi Liu et al.,
Cuscuta chinensis with Scopolamine drug had an evident antagonistic action to (1993)
other Invigorating-Qi I.V. 5 mg/kg; negative Scopolamine induced dysmnesia mice, and
drug control: could improve their memory.
P.O. normal saline
Seed Rats (in vivo) P.O. 50, 100 and 200 Positive control: 3 min Cuscuta chinensis with other 7 medicinal Chung et al.,
mg/day CMSD forebrain ischemia herbs (CMSD) had neuroprotective effects on (2006)
negative control: P.O. cultured primary neuron cell of neonatal rat,
vehicles with sham- and prevented ischemia-induced learning
operated disability and rescued hippocampal CA1
neurons from lethal ischemic damage in rats.
6 Antioxidant Seed SC50, IC50 The ethanol extract Positive control: They show potent antioxidant for preventing Yen et al.,
activity (in vitro) and its organic quercetin and kaemferol free radical damage to cell membranes (2008a)
fractions through scavenging of free radicals and
inhibition of the liquid peroxidation.
Seed IC50 (in vitro) Methanol extract and The ethyl acetate fraction of methanol extract Kwon et al.,
its organic fraction of Cuscuta chinensis seeds showed the (2000)
highest activity (EC50 value of 50 mg) by using
300 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

Table 4 (continued )

No. Activities Part of Model Formulation /dosage/ Control Result Reference


plant extract

the DPPH free radical scavenging method for


screening antioxidative effects
Seed MC3T3-E1 10, 20 and 40 mg/l Positive control: 0.1 mM The water extract can significantly inhibited Gao et al.,
cells (in vitro) water extract TBPH; negative control: the reactive oxygen species (ROS) generation, (2013)
DMSO 0.1% v/v malondialdehyde (MDA) production, and
increased the activity of superoxide
dismutase (SOD), GR, GST, and G6PD in
murine osteoblastic MC3T3-E1 cells tertiary
butyl hydroperoxide (TBHP) induced injury.
Seed PC 12 cells 25–250 mg/l Cuscuta Positive control: 0.3– Cuscuta chinensis flavonoids can protect PC12 Zhen et al.,
(in vitro) chinensis flavonoids 0.5 mm H2O2; negative cells against oxidative stress by its ability of (2006)
control: DMSO 0.1% v/v scavenging ROS and increasing the activity of
antioxidant enzyme.

7 Anti-aging Seed Mice (in vivo) P.O. 100, 200 and Positive control: S.C. 5% The Cuscuta chinensis polysaccharides had Cai et al.,
400 mg/kg/day of D-galactose 5 ml at the the anti-aging effects on senile mice model. (2005)
polysaccharides nape, vitamin E 200 mg/
kg/day; negative
control: S.C. distilled
water, vehicles

8 Effects on tumor Whole CCRF-CEM and 0.1–25 μg/ml water Negative control: culture Whole plant water extract has cytotoxic Zeraati
cells plant JM cells extract medium effect on human caucasian acute et al., (2010)
(in vitro) lymphoblastic leukemia (CCRF-CEM) cells.
Seed MCF-7 cells Cuscuta chinensis extract can stimulate Umehara
and T47D cells MCF-7 and T47D human breast cancer cell et al., (2004)
(in vitro) proliferation at a concentration of 10 μM
Seed Hela cells 1, 5, 10, 50, 100 μg/ml Positive control: 0.1 μM Isolated compounds from Cuscuta chinensis Yang et al.,
(in vitro) of its organic fraction 17-β-estradiol; negative seeds ethanol extract stimulated Hela cell via (2011)
and 1, 5, 10, 50 μM of control: DMSO 0.1% v/v estrogenic receptor ERα and ERβ
its isolated flavonoids

9 Renoprotective Seed Rats (in vivo) P.O. 250 mg/kg/day Positive control: renal The water extract 250 mg/kg/day oral Shin et al.,
aqueous extract arteries were clamped administration can recover rats renal (2011)
for 45 h; negative function parameters from ischemia/
control: operation reperfusion-induced ARF.
without clamping renal
arteries

10 Reproductive Seed Penile tissues 1–5 mg/ml ethanol Positive control: 1, 2, 3, 4 and 5 mg/ml of Cuscuta chinensis Sun et al.,
system (in vitro) extract 10 μmol/l phenylephrine extract has the effect on the rabbit penile (2013)
precontracted, 10 nmol/l corpus cavernosum (PCC) by relaxing PCC
sildenafil citrate; phenylephrine (10 μmol/l) induced
negative control: DMSO contraction in a dose dependent manner.
0.1%
Seed Rats (in vivo) P.O. 100 and 300 Negative control: P.O. 100 mg and 300 mg/day for 4 weeks oral Sohn et al.,
mg/kg of the herbal distilled water administration of KH-204 decresed blood (2008)
formula (KH-204) pressure and enhanced penile erection in
spontaneous hypertensive male rats.
Seed Human P.O. VPX (25 mg of Negative control: P.O. VPX can improve sexual abilities on patients Shah et al.,
(in vivo) Cuscuta chinensis placebo with erectile dysfunction. (2012)
seeds with other
herbal medicie) twice
daily
Seed Human Decoction with other Cuscuta chinensis improve sperm motility Peng et al.,
spermatozoa herbal medicine and help stabilized sperm membrane (1997)
(in vitro) function.
Seed Mice (in vivo) P.O. 33.8 mg/kg and Positive control: P.O. Seed of Cuscuta chinensis ethanol extract Yang et al.,
67.6 mg/kg of the 1.3 g/kg 33.8 mg/kg and 67.6 mg/kg can reverse the (2008)
ethanol extract metyltestoseterone or reduction of testosterone level, the androgen
6 g/kg Jinkui Shenqi receptor mRNA level and protein level
wan; negative control: P. induced by the hydrocortisone in the kidney
O. distilled water and testicle.
Seed Rats (in vivo) P.O. 5–10 mg/100 g of Negative control: P.O. Flavonoids from Cuscuta chinensis seeds (FSC) Wang et al.,
flavonoids normal saline improved the ovarian endocrine functions. (2002)

11 Prevention of Seed Pregnant rats P.O. 2.6 and 5.2 Positive control: S.C. Cuscuta chinensis total flavonoids regulate Ma et al.,
abortion (in vivo) mg/kg/day with S.C. 0.3 mg/kg/day the proliferation and apoptosis of the (2008b)
bromocriptine bromocriptine; negative deciduas and cytotrophoblasts and prevent
control: do nothing spontaneous abortion in rats.
Seed Pregnant rats P.O. 2.6 and 5.2 Positive control: S.C. Cuscuta chinensis total flavonoids can Ma et al.,
(in vivo) mg/kg/day with S.C. 0.3 mg/kg/day regulate endocrinological and immunological (2008a)
bromocriptine bromocriptine; negative network equilibrium on maternal-fetal face
control: do nothing and finally achieve prevention of fetal
abortion.
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 301

Table 4 (continued )

No. Activities Part of Model Formulation /dosage/ Control Result Reference


plant extract

12 Antimutagenic Whole Bacteria 0.1 mg of dry plant Positive control: Trp-P-1 Cuscuta chinensis methanol extract has the Nakahara
effect plant (in vitro) methanol extract carcinogen antimutagenic effect against Trp-P-1 in Ames et al., (2002)
test, ED90 values lower than 5 μL/plate

13 Antidiabetic Seed Mice (in vivo) P.O. 150, 300 and Positive control: I.V. 300, 600 mg/kg of Cuscuta chinensis Li et al.,
activity 600 mg/kg total 50 mg/kg alloxan, P.O. polysaccharides orally significantly lower the (2008a)
flavonoids acarbose 20mg/kg; high fasting blood glucose level, raise body
negative control: P.O. weight and immune organs.
normal saline
Seed Rats (in vivo) P.O. 100, 200 and Positive control: I.V. 200, 400 mg/kg orally of Cuscuta chinensis Xu et al.,
400 mg/kg 50 mg/kg streptomycin, polysaccharides significantly improve the (2011)
polysaccharides P.O. 13 mg/kg acar bose; weight loss of diabetic rats, reduce fasting
negative control: I.V. blood glucose and glycosylated serum
sodium citrate buffer protein, CCP 400 mg/kg significantly reduce
pH 4.5 CHO level, and CCP 400, 200, 100 mg/kg
could significantly reduce TG level.

14 Cardioprotection Seed Dogs (in vivo) I.V. 0.1 g/kg Cuscuta Cuscuta chinensis decoction in dogs can Jiangsu New
activity chinensis decoction reduce their blood pressure. Medical
College,
(1997)
Seed Dogs (in vivo) I.V. 20 mg/kg and Positive control: 20 mg/kg and 1ml/kg I.V. for 1 min of Liu et al.,
1 ml/kg of 3 different P.O. 20 mg/kg 3 different methods of extraction on mature (2004)
ethanol extract Diaoxinxuekang male hybrid dogs can increase coronary
traditional chinense blood flow and reduce myocardial oxygen
formula consumption.
Seed Rats (in vivo) 2 mg/g Cuscuta Positive control: Cuscuta chinensis seeds extract slowed down Han et al.,
chinensis seeds extract coronary artery ligated heart rate of MI /RI rats, declined ST level, (2011)
negative control: sham- reduced the degree of MI and the content of
operated I.V. normal CK, CK-MB, LDH, AST in blood serum.
saline

15 Antidepressant Arial part Mice (in vivo) I.P. 50 and 100 mg/kg Positive control: Methanolic extract had antidepressant Mokhtarifar
activity methanol extract I.P. 23 mg/kg effects by significantly reduced immobility et al., (2012)
imipramine and times in TST (50 mg/kg IP) and 50 and
fluoxetine 100 mg/kg IP in FST, imipramine and
fluoxetine (both 32 mg/kg IP), as positive
controls.

16 CNS depressant Whole Mice (in vivo) P.O. 1 g/kg aqueous The extract significantly decreased the motor Akbar et al.,
plant extract activity and the tonic and clonic phases of (1985)
electrically induced seizures in mice.

17 Effects on Seed AMMC 0.1, 1, 10 and 50 mg/ml Positive control: 10  8, Cuscuta chinensis water extract promoted Li et al., (
melanogenesis (in vitro) of water extract 10  7, 10  6, 10  5 mol/l melanogenesis of AMMC in dose-dependent 2008b)
8-methoxypsoralan manner, and increased the tyrosinase
negative control: culture activities.
medium
Seed Guinea pigs Apply 0.49, 0.93 and Positive control: apply 1. 86, 0. 93 g/kg externally applied on guinea Shen et al.,
(in vivo) 1.86 g/kg of Cuscuta 5% H2O2 1 ml on skin, pig skin significantly increased the skin (2012)
chinensis in alchohol methoxsalen 1 ml; melanin generation, and tyrosinase
preparation (0.25 g/g) negative control: apply production
alcohol on skin
Seed IC50 (in vitro) 0.5–20 mg/ml of Positive control: 100 The water extract of Cuscuta chinensis show Wang et al.,
water and ethanol μg/ml arbutin; negative inhibition of tyrosinase activity in a dose (2014)
extract control: solvent of dependent manner with IC50 value of 12
sample mg/ml and decrease the synthesis of
melanin.
Seed Zebrafish P.O. 0.5–20 mg/ml of Positive control: 100 Water extract of Cuscuta chinensis seeds Wang et al.,
(in vivo) water and ethanol μg/ml arbutin; negative demonstrate depigmenting activity in (2014)
extract control: solvent of zebrafish
sample

18 Anthelminthic Seed Goldfish P.O. 1% body weight of Negative control: DMSO The EC50 of Cuscuta chinensis methanol Huang et al.,
(in vivo) 125–300 mg/l organic extract at 48 h¼ 15.9 mg/L, by using in vivo (2013)
fraction anthelminthic efficacy assay.

19 Anti-nociceptive Seed Mice (in vivo) P.O. 20–500 mg/kg 100 and 500 mg/kg of Cuscuta chinensis seeds Liao et al.,
methanol extract methanol extract significantly reduce the (2014)
writhing response, at the dose of 20–500
mg/kg it can reduce licking time

20 Anti- Seed Mice (in vivo) P.O. 20–500 mg/kg The extract can significantly reduce Liao et al.,
inflammatory methanol extract λ-carrageenan-induce edema paw in the (2014)
time of four hours after injection with
λ-carrageenan.
302 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

effects (Yang et al., 2011). The results indicate that the in vitro 5.6. Antioxidant activities
effect of anti-osteoporosis showed the mechanisms of this activity
by promoting bone formation and inhibiting bone absorption, The ethyl acetate organic fraction from ethanol extract of the seeds
improving bone mass, balancing bone metabolism by acting as a of Cuscuta chinensis was observed to possess strongest antioxidant
phytoestrogen. effects with highest content of the flavonoid compounds kaempferol
and quercetin. Ethanol extract of Cuscuta chinensis seeds is an effective
antioxidant for free radical damage on cell membranes prevention
5.4. Immunological effects
through scavenging of free radicals and by inhibiting the liquid
peroxidation process (Yen et al., 2008a). The ethyl acetate fraction of
An ethanol extract of the dry seed of Cuscuta chinensis (EESC)
Cuscuta chinensis seeds methanol extract showed the highest activity
could be used as a vaccine adjuvant. Pan et al. (2005) found that EESC
(EC50 ¼50 mg) by using the DPPH free radical scavenging method for
significantly enhanced the mitogen- and ovalbumin (OVA)-stimulated
screening anti-oxidative effects, and there are 5 compounds isolated
splenocyte proliferation in OVA-immunized mice, and EESC could
from this fraction; methyl 4-hydroxy-3,5-dimethoxycinnamate, caffeic
significantly enhance a specific antibody and cellular response against
acid, quercetin, kaempferol and calycopteretin (EC50 values of 0.6, 8,
OVA in mice. Kaempferol, one of major flavonoids isolated from the
19, 17 and 12 mg, respectively) (Kwon et al., 2000). Gao et al. studied
dry matured seeds of Cuscuta chinensis methanol extract can decrease
about Cuscuta chinensis water extraction protecting murine osteoblas-
chemokines and cytokines which produced by LPS-stimulated den-
tic MC3T3-E1 cells against tertiary butyl hydroperoxide (TBHP)
dritic cells (DCs), and this reduction was not because of its cytotoxicity
induced injury. They found that the Cuscuta chinensis protects TBHP-
on DCs. Besides, kaempferol decreases the maturation of DC. An
treated MC3T3-E1 cells from death in a dose-dependent manner and
in vitro and in vivo experiment showed that kaempferol negated
it could suppress the malondialdehyde (MDA), reactive oxygen species
LPS-stimulated DCs ability to promote the antigen-specific T cell
(ROS) productions, and significantly enhance the Glutathione S-trans-
activation. Therefore, the results indicate that Cuscuta chinensis seeds
ferase, superoxide dismutase (SOD), glutathione reductase, and
have the immunosuppressive effects on DCs and that kaempferol
Glucose-6-phosphate dehydrogenase activities. In mitochondria, the
inhibits DC function, which suggests that kaempferol has potential in
release of the caspase 3 and Bax expression and cyto c were decreased,
treatment of autoimmune diseases and chronic inflammation (Lin
but the expression of antiapoptotic IDH2, Sirt3, and Bcl-2 was
et al., 2011). Lin et al. studied about the effect of immune enhance-
increased in the cells incubated with Cuscuta chinensis. Therefore they
ment, anti-fatigue and anoxia tolerance on mice of four kinds of
suggested that the treatment of Cuscuta chinensis with the TBHP
dodder seeds from Shandong Province. They found that Cuscuta
regulated the oxidative stress-induced apoptosis in MC3T3-E1 cells
chinensis had better effects than the other two kinds, and that the
because of its antioxidant abilities and functioning against
water extracts had better effects than the alcohol extracts in enhancing
mitochondria-dependent pathways (Gao et al., 2013). Pretreatment
phagocytosis of mice's macrophage, increasing the weights of thymus
of Cuscuta chinensis flavonoids (CF) with different concentrations for
and spleen of the immature mice and could prolong the survival time
0.5 h enhances the survival rate of PC12 cells, inhibits H2O2-induced
of mice swimming and oxygen lacking (Lin et al., 2003). From the
apoptosis and CF had the DPPH-generated scavenging free radicals
studies, Cuscuta chinensis, its extracts and isolated compounds, espe-
activity in a dose-dependent manner. These results suggest that CF can
cially kaempferol, have an impressive effect on the immune system.
protect PC12 cells against oxidative stress by its scavenging ROS ability
The aqueous extract showed immunosuppression effects while the
and increasing the antioxidant enzymes activity (Zhen et al., 2006).
ethanol extract showed immunostimulating effects. Further research
The in vitro antioxidant activities considered to be due to phenolic
works concerning the effects of Cuscuta chinensis on other immune
compounds, flavonoids and phenolic acids found in Cuscuta chinensis
organs are imperative.
which can act as free radical scavengers. The antioxidant activity could
be useful in preventing oxidative stress implicated with the develop-
5.5. Effect on neuronal system ment of many diseases such as cardiovascular or neurological diseases.

Glycoside from Cuscuta chinensis can induce neuronal differ- 5.7. Anti-aging activities
entiation with resulting outgrowth of neutrites and improve
acetylcholinesterase activity in rat phaeochromocytoma PC12 cells Cai et al. studied the effect of a polysaccharide from Cuscuta
in a dose-dependent manner, and glycoside inducing the PC12 chinensis (PCCL) in senile mice model. The results showed that
cells differentiation might be related to mitogen-activated protein polysaccharides from Cuscuta chinensis with oral administration of
kinase-mediated signaling pathway (Liu et al., 2003). Cuscuta 100, 200, 400 mg/kg/d (for 7 weeks) can raise Lipofuscin (LF),
chinensis, together with other invigorating-qi medicines, has an spleen index, thymus index, glutathione peroxidase and SOD in
effect on scopolamine-induced dysmnesia mice for antagonistic the liver and kidney. In the brain, LF and MDA contents were
action, and could improve their memory, and the acetylcholines- dropping which in senile mice model the results were found the
terase activity in the mice of the group that received the drug were opposite way, indicating that the PCCL can scavenge free radicals,
significantly less than those in the positive control group which has anti-lipid peroxidation and anti-aging activities (Cai et al.,
received scopolamine 5 mg/kg intraperitoneally and the erroneous 2005). Ethanol extract of Cuscuta chinensis showed anti-aging
times of the animal's reaction (Liu et al., 1993). Cuscuta chinensis effects in the D-galactose-induced rat aging model. The D-galactose
with other 7 medicinal herbs (CMSD) had neuroprotective effects aging Wistar rat model was performed by subcutaneous injection
on cultured primary neuron cell of neonatal rat. Chung et al. found of 48 mg/kg D-galactose in the cervico-dorsal region once daily for
that CMSD water extract protected the cell from Amyloid-β- 45 days. While Cuscuta chinensis was given by gavage, the results
induced cell death in a dose dependent manner, and the most showed that on day 30, Cuscuta chinensis significantly inhibited
effective concentration was 25–50 μg/mL. CMSD was also demon- the non-enzymatic glycosylation reaction of D-galactose-induced
strated to have protective effect from N-methyl-D-aspartate rat aging model (Li et al., 2013). These findings indicate that
receptor-mediated glutamate toxicity on primary cultured neu- Cuscuta chinensis has significant anti-aging effects in animals.
rons. In addition, CMSD oral administration in mice can also Moreover, Cuscuta chinensis is indicated in “Tai Ping Sheng Hui
recover the hippocampal CA1 neurons from lethal ischemic Fang” which are recorded anti-aging prescriptions. The research
damage and prevent learning disability because of ischemia- showed that Cuscuta chinensis could regulate immune function,
induced brain damage (Chung et al., 2006). prolong cell cycle, improve body metabolism and the function of
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 303

internal organs and stress ability, which express as anti-aging corpus cavernosum of spontaneous hypertensive male rats. They
effects (Yang and Huang, 1998). From these results, it has obvious found that the KH-204 herbal formulation enhances intracavernous
effects on anti-aging and can prove the claim of the anti-aging pressure and nitric oxide-cyclic guanosine monophosphate activity in
uses in ancient times. However, further research work and devel- penile tissues of spontaneous hypertensive male rats (Sohn et al.,
opment should be performed to provide more information about 2008). Moreover, 12 weeks treatment of Cuscuta chinensis in combi-
the adverse effects or toxicities. nation with other herbal medicines called VXP can improve sexual
functions of erectile dysfunction patients, by significantly improving
5.8. Effects on tumor cells IIEF-Erectile Function scores including orgasmic function, sexual
desire, intercourse satisfaction, and overall satisfaction, while there
Zeraati et al. (2010) studied about cytotoxic effect of Cuscuta was no difference in incidence of side effects and subject's rating for
chinensis whole plant water extract on human Caucasian acute tolerability against placebo group (Shah et al., 2012). Yang et al.
lymphoblastic leukemia (CCRF-CEM) and a human lymphocyte, discovered the effect of the total flavones from Cuscuta chinensis
Jurkat (JM) cells. The minimum effective concentration of the plant seeds on the kidney-yang deficiency male mouse. Total flavones from
extract was 1 μg/mL, and the stronger effect was shown in the Cuscuta chinensis seeds can reverse the reduction of testosterone
dose up to10 μg/mL. The extract has the IC50 value of 3 μg/mL in level, androgen receptor mRNA level and protein level induced by the
24 h and 2.5 μg/mL in 48 h, respectively. But in these doses the hydrocortisone in the kidney and testicle (Yang et al., 2008). Peng
extract did not have cytotoxic activity on JM cells. Seeds of Cuscuta et al. studied about the effects of Cuscuta chinensis on human sperm
chinensis extract can stimulate MCF-7 and T47D human breast motility in vitro and cytomembrane function. The results showed that
cancer cell proliferation at a concentration of 10 μM (Umehara the motility of sperm significantly improved and after incubation the
et al., 2004), and the Cuscuta chinensis seeds absolute ethanol function of sperm membrane became more stable (Peng et al., 1997).
extract show no cytotoxicity against T47D cells (Ahmed et al., Flavonoids from Cuscuta chinensis seeds (FSC) enhanced the ovarian
2014). Compounds isolated from the ethanol extract of Cuscuta endocrine functions in female rats exposed to psychological stress by
chinensis seeds promoted Hela cells (a human cervical cancer cell reducing the beta-endorphin content in hypothalamus and increas-
line) through estrogenic receptor ERα and ERβ, while quercetin, ing the LH content in anterior pituitaries and the basophilic cells,
kaempferol and isorhamnetin, and some other isolated com- which may be the mechanism of FSC in improving hypothalamus-
pounds showed more selectivity in ERβ (Yang et al., 2011). The pituitary-ovary axis (Wang et al., 2002). In order to investigate the
cytotoxic effects of Cuscuta chinensis on human cancer cell line effects of herbal medicines on small uterus (infantile uterus), 46
may also have the potential in anticancer application and might cases of patients with infantile uteruses were treated with a
support the use of this plant in prostate, bladder, gastro- compound formula containing Cuscuta chinensis, Rhizoma Curculigi-
esophageal and brain cancers (Alaoui-Jamali, 2010), except in the nis, Radix Morindae Officinalis, Radix Polgoni Multiflori, Radix Rehman-
human cell line that requires estrogen to grow because Cuscuta niae Praeparata, Cervus elaphus and Herba Epimedii. This drug formula
chinensis and some of its extracts can act as a phytoestrogen so can was taken together with diethylstilbestrol 1 mg dosage by starting on
be counter-productive in such cell lines. the 5th day of the menstrual cycle, only once a day for 20 days.
Following treatment, 37 cases exhibited complete recovery while
5.9. Renoprotective effects 9 cases showed improvement (Yin et al., 1992). The in vitro and
in vivo studies about the reproductive system of Cuscuta chinensis, its
Cuscuta chinensis seed water extract has a recovery effect in extracts and isolations showed potential improved effects in both
ischemia/reperfusion-induced acute renal failure (ARF) rats on the male and female reproductive system including improved fertility.
renal function parameters. In oral administration of 250 mg/kg/
day of the extract in male SD rats for four days, the results 5.11. Prevent abortion
indicated that the parameters of renal function including urinary
excretion rate, osmolality, potassium, sodium and chloride ions, Total flavonoids of Cuscuta chinensis regulated the deciduas and
creatinine clearance and solute-free water reabsorption were cytotrophoblasts proliferation and apoptosis, prevented sponta-
significantly improved and also ameliorated tubular damage in neous abortions by enhancing expression of proliferating cell
ischemia/reperfusion induced ARF rats (Shin et al., 2011). Although nuclear antigen on trophoblast and deciduas, heparin blinding
the renoprotective effect of Cuscuta chinensis against physical- epidermal growth factor on trophoblast, progesterone receptor on
induced kidney damage in rats was investigated and showed deciduas, and decreased expression of heparin blinding epidermal
satisfactory results, the mechanism and the chemical constituents growth factor on deciduas in pregnant rats (Ma et al., 2008b). Total
responsible for these activities are still unclear, therefore further flavonoids of Cuscuta chinensis could regulate endocrinological and
research work should be conducted. immunological network equilibration on maternal-fetal face,
finally achieving the prevention of abortion (Ma et al., 2008a).
5.10. Effect on reproductive system Moreover, Cuscuta chinensis with Radix Codonopsis Pilosulae, Colla
Corii Asini, Rhizoma Atractylodis Macrocephalae and Radix Dipsaci
The ethanol extract of Cuscuta chinensis has effects on the rabbit were used in treatment of 110 threatened abortion cases. Oral
penile corpus cavernosum (PCC) by relaxing PCC phenylephrine administration of the medicine once daily in the patients for 10
induced contraction in a dose dependent manner and significantly days, and the total effective rate was found to be 96.36% (Zhu et al.,
increases sildenafil-induced PCC relaxation. The penile cavernous 1987).
tissue incubated with the Cuscuta chinensis extracts show that the
cyclic guanosine monophosphate and cyclic adenosine monopho- 5.12. Anti-mutagenic activity
sphate levels in the PCC were raised significantly. Since the Cuscuta
chinensis extract presents a relaxing effect on penile cavernous tissue The methanol extract of Cuscuta chinensis has antimutagenic
by activating the nitric oxide-cyclic guanosine monophosphate path- effect against Trp-P-1 (3-amino-1,4-dimethyl-5H-pyrido[4,3-b]
way, it may improve erectile dysfunction conditions which do not indole) in Ames test, which has ED90 (suppressed 90% of the
completely respond to sildenafil (Sun et al., 2013). Sohn et al. also mutagenesis) values lower than 5 μL/plate of 0.1 mg dry plant
investigated the effects of the formulation of Cuscuta chinensis with material equivalent (Nakahara et al., 2002). From the results, we
other herbal medicines called KH-204 on the penile erection and also know that Cuscuta chinensis methanol extract may not cause
304 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

cancer (the mutagenic chemical may act as a carcinogen) and has 5.16. CNS depressant activities
an ability to prevent mutation but the Ames test is only one of the
initial screens for potential drugs to weed out possible carcino- The aqueous extract of the whole plant of Cuscuta chinensis was
gens, so further investigation should continue. studied for its CNS-depressant activity. Administer of 1 g/kg body
weight per os of the extract in mice significantly reduced the
motor activity and the tonic/clonic phases of electrically-induced
5.13. Anti-diabetic activities seizures. The narcosis and survival time under hypoxia in mice of
pentobarbitone were significantly intensified. The amphetamine-
Li et al. studied about Cuscuta chinensis polysaccharides (CCP) induced excitation was antagonized and the extract also exhibited
effects on diabetic mice induced by alloxan. Oral administration of marked analgesic activity. However, the extract did not affect the
300, 600 mg/kg CCP significantly decreases the high fasting blood reserpine-induced hypothermia and normal body temperature in
glucose level, increases weight of body and immune organs rats (Akbar et al., 1985). The results indicate that Cuscuta chinensis
(Li et al., 2008a,). Moreover, Xu et al. (2011) studied CCP effects has CNS depressant activity due to its potential effect against
in diabetic rat model. They found that after 15 days of treatment seizure and synergistic effect when combined with pentobarbi-
with oral administration of 200 and 400 mg/kg CCP in diabetic rats tone. Further research should be considered to evaluate the effects
could significantly reduce fasting blood glucose, glycosylate serum in human subjects.
protein and increase body weight; 400 mg/kg CCP could signifi-
cantly reduce CHO levels; and 400, 200, 100 mg/kg CCP could 5.17. Effect on melanin production
significantly reduce TG levels, but observed no obvious effect on
insulin levels. Moreover, Cuscuta chinensis has been used in anti- Li et al. studied about the Cuscuta chinensis aqueous extract on
diabetic prescriptions because it can invigorate kidney and supple- the differentiation of amelanotic melanocytes (AMMC) of outer
ment essence (Li et al., 2004; Chauhan et al., 2010). These root sheath from human hair follicles. The results showed that the
mentioned above results of preclinical investigations show that extract promoted melanogenesis of AMMC and increased the
Cuscuta chinensis polysaccharides show potential effect on redu- tyrosinase activities in a dose-dependent manner, but the effect
cing blood sugar in type-2 diabetes, but the mechanism of this was weaker than 8-methoxypsoralan (Li et al., 2008b). The doses
effect is still unclear. Therefore, more studies are needed to prove of 1.86, 0.93 g/kg Cuscuta chinensis preparation externally applied
clinical efficacy and reveal the exact mechanism of action. on guinea pig skin which significantly increased the skin melanin
generation and tyrosinase production, and high doses of Cuscuta
chinensis increased the activity of cholinesterase and reduced the
5.14. Cardiovascular activities activity of monoamine oxidase. It also showed good effects on
vitiligo treatment in guinea pigs (Shen et al., 2012). Furthermore,
Cuscuta chinensis seeds in the preparation of tincture and there is another study on melanogenesis effect of Cuscuta chinensis
decoction exhibit a contractile effect on toad heart model seeds water extract and ethanol extract in vitro and in vivo.
in vitro. The heart rate was shown to rise with the decoction Tyrosinase activity assay in mushroom and mouse B16F10 mela-
preparation but was shown to decrease with the tincture prepara- noma cell and depigmenting activity in zebrafish were performed.
tion. Administration of 0.1 g/kg bodyweight of Cuscuta chinensis The water extract showed the inhibition of tyrosinase activity in a
seeds decoction given to anaesthetized dogs by intravenous (I.V.) dose-dependent manner with IC50 value of 12 mg/mL (after
injection produced a reduction in their blood pressure (Jiangsu 2 weeks of preparation extract) while the ethanol extract showed
New Medical College, 1997). Liu et al. (2004) administered IV the opposite effect in tyrosinase activity, and the water extract
injection of Cuscuta chinensis seeds extract to mature male hybrid decreased the synthesis of melanin. They conclude that the
dogs and found that the extract could increase coronary blood contrast effect on melanin production is due to the different
flow as well as reduce myocardial oxygen consumption. Cuscuta extraction methods (Wang et al., 2014).
chinensis seeds extract significantly has a protective effect on
myocardial ischemia/reperfusion injury (MI/RI) rats (Han et al., 5.18. Anthelmintic activities
2011). The results show that the extract can slow down heart rate.
The level of ST significantly declined when blood is reperfused for Cuscuta chinensis can be used as an anthelmintic drug against
60, 90, 120 min. The degree of myocardial infarction was obviously Dactylogyrus intermedius (Monogenea) in goldfish (Carassius aur-
decreased and the content (U/L) of creatine kinase (CK), creatine atus). The methanol extract of Cuscuta chinensis has EC50 at
kinase (CK-MB), lactate dehydrogenase (LDH), aspartate amino- 48 h ¼15.9 mg/L, by using in vivo anthelmintic efficacy assay
transferase (AST) in blood serum were significantly reduced when (Huang et al., 2013). The work on anthelminthic activity is very
compared to control group. However, further studies are required little with only one monogean parasite being studied. Therefore,
to determine its specific interaction and exact mechanism of the studies on other models of animal and human subjects should
action. be considered for its effects against tapeworms, roundworms or
flukes.

5.15. Anti-depressant activities 5.19. Anti-nociceptive and anti-inflammatory activities

Mokhtarifar et al. studied the antidepressant effects of Cuscuta The methanol extract of Cuscuta chinensis seed was performed
chinensis methanol extract in mice by using force swimming test to provide anti-nociceptive and anti-inflammatory activities
(FST) and tail suspension test (TST). The results showed that in vivo by using the response of acetic acid-induced writhing and
32 mg/kg I.P. of imipramine and fluoxetine as the positive controls formalin-induced paw licking methods for evaluated anti-
significantly have the effects on both FST and TST. Administration nociceptive activities, and λ-carrageenan-induced paw edema in
of 50 mg/kg I.P. significantly decreased immobility times in TST mouse for evaluated anti-inflammatory activities. They found that
and 50 and 100 mg/kg I.P. of Cuscuta chinensis methanolic extract 100, 500 mg/kg of Cuscuta chinensis seeds methanol extract sig-
significantly reduced immobility times in FST (Mokhtarifar et al., nificantly reduces the writhing response. At the dose of
2012). 20–500 mg/kg, it can reduce licking time (early phase, 20 and
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 305

100 mg/kg and late phases, 100 mg/kg). In formalin test assay, the 6.2. Prevent abortion
extract can significantly reduce λ-carrageenan-induced edema
paw in the time of four hours after injection with λ-carrageenan. Cuscuta chinensis dried seeds 30 g, Semen soojiae Atricolor fruits
Moreover, it can reduce the level of IL-1β, IL-6, NF-κB, TNF-α, and 50 g, and Semen oryzae Glutinosae fruits 100 g are mixed together
COX-2. The mechanism of action may be due to the decrease in till they become porridge, and are usually taken (PCHMPD, 2010).
MDA and NO levels by the increasing activities of SOD, glutathione
peroxidase and glutathione reductase in the liver (Liao et al.,
2014). These pharmacological studies prove the ethnomedicinal 6.3. Male reproductive system disease
uses of Cuscuta chinensis as an anti-inflammatory agent and a pain
reliever. Cuscuta chinensis dried seeds 30 g, with some other traditional
Chinese medicine decocted with water. Oral administration for 20
days to 4 months can treat male infertility, chronic prostatitis,
5.20. Toxicological reports erectile dysfunction (Wang, 2000). In 19 cases of male infertility,
oral administration of Cuscuta chinensis dried seeds 9 g and fructus
Toxicity of Cuscuta chinensis is very low, as the LD50 of Cuscuta lycii dried mature fruits 30 g, decocted with water, divided into
chinensis seeds ethanol extract intradermal injection into mice was 3 times a day every day for 2 months (1 course of treatment),
found to be 2.465 g/kg. Gavage 30–40 g/kg of Cuscuta chinensis which found 89.5% of total efficiency (Wang, 2001).
seeds water extract in rats did not demonstrate any intoxicating
symptoms. Furthermore, when rats were gavaged with 4.15 g/kg of
6.4. Chyluria
Cuscuta chinensis seeds water extract or tincture once daily for 70
days, no development in pathological change or abnormality was
Fried Cuscuta chinensis dried seeds, Cervus Nippon Temminck
found (Jiangsu New Medical College, 1997). The acute toxicity study
horn, Codonopsis pilosula dried roots 12 g each, Tenodera sinensis
revealed the drug to be non-toxic up to an oral dose of 5 g/kg body
Saussure dried ootheca, Smilax corbularia dried rhizome, Angelica
weight of whole plant aqueous extract in mice (Akbar et al., 1985).
sinensis dried roots 9 g each, Chinemys reevesii carapace 21 g,
The toxicity of this plant against rat embryo was studied because of
Astragalus membranaceus dried roots, Meretrix meterrix Linnaeus
its ability to prevent abortion. A total of 60 SD pregnant rats were
shell podwder 30 g each, Poria cocos dried scerotium 18 g, Citrus
divided into 6 groups for high, medium and low dose of Cuscuta
reticulate dried fruits peel 6 g. If combined with hematuria, add
chinensis water extract (40 g/kg/d, 20 g/kg/d and 10 g/kg/d, respec-
Eclipta prostrate dried trunks 15 g, decocted with water for oral
tively) given by gavage for day 6–18, using normal saline 100 mL as
administration, and this preparation can treat chyluria or hema-
a negative control and cyclophosphamide injection 12.5 mg/kg on
turia which is caused by filariasis (Pan et al., 2008).
day 13 as a positive control group. No significant difference was
found in low, medium and high dose on embryo development of
appearance, body length, tail length, body weight, dry weight, 6.5. Chloasma faciei
placenta weight, skeletal development when compared to negative
control. The maximum tolerance dose of water extract of Cuscuta Cuscuta chinensis dried seeds, fresh and cooked Rehmannia
chinensis in SD rats is 80 g/kg, which belongs to non-toxic drugs glutinosa dried roots 15 g each, Ligustri lucidium dried mature
(Xia, 2012). Up to date, there are no reports on human toxicity or fruits, Polygonum multiflora dried roots 12 g each, Eclipta prostrate
side effects of Cuscuta chinensis. It can be noticed that pharmaco- dried trunks, Paeonia lactiflora Pall dried roots, Angelica sinensis
logical and toxicological studies have been conducted on only the dried roots 10 g each, Equus asinus skin, Lycium barbarum L. dried
seeds of Cuscuta chinensis. Studies on the fruit or whole plant which mature fruits 9 g each. If combined with anemia, add Codonopsis
contains some phytochemical constituents are very little, especially pilosula dried roots, Astragalus membranaceus dried roots 15 g
the fruits which show no pharmacological studies at all. Studies on each, Spatholobus subersctus Dunn dried trunks 30 g, Psoralea
the leaf and stem also indicate that the ethnomedicinal uses of corylifolia Linn. dried mature fruits 9 g, decocted with water for
these parts are still unknown. We suggest that further investiga- oral administration (Pan et al., 2008).
tions are needed to provide more information for ethnomedicinal
uses of this plant for their anti-inflammatory, pain relieving,
anthelminthic and lactation activities. 6.6. Glomerulonephritis

Dried Cuscuta chinensis seeds 30 g, decocted with water 300 ml,


oral administration 2 times a day for 3 months. In a 13 case study,
6. Clinical applications
12 cases were found to be effective (92.31%) (Xia, 2000).

Currently in China, Cuscuta chinensis seeds are usually pre-


scribed by doctors or traditional Chinese medicine practitioners 6.7. Nocturia
with a usual dosage of 9–15 g oral administration, and the
maximum dosage of 30 g each time (Ceng and Zhang, 2000) Using Jisheng, one of the Cuscuta chinensis seeds formulation,
without obvious side effects reported for supplementing or treat- oral administration 2 times day for 1 month (1 course of treat-
ing various diseases, as shown below. ment), 2–3 months, the total cure rate was 96.97% (You, 2005).

6.1. Female infertility 6.8. Treatment and prevent diabetes

Cuscuta chinensis dried seeds 25 g and Angelica sinensis dried Dried Cuscuta chinensis seeds, Lycium barbarum dried mature
roots 10 g, decocted with water for oral administration 3 times a fruits, cooked Rehmannia glutinosa dried roots 10 g each, Ilex
day. Start on the first day of menstruation and continue to the 18th pubescens dried roots 30 g, freas Zingiber officinale Rosc. roots
day (1 course of treatment). The duration of treatment is about 3 pieces, steaming and then usually drink the decoction
2–3 courses (Pan et al., 2008). (Song, 2010).
306 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

6.9. Herpes zoster Acknowledgments

Dried grounded seeds of Cuscuta chinensis seeds 50–100g, add This research was supported by the National Natural Science
some sesame oil mix until they become an ointment, and before Foundation of China (81374050) and (NSFC81125024), Program for
using add some saline, then apply on lesion once a day for 2–5 Innovative Research Team in Universities of Tianjin (TD12-5033)
days (Pan et al., 2008). In 98 cases, 2–5 day period of time, 100% and Tianjin Research Program of Application Foundation and
effective (Niu and Xia, 1994), and in another 49 cases, also found Advanced Technology (12JCQNJC08800) and we are thankful to
100% effective (Sun and Fu, 2000). Prof. Michael Heinrich and the other three anonymous reviewers
for their useful comments and suggestions on the manuscript.
6.10. Acne
References
Dried Cuscuta chinensis seeds 30 g in 500 ml water, apply on
the lesion, 1–2 times a day for 7 days (1 course of treatment), the Ahmed, HM.M., Yeh, J.Y., Tang, Y.C., Cheng, W.T.K., Ou, B.R., 2014. Molecular
duration of treatment is about 1–2 courses, in 50 cases, 94% screening of Chinese medicinal plants for progestogenic and anti-
progestogenic activity. Journal of Bioscience 39 (3), 453–461.
effective (Yu, 1996). Akbar, S., Nisa, M., Tariq, M., 1985. CNS depressant activity of Cuscuta chinensis Lam.
Pharmaceutical Biology 23, 91–94.
Alaoui-Jamali, M., 2010. Alternative and Complementary Therapies for Cancer:
6.11. Vitiligo Integrative Approaches and Discovery of Conventinal Drugs. Springer, Springer
Science þ Business Media, New York.
Bao, X., Wang, Z., Fang, J., Li, X., 2002. Structural features of an immunostimulating
Cuscuta chinensis seeds 9 g, soaked in 60 ml of 95% ethanol for
and antioxidant acidic polysaccharide from the seeds of Cuscuta chinensis.
2–3 day, then apply the solution on the lesion 2–3 times a day Planta Medica 68, 237–239.
(PCHMPD, 2010). Cai, X.G., Xu, A.X., Ge, B., Gao, X., Yang, S.H., 2005. Effects of a polysaccharide from
CCL on inhibiting oxygen free radical threshold of senile mice model. Acta
Summing up, the clinical applications from Cuscuta chinensis
Academiae Medicinae Militaris Tertiae 27, 1326–1328.
seeds preparation for the various diseases cited above have been Ceng, Z.L., Zhang, M., 2000. The Clinical Practice of Chinese Meteria Medica.
studied. Although Cuscuta chinensis alone or combined with other Acadamic Press, Beijing, pp. 427–428.
traditional medicines seem to show high efficiency against these Chauhan, A., Sharma, P.K., Srivastava, P., Karmar, N., Dudhe, R., 2010. Plants having
potential antidiabetic activity: a review. Der Pharmacia Lettre 2, 369–387.
disease, the clinical studies are very limited. Therefore, further Cheng, P.P., Shi, J., Du, P., Liu, D.H., Cao, X., Wen, X., 2013. Fatty acid in the Cuscuta
clinical trials are necessary to provide more information before it chinensis lam by capillary gas chromatography. Academic Periodical of Farm
can be used in medical practice worldwide. Products Processing 8, 116–118.
Chung, W.T., Koo, B.S., Choi, E.G., Kim, M.G., Lee, I.S., Kim, C.H., 2006. Neuroprotec-
tive effect of a Chuk-Me-Sun-Dan on neurons from ischemic damage and
neuronal cell toxicity. Neurochemical Research 31, 1–9.
7. Conclusion Committee for the Pharmacopoeia of PR China, 1995. Pharmacopeia of PR China,
Part I. China. Medical Science and Technology Press, P.R. China p. 269.
Cornwell, T., Cohick, W., Raskin, I., 2004. Dietary phytoestrogens and health.
Cuscuta chinensis is commonly used in traditional medicines as Phytochemistry 65, 995–1016.
a tonic and aphrodisiac in China and some Asian countries. It is Costea, M., Spence, I., Stefanovic, S., 2011. Systematics of Cuscuta chinensis species
complex (subgenus Grammica, Convolvulaceae): evidence for long-distance
reported to contain flavonoids, phenolic acids, steroids, hydroqui- dispersal and one new species. Organisms Diversity and Evolution 11, 373–386.
nones, volatile oils, lignans, alkaloids, polysaccharides, resin glyco- Cruz-Garcia, G.S., Price, L.L., 2011. Ethnobotanical investigation of ‘wild’ food plants
sides and fatty acids. About 3.0% of the total chemical constituents used by rice farmers in Kalasin, Northeast Thailand. Journal of Ethnobiology
and Ethnomedicine 7 (33), 1–20.
are flavonoids which have been linked to the pharmacological Du, X.M., Kohinata, K., Kawasaki, T., Guo, Y.T., Miyahara, K., 1998. Components of the
activities of its extracts, especially kaempferol that shows antiox- ether-insoluble resin glycoside-like fraction from Cuscuta chinensis. Phyto-
idant, estrogenic actions and impressive effect on immune chemistry 48, 843–850.
Editorial Committee of Flora of China, 2006. (Chinese Academy of Sciences). Flora of
response. The in vitro studies and in vivo models have provided China. Science Press, Beijing.
evidence for various ethnomedicinal uses and pharmacological Editorial Committee of Practical Chinese Herbal Medicine Picture Dictionary
activities which strongly indicate that Cuscuta chinensis is useful in (PCHMPD), 2010. Practical Chinese Herbal Medicine Picture Dictionary. Shang-
hai Scientific and Technical Publishers, Shanghai p. 452.
different diseases. Cuscuta chinensis extracts and its chemical
Gao, J.M., Li, R., Zhang, L., Jia, L.L., Ying, X.X., Duo, D.Q., Li, J.C., Li, H.B., 2013. Cuscuta
components show antioxidant effects, free radical scavenging chinensis seeds water extraction protecting murine osteoblastic MC3T3-E1 cells
activities, and can regulate immune response, protect hepatic, against tertiary butyl hydroperoxide induced injury. Journal of Ethnopharma-
cardiovascular, renal, neurological or skin diseases, delay aging cology 148, 587–595.
Gaudineau, C., Beckerman, R., Welbourn, S., Auclair, K., 2004. Inhibition of human
and improve physical properties. Most of the studies were per- P450 enzymes by multiple constituents of the Ginkgo biloba extract. Biochem-
formed only based on the extract. Therefore, the chemical and ical and Biophysical Research Communications 318, 1072–1078.
pharmacological studies should be conducted using the key Hajimehdipoor, H., Kondori, B.M., Amin, G.R., Adib, N., Rastegar, H., Shekarchi, M.,
2012. Development of a validated HPLC method for the simultaneous determi-
natural products reported from the species and combinations nation of flavonoids in Cuscuta chinensis Lam. by ultra-violet detection. DARU
thereof in order to clarify the pharmacological mechanism of Journal of Pharmaceutical Sciences 20, 1–6.
action and the metabolites responsible for the activities of Cuscuta Han, S.X., Yao, S.L., Li, Y.Y., Wang, T.S., 2011. Protective effects of Cuscuta chinensis
Lam. extract on myocardial ischemia/reperfusion injury in rats. Chinese
chinensis. Pharmacological Bulletin 27, 533–536.
Research work on Cuscuta chinensis has recently become He, X.H., Yang, W.Z., Meng, A.H., He, W.N., Guo, D.A., Ye, M., 2010. Two new lignan
popular, mainly focusing on its preparation, chemical constituents glycosides from the seeds of Cuscuta chinensis. Journal of Asian Natural
Products Research 12, 934–939.
analysis and pharmacological activities, but the information about Hou, D.Y., Li, T.C., Yu, B., 2003. Comparative study of volatile oil on Cuscuta 2 species.
the pharmacokinetics, toxicity or side effects of Cuscuta chinensis Journal of Chinese Mass Spectrometry Society 24, 343–345.
are insufficient, and no major side effects have yet been discov- Huang, A.G., Yi, Y.L., Ling, F., Lu, L., Zhang, Q.Z, Wang, G.X., 2013. Screening of plant
extracts for anthelmintic activity against Dactylogyrus intermedius (Monogenea)
ered. Further studies on the pharmacokinetics and toxicology are
in goldfish (Carassius auratus). Parasitology Research 112, 4065–4072.
needed in order to evaluate the uses of this plant, its extract and Jiangsu New Medical College, 1997. Pharmacopoeia of Chinese medicine. Shanghai
isolated compounds in clinical practice and validate its safety in People Hygiene Publisher, Shanghai.
humans. Moreover, additional research data and published clinical Kwon, Y.S., Chang, C.S., Kim, C.M., 2000. Antioxidative constituents from the seeds
of Cuscuta chinensis. Natural Product Science 6, 135–138.
trials would be necessary before it can be used in medical Lei, D.Q., 2000. Chinese Medicine. Shanghai Scientific and Technical Publishers,
practices worldwide. Shanghai.
S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308 307

Liao, G.I., Chen, M.Y., Kuoh, C.S., 2000. Cuscuta L. (Convolvulacea) in Taiwan. Patil, J.U, Birada, S.D., 2011. Folkoric medicinal plants of Hingoli district, Maharas-
Taiwania 45 (3), 226–234. thra. Indian Journal of Natural Products and Resources 2, 77–101.
Liao, J.C., Chang, W.T., Lee, M.S., Chiu, Y.J., Chao, W.J., Lin, Y.C., Lin, M.K., Peng, Y.H., Patil, S.J., Patil, H.M., 2012. Ethnomedicinal herbal recipes from Satpura Hill Ranges
2014. Antinociceptive and anti-inflammatory activities of Cuscuta chinensis of Shirpur Tahsil, Dhule, Maharashtra, India. Research Journal of Recent
seeds in mice. American Journal of Chinese Medicine 42 (1), 223–242. Sciences 1, 333–336.
Li, C.H., Deng, H.B., Li, D.D., Li, Z.H., 2013. Advances and challenges in screening Peng, L.X., 2010. Take the cuscuta as the example, the discussion traditional Chinese
traditional chinese anti-aging materia medica. Chinese Journal of Integrated medicine pharmacokinetic of related question research (Ph.D. thesis). Hubei
Medicine 19 (4), 243–252. University of Traditional Chinese Medicine, Hubei, China p. 97.
Li, D.Z., Peng, D.Y., Zhang, R., Xu, X.X., 2008a. Effects of Cuscuta chinensis Peng, S.J., Lu, R.K., Yu, L.H., 1997. Effect of Semen Cuacutae, Rhizoma Curculiginis,
polysaccharide on diabetic mice by alloxan. Anhui Medical and Pharmaceutical Radix Morindae, Officinalis on human spermatozoa's motility and membrane
Journal 12, 900–901. function in vitro. Chinese Journal of integrated traditional and Western
Li, S.J., 2009. Compendium of Materia Medica (Illustrated edition). Volume Publish- Medicine 17, 145–147.
ing Company, Beijing, pp. 263–265. Petel, J.N., Patel, N.K., 2010. Study of parasite hosts of the genus Cuscuta and its
Li, W.L., Cheng, H.C., Bukuru, J., Kimbe, N.D., 2004. Natural medicines used in the traditional uses in Palanpur Taluka, Gujarat, India. Ethnobotanical Leaflets 14,
traditional Chinese medical system for therapy of diabetes mellitus. Journal of 126–135.
Ethnopharmacology 92, 1–21. Qureshi, R., Bhatti, G.R., Memon, R.A., 2010. Ethnomedical uses of herbs from
Li, X.J., You, H.Y., Yang, J., Liu, B.M., You, G., Song, Y., 2008b. Aqueous extracts of northern part of Nara desert, Pakistan. Pakistan Journal of Botany 42, 839–851.
Cuscuta Chinensis Lam induces differentiat ion of amelanotic melanocytes of
Sam, H.V., Bass, P., Kebler, P., 2008. Traditional medical plants in Ben En national
human hair follicles. Chinese Journal of Dermatovenereology 22 (4–5), 13.
park, Vietnam. BLUMEA 53, 569–601.
Li, Z.L., 2013. Ben Cao Yuan Shi. People's Medical Publishing House, Beijing, pp.
Shah, G.R., Chaudhari, M.V., Patankar, S.B., Pensalwar, S.V, Sabale, V.P., Sonawane,
51–52.
N.A., 2012. Evaluation of a multi-herb supplement for erectile dysfunction: a
Lin, H.B., Lin, J.Q., Lin, J.Q., Lu, N., Yi, X.Y., 2003. Comparative study on immune
randomized double-blind, placebo-controlled study. BMC Complementary and
enhancement effects of four kinds of dodder seeds in Shandong Province.
Alternative Medicine 12, 155.
Journal of Chinese Integrative Medicine 1, 51–53.
Shekarchi, M., Kondori, B.M., Hajimehdipoor, H., Abdi, L., Mohsen, L., Pourfarzib, M.,
Lin, H.B., Lin, J.Q., Lu, N., Lin, J.Q., 2007. Study of quality control on Cuscuta chinensis
Amin, G., 2014. Finger printing and quantitative analysis of Cuscuta chinensis
and C. australia. Journal of Chinese Medicinal Materials 30, 1446–1449.
Lin, M.K., Yu, Y.L., Chen, K.C., Chang, K.C., Chang, W.T., Lee, M.S., Yang, M.J., Cheng, Flavonoid Content from different host by RP-HPLC. Food and Nutrition Sciences
H.C., Liu, H.C., Chen, Dz.C., Chu, C.L., 2011. Kaempferol from Semen cuscutae 5, 914–921.
attenuates the immune function of dendritic cells. Immunobiology 216, Shen, L., Huang, Y.Y., Wang, X.N., Du, J., Wang, Y.Y., Zhang, D.Q., 2012. Pharmaco-
1103–1109. logical effect of Cuscutae Semen by External useon experimental vitiligo in
Liu, J.H., Jiang, B., Bao, Y.M., An, L.J., 2003. Effect of Cuscuta chinensis glycoside on guinea pigs. Chinese Journal of Experimental Traditional Medical Formulae 18,
neuronal differentiation of rat pheochromocytoma PC12 cells. International 199–202.
Journal of Developmental Neuroscience 21, 277–281. Shin, S., Lee, Y.J., Kim, J.E., Lee, A.S., Kang, D.G., Lee, H.S., 2011. Effect of Cuscuta
Liu, Z.R., Luo, P.T., Fu, T.J, Ji, Y.Q., Wang, R.Z., 2004. The effect of 3 extraction chinensis on renal function in ischemia/reperfusion-induced acute renal failure
technique of Chinese dodder seed on cardiovascular activity. National Product rats. The American Journal of Chinese Medicine 39, 889–902.
Research and Development 16, 532–534. Shubhangi, P., Patil, D.A., 2012. Herbal haircare as revealed by people in Jalgaon
Liu, Z.Y., Yang, Y.G., Zheng, B., 1993. Effect of improving memory and inhibiting district, Maharashtra, India. Journal of Experimental Sciences 3, 32–34.
acetylcholinesterase activity by invigorating-qi and warming-yang recipe. Smittinan, T., 2011. Thai Plant Name. Botanical forest, Bureau of Forestry, Forest
Chinese Journal of Integrated Traditional and Western Medicine 11 (675–676), Department, Bangkok.
64613 11 (675–676), 646. Sohn, D.W., Kim, H.Y., Kim, S.D., Lee, E.J., Kim, H.S., Kim, J.K., Hwang, S.Y., Cho, Y.H.,
Loffler, C., Czygan, F.C., Proksch, P., 1997. Phenolic constituents as taxonomic Kim, S.W., 2008. Elevation of intracavernous pressure and NO-cGMP activity by
markers in the genus Cuscuta (Cuscutaceae). Biochemical Systematics and a new herbal formula in penile tissues of spontaneous hypertensive male rats.
Ecology 25, 297–303. Journal of Ethnopharmacology 120, 176–180.
Ma, H.X., You, Z.L., Wang, R.G., 2008a. Effect of total flavones from Cuscuta chinensis Song, L.R., 2010. The utility of Chinese herbal Medicine. Shanghai World Publishing
on expression of Th type-1/Th type-2 cytokines, serum P and PR in abortion Co., Ltd., Shanghai p. 452.
rats model. Journal of Chinese Medicinal Materials 31, 1201–1204. Sun, K., Zhao, C., Chen, X.F., Kim, H.K., Choi, B.R., Huang, Y.R., Park, J.K., 2013. Ex vivo
Ma, H.X., You, Z.L., Wang, X.Y., 2008b. Effect of total flavones from Cuscuta chinensis relaxation effect of Cuscuta chinensis extract on rabbit corpus cavernosum.
on expression of Fas/FasL, PCNA and HB-EGF in SD rats model with Asian Journal Andrology 15, 134–137.
bromocriptine-induced abortion. Journal of Chinese Medicinal Materials 31, Sun, W., Fu, D.N., 2000. Clinical observation on Cuscuta chinensis ointment in
1706–1709. treatment of 49 herpes zoster cases. Mordern Journal of Integrated Traditional
Mavlonov, G.T., Ubaidullaeva, Kh.A., Kadryaeva, G.V., Kuznetsova, N.N., 2008. and Western Medicine 9 (14), 1365.
Cytotoxic components of Cuscuta. Chemistry of Natural Compounds 44 (3), Teng, J.L., 2007. Chinese Materia Medica. People Medical Publishing House, Beijing,
409–410. pp. 547–548.
Miyahara, K., Du, X.M., Watanabe, M., Sugimura, C., Yahara, H., Nohara, T., 1996. The Plant List., 2013. Version 1.1. Published on the Internet. 〈http://www.theplant
Resin glycosides. XXIII. Two novel acylated trisaccharides related to resin list.org〉 (accessed 06.09.14.).
glycoside from the seeds of Cuscuta chinensis. Chemical and Pharmaceutical Umehara, K., Nemoto, K., Ohkubo, T., Miyase, T., Degawa, M., Noquchi, H., 2004.
Bulletin 44, 481–485. Isolation of a new 15-membered macrocyclic glycolipid lactone, Cuscutic
Mokhtarifar, N., Sharif, B., Naderi, N., Mosaddegh, M., Faizi, M., 2012. Evaluation of Resinoside a from the seeds of Cuscuta chinensis: a stimulator of breast cancer
anti-depressant effects of Cuscuta chinensis in experimental models. Research cell proliferation. Planta Medica 70, 299–304.
in Pharmaceutical Sciences 7, S826.
Wang, J., Wang, M., Ou, Y., Wu, Q., 2002. Effects of flavonoids from semen Cuscutae
Mou, X.Y., 2002. Shen Nong Ben Cao Jing Shu. Ancient Chinese Medical Publishing
on changes of beta-EP in hypothalamuses and FSH and LH in anterior
House, Beijing, pp. 216–217.
pituitaries in female rats exposed to psychologic stress. Journal of Chinese
Nakahara, K., Trakoontivakorn, G., Alzoreky, N.S., Onishi-Kameyama, M., Yoshida, M.,
Medicinal Materials 25, 886–888.
2002. Antimutagenicity of some edible Thai plants, and a bioactive carbazole
Wang, J.G., 2001. Cuscuta chinensis seeds in treatment of 19 cases male infertility.
alkaloid, mahanine, isolated from Micromelum minutum. Journal of Agricultural
Hebei Journal of Traditional Chinese Medicine 23 (1), 53.
and Food Chemistry 50, 4796–4802.
Wang, Q., 2000. Application of Cuscuta chinensis decoction on male reproductive
Ni, Z.M., 2005. Ben Cao Hui Yan. Shanghai Science and Technology Press, Shanghai,
system dieases. Jiangsu Journal of Traditional Chinese Medicine 21 (12), 43.
pp. 381–384.
Wang, T.J., An, J., Chen, X.H., Deng, Q.D., Yang, L., 2014. Assessment of Cuscuta
Niu, Z.M., Xia, C.Z., 1994. Cuscuta chinensis ointment in treatment of herpes zoster.
Liaoning Journal of Traditional Chinese Medicine 21 (1), 44. chinensis seeds' effect on melanogenesis: comparison of water and ethanol
Nisa, M., Akbar, S., Tariq, M., Hassain, Z., 1986. Effect of Cuscuta chinensis water fractions in vitro and in vivo. Journal of Ethnopharmacology 154, 240–248.
extract on 7,12-dimethylbenz(α)anthracene-induced skin papillomas and car- Wang, Z., Fang, J.N., Ge, D.L., Li, X.Y., 2000. Chemical characterization and
cinomas in mice. Journal of Ethnopharmacology 18, 21–31. immunological activities of an acidic polysaccharide isolated from the seeds
Pan, H.J., Sun, H.X., Pan, Y.J., 2005. Adjuvant effect of ethanol extract of Semen of Cuscuta chinensis Lam. Acta Pharmacologica Sinica 21, 1136–1140.
Cuscutae on the immune response to ovalbumin in mice. Journal of Ethno- Wang, Z., Liu, C., Fang, J., 2001. H3, an acid polysaccharide from Cuscuta chinensis.
pharmacology 99, 99–103. Journal of Chinese Pharmaceutical Sciences 10, 57–59.
Pan, S., Wang, X., Duan, W., Yu, Z., Zhang, L., Liu, W., 2014. Preparative isolation and Williamson, G., Barron, D., Shimoi, K., Terao, J., 2005. In vitro biological properties of
purification of Flavonoids from Cuscuta Chinensis Lam. by high-speed counter- flavonoid conjugates found in vivo. Free Radical Research 39, 457–469.
current chromatography. Journal of Liquid Chromatography and Related Tech- Xia, H.F., 2012. Study on embryonic developmental toxicity of water extract of
nologies 37 (15), 2162–2171. Cuscutae (Master thesis). Zunyi Medical Collage, Zunyi, China p. 62.
Pan, W.H., Xu, Z,C., Zhao, Y.Q., 2008. Research advancement of the pharmacologic Xia, M.M., 2000. Cuscuta chinensis seeds in treatment of 13 cases glomerulone-
action and clinical application of Cuscuta seed. Asia-Pacific Traditional Medi- phritis. Zhejiang Journal of Integrated Traditional and Western Medicine 10 (7),
cine 4 (4), 47–51. 439.
Patil, D.A., Patil, P.S., Ahirrao, Y.A., Asher, U.P., Dushing, Y.A., 2010. Ethnobotany of Xu, X.X., Li, D.Z., Peng, D.Y., Zhang, R., 2011. Effects of Cuscuta chinensis poly-
Buldhana district (Maharasthra: India): plants used veterinary medicine. saccharide on glucose-lipid metabolism in diabetic rats. Chinese Journal of
Journal of Phytology 2, 22–34. Experimental Traditional Medical Formulae 17, 232–234.
308 S. Donnapee et al. / Journal of Ethnopharmacology 157 (2014) 292–308

Yahara, S., Domoto, H., Sugimura, C., Nohara, T., Niiho, Y., Nagajima, Y., Ito, H., 1994. Yen, F.L., Wu, T.H., Lin, L.T., Lin, C.C., 2007. Hepatoprotective and antioxidant effects
An alkaloid and two lignans from Cuscuta chinensis. Phytochemistry 37, of Cuscuta chinensis against acetaminophen-induced hepatotoxicity in rats.
1755–1757. Journal of Ethnopharmacology 111, 123–128.
Yang, F.Y., Huang, J., 1998. “Tai Ping Sheng Hui Fang” in the anti-aging effects Yen, F.L., Wu, T.H., Lin, L.T., Lin, C.C., 2008b. Nanoparticles formulation of Cuscuta
medical research. Journal of Guiyang College of Traditional Chinese Medicine 2, chinensis prevent acetaminophen-induced hepatotoxicity in rats. Food and
7–8. Chemical Toxicology 46, 1771–1777.
Yang, H.M., Shin, H.K., Kang, Y.H., Kim, J.K., 2009a. Cuscuta chinensis extract Yin, L.M., Gao, S.Z., Zhou, J.M., Lee, Z.G., Guo, S.Z., Zhang, S.J., 1992. Integrating
promotes osteoblast differentiation and mineralization in human osteoblast- Chinese and western medicine to treat Infantile uterus. Herbal Journal of
like MG-63 cells. Journal of Medicinal Food 12, 85–92. Traditional Chinese Medicine 14, 40.
Yang, J.X., Wang, Y.L., Bao, Y., Guo, J., 2008. The total flavones from Semen cuscutae You, H.L., 2005. “Jisheng” Cuscuta chinensis tablet for treating nocturia in 60 cases.
reverse the reduction of testosterone level and the expression of androgen Lishizhen Medicine and Metria Medica Research 16 (12), 1315.
receptor gene in kidney-yang deficient mice. Journal of Ethnopharmacology Yu, G.T., 1996. Cuscuta chinensis solution in treatment of 50 cases acnes. Zhejiang
119, 166–171. Journal of Traditional Chinese Medicine 4, 179.
Yang, L., Chen, Q., Wang, F., Zhang, G., 2011. Antiosteoporotic compounds from seed Zeraati, F., Zamani, A., Goodarzi, M.T., Hashjin Malakouti, S.M., Razzaghi, K., 2010.
In Vitro Cytotoxic Effects of Cuscuta chinensis Whole Extract on Human Acute
of Cuscuta chinensis. Journal of Ethnopharmacology 135, 553–560.
Lymphoblastic Leukemia Cell Line. Iranian Journal of Medical Sciences 35,
Yang, M., Sun, J., Lu, Z., Chen, G., Guan, S., Liu, X., Jiang, B., Ye, M., Guo, D.A., 2009b.
310–314.
Phytochemical analysis of traditional Chinese medicine using liquid chromato-
Zhang, L., 2011. Ben Jing Feng Yuan. Chinese Medicine and Technology Publishing
graphy coupled with mass spectrometry. Journal of Chromatography A 1216,
House, Beijing, pp. 106–107.
2045–2062.
Zhang, S.X., 2013. Ben Cao Zheng Yi. Shanxi Science and Technology Press, Shanxi,
Yao, C.H., Tsai, H.M., Chen, Y.S., Liu, B.S., 2005. Fabrication and evaluation of a new
pp. 242–244.
composite composed of tricalcium phosphate, gelatin, and Chinese medicine as
Zhao, C.G., Si, S.L., Gao, W., Pang, J.P., 1990. Spectrometric analyses of microelements
a bone substitute. Journal of Biomedical Material Research part B Apply contained in 6 Chinese herbs for miscarriage prevention. China Journal of
Biomaterial 75, 277–288. Chinese Materia Medica 15, 43–44.
Ye, M., Li, Y., Yan, Y., Liu, H., Ji, X., 2002. Determination of flavonoids in Semen Zhen, G.H., Jiang, B., Bao, Y.M., Li, D,X., An, L.J., 2006. The protect effect of flavonoids
Cuscutae by RP-HPLC. Journal of Pharmaceutical and Biomedical Analysis 28, from Cuscuta chinensis in PC12 cells from damage induced by H2O2. Journal of
621–628. Chinese Medicinal Materials 29, 1051–1055.
Ye, M., Yan, Y., Guo, D., 2005. Characterization of phenolic compounds in Chinese Zheng, C., Mi, S.Q., Luo, S.P., Luo, Y.H., 2004. The determination of quercetin content
herbal drug Tu-Si-Zi by liquid chromatography coupled to electrospray ioniza- by semen cuscutacae intragastric administration in rat and its pharmacokinetic
tion mass spectrometry. Rapid Communication in Mass Spectrometry 19, study. Chinese Archive of Traditional Chinese Medicine 22 (6), 1148–1150.
1469–1484. Zheng, H.Z., Dong, Z.H., She, J., 1998. Tusizi, Modern Study of Traditional Chinese
Ye, M., Yan, Y., Ni, X., Qiao, L., 2001. Studies on the chemical constituents of the Medicine, 1st ed. Beijing Xue Yuan Press of the People's Republic of China,
herba of Cuscuta chinensis. Journal of Chinese Medicinal Materials 24, 339–341. Beijing, pp. 4110–4120.
Yen, F.L., Wu, T.H., Lin, L.T., Cham, T.M., Lin, C.C., 2008a. Concordance between Zhu, J.F., She, Y.C., Zhou, C.H., 1987. Experimental and clinical studies on the effect
antioxidant activities and flavonol contents in different extracts and fractions of of Shou Tai Wan and additives on threatened abortion. Journal of Integrated
Cuscuta chinensis. Food Chemistry 108, 455–462. Traditional and Western Medicine 7, 407–409.

View publication stats

Potrebbero piacerti anche