Academia.eduAcademia.edu
Journal of Ethnopharmacology 155 (2014) 1011–1028 Contents lists available at ScienceDirect Journal of Ethnopharmacology journal homepage: www.elsevier.com/locate/jep Review From arrow poison to herbal medicine – The ethnobotanical, phytochemical and pharmacological significance of Cissampelos (Menispermaceae) Deepak Kumar Semwal, Ruchi Badoni Semwal, Ilze Vermaak, Alvaro Viljoen n Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria 0001, South Africa art ic l e i nf o a b s t r a c t Article history: Received 31 March 2014 Received in revised form 22 May 2014 Accepted 24 June 2014 Available online 2 July 2014 Ethnopharmacological relevance: Cissampelos species have a rich history of traditional use, being used for both therapeutic and toxic properties. It is traditionally applied therapeutically in a diverse range of conditions and diseases including asthma, cough, fever, arthritis, obesity, dysentery, snakebite, jaundice and heart, blood pressure and skin-related problems. Conversely, it was traditionally included in preparations of curare applied as arrow poison during hunting to cause death of animals by asphyxiation. This review unites the ethnobotanical knowledge on Cissampelos with the phytochemistry and pharmacological activity which has been explored thus far. In addition, it identifies knowledge gaps and suggests further research opportunities. Methods: The available electronic literature on the genus Cissampelos was collected using database searches including Scopus, Google Scholar, Pubmed, Web of Science, etc. The searches were limited to peer-reviewed English journals with the exception of books and a few articles in foreign languages which were included. Results: The literature revealed that pharmacological activity including analgesic and antipyretic, antiinflammatory, anti-allergic, bronchodilator, immunomodulatory, memory-enhancing, antidepressant, neuroprotective, antimicrobial, antimalarial, antiparasitic, anti-ulcer, anticancer, anti-oxidant, cardiovascular, muscle-relaxant, hepatoprotective, antidiabetic, antidiarrhoeal, antifertility, and antivenom activity have been confirmed in vitro and/or in vivo for various Cissampelos species. Cissampelos pareira L. and Cissampelos sympodialis Eichl. are the most explored species of this genus and the smallest number of studies have been conducted on Cissampelos laxiflora Moldenke and Cissampelos tenuipes Engl. Many alkaloids isolated from Cissampelos such as warifteine, methylwarifteine, berberine, hayatin and hayatidin showed promising anti-allergic, immunosuppressive, antidepressant, anticancer, vasodilatory and muscle-relaxant activities. Conclusion: The plants of this genus are used in traditional medicine for the treatment of various ailments. These plants are a rich source of bioactive bisbenzylisoquinoline and aporphine alkaloids together with other minor constituents. Although these plants are reputable and revered in various traditional medicine systems, many have not yet been screened chemically or pharmacologically and so there is a vast amount of research still to be conducted to validate their traditional use. & 2014 Elsevier Ireland Ltd. All rights reserved. Keywords: Cissampelos pareira Cissampelos sympodialis Antidepressant Antidiabetic Curare Bisbenzylisoquinoline alkaloids Contents 1. 2. 3. 4. Introduction . . . . . . . . . . . Geographical distribution Botanical aspects . . . . . . . Ethnobotanical aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012 1012 1014 1014 Abbreviations: Ig, immunoglobulin; OVA, ovalbumin; PG, prostaglandin; PHF, polyherbal formulation; TGI, total growth inhibition; MIC, minimum inhibitory concentration; IZD, inhibition zone diameter; MLE, methanolic leaf extract; HLE, hydroalcoholic leaf extract; HRE, hydroalcoholic root extract; ELE, ethanolic leaf extract; ERE, ethanolic root extract; MRE, methanolic root extract; p.o., per oral; i.p., intra-peritoneal; i.v., intravenous n Corresponding author. Tel.: þ 27 12 382 6373; fax: þ27 12 382 6243. E-mail address: viljoenam@tut.ac.za (A. Viljoen). http://dx.doi.org/10.1016/j.jep.2014.06.054 0378-8741/& 2014 Elsevier Ireland Ltd. All rights reserved. 1012 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 5. Phytochemical studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1. Alkaloid constituents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2. Non-alkaloid constituents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6. Pharmacological activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.1. Analgesic and antipyretic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.2. Anti-inflammatory activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.3. Anti-allergic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.4. Bronchodilator activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.5. Immunomodulatory activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.6. Memory-enhancing activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.7. Antidepressant activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.8. Neuroprotective activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.9. Antimicrobial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.10. Antimalarial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.11. Antiparasitic activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.12. Anti-ulcer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.13. Anticancer activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.14. Anti-oxidant activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.15. Cardiovascular activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.16. Muscle-relaxant activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.17. Hepatoprotective activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.18. Antidiabetic activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.19. Antidiarrhoeal activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.20. Antifertility activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.21. Antivenom activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6.22. Miscellaneous activities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7. Toxicity studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8. Future perspectives and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1. Introduction The genus Cissampelos (Menispermaceae) has diverse traditional uses, being applied for its therapeutic as well as toxic effects. In the rainforests of South America, Cissampelos pareira (called Abuta), commonly known as the midwives' herb, has a rich history of use to treat all types of women's ailments including menstrual cramps, menorrhagia, uterine haemorrhage etc. This is due to its profound relaxant effect on smooth muscle (Singh et al., 2010; Arora et al., 2012). At the same time however, it was traditionally included in the preparation of curares, the well-known South American arrow poison used in hunting to cause death by asphyxiation. Again, this effect is due to its muscle-relaxant and neuromuscular blocking effect (Maurya et al., 2013). The alkaloid isolated from Cissampelos pareira, hayatin methiodide, showed an equal amount of curariform activity as compared to the well known d-tubocurarine (Bhattacharji et al., 1952; Taylor, 1996). The members of this genus mostly contain alkaloids including bisbenzylisoquinolines, berberines, morphines, and aporphines, etc. along with a moderate quantity of other constituents (Thornber, 1970; Rocha et al., 1984; Blasko and Cordell, 1988). Many alkaloids such as tropoloisoquinoline alkaloids isolated from the genus exhibited potent biological activities. Pareirubrines A and B (Morita et al., 1993a, 1993b) from Cissampelos pareira showed antileukemic activity, hayatin methiodide (Pradhan and De, 1953) and hayatinin methochloride (Basu, 1970) from Cissampelos pareira, and aporphine alkaloids (þ)-cissaglaberrimine and (þ )-trilobinine from Cissampelos glaberrima showed muscle-relaxant properties. Most of the plants including Cissampelos capensis, Cissampelos pareira and Cissampelos sympodialis have been recognised for their remarkable medicinal properties and these are being used in various indigenous medicine systems for antibacterial, anti-oxidant, antispasmodic, diuretic, hypotensive, muscle-relaxant, antiseptic, aphrodisiac, analgesic, anti-haemorrhagic and cardiotonic properties (Kaur et al., 2012). Based on traditional knowledge, some species of the genus were also studied clinically for malaria (Wilcox et al., 2005), 1015 1015 1018 1019 1019 1019 1019 1020 1020 1020 1020 1020 1020 1021 1021 1021 1021 1022 1022 1022 1023 1023 1023 1023 1023 1023 1023 1023 1024 1024 dengue (Bhatnagar et al., 2011), diabetes (Jannu et al., 2011), the treatment of ulcers (Nwafor and Akah, 2003) and many other diseases and conditions (Roy et al., 1952; Hemraj et al., 2012). The extremely diverse range of recorded traditional uses for Cissampelos species is in part due to its very wide geographical distribution. This review comprises of up-to-date information on the ethnobotany, phytochemistry and pharmacology of the genus Cissampelos. 2. Geographical distribution The genus Cissampelos has a wide global distribution spanning five continents as well as several islands (Fig. 1). Cissampelos pareira is the only species that has a pantropical distribution – a geographical distribution which includes the tropical areas of the three major continents; Africa, Asia and the Americas. It occurs in Asia (Indo-China, Southern China, Malaysia, Thailand, India and Pakistan), Africa (Sierra Leone east to Congo, Rwanda, Tanzania, south to northern Angola, Zambia), America (Brazil, Argentina, Peru, Mexico, Colombia and Florida), Australia, the West Indies, Comores, Mauritius, Seychelles and Madagascar. Cissampelos ovalifolia is found only in North and South America and Cissampelos sympodialis only in South America (Brazil). Cissampelos mucronata, Cissampelos owariensis and Cissampelos capensis is restricted to the African continent: Cissampelos mucronata is distributed throughout tropical Africa from Senegal east to Ethiopia and south to southern Africa; Cissampelos capensis has a small natural distribution in Namibia and the Cape Provinces (Eastern Cape, Western Cape and Northern Cape) of South Africa; Cissampelos owariensis is found from Sierra Leone east to Uganda and south to Angola, Zambia and Mozambique. Based on various resources including flora of America, Africa, India, Pakistan, China and Australia, the distribution of selected ethnomedicinally important species, D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 1013 Fig. 1. Geographical distribution of selected ethnomedicinally important Cissampelos species: (a) Cissampelos pareira; (b) Cissampelos ovalifolia and Cissampelos owariensis; (c) Cissampelos sympodialis, Cissampelos mucronata and Cissampelos capensis. 1014 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 i.e. Cissampelos pareira, Cissampelos mucronata, Cissampelos owariensis, Cissampelos capensis, Cissampelos sympodialis and Cissampelos ovalifolia is depicted in Fig. 1a–c (Chopra, 1958; Barbosa-Filho et al., 1997b; De Wet, 2006; ; Mosango, 2008; Muzila, 2008; Oyen, 2008a, 2008b). 3. Botanical aspects The genus name Cissampelos when directly translated means ivy–vine, derived from the Greek words for Ivy (Kissos) and vine (ampelos). The name refers to the ivy-like growth of this plant in green rambling branches and the vine or grape-like racemes of fruits (Sudhakaran, 2012). Rhodes (1975) has reported a comprehensive morphological description of 20 Cissampelos species with descriptive figures for all the plant parts. Cissampelos species are generally sub-herbaceous or suffrutescent twiners distributed throughout warmer parts of Asia, Africa, and America (Semwal et al., 2010). Their leaves are mostly cordate or reniform, often peltate and alternate. The flowers are in axillary racemes or clusters; staminate flowers with 4 free sepals and 4 fused petals, 2–5 anthers on a staminal column or disk; pistillate flowers with 1 fleshy sepal, 1 fleshy petal and a solitary carpel, with 3–5 lobed stigmas. The fruits are a subglobose drupe with a flattened and tuberculate stone. Wide rays, pitted tyloses and enlarged vessel pits near the perforation plates highlight the wood of these plants. Some of the plants have successive cambia; a single vascular cambium remained functional and showed normal secondary growth (Don, 1831; Rhodes, 1975; Tamaio et al., 2010). According to The Plant List (2013), Cissampelos is one of the major genera of Menispermaceae comprising 21 species. These plants are mostly climbers or lianes, having simple peltate or subpeltate and entire angular leaves. The flowers have 1–5 obovate sepals; petals and stamens are mostly connate. The fruits are hairy, or a glabrous drupe, with thin fleshy mesocarp. The leaves of Cissampelos sympodialis are peltate with deltoid blades and petioles swollen at extremities. The epidermis is hipostomatic with anticlinal walls of epidermal cells; mesophyll dorsiventral; vascular system formed by 6–7 free collateral bundles ring (Porto et al., 2008). In particular, Cissampelos pareira has a total of n ¼12 and 2n¼ 24 chromosomes, but in the male plant, these are unequal pairs, heterogametic with XY type of sex chromosomes (Mathew, 1958). Most of the plants have sieve-like rhizomes in their T.S. which are easily distinguishable in each species except Cissampelos hirta and Cissampelos mucronata. Last mentioned species are differenciated only by their leaf texture, colour and the presence of a geniculate pulvinus in Cissampelos hirta (De Wet et al., 2002). 4. Ethnobotanical aspects Ethnobotanical studies revealed that Cissampelos is one of the most widely and frequently used genera of the Menispermaceae family in Asian, American and African traditional medicine. These species are also used as curare for arrow poison in various parts of the world including South America (Quattrocchi, 1912). In Nigeria, the rhizomes of Cissampelos owariensis (Mosango, 2008) and Cissampelos mucronata (Muzila, 2008) are used in the preparation of arrow poison. Since ancient times, Cissampelos pareira has been used in Indian Ayurvedic medicine for preparing Pusyanug churn, Pathadi kwath, Mahayograj guggulu and Agnimukh churn (Rawat and Vashistha, 2011). Cissampelos pareira has been used for coughs, delirium, fever-cerrado habitants, madness, epilepsy, convulsions and also used as a stimulant, sedative, analgesic, febrifuge, anti-oxidant, tonic and narcotic in various parts of the globe (Mendes and Carlini, 2007; Giorgetti et al., 2011; Samanta and Bhattacharya, 2011). An infusion of the roots of Cissampelos pareira has been used to treat gastrointestinal disorders such as diarrhoea and dysentery, whereas topically, it is used for treating snake bites in Central America and Mexico (Morten, 1981; Leonti et al., 2001; Heinrich et al., 2014). In India, the plant is used for abortion, asthma, dysentery, fever, hydrocele, gonorrhoea, menstrual cycle regulation, bubo, tumour, piles and puerperal fever. The roots are specifically used as a diuretic, febrifuge, for heart trouble, dysentery, sores, snakebite and jaundice (Chopra, 1958; Kupchan et al., 1965; Siddiqui and Husain, 1994; Singh and Ali, 1994; Rana and Datt, 1997; Sharma et al., 2004; Basha and Sudarsanam, 2012; Sharma et al., 2012) as well as to prevent a threatened miscarriage and to stop uterine haemorrhage (Lewis, 1977). This plant, in combination with Piper nigrum L., Mimosa pudica L. and Hibiscus rosa-sinensis L., is used in different parts of India for birth control (Tiwari et al., 1982). The root decoction of Cissampelos pareira is used in malaria, pneumonia, and snake and dog bite (antidote) in India (Jain et al., 2005; Namsa et al., 2011). The root and leaves are used for helminthiasis (worm infestation) (Ramasubramaniaraja and Babu, 2010), against dyspepsia, diarrhoea, stomach ache, dropsy, cough, urinary difficulties like cystitis, dysentery, asthma, heart diseases and also as an anti-spasmodic; the leaves are used as an antiseptic against inflammation (Kakrani and Saluja, 2002; Rajan et al., 2002, 2003; Kufer et al., 2005; Kumar et al., 2006; Gupta et al., 2011; Nagarajan et al., 2011; Kaur et al., 2012). In Pakistan, the leaves of Cissampelos pareira are used to treat diarrhoea, and topically to treat abscesses and wounds (Abbasi et al., 2010; Haq et al., 2011). The roots and leaves of this plant are also used for snakebite, stomachache, diabetes and malaria in various countries including India, Mexico and Kenya (Shinwari and Khan, 1998; Galicia et al., 2002; Chhetri et al., 2005; Bora et al., 2007; Pattanaik et al., 2008; Rukunga et al., 2009). The tubers of Cissampelos pareira are used in pseudo-pregnancy in Malawi (Maliwichi-Nyirenda and Maliwichi, 2010). Cissampelos glaberrima and Cissampelos ovalifolia are used for coughs, delirium, fever-cerrado habitants, madness, stimulant, convulsions, epilepsy, sedative, analgesic, febrifuge, anti-oxidant, analgesic, as a tonic and narcotic (Giorgetti et al., 2011). The leaves of Cissampelos torulosa were used in diarrhoea, dysentery and sore throat complaints (Samie et al., 2005, 2009), whereas Cissampelos hirta Klotzsch. was used as an antidiarrhoeal in South Africa (De Wet et al., 2010). The leaves of Cissampelos tropaeolifolia DC. were used by the Q'eqchi to treat women's health complaints and to release the placenta (Michel et al., 2007). Cissampelos sympodialis is used in Brazil to treat several inflammatory disorders, bronchitis, asthma, rheumatism and gastrointestinal, urinary tract and skin infections (Machado et al., 2003; Moreira et al., 2003b; Correa et al., 2008; Costa et al., 2008; Porto et al., 2008; Feily and Namazi, 2009; Bezerra-Santos et al., 2012; Vieira et al., 2013). In South Africa, the root infusion of Cissampelos capensis is taken to treat heart and blood pressure problems (Olorunnisola et al., 2011) and the leaves are used to treat diabetes (Deutschlander et al., 2009; Afolayan and Sunmonu, 2010). The roots extract of Cissampelos capensis is used in obesity (Afolayan and Mbaebie, 2010), whilst the roots and rhizomes are used as emetic, purgative, tincture and also for dysentery, syphilis, snakebite, stomach and skin cancer (Van Wyk and Gericke, 2000; Van Wyk, 2008). In addition, it is used as a digestive and for endocrine, genitourinary and infection problems (De Wet and Van Wyk, 2008). The roots of Cissampelos mucronata were taken in Ethiopia for stomach ache, gastrointestinal complaints and to expel retained placenta (Giday et al., 2009; Tripathi et al., 2013), and the plant is used in liver diseases such as hepatitis (Mukazayire et al., 2011). The leaves and roots are used for gastrointestinal complaints, D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 menstrual problems, venereal diseases, malaria and wounds in Nigeria, Tanzania and Senegal (Tor-anyiin et al., 2003; Benoit-Vical, 2005; Nondo et al., 2011). The leaves and roots were used in Uganda for inducing labour and expelling the placenta (Mugisha and Origa, 2007), and as a tocolytic (uterine relaxant) agent (Nwafor et al., 2002). Cissampelos owariensis P. Beauvais ex D.C. is used in Nigeria for the management of various forms of female infertility problems (Elujoba, 1995). The decoction of Cissampelos owariensis (together with Heteranthera callifolia Rchb. Ex Kunth) is used against dementia-type loss of memory, whilst the plant is used for circulatory gynaecological problems, asthenia, diarrhoea, wounds and snakebite in various African countries. In association with Rauwolfia vomitoria Afzel., the plant is used for psychosis. In Benin, this species is used for cognitive disorders and in Tanzania, it is used for amnesia and psychosis (Hage et al., 2010). Apart from the medicinal uses, these plants are reported for various other properties such as Cissampelos pareira is used for augmenting milk production in dairy cows in many parts of India (Behera et al., 2013) and is also introduced for its ornamental value in various countries (Oyen, 2008a). The leaves of Cissampelos pareira are commonly used in food systems for various purposes including thickeners, gelling agents, texture modifiers and stabilisers in Asia (Vardhanabhuti and Ikeda, 2006). 5. Phytochemical studies Although, the genus Cissampelos comprises about 21 species, only a few have been phytochemically explored. A comprehensive literature survey revealed that alkaloids are the major constituents reported from the genus together with moderate levels of nonalkaloids. The chemical structures of the reported alkaloid and nonalkaloid constituents of the genus are depicted in Figs. 2 and 3, respectively. 5.1. Alkaloid constituents In 1840, Wiggers (1840) reported an amorphous bisbenzylisoquinoline alkaloid, pelosine (1) from the roots of a South American Cissampelos pareira species which was later found to be identical to that of l-curine (2) (Scholtz, 1896). During the 1950s, three bisbenzylisoquinoline alkaloids, hayatine or l-curine (2), hayatinine (3) and hayatidine (4), also known as (-)-4″-O-methylbebeerine or (-)-O-methylcurine, were reported from the Indian species (Bhattacharji et al., 1952; 1956) and their chemical structure and stereochemistry were described in the 1960s (Bhattacharji et al., 1962; Bhatnagar et al., 1967; Bhatnagar and Popli, 1967). Haynes et al. (1966) also isolated (þ þ)-4″-O-methylcurine (4) which was found stereochemically different to that previously reported. The roots and vines yielded d-isochondodendrine (5) and hayatine or l-curine (2) whereas a cytotoxic bisbenzylisoquinoline alkaloid, cissampareine (6) was also isolated from the plant. The stereochemistry of cissampareine (6) was confirmed by its methylation with diazomethane, which yielded O-methylcissampareine and by reduction with sodium borohydride, which afforded dihydrocissampareine (Kupchan et al., 1960, 1965, 1966). Boissier et al. (1965) reported two bisbenzylisoquinaline alkaloids, hayatine (2) also known as ( 7)-bebeerine or (7)-curine and ( þ)-isochondodendrine (5), whereas Anwer et al. (1968) isolated cyclanoline or cissamine (7) as chloride from the roots of Cissampelos pareira. Dwuma-Badu et al. (1975) reported isochondodendrine (5), dicentrine (8), dehydrodicentrine (9), cycleanine (10) and insularine (11) from the roots. Bhakuni et al. (1987) reported the biosynthetic pathway for bisbenzylisoquinaline alkaloids, (R,R)-bebeerine (2), 1015 hayatidine (4), (R,R)-isochondodendrine (5) and (R,R)-cycleanine (10), along with sepeerine (12) isolated from Cissampelos pareira. The study revealed that hayatidine (4) is biosynthesised stereospecifically by intermolecular oxidative coupling of (R)- and (S)-N-methylcoclaurine, whilst (R,R)-bebeerine (2), (R,R)-isochondodendrine (5) and (R,R)-cycleanine (10) are formed by oxidative dimerisation of (R)-N-methylcoclaurine. The study also confirmed the absolute configuration as ‘S' and ‘R' at the asymmetric centres C-1 and C-10 , respectively in hayatidine (4). Ahmad et al. (1992) reported five alkaloids, laudanosine (13), nuciferine (14), bulbocarpine (15), corytuberine (16) and magniflorine (17) (as hydrochloride) from the leaves and stems. Morita and coworkers (Morita et al., 1993a, 1993b) reported two tropoloisoquinoline alkaloids, pareirubrines A (18) and B (19) as antileukemic substances together with grandirubrine (20) and isoimerubrine (21) having similar skeletons from Cissampelos pareira. The conformation of tropolone ring in their structures was elucidated by NMR studies, whereas their solid-state tautomeric forms were examined by XRD analysis. In addition an azafluoranthene alkaloid, norimeluteine (22), as a cytotoxic substance together with norruffscine (23) was also reported from this source (Morita et al., 1993c) and a cytotoxic condensed tropone-isoquinoline alkaloid, pareitropone (24), was isolated from the roots (Morita et al., 1995). The plant roots contain berberine (25), reserpine (26) and cissampeline (1) which was found to be structurally similar to that of pelosine (Sharma et al., 2004; Stepp, 2004; Bafna and Mishra, 2010). Hullatti and Sharada (2010) isolated a principle marker compound l-bebeerine (2) in pure form to establish quality control parameters of Cissampelos pareira roots, since the concentration of bebeerine (2) has been suggested as a main criterion for the authentication of Cissampelos pareira (Singh et al., 2012). From the aerial parts of Cissampelos fasciculata Benth., a tropical American species and well-known repellant for leaf cutter ants, Acromyrmex octospinosus (Reich), a bisbenzylisoquinoline cissampentin (27) was isolated as an oil together with an aporphine alkaloid corydine (28). The authors could not explain the stereochemistry of cissampentin (27) at C-1 and C-10 , however, it was found to be a racemic mixture (Galinis et al., 1993). A tertiary aporphine alkaloid cissaglaberrimine (29), established as 1,2-methylenedioxy-3-hydroxyaporphine, was isolated from the stems and leaves of Cissampelos glaberrima A. St.-Hil. together with magnoflorine (30) and oxobuxifoline (31). The structure of magnoflorine (30) may have been confused with that of N,N-dimethyllindcarpine (30a) due to their closely related chemical structures (Barbosa-Filho et al., 1997a). A stephaoxocane (isoquinoline alkaloid bearing an oxocane ring) eletefine (32) was isolated from Cissampelos glaberrima roots. It appeared as a mixture of isomers with or without intramolecular H-bonding from O–H to the oxygen of the oxocane ether bridge. The hydroxy group at C-12 of eletefine (32) was oxidised with pyridinum dichromate to produce oxoeletefine (32a) which does not convert to a mixture. The study revealed that the property of always reverting to the equilibrium state mixture could be related to the hydroxy group at C-12 (Da-Cunha et al., 1998). In addition, the roots of the plant also afforded a quaternary aporphine alkaloid, (þ)-trilobinine (33) along with (þ )-cissaglaberrimine (29) (Cornelio et al., 1999). The rhizomes of Cissampelos ovalifolia D.C. yielded three tertiary bisbenzylisoquinoline bases possessing a p-xylyl moiety, named warifteine (34), methylwarifteine (35) and dimethylwarifteine (36), together with the corresponding dihydro compounds. Dimethylwarifteine (36) was found to be identical to a known alkaloid, O-methylcissampareine whereas methylwarifteine (35) was found to be isomeric with cissampareine (6) since both gave dimethylwarifteine (36) on permethylation (Gorinsky et al., 1972; Mukherjee and Keifer, 2003). Aguirre-Galvis (1995) phytochemically studied 1016 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Cissampelos ovalifolia and Cissampelos pareira species from India, Colombia and Guyana and reported various bisbenzylisoquinoline alkaloids including warifteine (34) and methylwarifteine (35). Moreover, a benzylisoquinoline alkaloid, (S)-6-methoxyjuziphine (37) was also reported from the South American antimalarial plant Cissampelos ovalifolia (Steele, 2000; Steele et al., 2002). A bisbenzylisoquinoline alkaloid, l-isochondodendrine (5) was reported from Cissampelos mucronata A. Rich. (Ferreira et al., 1965). Fig. 2. Chemical structures of alkaloid constituents isolated from the genus Cissampelos. D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Fig. 2. (continued) 1017 1018 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Fig. 3. Chemical structures of non-alkaloid constituents isolated from the genus Cissampelos. De Wet (2006) attempted a study based on the chemotaxonomical distribution of alkaloids from Cissampelos mucronata and found that the leaves contained dicentrine (8), salutaridine (38), reticuline (39) and pronuciferine (40), whereas the rhizomes contained dicentrine (8) and cycleanine (10) as major alkaloids. Two bisbenzylisoquinoline alkaloids, warifteine (34), methylwarifteine (35) and an aporphine alkaloid, laurifoline (41) were reported from the leaves and roots of Cissampelos sympodialis Eichl. (Cortes et al., 1995; Barbosa-Filho et al., 1997b; Aragao et al., 2001). The leaves of Cissampelos sympodialis yielded a 8,14dihydromorphinandienone alkaloid, milonine (42); its structure was established as (þ)-(9β,13β,14α)-5,6-didehydro-4-hydroxy-3, 6-dimethoxy-17-methylmorphinan-7-one which was found to be an isomer of (-)-8,14-dihydrosalutaridine by its absolute configuration (De Freitas et al., 1995). A bisbenzylisoquinoline alkaloid, roraimine (43) and an oxoaporphine alkaloid, liriodenine (44) were isolated from the roots of Cissampelos sympodialis. The NMR data of roraimine (43) were found to be analogous to that of warifteine (34) with the exception of chemical shifts of the carbonyl and adjacent carbons and protons (De Lira et al., 2002). Marinho et al. (2012) developed an analytical method for the simultaneous quantitation of the bioactive markers including warifteine (34), methylwarifteine (35) and milonine (42) from Cissampelos sympodialis leaves and applied the method to a phenological study of their relative concentrations. From the aerial parts of Cissampelos capensis, two anthelmintic aporphine alkaloids, (S)-dicentrine (8) and (S)-neolitsine (45) were isolated (Ayers et al., 2007). Furthermore, two aporphine alkaloids, bulbocapnine (15), dicentrine (8) and a morphinane alkaloid salutaridine (38) with four minor alkaloids, glaziovine (46), lauroscholtzine (47), crotsparine (48) and cycleanine (10) were isolated from the leaves of Cissampelos capensis, and from the stems, bulbocapnine (15), three bisbenzyltetrahydroisoquinoline alkaloids cycleanine (10), insularine (11), cissacapine (49), together with 12-Omethylcurine (4), dicentrine (8), reticuline (39) and insulanoline (50) were isolated. The rhizome of the plant furnished 12-O-methylcurine (4), cycleanine (10) and cissacapine (49) as the major alkaloids, whereas insularine (11), bulbocapnine (15), pronuciferine (40) and glaziovine (46) were obtained as minor alkaloids. The study suggested that the plant part, the developmental stage, the time of harvesting and the geographical distribution must be taken into account for chemotaxonomic purposes due to the chemical variations in different parts (De Wet et al., 2011). 5.2. Non-alkaloid constituents Apart from alkaloids as the major constituents of Cissampelos species, some non-alkaloidal constituents were reported. The roots of Cissampelos pareira contain d-quercitol (51), sterols, fixed oil and essential oil, which contains thymol (52) as a major constituent (Srivastava, 1956; Chowdury, 1972; Dwuma-Badu et al., 1975). The roots of Cissampelos glaberrima yielded four alkamides (small bioactive lipid signals), deca-2E,4E-dienoic acid isobutylamide (53), octa-2E,4E-dienoic acid isobutylamide (54), decen-2-oic acid isobutylamide (55) and decanoic acid isobutylamide (56), the last two being isolated as traces and identified using mass spectrometry (Rosario et al., 1996). Ramirez et al. (2003) reported a chalcone-flavone dimer, cissampeloflavone (57) elucidated as 2-(4-hydroxy-3-methoxyphenyl)-7-(4-methoxyphenyl)-6-(2-hydroxy-4,6-dimethoxybenzoyl) furano[3,2-g] benzopyran-4-one from the aerial parts of Cissampelos pareira. Singthong et al. (2005) extracted a pectin from Cissampelos pareira leaves which was a low methoxyl pectin consisting mainly of uronic (galacturonic) acid (58) ( 70–75%) and a small amount of neutral sugars. This pectin when studied for its rheological properties showed shear thinning flow behaviour. In addition, a well known flavonoid, 2-(3,4-dihydroxyphenyl)-3,5, D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 7-trihydroxy-4H-chromen-4-one (quercetin) (59) (Amresh et al., 2007a) and a saturated fatty acid, arachidic or eicosanoic acid (60) were also reported from Cissampelos pareira (Ramasubramaniaraja and Babu, 2010). The leaves of Cissampelos pareira have been reported to produce polysaccharides (hydrocolloids) (Vardhanabhuti and Ikeda, 2006), and pectins [(1-4)α-D-galacturonan] mainly composed of galacturonic acid with trace amounts of neutral sugars (Singthong et al., 2004). 6. Pharmacological activities 6.1. Analgesic and antipyretic activity The hydroalcoholic leaf extract (HLE) of Cissampelos sympodialis showed significant (po0.05) activity against acetic acid and formalin models of analgesia in mice. The extract reduced the number of abdominal contortions in the acetic acid test, whereas it inhibited the second phase in the formalin test at a dose of 200 mg/kg i.p. (Mendes de Oliveira et al., 2011). The hydroalcoholic root extract (HRE) from Cissampelos pareira showed resistance against mechanical pain in analgesymeter-induced pain in mice. The HRE lowered the writhing episodes in acetic acid-induced writhing (0.6%; i.p.) by protection of 22.73% and 51.63% at doses of 200 and 400 mg/kg, body weight, respectively. The HRE also exhibited protective effects against complete Freund's adjuvant-induced arthritis by 40.54% and 71.52% at similar doses (Amresh et al., 2007f; Arya et al., 2011). The extract from Cissampelos pareira and its polyherbal formulation (PHF) in combination with Pongamia pinnata (L.) Pierre and Vitex negundo var. negundo, showed remarkable analgesic effects against acetic acidinduced writhing in mice by 21.44 and 24.22 s at doses of 400 and 600 mg/kg, respectively, whereas aspirin, used as a positive control, exhibited an effect by 27.64 s at 300 mg/kg (Bansod et al., 2010, 2011). The aqueous extract of the PHF of Cissampelos pareira with Hemidesmus indicus (L.) R. Br. ex Schult., Rubia cordifolia L., Terminalia chebula (Gaertn.) Retz., Emblica officinalis Gaertn., Terminalia bellirica (Gaertn.) Roxb., Vitis vinifera L., Grewia asiatica L., Salvadora persica L., and Saccharum officinarum L., showed potent antipyretic and analgesic activity at a dose of 60 mg/kg/day and a lesser ulcer effect even at a very high dosage compared to that of aspirin (60 mg/kg/day) in human patients. The study was conducted on outpatients of the Institute of Post Graduate Ayurvedic Education and Research Hospital, Kolkata, India, and the study suggested that the PHF reduces body temperature and the level of PGE2 (Gupta et al., 2008a, 2008b). 6.2. Anti-inflammatory activity An aqueous fraction of the ethanolic leaf extract (ELE) of Cissampelos sympodialis Eichl., showed anti-inflammatory activity in mice and inhibited both 12-O-tetradecanoylphorbol 13-acetate and capsaicin-induced ear oedema by 58% and 37% respectively, at a dose of 100 mg/kg, i.p. The effective dose to inhibit carrageenan-induced rat paw oedema was 50 mg/kg (24%). The subcutaneous administration of 100 and 200 mg/kg in rats inhibited the carrageenan-induced neutrophil migration measurement after the administration of the irritant by 53% and 50% respectively (Batista-Lima et al., 2001). The extract from Cissampelos pareira and its PHF with Pongamia pinnata (L.) Pierre and Vitex negundo L., showed in vivo anti-inflammatory activity at doses of 400 and 600 mg/kg on carrageenan-induced hind paw oedema by 0.30 and 0.16 ml, respectively, whereas on formaldehyde-induced paw oedema, the activity was recorded at 0.15 and 0.07 ml, respectively when compared to aspirin (0.02 and 0.13 ml). The PHF (200, 400 and 600 mg/kg) showed anti-arthritic activity against Freund's complete adjuvant-induced arthritis in rats and reduced hind paw swelling and bodyweight along with a significant improvement in 1019 haematological parameters, whilst histopathology revealed a significant reduction in mononuclear infiltration, pannus formation and bone erosion (Bansod et al., 2010, 2011). The ethanolic extract of the aerial parts of Cissampelos pareira exhibited anti-inflammatory (paw oedema induced by carrageenan and arachidonic acid) and analgesic activity (abdominal writhes and hot plate) in rats and mice respectively, at a dose of 100 mg/kg, p.o. (Amresh et al., 2007b). The ethanolic root extract (ERE) of Cissampelos pareira showed antiinflammatory activity on acute, subacute and chronic rat models at doses of 200 and 400 mg/kg, p.o. The total protection produced for acute inflammation was 59.55% and 64.04% for carrageenan; 15.38% and 30.77% for histamine; 17.78% and 31.11% for 5-hydroxytryptamine, and 19.23% and 30.77% for PGE2-induced hind paw oedema, respectively. Similarly, in subacute inflammation, the protection was 38.36% and 47.95% for formaldehyde-induced hind paw oedema, whereas in chronic inflammation it was 15.02% and 19.19% in the cotton-pellet granuloma test (Amresh et al., 2007g). 6.3. Anti-allergic activity The extract from Cissampelos sympodialis (40 mg/kg, p.o.) and its alkaloid, warifteine (34) (50 μg/animal) showed anti-allergic activity on allergic eosinophilia in which two allergic inflammation models, asthma and allergic pleurisy, in actively sensitised Balb/c mice were used. The extract reduced pleural eosinophil influx triggered by allergen challenge and also affected the eosinophil activation by inhibiting new cytoplasmic lipid bodies formation and cysteinyl leukotriene secretion (Bezerra-Santos et al., 2006; Piuvezam et al., 2012). The extract and/or warifteine (34) from Cissampelos sympodialis inhibited allergen-induced airway hyperreactivity to inhale methacholine and IL-13 levels in the bronchoalveolar lavage on allergen-triggered lung remodelling in the murine model of asthma. Both the extract and warifteine decreased ovalbumin (OVA)-induced eosinophil tissue infiltration, mucous production and subepithelial fibrosis (Bezerra-Santos et al., 2012). The HLE (containing warifteine, methylwarifteine and milonine) from Cissampelos sympodialis showed promising activity in different animal models of asthma (Marinho et al., 2012), whereas warifteine and methylwarifteine produce a reversible, nonspecific and noncompetitive antagonism of histamine anti-allergic therapy (Gomes et al., 2012). Warifteine (34) isolated from Cissampelos sympodialis was studied to evaluate IgE production, leucocyte activation, thermal hyperalgesia, mast cell degranulation and scratching behaviour in a murine model of immediate allergic reaction. BALB/c mice treated with warifteine (0.4–10 mg/kg) one hour before OVA-sensitisation reduced OVA-induced paw oedema as well as the OVA-specific IgE serum titres. It also reduced death evoked by the IgE-dependent anaphylactic shock reaction at 30 min after intravenous OVA challenge. Thermal hyperalgesia evoked by IgE or an histamine/5-hydroxytryptamine challenge was inhibited in rats at a dose of 4.0 mg/kg. Warifteine (0.6 or 6.0 μg/mL) also decreased the IgEαDNP-BSA sensitised mast cell degranulation after DNP-BSA challenge measured by histamine release (Costa et al., 2008). Blomia tropicalis extract-induced allergy in mice was treated orally with an extract (400 mg/kg) containing a total alkaloid fraction of 8 mg/kg as well as 4 mg/kg of warifteine. All samples reduced the number of total cells and eosinophils in bronchoalveolar fluid (BAF) and eosinophil peroxidase (EPO) levels in the BAF. The samples also decreased the density of inflammatory cells in the lung (Cerqueira-Lima et al., 2010). ELE from Cissampelos sympodialis reduced food intake and bodyweight and also produced numerous alterations in the open-field test in female rats at doses of 45 and 225 mg/kg, p.o. ELE modified smooth muscle tone, leucocyte effects and cytokine secretion, which are the main parameters in asthma pathology (Almeida et al., 2005). The alcoholic leaf extract of Cissampelos sympodialis reduced eosinophil infiltration into the lung 1020 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 of OVA-sensitised mice. The inhaled extract inhibited eosinophil recruitment to the pleural cavity, bronchoalveolar lavage and peripheral blood. This treatment reduced the OVA-specific IgE serum titre and leucocyte infiltration in the peribronchiolar and pulmonary perivascular areas as well as mucous production. Flow cytometric analysis showed that the isolated alkaloid, methylwarifteine as well as the extract reduced the number of CD3 þT cells and eosinophil-like cells (Vieira et al., 2013). The aqueous fraction (10–300 μg/mL) of the ELE of Cissampelos sympodialis inhibited N-formyl-Met-Leu-Phe-induced release of lysozyme and myeloperoxidase from human neutrophils. Inhibition by the fraction, as well as by dibutyryl-cAMP and PGE2, was substantially greater when the cells were pre-treated with the phosphodiesterase inhibitor isobutyl methyl xanthine, indicating that the effect may be mediated by cAMP. Cyclic AMP dependent protein kinase-A activity was also increased by the fraction (1.5–100 μg/mL) (Thomas et al., 1999). The ERE from Cissampelos sympodialis reduced spontaneous tone and inhibited the contractions induced by submaximal concentrations of carbachol, histamine, PGF2α and substance P, in guinea-pig tracheal preparations with the IC50 value range being 13.9– 95.5 μg/mL. The extract improved the intracellular levels of cyclic AMP in guinea-pig bronchoalveolar leucocytes with IC50 range 1–100 μg/mL (Thomas et al., 1995). 6.4. Bronchodilator activity The HLE of Cissampelos sympodialis showed bronchodilator activity in a guinea pig model by inhibiting the spontaneous tone of the trachea with an IC50 value of 13.9 μg/mL. It was potentiated by 3-isobutyl-l-methylxanthine and blocked by timolol (β2-adrenoceptor blocking agent) with an IC50 4.6 μg/mL. However, no effect was noted on removal of the epithelium or addition of methylene blue. The extract also antagonised contractions induced by carbachol, capsaicin and arachidonic acid in normal trachea and by OVA in trachea obtained from sensitised guinea pigs with IC50 ranges of 34.1–70.5 μg/mL. In addition, the extract increased the preconvulsive time of animals exposed to an aerosol of histamine to 63.5 s at a dose of 100 mg/kg, i.p. (Thomas et al., 1997b). 6.5. Immunomodulatory activity The leaf extract of Cissampelos sympodialis exhibited an immunomodulatory effect on the murine model of OVA-induced allergy in BALB/c mice with doses ranging from 200 to 600 mg/kg, p.o. The extract at 400 or 600 mg/kg also reduced paw oedema induced by local OVA challenge. It increased the in vitro production of IFN-γ and IL-10 by Con-A stimulated cells (Bezerra-Santos et al., 2004). The hydroalcoholic extract of Cissampelos sympodialis leaves exhibited an immunomodulatory effect on B-lymphocyte function (Moreira et al., 2003b). The methanolic root extract (MRE) of Cissampelos pareira was studied for its immunomodulatory activity in mice and stimulatory activity on DTH response was found at 200–800 mg/kg dose. Higher doses of extract also offered protection against cyclophosphamide-induced myelosuppression by increasing total WBC count significantly (Bafna and Mishra, 2005). The berberine-containing alkaloidal fraction from Cissampelos pareira roots exhibited an immunosuppressive effect at doses of 25 and 50 mg/kg, p.o. and significantly lowered the humoral antibody titre. The fraction did not show any activity at higher doses, i.e. 75 and 100 mg/kg, p.o. (Bafna and Mishra, 2010). The extract of Cissampelos sympodialis used as an immunomodulator altered the activity of immune function through the dynamic regulation of informational molecules such as cytokines. The extract demonstrated modulation of IL-1, IL-6, TNF and IFN cytokines when tested in vitro as well as in vivo (Spelman et al., 2006). 6.6. Memory-enhancing activity The hydroalcoholic extract (400 mg/kg) of Cissampelos pareira considerably improved learning and memory of mice and significantly reversed amnesia induced by scopolamine (0.4 mg/kg, p.o.). The extract also decreased whole brain acetylcholinesterase activity when compared to piracetam (200 mg/kg) (Kulkarni et al., 2011). 6.7. Antidepressant activity The total tertiary alkaloid fraction (containing warifteine) from Cissampelos sympodialis reduced the total immobility time on two mouse models of depression (forced swim test and reserpine test), and reversed the reserpine-induced hypothermia, demonstrating an antidepressant effect in both models at a dose of 12.5 mg/kg (Mendonca-Netto et al., 2008). The hydroalcoholic leaf extract of Cissampelos sympodialis (containing warifteine) exhibited antidepressant effects together with anti-oxidant activity at dose ranges of 62.5–500 and 10–160 mg/kg, i.p. in the forced swimming test in mice (Zhang, 2004). The ELE of Cissampelos sympodialis was found to potentiate the toxicity of pentylenetetrazol in mice. Similar to imipramine, the extract also reduced the immobility period in the forced swimming test in mice and reversed the degree of ptosis and catalepsy induced by reserpine in rats (Almeida et al., 1998). The ERE of Cissampelos mucronata showed sedative activity, in mice, at up to 282.84 mg/kg (LD50). The extract progressively reduced ephedrine-induced spontaneous motor activity in rats and prolonged pentobarbitone-sleeping time in mice (Akah et al., 2002). 6.8. Neuroprotective activity Hexane, dichloromethane, ethylacetate and water extracts from aerial parts of Cissampelos owarensis exhibited activity on β-amyloid peptide production which is important in Alzheimer's disease treatment. The extracts were tested at non-toxic concentrations on Chinese hamster ovary (CHO) cells overexpressing the human neuronal β-amyloid peptide precursor to measure variations of APP processing. Cytotoxicity on CHO cells was recorded at IC50 values of 122.4, 20.5, 4200 and 4 200 mg/mL with chosen concentrations of 25, 6.25, 100 and 200 mg/mL for hexane, dichloromethane, ethylacetate and water extracts respectively (Hage et al., 2010). A combined extract of Cissampelos pareira and Anethum graveolens (1:5) exhibited protective action against age-related cognitive impairment in rats at doses of 2, 10, and 50 mg/kg. The research showed that this extract can be served as a food supplement for protection against mild cognitive impairment and the early phase of Alzheimer's disease (Thukham-mee and Wattanathorn, 2012). The methanolic extract from aerial parts of Cissampelos owarensis showed activity of 19.59% against acetylcholinesterase and 78.96% against butyrylcholinesterase with a test concentration of 42.5 μg/mL and physostigmine as the positive control (Elufioye et al., 2010). 6.9. Antimicrobial activity The methanolic extract from aerial parts of Cissampelos owarensis showed antimicrobial activity against Staphylococcus aureus, Staphylococcus pyogenes, Salmonella typhi, Escherichia coli, Shigella dysenteriae, Proteus vulgaris and Candida albicans with inhibition zone diameter (IZD), minimum inhibitory concentration (MIC) and minimum bactericidal concentrations (MBC) ranging from 18 to 27 mm, 6.25 to 50 mg/ml and 25 to 100 mg/ml, respectively (Hage et al., 2010). The extract from the whole D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 plant of Cissampelos pareira exhibited antifungal activity against Aspergillus niger and Saccharomyces cerevisiae by complete inhibition at concentrations of 500 and 1000 mg/ml in comparison to the positive controls ciprofloxacin and amphotericin B at a concentration of 3 mg/ml (Kumar et al., 2006). Dichloromethane and ethanolic extracts from Cissampelos mucronata aerial parts exhibited activity against bacteria including Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa, Salmonella typhi, Vibrio cholera, Bacillus anthracis, Streptococcus faecalis as well as against fungi such as Candida albicans and Cryptococcus neoformans. Moreover, its root extract showed remarkable larvicidal activity against Culex quinquefasciatus say (southern house mosquito) larvae. The dichloromethane root extract was found to be toxic with an LC50 of 59.6 μg/mL, whereas the ethanolic extract was found to be non-toxic at an LC50 of 100 μg/mL (Nondo et al., 2011). The total tertiary alkaloid fraction of Cissampelos capensis was found active against Bacillus subtilis with IC50 of 0.3 μg/mL. The alkaloids also inhibited bacteria (Pseudomonas aeruginosa, Proteus vulgaris and Escherichia coli, Bacillus subtilis, Staphylococcus aureus and Bacillus licheniformis) and fungi (Candida albicans, Candida eropiralis and Aspergillus niger) with IZD values ranging from 33 to 45 mm (Babajide et al., 2010). The leaf extract from Cissampelos torulosa was found to have weak antibacterial activity against Bacillus cereus and Staphylococcus aureus with MIC values of 412 mg/ml (Samie et al., 2005). The MLE of Cissampelos torulosa showed antibacterial activity against clinical strains of Campylobacter with MIC values ranging from 0.75 to 46 mg/mL (Samie et al., 2009). 6.10. Antimalarial activity The ethanolic extracts of Cissampelos andromorpha DC. and Cissampelos ovalifolia showed in vitro antimalarial activity with IC50 values of 104.1 and 37.4 mg/mL against a chloroquine-resistant strain and IC50 values of 166.6 and 34.8 mg/mL against a chloroquinesensitive strain of Plasmodium falciparum. Similarly, the total alkaloidal extracts showed activity with IC50 values of 1.5 and 3.3 mg/mL against the chloroquine-resistant strain, and an IC50 of 13.6 and 1.0 mg/mL against the chloroquine-sensitive strain (Fischer et al., 2004). The MRE from Cissampelos mucronata exhibited in vitro antiplasmodial activity against chloroquine-sensitive (D6) and chloroquine-resistant (W2) Plasmodium falciparum strains with IC50 values of 1.5 and 1.1 μg/mL, respectively. It was found the MRE also inhibited the enzyme tyrosine kinase p56lck (Tshibangu et al., 2002, 2003). The root extract of Cissampelos mucronata produced a significant reduction of parasitaemia (59% suppression) for the 4-day suppressive test in Plasmodium berghei-infected mice at a dose of 500 mg/kg, p.o. (Gessler et al., 1995), whereas the ethanolic extract showed activity against Plasmodium falciparum in vitro with IC50 values o10 μg/mL (Gessler et al., 1994; Benoit-Vical et al., 2008). The ERE of Cissampelos pareira inhibited the propagation of the rodent parasite Plasmodium berghei in vivo on BALB/c mice (Jannu et al., 2011). The hydromethanolic extract of Cissampelos pareira showed significant anti-plasmodial activity against chloroquine-sensitive (NF54) and chloroquine-resistant (ENT30) Plasmodium falciparum strains in vitro using the 3-hypoxanthine assay with an IC50 value of 5.85 μg/mL (Rukunga et al., 2009). An ethanolic root extract of Cissampelos pareira showed a significant inhibition of Plasmodium berghei with an oral dose of 500 mg/kg in mice. The mean parasitaemia in treated mice was found to 11.64% in comparison to the control, cloroquine (no parasitaemia) (Singh and Banyal, 2011). 6.11. Antiparasitic activity The alkaloidal extract from the leaves of Cissampelos ovalifolia produced an in vitro antiparasitic effect against Leishmania chagasi and Trypanosoma cruzi parasites with an EC50 value of 63.88 μg/mL. 1021 The extract reduced the number of infected macrophages at 25 μg/mL by 86.1% and 89.8%, respectively (Tempone et al., 2005). The aqueous fraction of the ELE of Cissampelos sympodialis exhibited both immunosuppressive and anti-inflammatory effects by inhibiting cyclic nucleotide phosphodiesterase activity and increased cAMP levels in intact smooth cell cultures, pig bronchoalveolar leucocytes and murine B cells. Normal and thioglycolate-elicited mice peritoneal macrophages were infected in vitro with the protozoan Trypanosoma cruzi DM28c clone. The ELE improved Trypanosoma cruzi growth and induced a 75% decrease in nitric oxide production with doses ranging between 13 and 100 μg/mL (Moreira et al., 2003a; Feily and Namazi, 2009). Cissampeloflavone (57) isolated from Cissampelos pareira showed excellent activity against Trypanosoma cruzi and Trypanosoma brucei rhodesiense (Ramirez et al., 2003). Warifteine (34) isolated from the leaves and roots of Cissampelos sympodialis showed growth inhibitory activity (anti-leishmanicidal effect) against Leishmania chagasi promastigotes in axenic cultures and the occurrence of drug-induced ultrastructural changes in the parasite with an MIC value of 0.08 mg/mL (Da Silva et al., 2012). The methanolic extract from the whole plant of Cissampelos torulosa showed in vitro antiamoebic activity against Entamoeba histolytica with IC50 and IC90 values of 410 mg/mL (Samie et al., 2009). 6.12. Anti-ulcer activity The ERE from Cissampelos mucronata, exhibited an anti-ulcer effect on indomethacin-, histamine- and stress-induced ulcer models in rats. The acute toxicity evaluation showed the LD50 value to be 288.53 mg/kg, p.o. The activity was found to be significant (po0.05) against indomethacin- and histamine-induced ulcers. This research revealed cytoprotective and antispasmodic mechanisms of action responsible for the activity (Nwafor and Okoye, 2005). The MLE of Cissampelos mucronata showed anti-ulcer activity at a dose of 450 mg/kg on isolated guinea pig ileum and inhibited contractions evoked by acetylcholine, histamine and serotonin. It showed varying degrees of protection against ulcers induced by indomethacin when compared to that of the positive control cimetidine (100 mg/kg, i.p.) and the LD50 value was calculated as 8.5 g/kg, p.o. (Akah and Nwafor, 1999; Nwafor and Akah, 2003). The ERE of Cissampelos pareira and its constituent quercetin, showed protective effects against ulceration at doses of 25–100 mg/kg p.o. in various acute and chronic ulcers in rats. The extract demonstrated significant (Po0.05 to Po0.001) protection against ethanol-, aspirin-, cold-restraint stress and pylorus ligation-induced acute gastric ulcers. The extract also reduced the ulcer index with decreased perforations in acetic acid-induced chronic ulcers (Amresh et al., 2007e). 6.13. Anticancer activity The HRE of Cissampelos pareira showed activity against forestomach cancer and carcinogen metabolising phase I and phase II enzymes along with anti-oxidant enzymes. The extract reduced the tumour incidence, the mean number of tumours and the tumour multiplicity on benzo(a)pyrene-induced gastric cancer in mice. The enhanced glutathione S-transferase level and enzyme activities involved in xenobiotic metabolism and maintaining antioxidant status of cells was due to a chemopreventive efficacy of the extract against chemotoxicity (Amresh et al., 2007c). The ethanolic extract (containing quercetin) of Cissampelos pareira showed protective effects on benzo(a)pyrene induced gastric cancer, tumour multiplicity and micronucleus polychromatic erythrocytes in mice (Amresh et al., 2007a). The crude alkaloid extracts from the leaves and rhizomes of Cissampelos capensis, Cissampelos hirta, Cissampelos mucronata and Cissampelos torulosa, showed cytotoxicity against MCF7 (breast), UACC62 (melanoma) and TK10 (renal) cancer cell lines. Leaves and rhizomes of 1022 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Cissampelos capensis showed activity with GI50 values of 16, 19 and 16 μg/mL with a TGI value of 37.5 μg/mL each; the rhizomes of Cissampelos hirta showed activity with GI50 values of 6.25, 12.5 and 12.5 μg/mL and TGI of 25, 18 and 25 μg/mL; Cissampelos mucronata showed activity with GI50 values of o6.25, 6.25 and 9 μg/mL with TGI of 18, 15 and 24 μg/mL, whilst Cissampelos torulosa showed activity with GI50 values of 12.5, 12.5 and 9 μg/mL and TGI of 37.5, 28 and 50 μg/mL against breast, melanoma and renal cancer cell lines, respectively (De Wet et al., 2009). The MLE of Cissampelos torulosa showed in vitro cytotoxic activity against Vero cells with a median inhibitory concentration of 206.4 mg/mL (Samie et al., 2009). The ethanolic extract of Cissampelos mucronata showed cytotoxic activity in human carcinoma cell lines in vitro (Gessler et al., 1995). An azafluoranthene alkaloid, norimeluteine (Morita et al., 1993c), tropone-isoquinoline alkaloid, pareitropone (Morita et al., 1995), bisbenzylisoquinoline alkaloid and cissampareine (Kupchan et al., 1965) isolated from Cissampelos pareira, exhibited cytotoxic activity against P-388 cells, whereas cissampeloflavone (57) had a low toxicity to the human KB cell line (Ramirez et al., 2003). Warifteine (34) and milonine (42) isolated from the leaves of Cissampelos sympodialis exhibited cytotoxic effects in cultured hepatocytes and V79 fibroblasts in vitro. The IC50 values determined in the three assays (nucleic acid content, tetrazolium reduction and neutral red uptake) were about 100 and 400 μM after milonine treatment whereas a dose ranging from 10 to 35 μM were obtained for warifteine in the viability tests evaluated in V79 cells and hepatocytes. Cimetidine (1.0 mM), a traditional cytochrome P450 inhibitor, did not protect the cells from the toxic action of warifteine or milonine (Melo et al., 2003). The aqueous fraction of the ELE of Cissampelos sympodialis decreased the lymphocyte proliferative response of concanavalin-A-activated BALB/c spleen cells (proliferation and cytokine secretion) in the presence of the mitogen (5 μg/mL) at concentration ranges of 6.25–50 μg/mL, in vitro. It reduced the levels of secreted IFN-γ and increased the production of both IL-10 and IL-4. The increased IL-10 production down-regulates IFN-γ secretion and T cell proliferative responses (Piuvezam et al., 1999). Warifteine from Cissampelos sympodialis inhibited the proliferative response and Ig secretion on B-lymphocytes by blocking B cell function in vitro and in vivo. Warifteine also induced an increase in cAMP and its effect on LPS-induced proliferation was mimicked by the control adenyl cyclase activator, forskolin. In vivo Ig production induced by the TI-2 antigen TNP-ficoll was also inhibited by warifteine (Rocha et al., 2010). The hydroalcoholic extract of Cissampelos sympodialis leaves inhibited the in vitro proliferative response of resting B cells induced by LPS, anti-delta-dextran and anti-IgM with IC50 values of 17.2, 13.9 and 24.3 μg/mL, respectively. The extract inhibited B cell function through an increase in intracellular cAMP levels and inhibited Ig secretion (Moreira et al., 2003b). 6.14. Anti-oxidant activity The ERE of Cissampelos pareira (containing polyphenols) showed anti-oxidant activity in the 2, 2-diphenyl-1-picrylhydrazyl (DPPH) assay at doses ranging between 50 and 400 μg/kg in vitro. The extract showed potent protective effects in an acute oxidative tissue injury on benzo(a)pyrene-induced gastric toxicity in mice at doses of 50 and 100 mg/kg (Amresh et al., 2007d; Hussain et al., 2010). The methanolic extract from aerial parts of Cissampelos owarensis had 91% anti-oxidant activity at 125 μg/mL (Habila et al., 2011). The alkaloidal fraction from Cissampelos pareira roots exhibited potent anti-oxidant activity by scavenging the stable free radical DPPH, superoxide ion and by inhibiting lipid peroxidation in rat liver homogenate induced by iron/ADP/ascorbate complex. The fraction scavenged the superoxide radical generated from the riboflavinNBT-light system in vitro with IC50 31.99 μg/mL when compared to the positive control ascorbic acid which showed activity with IC50 value of 23.52 μg/mL (Bafna and Mishra, 2010). 6.15. Cardiovascular activity The aqueous fraction of ELE from Cissampelos sympodialis exhibited cardiovascular effects on conscious, free-moving rats. The extract increased mean arterial pressure by 7, 16, 33, 43 and 38 mmHg at doses of 0.5, 1, 2, 4 and 8 mg/kg, i.v. respectively and decreased heart rate significantly. The hypertensive effect was exhibited at doses of 2 and 4 mg/kg after cardiac autonomic blockade by atenolol and atropine at a dose of 2 mg/kg. The extract significantly increased blood pressure in experimental rats and improved heart rate and contractility in isolated perfused atrial preparations (Medeiros et al., 1998). The ERE from Cissampelos pareira exhibited cardioprotective activity (po0.05) on isoproterenol-induced cardiac dysfunction in rats. The ERE improved the heart weight/body weight ratio, serum calcineurin, nitric oxide, lactate dehydrogenase and thiobarbituric acid reactive substance levels (Singh et al., 2013). The hydroalcoholic extract of Cissampelos sympodialis produced contractions (EC50 value of 76.6 μg/mL) in the presence of functional endothelium whereas in the absence of functional endothelium, the concentration–response curves were shifted to the left (EC50 values of 1.3 μg/mL) without modification of its maximal contractile effect. In the presence of L-NAME (300 μM) and indomethacin (10 mM), the concentration–response curves produced were shifted to the left (EC50 values of 21.8 and 24.3 μg/mL, respectively). The contractions induced by the extract in the rat aorta were due to activation of α-adrenoceptors (Freitas et al., 2000). Warifteine from Cissampelos sympodialis caused vasorelaxation of the rat thoracic aorta. Warifteine (1 pmol/L-10 μmol/L) induced relaxation (pD2 ¼9.40) of both endothelium-intact aortic rings precontracted with noradrenaline (10-100 μmol/L) and PGF2α (1-10 mmol/L)-precontracted rings (pD2 ¼9.2). In vascular myocytes, warifteine (100 nmol/L) significantly increased whole-cell K þ currents (at 70 mV) (Assis et al., 2013). The regulation of intracellular Ca2þ as a mechanism of spasmolytic activity of warifteine (from leaves of Cissampelos sympodialis) was studied in the rabbit aorta. Warifteine (pD'24.12) antagonised the KCl-induced contractions in a noncompetitive and reversible manner mediated by Ca2þ entry which was found similar to verapamil (pD'26.89). Noradrenaline-induced sustained contractions were also inhibited by warifteine (IC50 6.03  10 5 M) as compared to the standard, sodium nitroprusside (IC50 1.9  10 8 M) (De Freitas et al., 1996). Warifteine (from Cissampelos sympodialis) produced reversible, nonspecific, noncompetitive antagonism of histamine-, carbachol- and bradykinin-induced contractions of guineapig ileum with pD'2 values of 4.90, 4.95 and 5.03 respectively. Oxytocin- and bradykinin-induced contractions of the rat uterus was antagonised by pD'2 4.30 and 3.76, respectively and inhibited spontaneous tone and carbachol-induced sustained contractions in guinea pig trachea with IC50 values of 1.1  10 5 M and 2.9  10 5 M, respectively (Cortes et al., 1995). 6.16. Muscle-relaxant activity The ELE from Cissampelos sympodialis relaxed the spontaneous tone of guinea pig tracheal smooth muscle rings with an IC50 value of 20.5 μg/mL, inhibited cyclic nucleotide phosphodiesterase activity in isolated smooth muscle homogenates and stimulated an increase in intra-cellular cAMP synthesis in intact cultured smooth muscle cells (Thomas et al., 1997a). The quaternary alkaloid fraction (Kupchan et al., 1960) as well as hayatin methiodide (Pradhan and De, 1953) and hayatinin methochloride (Sur and Pradhan, 1964; Basu, 1970) from Cissampelos pareira, exhibited muscle-relaxant properties and were recognised as curariform drugs. Two aporphinic alkaloids (þ )-cissaglaberrimine and (þ)-trilobinine isolated from the root bark of Cissampelos glaberrima showed relaxant D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 effects in guinea pig tracheal preparations. The alkaloids reduced the spontaneous tone and inhibited the contractions induced by carbachol and histamine. Trilobinine was 6 times more potent than cissaglaberrimine in reducing the spontaneous tone and it was 1.5 times more potent in antagonising the effects of carbachol and histamine. The inhibitory effect of cissaglaberrimine in contractions induced by histamine was not attenuated in the presence of timolol (10 μM) (Cornelio et al., 1999). The aqueous leaf extract from Cissampelos mucronata showed uterine relaxant and antiabortifacient properties. The extract was found to have deleterious effects on the blood vessels of the kidneys of wistar rats. The photomicrographs of the kidneys of the rats which had received 1 ml extract for 2 weeks, showed ruptured blood vessels with distorted cytoplasm and the basement membrane of the sections treated with 0.6 ml extract had collapsed (Falana et al., 2011). The ERE of Cissampelos mucronata displayed significant in vitro relaxant activity on isolated gravid and non-gravid rat uterine smooth muscles (Nwafor et al., 2002). 6.17. Hepatoprotective activity The HRE of Cissampelos pareira showed significant hepatoprotective action against CCl4-induced hepatotoxicity in rats at doses of 100, 200 and 400 mg/kg. The catalase levels for anti-oxidant superoxide dismutase (SOD) enzymes were increased at doses of 200 and 400 mg/kg. At similar doses, it decreased cholesterol levels and increased triglyceride levels when compared to silymarin (Surendran et al., 2011). 6.18. Antidiabetic activity The HLE (200 and 400 mg/kg, p.o.) of Cissampelos pareira showed antidiabetic activity on streptozotocin-induced diabetic rats and significantly decreased fasting blood glucose and increased the body weight of rats compared to glibenclamide (5 mg/kg) (Jannu et al., 2011). 1023 exhibited activity on snake proteins from B. diporus (a venomous pit viper) (Badilla et al., 2008; Camargo et al., 2011; Dey and De, 2012). 6.22. Miscellaneous activities The hydroalcoholic extract of Cissampelos sympodialis leaves was evaluated to determine the possible toxic effects on the development of the offspring of pregnant female rats. The duration of pregnancy, weight gain, litter size, body weight of the pups, righting reflex, eye opening, hind paws supporting, body lifting and external malformation occurrence were found normal, with no signs of side effects after treatment (Maior et al., 2003). Warifteine (34) hydrochloride isolated from Cissampelos ovalifolia exhibited potent neuromuscular blocking and local anaesthetic activities (Gorinsky et al., 1972). The methiodide of hayatine isolated from Cissampelos pareira, was shown to possess powerful neuromuscular blocking activity comparable to that of d-tubocurarine chloride (Kupchan et al., 1965). The alcoholic and aqueous extract of Cissampelos pareira showed anthelmintic activity against earthworms at doses of 5, 10, 25, 50 and 100 mg/mL; both extracts were found to paralyse (vermifuge) as well as kill the earthworms (vermicide) (Shukla et al., 2012). The aqueous extract of Cissampelos mucronata was found to have significant molluscicidal activity on 2-week old Lymnaea natalensis Krauss (snail) with upper and lower fiducial limits of LC50 (Kela et al., 1989). Two aporphines, (S)-dicentrine and (S)-neolitsine, from the aerial parts of Cissampelos capensis showed activity on larval motility, with EC90 values of 6.3 and 6.4 μg/mL, respectively. The dicentrine reduced the worm counts in mice by 67% at 25 mg/kg, p.o. (Ayers et al., 2007). The aqueous extract of Cissampelos glaberrima affected the development of Plutella xylostella (diamondback moth) and caused 93.3% mortality of larvae and 66.7% of pupae (Torres et al., 2001). The extract from Cissampelos pareira was reported for its anticonvulsant activity in vivo/in vitro (Adesina, 1982; Quintans et al., 2008). 6.19. Antidiarrhoeal activity 7. Toxicity studies The ERE of Cissampelos pareira exhibited antidiarrhoeal activity with doses ranging between 25 and 100 mg/kg, p.o. and a decrease in the total number of faecal droppings by 29–60% in castor oilinduced diarrhoea. The extract produced a significant (p o0.01) reduction in intestinal fluid accumulation by 26–59% (Amresh et al., 2004). 6.20. Antifertility activity The leaf extract of Cissampelos pareira was studied to evaluate its antifertility effect and the findings showed that it altered the oestrous cycle pattern in female mice, prolonged the length of the oestrous cycle with significant increase in the duration of dioestrus stage, and reduced the number of litters of albino mice. The analysis of the principal hormones involved in oestrous cycle regulation showed that the extract altered gonadotropin release (Luteinising hormone, Follicle stimulating hormone and prolactin) and estradiol secretion with an LD50 of 7.3 g/kg, p.o. (Ganguly et al., 2007). The hydroalcoholic extract of Cissampelos pareira was evaluated for acute and subacute toxicity and produced neither mortality, nor changes in behaviour, or any other physiological effects in animals at a dose of 2 g/kg, p.o. for a period of 28 days (Amresh et al., 2008). The ethanolic extract of the aerial parts of Cissampelos pareira was determined to be safe up to a dose of 2000 mg/kg (LD50) (Amresh et al., 2007b). The ERE of Cissampelos mucronata was found to be a sedative in mice at a dose of 288.53 mg/kg (Akah et al., 2002), an acute lethality test showed an LD50 value of 282.84 mg/kg, p.o. (Nwafor and Okoye, 2005), and an LD50 value of 8.5 g/kg p.o was recorded for the MLE (Nwafor and Akah, 2003). In female mice, the acute toxicity of the leaf extract of Cissampelos pareira was found at an LD50 of 7.3 g/kg, p.o. (Ganguly et al., 2007). The tuber juice of Cissampelos ovalifolia has been used as a refreshment but due to its toxic effects, the use of this plant is restricted in various cultures in Brazil (Rodrigues, 2007). 8. Future perspectives and conclusions 6.21. Antivenom activity The aqueous leaf extract of Cissampelos pareira was studied to neutralise the haemorrhagic and proteolytic activities of the venom of Bothrops asper (a venomous pit viper) and produced total inhibition by injecting a mixture of extract and venom into the skin of mice. The aqueous, alcoholic and hexane extracts also The genus Cissampelos has been and is still used to treat a diverse range of ailments in folk medicine across many centuries, countries and continents. This genus is a rich source of many bioactive alkaloids including bisbenzylisoquinolines and aporphines (Bhakuni et al., 1987) and the aporphine alkaloids specifically are gaining popularity due to their promising anticancer, 1024 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 antiplatelet, vasodilator and antiprotozoal activities (Semwal and Semwal, 2013). The chemistry and biological activity of some species of the genus, including Cissampelos pareira, Cissampelos sympodialis, Cissampelos capensis and Cissampelos glaberrima, are well known. However, many other species including Cissampelos andromorpha DC., Cissampelos friesiorum Diels and Cissampelos nigrescens Diels have not been phytochemically or pharmacologically explored. Consequently, a broad field of future research is awaiting the researcher to discover lead molecules or purified fractions, which may have promising biological activity. In addition, many species are very well known in folk medicine, but no scientific validation for its use has been reported. There is a need for proper documentation of traditional knowledge to lead to the selection of potent as well as authentic medicines to provide a solid basis for further research (Heinrich, 2000). The polyherbal formulation, rather than an individual species extract or isolated compound, is one of the most interesting concepts in the field of herbal medicines due to its potent and remarkable synergistic biological activity. Some research groups attempted the polyherbal formulation concept with Cissampelos species and found promising antipyretic and analgesic activities (Gupta et al., 2008a; Bansod et al., 2010). Based on their research, it can be stated that the synergy of plant extracts showed more potent action compared to that of individual compounds or extracts as is frequently the case and there is a need for further work in this field. Isolated compounds, including warifteine as well as plant extracts from the genus, showed very good bioactivity (Gorinsky et al., 1972) but many of the mechanisms of action is still not well defined. Therefore, a detailed study is needed to clarify the structure–activity relationships or mechanism of action to determine the standard dose and to minimise the side-effects. Ideally, traditional knowledge should be translated into basic scientific studies including biological activity to determine efficacy, toxicity studies to dermine safety, isolation of active compounds and biomarkers to enable quality control and elucidation of the mechanism of action for combinations of plants, single extracts and isolated compounds before eventually moving into the clinical trial phase to validate traditional uses. Acknowledgements The work was financially supported by the National Research Foundation and the Tshwane University of Technology, South Africa. References Abbasi, A.M., Khan, M.A., Ahmad, M., Zafar, M., Jahan, S., Sultana, S., 2010. Ethnopharmacological application of medicinal plants to cure skin diseases and in folk cosmetics among the tribal communities of North-West Frontier Province, Pakistan. Journal of Ethnopharmacology 128, 322–335. Adesina, S.K., 1982. Studies on some plants used as anticonvulsants in Amerindian and African traditional medicine. Fitoterapia 53, 147–162. Afolayan, A.J., Mbaebie, B.O., 2010. Ethnobotanical study of medicinal plants used as anti-obesity remedies in Nkonkobe Municipality of South Africa. Pharmacognosy Journal 2, 368–373. Afolayan, A.J., Sunmonu, T.O., 2010. in vivo studies on antidiabetic plants used in South African herbal medicine. Journal of Clinical Biochemistry and Nutrition 47, 98–106. Aguirre-Galvis, L.E., 1995. Phytochemical notes on Cissampelos. Acta Biologica Colombiana 9, 85–105. Ahmad, R., Arif Malik, M., Zia-Ul-Haq, M., 1992. Alkaloids of Cissampelos pareira. Fitoterapia 63, 282. Akah, P.A., Nwafor, S.V., 1999. Studies on anti-ulcer properties of Cissampelos mucronata leaf extract. Indian Journal of Experimental Biology 37, 936–938. Akah, P.A., Nwafor, S.V., Okoli, C.O., Egbogha, C.U., 2002. Evaluation of the sedative properties of the ethanolic root extract of Cissampelos mucronata. Bollettino Chimico Farmaceutico 141, 243–246. Almeida, R.N., Melo-Diniz, M.F.F., Medeiros, I.A., Quintans-Junior, L.J., Navarro, D.S., Falcao, J.M., Duarte, J.C., Barbosa-Filho, J.M., 2005. Anorectic and behavioural effects of chronic Cissampelos sympodialis treatment in female and male rats. Phytotherapy Research 19, 121–124. Almeida, R.N., Navarro, D.S., De Assis, T.S., De Medeiros, I.A., Thomas, G., 1998. Antidepressant effect of an ethanolic extract of the leaves of Cissampelos sympodialis in rats and mice. Journal of Ethnopharmacology 63, 247–252. Amresh, G., Kant, R., Rao, C.V., Singh, P.N., 2007a. Chemomodulatory influence of Cissampelos pareira (L.) Hirsuta on gastric cancer and antioxidant system in experimental animal. Acta Pharmaceutica Sciencia 49, 71–83. Amresh, G., Rao, C.V., Singh, P.N., 2007c. Evaluation of Cissampelos pareira against gastric cancer and enzymes associated with carcinogen metabolism. Pharmaceutical Biology 45, 595–603. Amresh, G., Rao, C.V., Singh, P.N., 2007d. Antioxidant activity of Cissampelos pareira on benzo(a)pyrene-induced mucosal injury in mice. Nutrition Research 27, 625–632. Amresh, G., Reddy, G.D., Rao, C.V., Shirwaikar, A., 2004. Ethnomedical value of Cissampelos pareira extract in experimentally induced diarrhea. Acta Pharmaceutica 54, 27–35. Amresh, G., Reddy, G.D., Rao, C.V., Singh, P.N., 2007g. Evaluation of antiinflammatory activity of Cissampelos pareira root in rats. Journal of Ethnopharmacology 110, 526–531. Amresh, G., Singh, P.N., Rao, C.V., 2007f. Antinociceptive and antiarthritic activity of Cissampelos pareira roots. Journal of Ethnopharmacology 111, 531–536. Amresh, G., Singh, P.N., Rao, C.V., 2008. Toxicological screening of traditional medicine Laghupatha (Cissampelos pareira) in experimental animals. Journal of Ethnopharmacology 116, 454–460. Amresh, G., Zeashan, H., Gupta, R.J., Kant, R., Rao, C.V., Singh, P.N., 2007e. Gastroprotective effects of ethanolic extract from Cissampelos pareira in experimental animals. Journal of Natural Medicines 61, 323–328. Amresh, G., Zeashan, H., Rao, C.V., Singh, P.N., 2007b. Prostaglandin mediated antiinflammatory and analgesic activity of Cissampelos pareira. Acta Pharmaceutica Sciencia 49, 153–160. Anwer, F., Popli, S.P., Srivastava, R.M., Khare, M.P., 1968. Studies in medicinal plants. Part III. Protoberberine alkaloids from the roots of Cissampelos pareira Linn. Experientia 24, 999. Aragao, C.F.S., Filho, J.M.B., Macedo, R.O., 2001. Thermal characterization of warifteine by means of TG and a DSC photovisual system. Journal of Thermal Analysis and Calorimetry 64, 185–191. Arora, M., Sharma, T., Devi, A., Bainsal, N., Siddiqui, A.A., 2012. An inside review of Cissampelos pareira Linn: a potential medicinal plant of India. International Research Journal of Pharmacy 3, 38–41. Arya, V., Gupta, V.K., Kaur, R., 2011. A review on plants having anti-arthritic potential. International Journal of Pharmaceutical Sciences Review and Research 7, 131–136. Assis, A.C.L., Araujo, I.G.A., Lima, R.P.C., Almeida, M.M., Marinho, A.F., Barbosa-Filho, J.M., Cruz, J.S., Silva, D.F., Medeiros, I.A., 2013. Warifteine, a bisbenzylisoquinoline alkaloid, induces relaxation by activating potassium channels in vascular myocytes. Clinical and Experimental Pharmacology and Physiology 40, 37–44. Ayers, S., Zink, D.L., Mohn, K., Powell, J.S., Brown, C.M., Murphy, T., Brand, R., Pretorius, S., Stevenson, D., Thompson, D., Singh, S.B., 2007. Anthelmintic activity of aporphine alkaloids from Cissampelos capensis. Planta Medica 73, 296–297. Babajide, O.J., Mabusela, W.T., Green, I.R., Ameer, F., Weitz, F., Iwuoha, E.I., 2010. Phytochemical screening and biological activity studies of five South African indigenous medicinal plants. Journal of Medicinal Plants Research 4, 1924–1932. Badilla, B., Chaves, F., Jimenez, S., Rodriguez, G., Poveda, L.J., 2008. Effects of an extract of Cissampelos pareira on the hemorrhagic and proteolytic activities from Bothrops asper venom. Pharmacognosy Magazine 4, 27–31. Bafna, A., Mishra, S., 2010. Antioxidant and immunomodulatory activity of the alkaloidal fraction of Cissampelos pareira Linn. Scientia Pharmaceutica 78, 21–31. Bafna, A.R., Mishra, S.H., 2005. Immunomodulatory activity of methanol extract of roots of Cissampelos pareira Linn. Ars Pharmaceutica 46, 253–262. Bansod, M.S., Kagathara, V.G., Pujari, R.R., Patel, V.B., Ardeshna, H.H., 2011. Therapeutic effect of a polyherbal preparation on adjuvant induced arthritis in wistar rats. International Journal of Pharmacy and Pharmaceutical Sciences 3, 186–192. Bansod, M.S., Kagathara, V.G., Somkuwar, A.D., 2010. Evaluation of analgesics and anti-inflammatory activity of a poly-herbal formulation. International Journal of PharmTech Research 2, 1520–1527. Barbosa-Filho, J.M., Agra, M.F., Thomas, G., 1997b. Botanical, chemical and pharmacological investigation on C. sympodialis from Paraíba (Brazil). Ciência e Cultura 49, 386–394. Barbosa-Filho, J.M., da-Cunha, E.V.L., Cornelio, M.L., Da Silva Dias, C., Gray, A.I., 1997a. Cissaglaberrimine, an aporphine alkaloid from Cissampelos glaberrima. Phytochemistry 44, 959–961. Basha, S.K., Sudarsanam, G., 2012. Traditional use of plants against snakebite in Sugali tribes of Yerramalais of Kurnool district, Andhra Pradesh, India. Asian Pacific Journal of Tropical Biomedicine 2, S575–S579. Basu, D.K., 1970. Studies on curariform activity of hayatinin methochloride, an alkaloid of Cissampelos pareira. Japanese Journal of Pharmacology 20, 246–252. Batista-Lima, K.V., Ribeiro, R.A., Balestieri, F.M.P., Thomas, G., Piuvezam, M.R., 2001. Anti-inflammatory activity of Cissampelos sympodialis Eichl. (Menispermaceae) leaf extract. Acta Farmaceutica Bonaerense 20, 275–279. Behera, P.C., Tripathy, D.P., Parija, S.C., 2013. Shatavari: potentials for galactogogue in dairy cows. Indian Journal of Traditional Knowledge 12, 9–17. D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Benoit-Vical, F., 2005. Ethnomedicine in malaria treatment. IDrugs 8, 45–52. Benoit-Vical, F., Soh, P.N., Salery, M., Harguem, L., Poupat, C., Nongonierma, R., 2008. Evaluation of Senegalese plants used in malaria treatment: focus on Chrozophora senegalensis. Journal of Ethnopharmacology 116, 43–48. Bezerra-Santos, C.R., Balestieri, F.M.P., Rossi-Bergmann, B., Pecanha, L.M.T., Piuvezam, M.R., 2004. Cissampelos sympodialis Eichl. (Menispermaceae): oral treatment decreases IgE levels and induces a Th1-skewed cytokine production in ovalbumin-sensitized mice. Journal of Ethnopharmacology, 95, pp 191–197. Bezerra-Santos, C.R., Vieira-de-Abreu, A., Barbosa-Filho, J.M., Bandeira-Melo, C., Piuvezam, M.R., Bozza, P.T., 2006. Anti-allergic properties of Cissampelos sympodialis and its isolated alkaloid warifteine. International Immunopharmacology 6, 1152–1160. Bezerra-Santos, C.R., Vieira-De-Abreu, A., Vieira, G.C., Filho, J.R., Barbosa-Filho, J.M., Pires, A.L., Martins, M.A., Souza, H.S., Bandeira-Melo, C., Bozza, P.T., Piuvezam, M.R., 2012. Effectiveness of Cissampelos sympodialis and its isolated alkaloid warifteine in airway hyperreactivity and lung remodeling in a mouse model of asthma. International Immunopharmacology 13, 148–155. Bhakuni, D.S., Jain, S., Chaturvedi, R., 1987. The biosynthesis of the alkaloids of Cissampelos pareira Linn. Tetrahedron 43, 3975–3982. Bhatnagar, A.K., Bhattacharji, S., Roy, A.C., Popli, S.P., Dhar, M.L., 1967. Chemical examination of the roots of Cissampelos pareira Linn. IV. Structure and stereochemistry of hayatin. Journal of Organic Chemistry 32, 819–820. Bhatnagar, A.K., Popli, S.P., 1967. Chemical examination of the roots of Cissampelos pareira Linn. Part V. structure and stereochemistry of hayatidin. Experientia 23, 242–243. Bhatnagar, P.K., Katiyar, C.K., Khanna, N., Upadhyay, D.J., Swaminathan, S., Srinivas, K., Sharma, N., Kanaujia, A., Sood, R., Singhal, S., Shukla, G., Duggar, R., Pareek, P.K., Singh, Y., Khan, S. Raut, R., 2011. Anti dengue activity of Cissampelos pareira extracts. Patent US20120107424 (published on 30 November). Bhattacharji, S., Roy, A.C., Dhar, M.L., 1962. Chemical examination of the roots of Cissampelos pareira. Part II-isolation & structure of hayatinin. Journal of Scientific and Industrial Research—B 21, 428–433. Bhattacharji, S., Sharma, V.N., Dhar, M.L., 1952. Chemical examination of the roots of Cissampelos pareira Linn. Journal of Scientific and Industrial Research—B 11, 81–82. Bhattacharji, S., Sharma, V.N., Dhar, M.L., 1956. Chemical examination of the roots of Cissampelos pareira. Journal of Scientific and Industrial Research—B 15, 363–368. Blasko, G., Cordell, G.A., 1988. Morphinandienone alkaloids. Heterocycles 27, 1269–1300. Boissier, J.R., Combes, G., Pernet, R., Dumont, C., 1965. Contribution a letude des alcaloides de quelques Menispermacees de Madagascar: Cissampelos pareira, Cyclea madagascariensis, Anisocyclea grandidieri et Spirospermum penduliflorum. Lloydia 28, 191–198. Bora, U., Sahu, A., Saikia, A.P., Ryakala, V.K., Goswami, P., 2007. Medicinal plants used by the people of Northeast India for curing malaria. Phytotherapy Research 21, 800–804. Camargo, F., Torres, A.M., Ricciardi, G., Ricciardi, A., Dellacassa, E., 2011. SDS PAGE: a useful tool for preliminary screening of anti-snake activity of plant extracts. Boletin Latinoamericano y del Caribe de Plantas Medicinales y Aromaticas 10, 429–434. Cerqueira-Lima, A.T., Alcantara-Neves, N.M., de Carvalho, L.C.P., Costa, R.S., BarbosaFilho, J.M., Piuvezam, M., Russo, M., Barboza, R., de Jesus Oliveira, E., Marinho, A., Figueiredo, C.A., 2010. Effects of Cissampelos sympodialis Eichl. and its alkaloid, warifteine, in an experimental model of respiratory allergy to Blomia tropicalis. Current Drug Targets 11, 1458–1467. Chhetri, D.R., Parajuli, P., Subba, G.C., 2005. Antidiabetic plants used by Sikkim and Darjeeling Himalayan tribes, India. Journal of Ethnopharmacology 99, 199–202. Chopra, R.N., 1958. Indigenous Drugs of India, 2nd edition V.N. Dhur and Sons, Calcutta. Chowdury, A.R., 1972. Chemical investigation on Cissampelos pareira Linn. Science and Culture 38, 358–359. Cornelio, M.L., Barbosa-Filho, J.M., Cortes, S.F., Thomas, G., 1999. Tracheal relaxant activity of cissaglaberrimine and trilobinine, two aporphinic alkaloids from Cissampelos glaberrima. Planta Medica 65, 462–464. Correa, M.F.P., De Melo, G.O., Costa, S.S., 2008. Natural products from plant origin potentially usefull in the asthma therapy. Brazilian Journal of Pharmacognosy 18, 785–797. Cortes, S.F., De Alencar, J.L., Thomas, G., Barbosa Filho, J.M., 1995. Spasmolytic actions of warifteine, a bisbenzylisoquinoline alkaloid isolated from the root bark of Cissampelos sympodialis Eichl. (Menispermaceae). Phytotherapy Research 9, 579–583. Costa, H.F., Bezerra-Santos, C.R., Barbosa Filho, J.M., Martins, M.A., Piuvezam, M.R., 2008. Warifteine, a bisbenzylisoquinoline alkaloid, decreases immediate allergic and thermal hyperalgesic reactions in sensitized animals. International Immunopharmacology 8, 519–525. Da Silva, E.C., Rayol, C.D., Medeiros, P.L., Figueiredo, R.C.B.Q., Piuvezan, M.R., Brabosa-Filho, J.M., Fernandes Marinho, A., Silva, T.G., Militao, G.C.G., Cassilhas, A.P.P., De Andrade, P.P., 2012. Antileishmanial activity of warifteine: a bisbenzylisoquinoline alkaloid isolated from Cissampelos sympodialis Eichl. (Menispermaceae). The Scientific World Journal (Article no. 516408). Da-Cunha, E.V.L., Cornelio, M.L., Barbosa-Filho, J.M., Braz-Filho, R., Gray, A.I., 1998. Eletefine, a stephaoxocane alkaloid from Cissampelos glaberrima. Journal of Natural Products 61, 1140–1142. De Freitas, M.R., Cortes, S.D.F., Thomas, G., Filho, J.M.B., 1996. Modification of Ca2 þ metabolism in the rabbit aorta as a mechanism of spasmolytic action 1025 of warifteine, a bisbenzylisoquinoline alkaloid isolated from the leaves of Cissampelos sympodialis Eichl. (Menispermaceae). Journal of Pharmacy and Pharmacology 48, 332–336. De Freitas, M.R., De Alencar, J.L., da-Cunha, E.V., Barbosa-Filho, J.M., Gray, A.I., 1995. Milonine, an 8,14-dihydromorphinandienone alkaloid from leaves of Cissampelos sympodialis. Phytochemistry 40, 1553–1555. De Lira, G.A., De Andrade, L.M., Florencio, K.C., Da Silva, M.S., Barbosa-Filho, J.M., Leitao da-Cunha, E.V., 2002. Roraimine: a bisbenzylisoquinoline alkaloid from Cissampelos sympodialis roots. Fitoterapia 73, 356–358. De Wet, H., 2006. An Ethnobotanical and Chemotaxonomic Study of South African Menispermaceae. (PhD thesis). Department of Botany, University of Johannesburg. Available at: 〈https://ujdigispace.uj.ac.za/bitstream/handle/10210/233/ Chapter6.pdf?sequence=6〉. De Wet, H., Fouche, G., Van Heerden, F.R., 2009. in vitro cytotoxicity of crude alkaloidal extracts of South African Menispermaceae against three cancer cell lines. African Journal of Biotechnology 8, 3332–3335. De Wet, H., Nkwanyana, M.N., Van Vuuren, S.F., 2010. Medicinal plants used for the treatment of diarrhoea in northern Maputaland, KwaZulu-Natal Province, South Africa. Journal of Ethnopharmacology 130, 284–289. De Wet, H., Tilney, P.M., Van Wyk, B.E., 2002. Vegetative morphology and anatomy of Cissampelos in South Africa. South African Journal of Botany 68, 181–190. De Wet, H., Van Heerden, F.R., Van Wyk, B.E., 2011. Alkaloidal variation in Cissampelos capensis (Menispermaceae). Molecules 16, 3001–3009. De Wet, H., Van Wyk, B.E., 2008. An ethnobotanical survey of southern African Menispermaceae. South African Journal of Botany 74, 2–9. Deutschlander, M.S., Lall, N., Van De Venter, M., 2009. Plant species used in the treatment of diabetes by South African traditional healers: an inventory. Pharmaceutical Biology 47, 348–365. Dey, A., De, J.N., 2012. Phytopharmacology of antiophidian botanicals: a review. International Journal of Pharmacology 8, 62–79. Don, G., 1831. A General System of Gardening and Botany, vol. 1. Longman and Co., London, pp. 109–112. Dwuma-Badu, D., Ayim, J.S.K., Mingle, C.A., Tackie, A.N., Slatkin, D.J., Knapp, J.E., Schiff Jr., P.L., 1975. Alkaloids of Cissampelos pareira. Phytochemistry 14, 2520–2521. Elufioye, T.O., Obuotor, E.M., Sennuga, A.T., Agbedahunsi, J.M., Adesanya, S.A., 2010. Acetylcholinesterase and butyrylcholinesterase inhibitory activity of some selected Nigerian medicinal plants. Brazilian Journal of Pharmacognosy 20, 472–477. Elujoba, A.A., 1995. Female infertility in the hands of traditional birth attendants in South-Western Nigeria. Fitoterapia 66, 239–248. Falana, B.A., Caxton-Martins, E.A., Ofusori, D.A., 2011. Microanatomical effects of aqueous extract of the leaves Cissampelos mucronata on the kidneys of adult female wistar rats (Rattus norvegicus). Scientific Research and Essays 6, 2619–2623. Feily, A., Namazi, M.R., 2009. Cissampelos sympodialis Eichl (Menispermaceae) leaf extract as a possible novel and safe treatment for psoriasis. Sao Paulo Medical Journal 127, 241–242. Ferreira, M.A., Prista, L.N., Alves, A.C., Roque, A.S., 1965. Estudo QuUmico De Cissampelos mucronata A. Rich 1. Isolamento de d-isochondodendrina. Garcia de Orta 13, 395–406. Fischer, D.C.H., Gualda, N.C.D.A., Bachiega, D., Carvalho, C.S., Lupo, F.N., Bonotto, S.V., Alves, M.D.O., Yogi, A., Santi, S.M.D., Avila, P.E., Kirchgatter, K., Moreno, P.R.H., 2004. in vitro screening for antiplasmodial activity of isoquinoline alkaloids from Brazilian plant species. Acta Tropica 92, 261–266. Freitas, M.R., Lemos, V.S., Queiroga, C.E.G., Thomas, G., Medeiros, I.A., Cortes, S.F., 2000. Mechanisms of the contractile effect of the hydroalcoholic extract of Cissampelos sympodialis Eichl. in the rat aorta. Phytomedicine 7, 63–67. Galicia, E.H., Aguilar-Contreras, A., Aguilar-Santamaria, L., Roman-Ramos, R., Chavez-Miranda, A.A., Garcia-Vega, L.M., Flores-Saenz, J.L., Alarcon-Aguilar, F.J., 2002. Studies on hypoglycemic activity of Mexican medicinal plants. Proceedings of the Western Pharmacology Society 45, 118–124. Galinis, D.L., Wiemer, D.F., Cazin, J., 1993. Cissampentin: a new bisbenzylisoquinoline alkaloid from Cissampelos fasciculata. Tetrahedron 49, 1337–1342. Ganguly, M., Kr. Borthakur, M., Devi, N., Mahanta, R., 2007. Antifertility activity of the methanolic leaf extract of Cissampelos pareira in female albino mice. Journal of Ethnopharmacology 111, 688–691. Gessler, M.C., Nkunya, M.H.H., Mwasumbi, L.B., Heinrich, M., Tanner, M., 1994. Screening Tanzanian medicinal plants for antimalarial activity. Acta Tropica 56, 65–77. Gessler, M.C., Tanner, M., Chollet, J., Nkunya, M.H.H., Heinrich, M., 1995. Tanzanian medicinal plants used traditionally for the treatment of malaria: in vivo antimalarial and in vitro cytotoxic activities. Phytotherapy Research 9, 504–508. Giday, M., Asfaw, Z., Woldu, Z., 2009. Medicinal plants of the Meinit ethnic group of Ethiopia: an ethnobotanical study. Journal of Ethnopharmacology 124, 513–521. Giorgetti, M., Rossi, L., Rodrigues, E., 2011. Brazilian plants with possible action on the central nervous system – a study of historical sources from the 16th to 19th century. Brazilian Journal of Pharmacognosy 21, 537–555. Gomes, A.P.B., Freire, F.D., Aragao, C.F.S., 2012. Determination of vapor pressure curves of warifteine and methylwarifteine by using thermogravimetry. Journal of Thermal Analysis and Calorimetry 108, 249–252. Gorinsky, C., Luscombe, D.K., Nicholls, P.J., 1972. Neuromuscular blocking and local anaesthetic activities of warifteine hydrochloride, an alkaloid isolated from Cissampelos ovalifolia D.C. Journal of Pharmacy and Pharmacology 24 (Suppl. 147), P-148. 1026 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Gupta, A., Pandey, S., Shah, D.R., Seth, N.R., Yadav, J.S., 2011. Pharmacognostical and phytochemical evaluation of leaves of Cissampelos pareira. Pharmacognosy Journal 3, 25–28. Gupta, M., Shaw, B.P., Mukerjee, A., 2008a. Studies on antipyretic-analgesic and ulcerogenic activity of polyherbal preparation in rats and mice. International Journal of Pharmacology 4, 88–94. Gupta, M., Shaw, B.P., Mukherjee, A., 2008b. Evaluation of antipyretic effect of a traditional polyherbal preparation: a double-blind, randomized clinical trial. International Journal of Pharmacology 4, 190–195. Habila, J.D., Bello, I.A., Dzikwe, A.A., Ladan, Z., Sabiu, M., 2011. Comparative evaluation of phytochemicals, antioxidant and antimicrobial activity of four medicinal plants native to Northern Nigeria. Australian Journal of Basic and Applied Sciences 5, 537–543. Hage, S., Kienlen-Campard, P., Octave, J.N., Quetin-Leclercq, J., 2010. in vitro screening on β-amyloid peptide production of plants used in traditional medicine for cognitive disorders. Journal of Ethnopharmacology 131, 585–591. Haq, F., Ahmad, H., Alam, M., 2011. Traditional uses of medicinal plants of Nandiar Khuwarr catchment (District Battagram), Pakistan. Journal of Medicinal Plants Research 5, 39–48. Haynes, L.J., Herbert, E.J., Plimmer, J.R., 1966. ( þ þ)-4″-O-methylcurine from Cissampelos pareira L. Journal of the Chemical Society C: Organic, 615–617. Heinrich, M., 2000. Ethnobotany and its role in drug development. Phytotherapy Research 14, 479–488. Heinrich, M., Haller, B.F., Leonti, M., 2014. A perspective on natural products research and ethnopharmacology in Mexico: the eagle and the serpent on the prickly pear Cactus. Journal of Natural Products 77, 678–689. Hemraj, Upmanyu, Gupta, N., Jindal, A., Jalhan, S., A., 2012. Pharmacological activities of Stephania glabra, Woodfordia fruticosa and Cissempelos pareira – a review. International Journal of Pharmacy and Pharmaceutical Sciences 4, 16–23. Hullatti, K.K., Sharada, M.S., 2010. Comparative phytochemical investigation of the sources of ayurvedic drug Patha: a chromatographic fingerprinting analysis. Indian Journal of Pharmaceutical Sciences 72, 39–45. Hussain, I., Khan, H., Khan, M.A., 2010. Screening of selected medicinal plants for the antioxidant potential. Pakistan Journal of Scientific and Industrial Research 53, 338–339. Jain, A., Katewa, S.S., Galav, P.K., Sharma, P., 2005. Medicinal plant diversity of Sitamata wildlife sanctuary, Rajasthan, India. Journal of Ethnopharmacology 102, 143–157. Jannu, V., Sai Vishal, D., Ranjith Babu, V., Harisha, B., D., Ravi Chandra Sekhara Reddy, 2011. Antidiabetic activity of hydro-alcoholic extract of Cissampelos pareira linn. leaves in streptozotocin induced diabetic rats. International Journal of Pharmacy and Technology 3, 3601–3611. Kakrani, H.N., Saluja, A.K., 2002. Traditional treatment of gastro-intestinal tract disorders in Kutch district, Gujarat state, India. Journal of Natural Remedies 2, 71–75. Kaur, A., Nain, P., Nain, J., 2012. Herbal plants used in treatment of rheumatoid arthritis: a review. International Journal of Pharmacy and Pharmaceutical Sciences 4, 44–57. Kela, S.L., Ogunsusi, R.A., Ogbogu, V.C., Nwude, N., 1989. Susceptibility of two-week old Lymnaea natalensis to some plant extracts. Revue d“elevage et de medecine veterinaire des pays tropicaux 42, 189–192. Kufer, J., Forther, H., Poll, E., Heinrich, M., 2005. Historical and modern medicinal plant uses – the example of the Ch’orti’ Maya and Ladinos in Eastern Guatemala. Journal of Pharmacy and Pharmacology 57, 1127–1152. Kulkarni, P.D., Ghaisas, M.M., Chivate, N.D., Sankpal, P.S., 2011. Memory enhancing activity of Cissampelos pareira in mice. International Journal of Pharmacy and Pharmaceutical Sciences 3, 206–211. Kumar, V.P., Chauhan, N.S., Padh, H., Rajani, M., 2006. Search for antibacterial and antifungal agents from selected Indian medicinal plants. Journal of Ethnopharmacology 107, 182–188. Kupchan, S.M., Kubuta, S., Fujita, E., Kobayashi, S., Block, J.H., Telang, S.A., 1966. Tumor inhibitors. XV. The structure and configuration of cissampareine, a novel bisbenzylisoquinoline alkaloid. Journal of the American Chemical Society 88, 4212–4218. Kupchan, S.M., Patel, A.C., Fujita, E., 1965. Tumor inhibitors VI. Cissampareine, new cytotoxic alkaloid from Cissampelos pareira. Cytotoxicity of bisbenzylisoquinoline alkaloids. Journal of Pharmaceutical Sciences 54, 580–583. Kupchan, S.M., Yokoyama, N., Beal, J.L., 1960. Menispermaceae alkaloids. I. The alkaloids of Cissampelos pareira Linn. and the origin of radix pareirae brave. Journal of the American Pharmaceutical Association 49, 727–731. Leonti, M., Vibrans, H., Sticher, O., Heinrich, M., 2001. Ethnopharmacology of the Popoluca, Mexico: an evaluation. The Journal of Pharmacy and Pharmacology 53, 1653–1669. Lewis, W.H., 1977. Medical Botany. John Wiley and Sons, Inc., New York, p. 324. Machado, T.B., Pinto, A.V., Pinto, M.C.F.R., Leal, I.C.R., Silva, M.G., Amaral, A.C.F., Kuster, R.M., Netto-dosSantos, K.R., 2003. in vitro activity of Brazilian medicinal plants, naturally occurring naphthoquinones and their analogues, against methicillin-resistant Staphylococcus aureus. International Journal of Antimicrobial Agents 21, 279–284. Maior, F.N.S., Siqueira, J.D.S., Barbosa, M.D.D.S., De Almeida, R.N., 2003. Postnatal development of offsprings exposed to hydro alcoholic extract of Cissampelos sympodialis Eichl., during the pregnancy. Acta Farmaceutica Bonaerense 22, 321–325. Maliwichi-Nyirenda, C.P., Maliwichi, L.L., 2010. Medicinal plants used for contraception and pregnancy-related cases in Malawi: a case study of Mulanje District. Journal of Medicinal Plants Research 4, 2121–2127. Marinho, A.F., Barbosa-Filho, J.M., Oliveira, E.J., 2012. A validated method for the simultaneous quantitation of bioactive alkaloid markers in the leaf ethanolic extract of Cissampelos sympodialis Eichl.: a phenological variation study. Phytochemical Analysis 23, 426–432. Mathew, P.M., 1958. Studies on the Menispermaceae. Proceedings of the Indian Academy of Sciences—Section B 47, 274–286. Maurya, S.K., Periasamy, M., Bal, N.C., 2013. High gender-specific susceptibility to curare – a neuromuscular blocking agent. Biological Research 46, 75–78. Medeiros, I.A., Pires, S.L.S., Almeida, R.N., Thomas, G., 1998. Cardiovascular effects of an aqueous fraction of the ethanol extract of the leaves of Cissampelos sympodialis Eichl. in the rat. Phytomedicine 5, 97–102. Melo, P.S., De Medeiros Cavalcante, H.M., Barbosa-Filho, J.M., De Fatima Formiga Melo Diniz, M., De Medeiros, I.A., Haun, M., 2003. Warifteine and milonine, alkaloids isolated from Cissampelos sympodialis Eichl: cytotoxicity on rat hepatocyte culture and in V79 cells. Toxicology Letters 142, 143–151. Mendes de Oliveira Jr, W., Batista Benedito, R., De Menezes Patricio Santos, C.C., Dias Rodrigues, L.T., Fernandes Marinho, A., Soares Lima de Morais, L.C., De Fatima Formiga Melo Diniz, M., Nobrega de Almeida, R., 2011. Analgesic effect of hydroalcoholic extract of Cissampelos sympodialis Eichl leaves. Boletin Latinoamericano y del Caribe de Plantas Medicinales y Aromaticas 10, 333–337. Mendes, F.R., Carlini, E.A., 2007. Brazilian plants as possible adaptogens: an ethnopharmacological survey of books edited in Brazil. Journal of Ethnopharmacology 109, 493–500. Mendonca-Netto, S., Varela, R.W.B., Fechine, M.F., Queiroga, M.N.G., Souto-Maior, F.N., Almeida, R.N., 2008. Antidepressant effects of total tertiary alkaloid fraction of Cissampelos sympodialis Eichler in rodents. Brazilian Journal of Pharmacognosy 18, 165–169. Michel, J., Duarte, R.E., Bolton, J.L., Huang, Y., Caceres, A., Veliz, M., Soejarto, D.D., Mahady, G.B., 2007. Medical potential of plants used by the Qeqchi Maya of Livingston, Guatemala for the treatment of women’s health complaints. Journal of Ethnopharmacology 114, 92–101. Moreira, M.S.A., Freire-de-Lima, C.G., Trindade, M.N., Castro-Faria-Neto, H.C., Piuvezam, M.R., Peçanha, L.M.T., 2003a. Cissampelos sympodialis Eichl (Menispermaceae) leaf extract induces interleukin-10-dependent inhibition of Trypanosoma cruzi killing by macrophages. Brazilian Journal of Medical and Biological Research 36, 199–205. Moreira, M.S.A., Piuvezam, M.R., Pecanha, L.M.T., 2003b. Modulation of B-lymphocyte function by an aqueous fraction of the ethanol extract of Cissampelos sympodialis Eichl (Menispermaceae). Brazilian Journal of Medical and Biological Research 36, 1511–1522. Morita, H., Matsumoto, K., Takeya, K., Itokawa, H., 1993b. Conformation of tropolone ring in antileukemic tropoloisoquinoline alkaloids. Chemical and Pharmaceutical Bulletin 41, 1478–1480. Morita, H., Matsumoto, K., Takeya, K., Itokawa, H., 1993c. Azafluoranthene alkaloids from Cissampelos pareira. Chemical and Pharmaceutical Bulletin 41, 1307–1308. Morita, H., Matsumoto, K., Takeya, K., Itokawa, H., Iitaka, Y., 1993a. Structures and solid state tautomeric forms of two novel antileukemic tropoloisoquinoline alkaloids, Pareirubrines A and B, from Cissampelos pareira. Chemical and Pharmaceutical Bulletin 41, 1418–1422. Morita, H., Takeya, K., Itokawa, H., 1995. A novel condensed tropone-isoquinoline alkaloid, pareitropone, from Cissampelos pareira. Bioorganic and Medicinal Chemistry Letters 5, 597–598. Morten, J.F., 1981. Atlas of Medicinal Plants of Middle America, Bahamas to Yucatan, 1st Ed. Charles C Thomas Publisher Ltd., Springfield, IL. Mosango, D.M., 2008. Cissampelos owariensis P.Beauv. ex DC. In: Schmelzer, G.H., Gurib-Fakim, A. (Eds.), Prota 11(1): Medicinal Plants/Plantes médicinales 1. [CD-Rom]. PROTA, Wageningen, Netherlands. Mugisha, M.K., Origa, H.O., 2007. Medicinal plants used to induce labour during childbirth in western Uganda. Journal of Ethnopharmacology 109, 1–9. Mukazayire, M.J., Minani, V., Ruffo, C.K., Bizuru, E., Stevigny, C., Duez, P., 2011. Traditional phytotherapy remedies used in Southern Rwanda for the treatment of liver diseases. Journal of Ethnopharmacology 138, 415–431. Mukherjee, R., Keifer, P.A., 2003. Warifteine and methylwarifteine: 1H and 13C assignments by two-dimensional NMR spectroscopy. Magnetic Resonance in Chemistry 41, 213–218. Muzila, M., 2008. Cissampelos mucronata A.Rich. In: Schmelzer, G.H., Gurib-Fakim, A. (Eds.), Prota 11(1): Medicinal Plants/Plantes médicinales 1. [CD-Rom]. PROTA, Wageningen, Netherlands. Nagarajan, K., Chauhan, N., Mittal, A., Singh, V., Bodla, R.B., Tiwari, R.K., 2011. Phytochemical extraction, optimization and physico-chemical characterization of two bioactive isolates from the leaves and stem of Cissampelos pareira. Der Pharma Chemica 3, 327–337. Namsa, N.D., Mandal, M., Tangjang, S., 2011. Anti-malarial herbal remedies of northeast India, Assam: an ethnobotanical survey. Journal of Ethnopharmacology 133, 565–572. Nondo, R.S.O., Mbwambo, Z.H., Kidukuli, A.W., Innocent, E.M., Mihale, M.J., Erasto, P., Moshi, M.J., 2011. Larvicidal, antimicrobial and brine shrimp activities of extracts from Cissampelos mucronata and Tephrosia villosa from coast region, Tanzania. BMC Complementary and Alternative Medicine 11 (article no. 33). Nwafor, S.V., Akah, P.A., 2003. Effect of methanolic leaf extract of Cissampelos mucronata A. Rich against indomethacin induced ulcer in rats. Indian Journal of Experimental Biology 41, 181–183. Nwafor, S.V., Akah, P.A., Okoli, C.O., Ndu, O.O., Ichu, E.O., 2002. Uterine relaxant property of the ethanolic root extract of Cissampelos mucronata. Journal of Natural Remedies 2, 59–65. D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Nwafor, S.V., Okoye, C.F., 2005. Antiulcer properties of the ethanol root extract of Cissampelos mucronata. Pharmaceutical Biology 43, 396–403. Olorunnisola, O.S., Bradley, G., Afolayan, A.J., 2011. Ethnobotanical information on plants used for the management of cardiovascular diseases in NKonkobe municipality, South Africa. Journal of Medicinal Plants Research 5, 4256–4260. Oyen, L.P.A., 2008a. Cissampelos pareira L. In: Schmelzer, G.H., Gurib-Fakim, A. (Eds.), Prota 11(1): Medicinal Plants/Plantes medicinales 1. [CD-Rom]. PROTA, Wageningen, Netherlands. Oyen, L.P.A., 2008b. Cissampelos capensis L.f. In: Schmelzer, G.H., Gurib-Fakim, A. (Eds.), Prota 11(1): Medicinal Plants/Plantes médicinales 1. [CD-Rom]. PROTA, Wageningen, Netherlands. Pattanaik, C., Sudhakar Reddy, C., Murthy, M.S.R., 2008. An ethnobotanical survey of medicinal plants used by the Didayi tribe of Malkangiri district of Orissa, India. Fitoterapia 79, 67–71. Piuvezam, M.R., Bezerra-Santos, C.R., Bozza, P.T., Bandeira-Melo, C., Vieira, G., Costa, H.F., 2012. Cissampelos sympodialis (Menispermaceae): a novel phytotherapic weapon against allergic diseases? (Chapter 24). In: Pereira, C. (Ed.), Allergic Diseases – Highlights in the Clinic, Mechanisms and Treatment. InTech, Croatia, ISBN: 978-953-51-0227-4, pp. 477–498. Piuvezam, M.R., Pecanha, L.M.T., Alexander, J., Thomas, G., 1999. Cissampelos sympodialis Eichl. leaf extract increases the production of IL-10 by concanavalin-A-treated BALB/c spleen cells. Journal of Ethnopharmacology 67, 93–101. Porto, N.M., Basilio, I.J.L.D., Agra, M.D.F., 2008. Pharmacobotanical study of the leaves of Cissampelos sympodialis Eichl., (Menispermaceae). Brazilian Journal of Pharmacognosy 18, 102–107. Pradhan, S.N., De, N.N., 1953. Hayatin methiodide: a new curariform drug. British Journal of Pharmacology and Chemotherapy 36, 399–405. Quattrocchi, U., 1912. CRC World Dictionary of Medicinal and Poisonous Plants. CRC Press, pp. 965–969. Quintans Jr, L.J., Almeida, J.R.G.S., Lima, J.T., Nunes, X.P., Siqueira, J.S., De Oliveira, L.E. G., Almeida, R.N., De Athayde-Filho, P.F., Barbosa-Filho, J.M., 2008. Plants with anticonvulsant properties—a review. Brazilian Journal of Pharmacognosy 18, 798–819. Rajan, S., Jayendran, M., Sethuraman, M., 2003. Medico-ethnobotany: aA study on the Kattunayaka tribe of Nilgiri hills, Tamil Nadu. Journal of Natural Remedies 3, 68–72. Rajan, S., Sethuraman, M., Mukherjee, P.K., 2002. Ethnobiology of the Nilgiri Hills, India. Phytotherapy Research 16, 98–116. Ramasubramaniaraja, R., Babu, N.M., 2010. Antihelminthic studies and medicinal herbs – an overview. International Journal of Pharmaceutical Sciences Review and Research 5, 39–47. Ramirez, I., Carabot, A., Melendez, P., Carmona, J., Jimenez, M., Patel, A.V., Crab, T.A., Blunden, G., Cary, P.D., Croft, S.L., Costa, M., 2003. Cissampeloflavone, a chalcone–flavone dimer from Cissampelos pareira. Phytochemistry 64, 645–647 (1421). Rana, T.S., Datt, B., 1997. Ethnobotanical observation among Jaunsaris of JaunsarBawar, Dehra Dun (U.P.), India. Pharmaceutical Biology 35, 371–374. Rawat, R., Vashistha, D.P., 2011. Common herbal plant in Uttarakhand, used in the popular medicinal preparation in Ayurveda. International Journal of Pharmacognosy and Phytochemical Research 3, 64–73. Rhodes, D.G., 1975. A revision of the genus Cissampelos. Phytologia 30, 415–484. Rocha, A.I., Luz, A.I.R., Silva, M.F., 1984. A presenca de alcaloides em especies botanicas da Amazonia. Menispermaceae. Acta Amazonica 14, 245. Rocha, J.D.B., Decote-Ricardo, D., Redner, P., Lopes, U.G., Barbosa-Filho, J.M., Piuvezam, M.R., Arruda, L.B., Pecanha, L., Maria Torres, 2010. Inhibitory effect of the alkaloid warifteine purified from Cissampelos sympodialis on B-lymphocyte function in vitro and in vivo. Planta Medica 76, 325–330. Rodrigues, E., 2007. Plants of restricted use indicated by three cultures in Brazil (Caboclo-river dweller, Indian and Quilombola). Journal of Ethnopharmacology 111, 295–302. Rosario, S.L., Da Silva, A.J.R., Parente, J.P., 1996. Alkamides from Cissampelos glaberrima. Planta Medica 62, 376–377. Roy, P.K., Dutta, A.T., Ray, G.K., Mukerji, B., 1952. A preliminary note on the pharmacological action of the total alkaloids isolated from Cissampelos pareira Linn. (false Pareira brava). The Indian Journal of Medical Research 40, 95–99. Rukunga, G.M., Gathirwa, J.W., Omar, S.A., Muregi, F.W., Muthaura, C.N., Kirira, P.G., Mungai, G.M., Kofi-Tsekpo, W.M., 2009. Anti-plasmodial activity of the extracts of some Kenyan medicinal plants. Journal of Ethnopharmacology 121, 282–285. Samanta, J., Bhattacharya, S., 2011. Cissampelos pareira: a promising antifertility agent. International Journal of Research in Ayurveda and Pharmacy 2, 439–442. Samie, A., Obi, C.L., Bessong, P.O., Namrita, L., 2005. Activity profiles of fourteen selected medicinal plants from Rural Venda communities in South Africa against fifteen clinical bacterial species. African Journal of Biotechnology 4, 1443–1451. Samie, A., Obi, C.L., Lall, N., Meyer, J.J.M., 2009. In-vitro cytotoxicity and antimicrobial activities, against clinical isolates of Campylobacter species and Entamoeba histolytica, of local medicinal plants from the Venda region, in South Africa. Annals of Tropical Medicine and Parasitology 103, 159–170. Scholtz, M., 1896. Ueber Bebirin. Berichte der deutschen chemischen Gesellschaft 29, 2054–2058. Semwal, D.K., Badoni, R., Semwal, R., Kothiyal, S.K., Singh, G.J.P., Rawat, U., 2010. The Genus Stephania (Menispermaceae): chemical and pharmacological perspectives. Journal of Ethnopharmacology 132, 369–383. Semwal, D.K., Semwal, R.B., 2013. Ethnobotany, pharmacology and phytochemistry of the genus Phoebe (Lauraceae). Mini-Reviews in Organic Chemistry 10, 12–26. 1027 Sharma, J., Gairola, S., Gaur, R.D., Painuli, R.M., 2012. The treatment of jaundice with medicinal plants in indigenous communities of the sub-Himalayan region of Uttarakhand, India. Journal of Ethnopharmacology 143, 262–291. Sharma, P.K., Chauhan, N.S., Lal, B., 2004. Observations on the traditional phytotherapy among the inhabitants of Parvati valley in western Himalaya, India. Journal of Ethnopharmacology 92, 167–176. Shinwari, M.I., Khan, M.A., 1998. Multiple dimensions of ethnobotany and its present status in Pakistan. Hamdard Medicus 42, 5–10. Shukla, P., Shukla, P., Gopalakrishna, B., 2012. Investigation of in-vitro anthelmintic activity of Cissampelos pareira linn against Pheretima posthuma. International Journal of Pharmaceutical Sciences 3, 265–267. Siddiqui, M.B., Husain, W., 1994. Medicinal plants of wide use in India with special reference to Sitapur district (Uttar Pradesh). Fitoterapia 65, 3–6. Singh, A., Duggal, S., Singh, J., Katekhaye, S., 2010. An inside preview of ethnopharmacology of Cissampelos pareira Linn. International Journal of Biological Technology 1, 114–120. Singh, A., Ashish, S., Katekhaye, S., 2012. Rajpatha ethnomedicine of controversial origin. International Journal of Current Pharmaceutical Review and Research 3, 86–90. Singh, B.K., Pillai, K.K., Kohli, K., Haque, S.E., 2013. Effect of Cissampelos pareira root extract on isoproterenol-induced cardiac dysfunction. Journal of Natural Medicines 67, 51–60. Singh, V., Banyal, H.S., 2011. Antimalarial effect of Tinospora cordifolia (Willd.) Hook. f. & Thoms and Cissampelos pareira L. on Plasmodium berghei. Current Science 101, 1356–1358. Singh, V.K., Ali, Z.A., 1994. Folk medicines in primary health care: common plants used for the treatment of fevers in India. Fitoterapia 65, 68–74. Singthong, J., Cui, S.W., Ningsanond, S., Douglas Goff, H., 2004. Structural characterization, degree of esterification and some gelling properties of Krueo Ma Noy (Cissampelos pareira) pectin. Carbohydrate Polymers 58, 391–400. Singthong, J., Ningsanond, S., Cui, S.W., Douglas Goff, H., 2005. Extraction and physicochemical characterization of Krueo Ma Noy pectin. Food Hydrocolloids 19, 793–801. Spelman, K., Burns, J.J., Nichols, D., Winters, N., Ottersberg, S., Tenborg, M., 2006. Modulation of cytokine expression by traditional medicines: a review of herbal immunomodulators. Alternative Medicine Review 11, 128–150. Srivastava, G.S., 1956. Propagation of Cissampelos pareira L. by root cuttings. Science and Culture 21, 601–603. Steele, J.C.P., 2000. The Pharmacological Evaluation of Plants Used Traditionally for the Treatment of Malaria by Indigenous People of South America (Ph.D. thesis). University of London. Steele, J.C.P., Phelps, R.J., Simmonds, M.S.J., Warhurst, D.C., Meyer, D.J., 2002. Two novel assays for the detection of haemin-binding properties of antimalarials evaluated with compounds isolated from medicinal plants. Journal of Antimicrobial Chemotherapy 50, 25–31. Stepp, J.R., 2004. The role of weeds as sources of pharmaceuticals. Journal of Ethnopharmacology 92, 163–166. Sudhakaran, M.V., 2012. Histo-morphological, fluorescent and powder microscopic characterization of Cissampelos pareira Linn. Pharmacognosy Journal 34, 57–68. Sur, R.N., Pradhan, S.N., 1964. Studies on Cissampelos alkaloids. I. action of hayatin derivatives. Archives Internationales de Pharmacodynamie et de Therapie 152, 106–114. Surendran, S., Bavani Eswaran, M., Vijayakumar, M., Rao, C.V., 2011. in vitro and in vivo hepatoprotective activity of Cissampelos pareira against carbontetrachloride induced hepatic damage. Indian Journal of Experimental Biology 49, 939–945. Tamaio, N., Joffily, A., Braga, J.M.A., Rajput, K.S., 2010. Stem anatomy and pattern of secondary growth in some herbaceous vine species of Menispermaceae. Journal of the Torrey Botanical Society 137, 157–165. Taylor, L., 1996. Tropical plant database file for Abuta (Cissampelos pareira). Raintree. Available on: 〈http://www.rain-tree.com/abuta.htm〉 (accessed 14.03.14). Tempone, A.G., Treiger Borborema, S.E., De Andrade Jr, H.F., Gualda, De Amorim, Yogi, N.C., Salerno Carvalho, A., Bachiega, C., Lupo, D., Bonotto, F.N., Fischer, D.C.H., S.V., 2005. Antiprotozoal activity of Brazilian plant extracts from isoquinoline alkaloid-producing families. Phytomedicine 12, 382–390. The Plant List, 2013. Version 1.1. Published on the Internet; 〈http://www.theplant list.org/1.1/browse/A/Menispermaceae/Cissampelos/〉 (accessed 1st January). Thomas, G., Araujo, C.C., De Agra, F.M., De Diniz, F.F.M., Bachelet, M., Vargaftig, B.B., 1995. Preliminary studies on the hydroalcoholic extract of the root of Cissampelos sympodialis Eichl in guinea-pig tracheal strips and bronchoalveolar leucocytes. Phytotherapy Research 9, 473–477. Thomas, G., Araujo, C.C., Duarte, J.C., De Souza, D.P., 1997b. Bronchodilator activity of an aqueous fraction of an ethanol extract of the leaves of Cissampelos sympodialis Eichl. (Menispermaceae) in the guinea pig. Phytomedicine 4, 233–238. Thomas, G., Burns, F., Pyne, S., Pyne, N.J., 1997a. Characterization of an extract from the leaves of Cissampelos sympodialis Eichl. on the spontaneous tone of isolated trachea. Phytotherapy Research 11, 496–499. Thomas, G., Selak, M., Henson, P.M., 1999. Effects of the aqueous fraction of the ethanol extract of the leaves of Cissampelos sympodialis Eichl. in human neutrophils. Phytotherapy Research 13, 9–13. Thornber, C.W., 1970. Alkaloids of the Menispermaceae. Phytochemistry 9, 157–187. Thukham-mee, W., Wattanathorn, J., 2012. Evaluation of safety and protective effect of combined extract of Cissampelos pareira and Anethum graveolens (PM52) against age-related cognitive impairment. Evidence-based Complementary and Alternative Medicine (Article no. 674101). 1028 D.K. Semwal et al. / Journal of Ethnopharmacology 155 (2014) 1011–1028 Tiwari, K.C., Majumder, R., Bhattacharjee, S., 1982. Folklore information from Assam for family planning and birth control. International Journal of Crude Drug Research 20, 133–137. Tor-anyiin, T.A., Sha’ato, R., Oluma, H.O.A., 2003. Ethnobotanical survey of antimalarial medicinal plants amongst the Tiv people of Nigeria. Journal of Herbs, Spices and Medicinal Plants 10, 61–74. Torres, A.L., Barros, R., De Oliveira, J.V., 2001. Effects of plant aqueous extracts on the development of Plutella xylostella (L.) (Lepidoptera: Plutellidae). Neotropical Entomology 30, 151–156. Tripathi, V., Stanton, C., Anderson, F.W.J., 2013. Traditional preparations used as uterotonics in Sub-Saharan Africa and their pharmacologic effects. International Journal of Gynecology and Obstetrics 120, 16–22. Tshibangu, J.N., Chifundera, K., Kaminsky, R., Wright, A.D., Konig, G.M., 2002. Screening of African medicinal plants for antimicrobial and enzyme inhibitory activity. Journal of Ethnopharmacology 80, 25–35. Tshibangu, J.N., Wright, A.D., Konig, G.M., 2003. HPLC isolation of the anti-plasmodially active bisbenzylisoquinone alkaloids present in roots of Cissampelos mucronata. Phytochemical Analysis 14, 13–22. Van Wyk, B.E., 2008. A review of Khoi-San and Cape Dutch medical ethnobotany. Journal of Ethnopharmacology 119, 331–341. Van Wyk, B.E., Gericke, N., 2000. People’s Plants. A Guide to Useful Plants of Southern Africa. Briza Publications, Pretoria p. 124. Vardhanabhuti, B., Ikeda, S., 2006. Isolation and characterization of hydrocolloids from monoi (Cissampelos pareira) leaves. Food Hydrocolloids 20, 885–891. Vieira, G.C., De Lima, J.F., De Figueiredo, R.C.B.Q., Mascarenhas, S.R., Bezerra-Santos, C.R., Piuvezam, M.R., 2013. Inhaled Cissampelos sympodialis down-regulates airway allergic reaction by reducing lung CD3þ T cells. Phytotherapy Research 27, 916–925. Wiggers, A., 1840. Ueber das Pelosin. Justus Liebigs Annalen der Chemie 33, 81–92. Wilcox, M.L., Bodeker, G., Pittler, M.H., 2005. Clinical trials of traditional herbal medicines for treating malaria. Focus on Alternative and Complementary Therapies 10, 27–28. Zhang, Z.J., 2004. Therapeutic effects of herbal extracts and constituents in animal models of psychiatric disorders. Life Sciences 75, 1659–1699.