Academia.eduAcademia.edu
Iranian Journal of Basic Medical Sciences ijbms.mums.ac.ir The genus Cuscuta (Convolvolaceac): An updated review on indigenous uses, phytochemistry, and pharmacology Shazia Noureen 1, Sobia Noreen 1*, Shazia Akram Ghumman 2, Fozia Batool 1, Syed Nasir Abbas Bukhari 3 1 Department of Chemistry, University of Sargodha, Sargodha-40100, Pakistan College of Pharmacy, University of Sargodha, Sargodha-40100, Pakistan 3 Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Aljouf, Sakaka2014, Saudi Arabia 2 ARTICLE INFO ABSTRACT Article type: Review article Cuscuta, commonly known as dodder, is a genus of family convolvolaceace. Approximately 170 species of Cuscuta are extensively distributed in temperate and subtropical areas of the world. Species of this genus are widely used as essential constituents in functional foods and traditional medicinal systems. Various parts of many members of Cuscuta have been found efficacious against a variety of diseases. Phytochemical investigations have confirmed presence of biologically active moieties such as flavonoids, alkaloids, lignans, saponines, phenolics, tannins, and fatty acids. Pharmacological studies and traditional uses of these plants have proved that they are effective antibacterial, antioxidant, antiostioporotic, hepatoprotective, anti-inflammatory, antitumor, antipyretic, antihypertensive, analgesic, anti hair fall, and antisteriogenic agents. Article history: Received: Oct 23, 2018 Accepted: May 10, 2019 Keywords: Bioactive Cuscuta Folk medicines Pharmacological activities Phytochemicals ►Please cite this article as: Noureen Sh, Noreen S, Ghumman ShA, Batool F, Bukhari SNA. The genus Cuscuta (Convolvolaceac): An updated review on indigenous uses, phytochemistry, and pharmacology. Iran J Basic Med Sci 2019; 22:1225-1252. doi: 10.22038/ijbms.2019.35296.8407 Introduction Plant-based medicines are an integral part of virtually all cultures since immemorial times. The journey of information from prehistoric texts to various indigenous folklores and modern preparations have witnessed the presence of bioactive moieties with therapeutic potential in these herbs (1-4). The immense population of current allopathic products is embedded in nature. More than half of the clinically approved drugs in the world are either natural products or their modifications. Higher plants being an endless reservoir contribute above one fourth. The remarkable resurgence of interest in nature to explore pharmaceutical and nutraceutical agents is still marching towards new horizons (5-7). Ever growing consumption of natural products by local masses has forcefully motivated the scientists to acquire systematic, elaborated, and practical knowledge about their constituents by using advanced technologies (8). Herbal products, both as purified compounds and in the form of standard extracts, offer infinite odds for novel pharmaceutical products due to the matchless accessibility to different chemical species (9). Targetbased phytochemicals have transfigured the medicinal industry because these are not only directly utilized for treatment purposes but also act as leads and standard template for synthetics drugs (10-11). Therefore, modern scientific investigations are turning towards traditional medicines to look for new windows of opportunities giving rise to superior pharmacologically active agents against diseases (12). The genus Cuscuta L. commonly known as dodder is one of the essential herbal constituents of pharma foods and curative tonics that are frequently prescribed to nourish various body parts. It is used to enhance the nutritional value of porridge and alcoholic beverages (13). The genus has a rich history of folk medicinal uses, and numerous phytoconstituents of therapeutic value have been isolated and identified (14). Various species are indigenously used to cure fits, melancholy, insanity (15), fertility problems (16), tumors (17), scabies, eczema (18), chronic ulcer, jaundice, inflammation (19), chest pain (20), fever, itching (21), osteoporosis (22), diarrhea, oedema, stomach ache, infections, measles, sores, kidney problems (23), sprain (24), alleviation of high blood pressure, leucorrhoea (25), obesity (26), migraine, amnesia, epilepsy, and constipation (27). Pharmacological analysis of various Cuscuta species unveiled their antitumor, antimicrobial (28-31), hepatoprotective (32-33), anticonvulsant (34), immunostimulatory, antioxidant (14, 35-37), α-glucosidase inhibition (38), psychopharmacological (39), hair-growth promoting (40-41), anti-steroidogenic (42), anti-inflammatory (43-44), diuretic (45), analgesic (46), antipyretic (47-48), anti-HIV (49), antidiabetic (50), neuroprotective (51), antiulcer (52), antispasmodic, heamodynamic, bradycardia1, antihypertensive, cardiotonic, and muscle relaxant activities (53). Cuscuta species are rich in bioactive constituents that exhibit a wide variety of pharmacological activities. Presence of a good deal of valuable components, broad range of biological attributes and remedial value of these plants in folk medicinal systems gives stimulation toward the concept that this genus can play an important role in discovery of new and more efficient therapeutic agents. This review is an effort to edify knowledge of its phytochemical richness, pharmacological and biological significance, and folk medicinal uses, which will enhance its value as a potent pharmaceutical precursor. *Corresponding author: Sobia Noreen. Department of Chemistry, University of Sargodha, Sargodha, 40100, Pakistan. Tel: +923018434400; Email: sobianoreen@ uos.edu.pk Noureen et al. An overview of the genus Cuscuta Methods C. capitata Roxb (80). Cuscuta species are holophrastic, annual or perennial, herbaceous vines. The thread-like slender, twining stems have orange, red, or yellow color. Majority of the members have achlorophyllous, scaly leaves while some of them are with reduced synthetic apparatus and can perform localized and limited photosynthesis. Bisexual flowers in multiple colors like cream, yellow, white, and pink are pollinated by insects. Roots are absent, and haustoria are used to suck water and nutrients. Several morphological and physiological simplifications, for instance absence of cotyledons or radicles in their embryos, scaly leaves without vascular tissue and haustoria represent an adaptation to parasitism. They are obligate parasitic plants (54, 61, 81-84). These stem and leaf parasites depend entirely on their host plant, thus reducing the growth and yield of the host. They mostly infect many broadleaf crops, ornamentals plants, weeds, and a few monocot crops. Some of the species are strictly host-specific while others thrive on diverse hosts (85, 86). The usual growing season is early summer; germination starts in May, parasites invade the host by haustoria and may wither and die in the absence of a suitable host within two weeks (87). Flowering starts in June and seed production in November (88). This review on Cuscuta genus has been written according to the information collected from various scientific databases such as Scopus, Researchgate, Web of Science, ScienceDirect, and PubMed up to August 2018. Distribution and botanical description Cuscuta, a flowering parasitic genus was previously placed in the Convolvulaceae family, but later it was segregated as the separate family Cuscutaceae (54-57). Global distribution record indicates that most of the species are concentrated in tropical and subtropical areas and fewer in temperate regions. This parasitic genus is known by many common names such as dodder, gold-thread, hair-weed, devil’s hair, hell-vine, stranglevine, love-vine, pull-down, etc. in different regions of the world. The number of species documented by various authors varies from 100 to 170 (58-66). Medicinally important species are C. reflexa Roxb. (67), C. chinesis Lam. (68), C. japonica Choisy (69), C. australis R. Br. (70), C. europaea Linn. (71), C. gigantea Griff. (72), C. hyalina Roth. (73), C. campestris Yuncker. (47), C. racemosa Mart. (52), C. pedicellata Ledeb. (74), C. epithymum L. (75), C. kilimanjari Oliv. (76), C. kotschyana Boiss. (77), C. mitraeformis Engelm. (78), C. tinctoria Mart (79), and Table 1. Common names and global distribution of some medicinally important Cuscuta species Name C. reflexa Common name Distribution References Hell weed, devil's gut, beggar weed, Pakistan, India, China, E. Asia, Afghanistan, (27, 29, 89-90) strangle tare, scald weed, dodder of Bangladesh, thyme, greater dodder, lesser dodder C. chinesis Chinese dodder Ethiopia, Kazakhstan, Kyrgyzstan, Tajikistan, (68, 91) Turkmenistan, Uzbekistan, Mongolia; Russia, China, Iran, Iraq, Afghanistan, India, Sri Lanka, Indonesia, Korea, Japan, Taiwan, Thailand, Australasia, C. japonica Japanese dodder Korea (92-93) C. australis Australian dodder, Omonigelegele, Taiwan, Africa, Japan, Australia, Madagascar, (23, 70, 94-96) southern dodder Europe, Asia, Senegal, Ethopia, C. europaea ……………. India, Romania, Bulgaria, Iran (97-99) C.gigantea …………….. Pakistan, China, Afghanistan, Tajikistan. (62, 72) C. hyaline …………….. Pakistan, Ethiopia, Sudan, Kenya, Uganda, (100) Burundi, Rwanda, Zimbabwe,India,Botswana, Namibia, South Africa, C. planif'lora Small seed dodder, red dodder North Africa, Southwestern and southern Asia, (23, 101-102) Ethiopia, Madagascar, Angola C. campestris C. racemosa Field dodder, common dodder, prairie Saudi Arabia, Nigeria, South America, Europe, (81, 86, 103- dodder, yellow dodder, gewone dodder, Asia, Africa, Australia, Taiwan 105) Chilean dodder, lead-vine, golden Brazil, Chile (52, 106) Pakistan, Egypt, Qatar, Saudi Arabia, UAE, Iran (26, 99, 107- thread C. pedicellata Clover dodder 109) C. epithymum Common dodder, Pakistan, Ireland, Iran, Poland (95, 106, 110- Clover dodder, lesser dodder, flax 112) dodder C. kilimanjari Dodder Sudan, Etopia, Congo, Malawi, Zimbabwe, (23,95) Mozambique, Limpopo, Madagascar C. monogyna Eastern dodder Iran (113) C. approximata Alfalfa dodder Turkey, Iran (14, 114-115) Smooth seed alfalfa dodder C. kotschyana …………….. Iran, (99) C. capitata …………….. India, Nepal (80,116) C. mitraeformis .……………. México (78) C: Cuscuta 1226 Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta Noureen et al. Medicinal uses The local inhabitants of rural areas are aware of inherent properties of various plants. They preferentially use these herbs and their products to treat multiple types of diseases due to their handiness and low cost (117). Potentially useful plants have been acknowledged and sequentially conveyed throughout the centuries in all societies. Some of them are used through self-medication, while others are recommended by traditional healers (118). Plant utilization as medicine ranges from the direct administration of the leaves, seeds, barks, roots, and stems to the extracts and decoctions from different parts of the plants (119). Many Cuscuta species being rich sources of diverse phytochemicals are popular components of various folk medicinal systems. Cuscuta species are used in traditional medicine as a purgative, diaphoretic, anthelmintic, diuretic, and tonic as well as a treatment for itching and bilious disorders (120, 121). Seeds, stem, and whole plant are utilized as prescription to treat different types of ailments. Medicinal uses of several parts of Cuscuta members are given in Table 2. C. reflexa is a treasured medicinal herb and widely used in conventional medicinal system of various Asian countries including China, India, Bangladesh, and Thailand for treating multiple disorders (122). It is called a miracle therapeutic plant in the ethnobotany, and a wide array of chemical compounds has been isolated with diverse medicinal properties (123). C. reflexa whole plant is used to treat conjunctivitis, respiratory disorders, piles, ulcers, and stomach problems (124). The paste of whole plant mixed with latex Carica papaya causes abortions (125). In rural areas of India its juice is used against jaundice. Paste of plant is effective to Table 2. Traditional medicinal uses of some Cuscuta species Species Plant part Preparation Traditional use References WP* Paste Treatment of swollen testicles, gout and joint pain, (67, 125, 127- causes abortion, anti-rheumatic, analgesic 128, 132, 169- Infection treatment (149) C. reflexa 170) Maceration Infusion Anti-poisonous (142) Juice Antiseptic, useful in itching skin and jaundice (127, 171) Powder Anti-fertility agent, astringent, diaphoretic (136) Pills Anti-tuberculosis (89) Decoction Useful in skin disease, used for jaundice, cough, blood (171-172) --------- Antidiarrheal, anti-inflammatory, anti-ulcer, purgative, (124, 131, 144, antidandruff, conjunctivitis, analgesic, 150, 169, 173- hepatoprotective, useful in cough, cephalagia, fever, 175) purification, bronchitis, fever, sex stimulation leucorrhoea, and paralysis, respiratory disorders, piles, stomach problem, constipation, spleen diseases, helminthiasis, fracture joining Stem Decoction Hepatoprotective, antidiarrheal, useful in constipation, (144, 169) stomach disorders, urinary tract infections, jaundice, epilepsy, cholera, asthma Paste Anti-hair fall, anti-rheumatic, useful in skin diseases (29, 128, 144) Juice Jaundice treatment (126, 176) Crushed Blood purifier, purgative, good for brain, fever, (135, 138) anthrax in cattle Seeds --------- Effective in bilious disorders and fever Decoction Cause abortion (133-134) (144) --------- Carminative, anthelmintic, alterative, emmenagogue, (129, 170) sedative, diuretic, useful in ulcer, liver disorders Leaves C. chinensis Poultice Pain reliever (177) Extract Cold treatment (178) Juice Anti-hypertensive, anti-diarrheal, useful in jaundice. (179) --------- Effective in scabies, eczema, inducing sterility (18, 180) Fruits --------- Antipyretic, cough reliever (67) WP Juice Anti-ulcer, anti-inflammatory, wound healer, jaundice (19) treatment Seeds Dressing Useful in painful inflammations (151) Paste Anti-ulcer and wound healer (151) --------- Carminative, tonic, diuretic, sedative, (158) diaphoretic Stem Paste Joining fractures (155) --------- Expectorant, carminative, tonic, anthelmintic, (158) purgative, diaphoretic, anti-inflammatory, analgesic C. japonica Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Leaves --------- Antihypertensive (93) 1227 Noureen et al. An overview of the genus Cuscuta Continued Table 2. C. australis --------- --------- Laxative, anthelmintic, astringent, emollient, sedative, (23) sudorific, liver and kidney tonic, useful in sores and measles C. austrais seeds Decoction Brain tonic (181) C. europaea Sap --------- Carminative (71) WP Extract Anti-psoriasis (71) Juice Useful in skin diseases (167) --------- Laxative, diuretic, analgesic (116, 166) --------- Juice Antipoisnous (72, 164) --------- --------- Anti-septic (116) WP --------- Purgative, useful externally against itching and (21) Seed, vegetative pant C. gigantea C. hyalina internally in protracted fevers C. planif'lora WP Infusion Sores washers (21) --------- Abortion treatment (73) --------- Antiulcer, against culex mosquito, (23) --------- Carminative, laxative (130) Stem --------- Anti-diarrheal (23) C. campestris WP Decoction Purgative, useful in constipation, (105) C. racemosa --------- Anti-inflammatory, diuretic, effective in the stomach (52) poultice --------- and hepatic disorders and fresh wounds C. pedicellata --------- --------- Anti-obesity (26) Stem --------- Purgative, wound healer, anti-inflammatory, (168) WP --------- Diuretic, laxative, liver and kidney tonic, to treat antihypertensive, useful in Stomachache C. epithymum (163, 182) sciatica, scurvy and scrofula derma C. kilimanjari --------- Astringent, Laxative, detersive (75) Extract Scleroderma treatment (162) Stem --------- Useful in epilepsy (183) Stem Sap Useful inear, nose and throat diseases (76) --------- Effective in stomach ache, edema, veterinary (23) WP C. capitata treatment, agalactia --------- Sap Treatment of ringworm and warts (79) WP Powder Reduces irritation of bladder and improves urinary (80) function C. approximata C: Cuscuta; *Whole plant Useful in kidney problems (116) WP --------- Laxative, carminative, hepatoprotective (130) --------- --------- Useful in sin disease (116) treat headache, gout, and rheumatism (67, 126-128). Plant juice mixed with other decoctions is purgative. Seeds of C. reflexa are carminative, anthelmintic, alterative, emmenagogue, sedative, and diuretic. It is effective against warts (116, 129). Leaves are used to treat eczema, scabies, cold, and to induce sterility (18, 130). Rabha tribes of west Bengal use the whole plant to treat leucorrhoea (131). It is applied internally to cure protracted fevers and externally on itchy skin. The plant is frequently used in Ayurvedic medicine to give relief in urinating difficulties, muscle pain, and coughs (132, 133). Pills prepared from the dried plant are used for treatment of tuberculosis (89). Its stem is a blood purifier, good for brain and fever (134-135). Tribal people use its various parts to treat fits, insanity, melancholy, and to control fertility (15). It is commonly used in veterinary medicines as poultice and sprains. The powder is used as astringent and diaphoretic for cattle (136-137). 1228 C. reflexa stems are crushed with Clerodendrum viscosum leaves and fed to cattle to treat anthrax (138). The plant is used for skin infections and dandruff (139140). The paste of whole plant with Achyranthes aspera is used to control excessive bleeding during menstruation (141). It is also used for treatment of bone fracture and body pain (142). In folk medicine of Bangladesh, it is used to cure tumors (17). The Tripura community of Bangladesh and Satar tribes in Nepal use this plant to cure edema, body ache and for maintenance of liver function. It is used for treating constipation, spleen diseases, diarrhea, and inflammation. Paste mixed with sesame oil is applied for curing hair fall. The decoction of stem is used to cure diarrhea, cholera, and asthma, while decoction of seeds causes depression, nausea, and vomiting (29, 143-145). Whole plant powder is used to treat jaundice by tribal people of nallamalais in Andhra Pradesh (146). It is also used as expectorant, aphrodisiac, is useful Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta in vomiting, and purifies the blood (32). C. reflexa is an essential constituent of several medical compositions, which are used in the treatment of migraine, headache, chronic catarrh, epilepsy, amnesia, and to prolong fever (27, 147-148). Maceration of whole plant is used to treat infections (149). The whole plant is also useful in cephalagia, paralysis, stomach pain and helminthiasis (89, 150). C. chinesis Lam. also known as Chinese dodder or TuSi-Zi, also has a wide range of uses. It has been mentioned in various old Chinese scripts and recommended by many herbal practitioners (68). Besides China it is also a famous prescription in many other countries. In Pakistan dressing made of plant is used on painful inflammations. Moreover, paste is useful for chronic ulcers and wounds (151). In traditional Indian system, leaves and stems are used to enhance lactation (152). In Vietnam people use whole plant in back pain and constipation (153). In Korea, seeds with other herbal prescriptions are effective to improve sexual function and health (154). Stem paste of C. chinensis is applied to fractured bone to promote the joining (155). Whole plant juice is used to treat inflammation and jaundice (19, 156). A lotion prepared from stem is used to treat sore heads and inflamed eyes. It has been found useful in the treatment of impotence, nocturnal emissions, dizziness, lumbago, leucorrhoea, decreased eyesight, abortion, and chronic diarrhea (133). C. chinensis is used in treatment of mania, epilepsy, and insanity (157). Its stem and seeds are considered tonic, expectorant, purgative, sedative, diuretic, diaphoretic, carminative, anthelmintic, and advantageous in muscles and joints pain (158-159). Prescriptions containing C. chinensis are used to treat impairment of sexual function, cure cardiovascular diseases and osteoporosis, treatment of premature ejaculation, to treat lower abdominal and back pain, infertility, wet dreams, impotence, urinary retention, and urinary incontinence (68). It is also used to cure melisma, freckles and considered as antidandruff agent (160-161). C. epithymum is a mild diuretic and used to treat sciatica and scurvy. The fresh plant is applied to the skin against scrofula derma and scleroderma. It is associated with the health of liver and kidneys and used in various formulas. It is considered a mild laxative (162-163). The whole plant is dried and used as astringent and detersive (75). Whole plant decoction of C. campestris is used as purgative and poultice (105). The sap of C. tinctoria is used to cure ringworm and warts (79). Juice of C. gigantea plant is famous as an anti-poisonous agent (140, 164). The sap of C. europaea is used as a carminative, and the extract is applied to treat psoriasis (165). Seeds and vegetative parasitic plant is used as laxative, diuretic, and pain reliever and is poisonous. The juice is used for skin treatment (166-167). C. capitata whole plant reduces irritation of bladder and improves urinary function (80). C. hyaline is used to treat chest pain (20, 24). Its infusion is used as sores washer and to prevent abortion (21, 73). It is antiulcer and used against culex mosquito. C. australis is used as laxative, anthelmintic, astringent, for treatment of sores, measles and as kidney and liver tonic, emollient, sedative, and sudorific (23). Leaves of C. japonica are considered antihypertensive Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. (93). The sap of C. kilimanjari collected from stems is directly installed to treat ear, nose, and throat diseases in central Kenya. The whole plant is used to treat stomach ache, edema, agalactia, and in veterinary medicines (23, 76). C. pedicellate is used for treatment of obesity, stomachache, to cure wounds, hypertension, as purgative, and anti-inflammatory agent (26, 168). The whole plant of C. planiflora is carminative and laxative, and the stem is anti-diarrheal (23, 130). C. racemosa has anti-inflammatory and diuretic effects, is also used for stomach and hepatic complaints and treatment of fresh wounds (52). Phytochemistry Exploration of nature’s garden of medication to expose more acceptable solutions with safety is a subject of interest from prehistoric era as more than half of world population still relies on medicinal plants to sustain life. The capability of these odds to appease and treat various diseases and infirmity is undoubted. The curative plants are extensively used in pharmaceuticals, food industry mostly as functional food, agricultural, and cosmetics. Various herbs, their extracts, and prescriptions are loaded with different biologically active constituents particularly alkaloids, steroids, saponins, flavonoids, and terpenoids that are responsible for their therapeutic outcomes (27, 184-189). Phytochemical screening of ever more medicinal plants is extremely momentous in detecting and identifying innovative sources of healing as well as commercially important compounds (190). Genus Cuscuta is rich in many phytoconstituents representing a varied spectrum of secondary metabolites including flavonoids, alkaloids, lignans, polysaccharides, steroids, volatile oils, and resin glycosides (191-199). In a comparative study it was suggested that the plants in the Cuscuta species are blessed with almost same soluble phenolic secondary metabolites as Chlorogenic acid,3,5-dicaffeoylquinic acid, 4,5-dicaffeoylquinic acid, hyperoside, quercetin, astragalin, kaempferol-3O-galactoside, and quercetin-3-O-glucoside but with varying quantities (200). Chemical constituents of Cuscuta species are hostdependent. For instance, a large number of alkaloids identified in these parasitic plants are the same as those found in their alkaloid containing hosts except a very few (201). These species can synthesize flavonoids, while the study of relation between flavonoids of host and parasite is under consideration. Preliminary determination indicates that flavonoid content of various Cuscuta samples growing on different hosts is quite different (202). The most thoroughly characterized species of this genus are C. reflexa and C. chinensis (6768, 203). Essential component of many medicinal compositions of C. reflexa has an extensively varied array of phytochemicals identified as phenolic compounds, flavonoids, alkaloids, phytosterols, amarbelin, betasterol, stigmasterol, glycosides, saponins, cuscutine, myricetin, dulcitol, coumarin, cuscutamine, luteolin, bergenin, proteins, fixed oils, fats, and carbohydrates (27, 67, 204). This genus is a source of many novel metabolites. Qualitative analysis of methanolic extract of C. reflexa isolated two new compounds named as 7’-(3’,4’-dihydroxyphenyl)-N-[(4-methoxyphenyl)ethyl] 1229 Noureen et al. An overview of the genus Cuscuta glucopyranoside), cuscutoside C (2′-hydroxyl asarinin 2′-O-β-D-glucopyranoside), cuscutoside D (2′-hydroxyl a s a r i n i n 2 ′ - O - β - D - a p i o f u ra n o s y l - ( 1 → 2 ) - [ β - D glucopyranosyl-(1 → 6)]-β-D-glucopyranoside) and neosesamin (188, 193, 210). C. chinensis and C. australis are used to prepare the famous Chinese herbal prescription Tu-Si-Zi. Phytochemical analysis was done to compare the phenolic constituents of both plants. Principal compounds of C. australis were kaempferol and astragalin while hyperoside was predominant in C. chinensis (211). Several Phytoestrogens were isolated and identified from C. chinensis. Ethanolic extract of seeds afforded three new lignans named cuscutaresinols A−C (212). In another investigative study, four new glycosidic acids called cuscutic acids A-D were isolated from the alkaline hydrolysate of the ether-insoluble resin glycoside (191). Up till now bulk of the phytochemical investigations on C. chinensis targeted the seeds while other parts of the plant have had much less attention by the researchers. An ether insoluble resin glycoside fraction was separated from seeds of C. australis and identification and characterization of resin matrix revealed the presence of three new glycosidic acids, cuscutic acids A1−A3 (213). C. racemosa like other species of the genus offers flavonoids as the chief constituent along with tannins. In another experiment alkaloids, flavonoids, tannins, and saponins have been identified (52, 214). propenamide and 7’-(4’-hydroxy,3’-methoxyphenyl)-N[(4-butylphenyl)ethyl]propenamide (38). From aerial parts of same plant two novel tetrahydrofuran derivatives, namely Swarnalin and Cis-swarnelin were separated (205) while a flavanon, reflexin chemically named as 5-hydroxy-7-methoxy-6-(2,3-epoxy-3-methylbutyl)flavanone, was isolated from the stem (206). Moreover, 3’-methoxy-3,4’,5,7-tetrahydroxy flavone and 3’-methoxy4’,5,7-trihydroxy flavone-3-glucoside were isolated from whole plant (207). An antiviral protein with molecular weight about 14,000–18,000 Daltons was separated and evaluated against several isometric and anisometric viruses (208). Phytochemical investigations of C. chinensis have shown that flavonoids, alkaloids, poly-saccharides, steroids, lignans, and volatile oils are mostly reported in its various parts (68). The active moieties responsible for various pharmacological activities of the C. chinensis mostly include flavonoids, lignans, quinic acid, and polysaccharide. Flavonoids are the prime biologically active components in C. chinesis. Additionally, quercetin, kaempferol, and hyperoside can serve as an index to evaluate the quality of the crude drug (209). C. chinensis extract afforded four new lignans cuscutoside A (2′-hydroxyl asarinin 2′-O-β-Dapiofuranosyl-(1→2)-β-D-glucopyranoside), cuscutoside B (2′-hydroxyl asarinin 2′-O-β-xylopyranosyl-(1→6)-βTable 3. Phytochemical profile of various Cuscuta species Name Plant part Solvent Extraction Separation Phytochemicals References 7’-(3’,4’-dihydroxyphenyl)-N-[(4- (38) technique C. reflexa WP MeOH Maceration CC methoxyphenyl)ethyl]propenamide 7’-(4’-hydroxy,3’-methoxyphenyl)-N-[(4butylphenyl)ethyl]propenamide 6,7-dimethoxy-2H-1-benzopyran-2-one 2-(3-hydroxy-4-methoxyphenyl)-3,5dihydroxy-7-O-β-D- glucopyranoside-4H-1benzopyrane-4-one, 3-(3,4-dihydroxyphenyl)-2-propen-1ethanoate 6,7,8-trimethoxy-2H-1-benzopyran-2-one 3-(4-O- β-D-glucopyranoside-3, dimethoxyphenyl) -2-propen-1-ol -------- -------- HPLC Kaempferol (215) Quercetin Lupeol ß-sitosterol Aq. EtOH Soxhlet TLC Gallic acid (53) Quarcetin EtOH -------- VLC Odoroside H (216) 21-hydroxyodoroside H Neritaloside Strospeside 16-_-hydroxydigitoxin N-trans and cis feruloyl tyramines Ethyl caffeate Coumarins Ursolic acid _-sitosterol Glucoside 4-O-p-coumaroyl-_-D-glucoside n-hex Soxhlet GC-MS Heneicosanoic acid (217) Pentadecanoic acid Hexadecenoic acid Heptadecanoic acid 1230 Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 EA tion MS An overview of the genus Cuscuta Noureen et al. Continued Table 3. Pentadecanoic acid Hexadecenoic acid Heptadecanoic acid Octadecanoic acid Stem EA Maceration GC-MS 1, 2, 3 Propanetriol, 1- acetate, (218) Benzofuran 2, 3, dihydro Glycerol 1, 2- diacetate 1 H- 1, 2, 4-triazol-5-amine 1- ethyl2-methoxy-4-vinylphenol Triacetin D - glucitol, 4 - O-hexyl 3,4,5-trimethoxy cinnamic acid Hexadecanoic acid, ethyl ester 3,6 -di methoxy phenanthrene 3, 5 - di - tert -Butyl -4 -hydroxyanisol Vanillin 3 – aminopyrrolidine Cetene Sarcosine, N -isobutyryl, tetradecyl ester 4 - ((1E) – 3 – hydroxyl -1-propenyl)-2methoxy phenol 1,5-diphenyl-2H-1,2,4-triazoline-3-thione 1-octadecene Heptanamide, N-(1-cyclohexylethyl)-2methyl Scoparone Hexadecanoic acid, ethyl ester 3’-Methyl-2-benzylidene-coumaran-3-one Pet Eth Soxhlet CC 5-hydroxy-7-methoxy-6-(2,3-epoxy-3- (206) methylbutyl)-flavanone (reflexin) Isorhamnetin (122) Isorhamnetin-3-O-glucoside Isorhamnetin-3-O-robinobioside MeOH Maceration GC-MS 2-Methoxy-4-vinyl phenol (219) Benzofuran-2,3-dihydro 3,5-di-tert-Butyl-4-hydroxyanisole Hexatriacontane n-Hexadecanoic acid Scoparone Hexadecanoic acid methyl ester 1,3-Benzenediamine, N, N, N′, N′ tetramethyl- Phenol, 4(3-hydroxy1propenyl), 2-methoxy Phenol, 2,4 bis (1,1dimethylethyl); 2,3,5,6Tetramethyl para phenylene diamine Retinoic acid-5,6-epoxy-5,6-dihydro 2,4-Dihydroxy2,5-dimethyl-3(2H) furan-3-one 2,3-dihydro-3,5-dihydroxy-6-methyl-2Propyl-tetrahydro-pyran-3-ol Pregn-4-ene-18-oic acid AP MeOH Maceration RHPLC Swarnalin HPLC Cis-swarnelin (205) Coumarin 5, 6, 7-trimethoxycoumarin ------ Water -------- --------- Aromadendrin (49) Taxifolin Aromadendrin-7-0-β-D-glucopyranoside 3,5,7,8,4'-pentahydroxyflavanone Taxifolin-7-0-β-D-glucopyranoside Coccinoside B Pruning 3-O-dicaffeoyl quinic acid 3-4-O-dicaffeoyl quinic acid 3, 4, 5-O-Tricaffeoylquinic acid ------- DCM Maceration HPLC Violaxanthin (220) Lutein Lycopene β, ψ-carotene Rubixanthin β, β – carotene Esterified rubixanthin Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 ater 0% eOH tion β ight ----------Cuscutoside A (2′ hydroxyl asarinin 2′ O-β- 1231 Noureen et al. An overview of the genus Cuscuta Continued Table 3. β, ψ Rubixanthin β, β – carotene Esterified rubixanthin Lutein violaxanthin Fil. Water Maceration β-cryptoxanthin CC An antiviral protein with molecular weight (219) about 14,000---18,000 daltons C. chinesis Fruit 50 % ----------- CC Cuscutamine (194) Cuscutoside A (2′-hydroxyl asarinin 2′-O-β- MeOH D-apiofuranosyl-(1 → 2)-β-Dglucopyranoside Cuscutoside B (2′-hydroxyl asarinin 2′-O-βxylopyranosyl-(1 → 6)-β-glucopyranoside Hyperoside Astragalin Quercetin Quercetin-3-O-apiosyl (1-2)-galactoside Pinoresinol-4-O-glucoside Einoresinol Epipinoresinol) p-coumaric acid Caffeic acid Chlorogenic acid Arbutin Stem Pet. eth -------- β-sitosterol --------- CF (221) d-sesamin 9(R) - hydroxy-d-sesamin D-pinoresinol daucosterol Seed Pet. eth Reflux Cuscutoside C (2′-hydroxyl asarinin 2′-O-β- CC (196) D-glucopyranoside) Cuscutoside D (2′-hydroxyl asarinin 2′-O-βD-apiofuranosyl-(1 → 2)-[β-Dglucopyranosyl-(1 → 6)]-β-D- glucopyranoside --------- -------- --------- Neo-sesamin (210) Kaempferol Kaempferol-3-O-β-D-glucopyranoside 4', 4, 6-trihydroxyaurone Quercetin Hyperoside Palmitic acid Stearic acid β-sitosterol Daucosterol --------- -------- RHPLC quercetin 3-O-β-D-galactoside-7-O-β-D- (218) glucoside quercetin 3-O- β-D-apiofuranosyl-(1etin3Dgalactoside hyperoside quercetin kaempferol Ether Saponi- Water fication CC A trisaccharide (194) Four new glycosidic acids (cuscutic acids AD) Acetic acid Propionic acid 2-methylbutyric acid Tiglic acid Nilic acid Convolvulinolic acid Jalapinolic acid 95 % -------- Cuscutaresinols A−C CC EtOH (212) (+)-sesamin (+)-xanthoxylol 9-hydroxysesamin (+)-pinoresinol Kaempferol Isorhamnetin 5% 1232 -------- H hex Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 -------- GC Cuscutaresinols A−C An overview of the genus Cuscuta Noureen et al. Continued Table 3. (+)-pinoresinol Kaempferol Isorhamnetin 95 % -------- CC Kaempferol EtOH (22) Quercetin astragalin, isorhamnetin hyperoside n-hex -------- Capillary GC Sixteen fatty acids including (222) Palmitic acid Linoleic acid Oleic acid Linolenic acid MeOH -------- --------- Methyl 4-hydroxy-3,5dimethoxycinnamate, (223) Caffeic acid Quercetin Kaempferol Calycopteretin EtOH -------- CC neocuscutosides (224) A, B and C ------ --------- -------- --------- Octadecyl (E)-p-coumarate (225) Methyl 3-O-β-D-glucopyranosyl-5hydroxycinnamate Quercetin-3-O-(6″-galloyl) β-D-glucoside Kaempferol Astragalin Hyperoside Astragalin 6″-O-gallate β-sitosterol Daucosterol C. japonica Seed MeOH -------- FCC 3, 5-Di-O-caffeoylquinic acid (226) 3, 4-Di-O-caffeoylquinic acid Methyl 3, 5-Di-O-caffeoylquinate Methyl 3, 4-Di-O-caffeoylquinate C. australis Stem 80 % -------- CC acetone α-caroten-5 (227) 6-epoxide β-and γ-carotene Xanthophylls Taraxanthin Lutein Kaempferol Seed --------- -------- GC Cuscutic acids A1−A3 (217) Acetic acid Isobutyric acid 2-methylbutyric acid Tiglic acid Nilic (3-hydroxy-2-methylbutyric) acid --------- -------- --------- β –sitosterol (228-229) Sesamin Hexadecanoic acid Hexadecanoic acid Kaempferol Quercetin Astragloside Hyperoside caffeic acid Quercetin-3-O-β-D-galactopyranosyl-β-Dapiopyranoside C. europaea ------ --------- -------- --------- Glycoside (166) Flavonoids C. campestris AP MeOH Maceration HPLC Sinapic acid (14) Quercetin Hesperidin Eugenol C. racemosa WP 70 % Percolation TLC Flavonoids EtOH (214) Tannins Flavonol (4’methoxyquercetin) eOH d Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 1233 α β --------- -------- --------- er Noureen et al. An overview of the genus Cuscuta Continued Table 3. EtOH Tannins Flavonol (4’methoxyquercetin) MeOH Socked DCCC Kaempherol (230) Quercetin Pinoresinol 9-α-hydroxysesamin 9-β-hydroxysesamin Acuminatolide C. pedicellata WP --------- -------- --------- EtOH -------- CC Quercetin 5, 7, 3’, 4’-tetramethyl ether (231) Naringenin (26) Kaempferol Aromadenderin Quercitin 3,5,7,30,50-pentahydroxy flavanone, Naringenin -7-O-b-D-glucoside Aromadenderin -7-O-b-D-glucoside, Taxifolin -7-O-b-D-glucoside, Kaempferol -3-O-b- D-glucoside Quercitin -3-O-b-D-glucoside Seed Pet. eth Soxhlet CC Quercetin (232) Kaempferol Genkwanin Astragalin Palmitic acid C. epithymum WP MeOH Soxhlet --------- Alkaloids (182) Carbohydrates Flavonoids Glycosides Phytosterols Triterpenoids C. approximata AP MeOH Maceration HPLC Gallic acid (14) Catechin Caffeic acid Chloregenic acid, Quercetin Coumarin, Vanilin, Eugenol C. monogyna AP MeOH Maceration HPLC Sinapic acid (14) Catechin Caffeic acid Chloregenic acid Rutin Coumarin Vanilin Hesperidin Ellagic acid C. mitraeformis Stem n-hex -------- GC-FID Nonanal GC-MS Thymol (78) HPLC-DAD Eugenol β- carotene -------- Quercetin Lutein C. kotschyana ------ -------- -------- (233) kaempferol C: Cuscuta; WP: whole plant; AP: aerial parts; Fil: filament; Aq: aqueous; MeOH: methanol; EtOH: ethanol; Pet. eth: petroleum ether; n-hex: n-hexan; EA: ethyl acetate; DMC: dichloromethane; CC: column chromatography; HPLC: high performance liquid chromatography; RHPLC: reverse phase high performance liquid chromatography; TLC: thin layer chromatography; VLC: vacuum liquid chromatography; GC-MS: gas chromatographymass spectrometry; FCC: Flash Column Chromatography; DCCC: Droplet counter-current chromatography; FID: flame ionization detector; DAD: diode array detector Pharmacological attributes Impressive medicinal background of Cuscuta species has attracted the attention of many pharmacological researchers. A good deal of biological attributes has been studied and is listed in tabular form in Table 4. Antioxidant Medicinally important plants are endless reservoirs of antioxidants that enhance the antioxidant capacity of 1234 the body, which lead to a reduced risk of many diseases (234-235). Although a diverse population of synthetic analogs is commercially available due to side effects (liver impairment and carcinogenesis) blind reliance on these formulations has been over. Therefore, plants can play a key role to fulfill prerequisite for exploration of effective, biocompatible, and economic antioxidants (236). Many investigators have employed different Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta Noureen et al. qualitative and quantitative approaches to detect antioxidants in various Cuscuta species. Stem collected from different hosts and extracted with various solvents (100% methanol, 80% methanol, 100% ethanol, 80% ethanol, water, and n-hexane) were analyzed for quantity of phenolics and flavonoids content. Their antioxidant capacity was measured by using a variety of assays including reducing power, DPPH scavenging activity, percent inhibition of linoleic acid peroxidation and δ-tocopherol. It was observed that there was a strong correlation between amount of total phenolics and antioxidant capacity (13). C. reflexa has been reported for its antioxidant potential (37, 237). Free radical scavenging capacity of methanolic extract of C. reflexa was evaluated by DPPH and reducing power assays. Results of DPPH assay, illustrated as IC50 value demonstrated its antioxidant activity 359.48 μg/ml as compared to 9.22 μg/ml value for ascorbic acid used as standard. The reducing power of extract was found dose-dependent and increased by increasing concentration (35). Ethyl acetate fraction of ethanolic extract of C. reflexa was significantly antioxidant. Activity may be related to presence of flavonoids, alpha tocopherol, and rutin, which were confirmed in preliminary phytochemical screening (238). Table 4. Pharmacological attributes exhibited by Cuscuta species Species Activity Plant Method Extract type Test applied Testing model Effective part C. reflexa Antioxidant St Reference dose/conc. Soxhlet L MeOH DPPH and FRAP assay ------------- 600 μg/ml (35) EtOH Non-Enzymatic Glycolysation Hemoglobin ------------ (238) of Haemoglobin Antibacterial Fl ---------- MeOH DPPH assay ------------- ------------ (323) L Soxhlet 50% EtOH Disc diffusion Escherichia coli ------------ (29) method Staphylococcus aureus Cup plate method Staphylococcus aureus 125 μg/ml (259) Bacillus subtilis 16 to 512 (235) Staphylococcus lutea μg/ml St ---------- MeOH Escherichia coli Bacillus punilus Salmonella typhi Salmonella thyphimurium Salmonella boydii Salmonella sonnei Salmonella dysenteriae Pseudomonas aeruginosa Klebsiella pneumoniae Vibrio cholerae WP ---------- DCM pet. eth Disc diffusion method Xanthomonas campestris Escherichia coli Klebsiella pneumoniae Proteus vulgaris Proteus denitrificans Soaked EtOH Agar well diffusion assay Bacillus subtilis Staphylococcus aureus 500 μg/ml (324) ----------- (260) ----------- (29) 30% (w/v) (107) 0.1 ml bolus (283) Escherichia coli Salmonella typhi ------- ---------- MeOH Agar well diffusion Staphylococcus epidermidis, Staphylococcus aureus Escherichia coli Pseudomonas sp. Klebsiella pneumoniae Antifungal L soxhlet 50% EtOH ------------------ Aspergillus niger Candida albicans ------- ---------- Water well diffusion method Aspergillus alternate Aspergillus niger Fusarium solani Fusarium oxysporium Macrophomina phaseolina Antihypertensive WP Socked EtOH ----------------- Wistar rats injection Psychopharmaco St Soxhlet Pet. eth logical effect Anti- General and exploratory Swiss albino mice ----------- (39) behavior study St Suc. Ex MeOH Pet. eth Membrane stabilizing activity Red blood cells ----------- (44) Soxhlet EtOH water Percentage volume reduction Albino rats 200, 400 (254) inflammatory mg/kg ivity WP c. AP ---------- er ell ---- 253) kg 5) ters kg 242) ters ---- 2) g/kg 5) lyte r Iran J Basic Med Sci, Vol. 22, No. 11, Nov ective2019WP Ex AP hlet ol WP Ex CF ntic 1235 Noureen et al. An overview of the genus Cuscuta Continued Table 4. Soxhlet EtOH water Percentage volume reduction Albino rats 200, 400 (254) mg/kg WP Decoc. Water SQ-RT-PCR analysis Murine macrophage cell -------------- (253) Wister rats 300 mg/kg (45) line RAW264.7 Diuretic activity AP Hepatoprotective Antitumor/antic ---------- EtOH Urine volume and electrolyte water content WP Suc. Ex Aq. Biochemical parameters Albino rats 200 mg/kg (242) AP Soxhlet Methanol Biochemical parameters Albino rats -------------- (32) WP Suc. Ex MeOH CF ------------------ Swiss albino mice 40 mg/kg (15) ------------- (253) Hep 3B cell line ------------- (325) Swiss albino mice ------------- (42) ---------------------- 2% extract in (40) ancer MCF-7 cancer cell line Decoc. Water MTT assay Human hepatocellular DAPI staining carcinoma cell line Hep3B Annexin V staining SQ-RT-PCR analysis ------- ---------- CF Annexin V-FITC Apoptotic assay PARP cleavage Caspase activation Antisteroidogenic St Soxhlet MeOH Ovary and uterus weight Biochemical parameters Hair growth St Soxhlet Pet. eth Visual observation Skin biopsy vehicle Histopathological Swiss albino rats 250 mg/kg (313) Long Evans rats and Swiss 50-200 mg/kg (50) albino mice bw observation Antidiabetic St Macera. MeOH Oral glucose tolerance test CF Antimutagenic AP Macera. MeOH water Oral glucose tolerance test Swiss albino rats 400 mg/kg (245) St Soxhlet MeOH Ames test Salmonella typhimurium ------------- (122) TA 98 and TA 100 Anthelmintic WP ---------- Pet. eth CF ------------------ Pheritima posthuma 20-50 mg/ml (44) Elevated plus-maze Swiss albino mice 400 mg /kg (305) MeOH Anxiolytic effect WP Macera. MeOH Light and dark chamber Anti-arthritic St ---------- 70% MeOH Percentage inhibition of Sprague–Dawley rats 600 mg/kg (321) Sprague–Dawley rats 600 mg/kg (321) Albino mice 200 and 400 (238) oedema Percentage inhibition of protein denaturation Nephroprotective St ---------- 70% MeOH Biochemical parameters and pathological syptoms Anticonvulsant L Macera. EOH Delay the onset of convulsions Genotoxic effects ------- ---------- MeOH mg/kg Root growth, root apical Allium cepa L. meristem mitotic index (MI), Allium sativum L. ------------- (326) chromosomal aberrations C. chinensis anti-histaminic ------ --------- EtOH Albino rats 100 mg/kg (327) Anticancer WP Soaked Water Histological study Swiss albino mice 1 g/kg (30 Neuronal Sd Percola. MeOH Neurite assay Rat pheochromocytoma 200 mg/l (277) ICR mice 200 µg (272) Wistar-albino rats 125 and 250 (33) differentiation Adjuvant effect PC12 cells Sd ---------- 70% EtOH Splenocyte proliferation assay Indirect ELISA Hepatoprotective Sd Decoc. EtOH Liver function markers and histopathological study Antioxidant Sd Decoc. EtOH Antioxidant enzyme levels mg/kg Wistar-albino rats 125 and 250 (33) mg/kg Antiosteoporotic Sd ---------- 95% EtOH Alkaline phosphatases activity UMR-106 cells ------------- (22) Alamar-Blue cell proliferation assay Reporter assays Improve erectile Sd ---------- ---------- Radioimmunoassay New Zealand white rabbits 1-5 mg/ml (288) Sd ---------- 80% EtOH Griess assay Mouse microglia line BV-2 ------------- (255) ELIZA cells dysfunction Antiinflammatory Anti-apoptosis Sd ---------- 95% EtOH Annexin V-FITC method SD rats ------------- (303) Effect on Sd Hot Ex EtOH water Melanin contents and B16F10 mouse melanoma ------------- (160) tyrosinase activity cells Methyl tetrazolium bromide Human Acute 3 µg/ml in 24 (267) test Lymphoblastic Leukemia hr Melanogenesis Zebrafish Cytotoxic 1236 WP ---------- sive d ---------- sis d t Ex ---------- ml Vol. 22, 226)No. 11, Nov 2019 Iran J Basic Med Sci, er ay oma --- 9) An overview of the genus Cuscuta Noureen et al. Continued Table 4. Light and dark chamber Anti-arthritic St ---------- 70% MeOH Percentage inhibition of Sprague–Dawley rats 600 mg/kg (321) Sprague–Dawley rats 600 mg/kg (321) Albino mice 200 and 400 (238) oedema Percentage inhibition of protein denaturation Nephroprotective St ---------- 70% MeOH Biochemical parameters and pathological syptoms Anticonvulsant L Macera. EOH Delay the onset of convulsions Genotoxic effects ------- ---------- MeOH mg/kg Root growth, root apical Allium cepa L. meristem mitotic index (MI), Allium sativum L. ------------- (326) chromosomal aberrations C. chinensis anti-histaminic ------ --------- EtOH Albino rats 100 mg/kg (327) Anticancer WP Soaked Water Histological study Swiss albino mice 1 g/kg (30 Neuronal Sd Percola. MeOH Neurite assay Rat pheochromocytoma 200 mg/l (277) ICR mice 200 µg (272) Wistar-albino rats 125 and 250 (33) differentiation Adjuvant effect PC12 cells Sd ---------- 70% EtOH Splenocyte proliferation assay Indirect ELISA Hepatoprotective Sd Decoc. EtOH Liver function markers and histopathological study Antioxidant Sd Decoc. EtOH Antioxidant enzyme levels mg/kg Wistar-albino rats 125 and 250 (33) mg/kg Antiosteoporotic Sd ---------- 95% EtOH Alkaline phosphatases activity UMR-106 cells ------------- (22) Alamar-Blue cell proliferation assay Reporter assays Improve erectile Sd ---------- ---------- Radioimmunoassay New Zealand white rabbits 1-5 mg/ml (288) Sd ---------- 80% EtOH Griess assay Mouse microglia line BV-2 ------------- (255) ELIZA cells dysfunction Antiinflammatory Anti-apoptosis Sd ---------- 95% EtOH Annexin V-FITC method SD rats ------------- (303) Effect on Sd Hot Ex EtOH water Melanin contents and B16F10 mouse melanoma ------------- (160) tyrosinase activity cells Methyl tetrazolium bromide Human Acute 3 µg/ml in 24 (267) test Lymphoblastic Leukemia hr Melanogenesis Zebrafish Cytotoxic WP ---------- ---------- Cell Line C. japonica Antihypertensive Sd ---------- EA Plasma ACE activity Rats 400 mg/ml (226) Tyrosinase activity assay B16F10 mouse melanoma ------------- (69) melanin contents cells (CRL 6323) 50 and 100 (315) MtOH Melanogenesis Sd Hot Ex Water inhibition cAMP assay Western blot analysis Memory Sd Sonicat. Water enhancing Novel object recognition test ICR mice The step-through passive mg/kg/day avoidance test Immunohistochemistry Melasma AP Heating Water elimination C. australis Hepatoprotective Melasma Area Severity Index Patients 4.8 g/day (311) Wistar rats 125 and 250 (70) degree of hyperpigmentation St Soxhlet EtOH Hepatic injury markers mg/kg C. europaea Antibacterial WP Shaking EtOH Agar well method S. aureus 20 mg/ml (263) 500 mg (308) E. coli C. planif'lora Antidepressant AP ---------- ---------- Triple-blind controlled Depression patients clinical trial C. campestres Analgesic WP ---------- 95% EtOH capsule Writhing Test Albino mice 50 and 100 (47) mg/kg. Cold MeOH Macera. Antipyretic WP ---------- Writhing Test Swiss Albino mice 400 mg/kg (46) Albino mice 50 and 100 (47) Heat conduction method 95% EtOH electric thermocouple mg/kg Antiiflammatory WP ---------- 95% EtOH Volume plethysmographically Albino mice 100 mg/kg (47) CNS-depressant WP ---------- 95% EtOH Behavioural study Albino mice 50 and 100 (47) mg/kg Anticancer WP ---------- EA --------------- Hepatocellularcarcinoma MeOH AP Macera. MeOH ----------- (270) ----------- (271) cell line RT PCR analysis MCF 10A, MCF-7 and MDAMB-231 cell lines al AP king ective WP ---------- tOH s and Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 ial WP ty WP /kg 264) nd 104) 1237 kg la. tOH ium l 214) ents kg 6) Noureen et al. An overview of the genus Cuscuta Continued Table 4. AP Macera. MeOH RT PCR analysis MCF 10A, MCF-7 and MDA- ----------- (271) 1000 mg/kg (264) 20, 100 and (104) MB-231 cell lines Antiviral AP Shaking MeOH RT-PCR analysis Peripheral blood mononuclear cell Hepatoprotective WP ---------- 75% EtOH Biochemical parameters and Mice histological C. racemosa Antimicrobial WP C. pedicellata Anti-obesity WP Antioxidant Sd Percola. Soxhlet 500 mg/kg 70% EtOH Dilution in a liquid medium Staphylococcus aureus 2 mg/ml (214) EtOH Biochemical measurements Albino rats 400 mg/kg (26) MeOH DPPH assay ------------------ -------------- (232) Water ----------------- Xanthomonas campestris -------------- (74) MeOH Agar well diffusion Staphylococcus aureus Pseudomonas aeruginosa 100 μl (168) method 67 μl (168) Macera. Antibacterial L Decoc. infusion L ---------- St Fr Klebsiella pneumonia Acinetobacter baumannii Antifungal L ---------- MeOH Agar tube dilution method Aspergillus fumigatus, St Aspergillus flavus Fr Rhizopus oryzae Cytotoxic L potential St ---------- MeOH ---------- MeOH Brine shrimp assay -------------------- -------------- (168) Albumin denaturation, -------------------- 200 μg/ml (168) Fr C. epithymum C. kotschyana anti- L inflammatory St membrane stabilization Fr proteinase inhibitory assays Hepatoprotective Anticancer WP Soxhlet MeOH Blood serum parameters Wistar albino rats 400 mg/kg BW (182) Sd Decoc. 2N HCl EA MTT assay MCf-7 cell line 100 μg/ml (77) St C. mitraeformis Annexin V Antioxidant St Hydro. dil Antimicro-bial St Hydro. dil n-hex DPPH assay --------------------- -------------- (78) n-hex Broth microdilution method Clavibacter michiganensis -------------- (78) 100 mg/kg (257) aceton Erwinia carotovora Pseudomonas syringae C. arvensis Analgesic activity WP ---------- n-hex, Writhing test Swiss albino mice DCM EA MeOH water WP whole plant; AP aerial parts; Fl flower; St stem; Sd seed;Fr fruit; L leaves; Aq. aqueous; MeOH methanol; EtOH ethanol; Pet. eth petroleum ether; n-hex n-hexan; EA ethyl acetate; DMC dichloromethane; CF chloroform; Macera maceration; Decoc decoction; Hydro. Dil hydro distillation; Percola percolation; Sonicat sonication; Hot Ex hot extraction; Suc. Ex successive extraction Seed oil of C. pedicellata was extracted with petroleum ether (pet. ether) and lipid contents were saponified to separate unsaponifiable materials and fatty acids. The extract was fractionated by using various solvents, and antioxidant activity of all extracts (pet. ether, unsaponified, fatty acids, 70 % methanol, ethyl acetate, and chloroform) was appraised by DPPH free radical assay. The methanol extract was found most potent (230). In another study, a correlation was established between antioxidant activity and total phenolic content of aerial parts of three Iranian Cuscuta species. C. approximate, C. monogyna and C. campestris were estimated by using DPPH microplate method. The highest concentration of phenolic compounds was found in C. monogyna and C. approximata. TPC of 1238 plant methanolic extracts was determined. Methanolic extracts of C. approximata and C. monogyna contain highest amounts of total phenolic, 56.67 mg/g and 49.59 mg/g, respectively, while antioxidant potential was in the order C. monogyna ˃ C. approximate ˃ C. campestris (14). Ethyl acetate fraction of ethanolic extract of C. chinensis seeds possesses strongest antioxidant effect with kaempferol and quercetin as its main constituents. It hunts free radicals and inhibits liquid peroxidation (198, 239). The same fraction of methanolic extract was ascertained as an effective antioxidant by DPPH free radical scavenging assay (222). Moreover, aqueous extract of C. chinensis can protect murine osteoblastic MC3T3-E1 cells against tertiary butyl hydroperoxide induced injury because of its oxidation Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta stress management potential and functioning against mitochondria-dependent pathways (240). In another experiment, flavonoids of C. chinensis were evaluated for their protective effect against oxidative stress. The survival rate of PC12 cells having H2O2-induced apoptosis was measured. The protective effect was possibly due to scavenging of reactive oxidative species and enhanced activity of antioxidant enzyme (241). Essential oils and carotenoids separated from C. mitraeformis also showed antioxidant activity (78). These results suggest that Cuscuta plants are enriched with highly important natural antioxidants that may be used in development of functional foods and drugs effective against diseases caused by oxidative stress. Isolation, identification and possible synergism among various components may be the subject of interest for further studies. Hepatoprotective Anti-hepatotoxic drug designing is a major thrust area seeking the attention of natural product researchers because synthetic formulations have serious side effects. C. epithymum is traditionally used as a liver tonic. C. epithymum whole plant extracted in methanol exhibited appreciably high hepatoprotective effect against CCl4 induced hepatotoxicity in albino rats. Elevated serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP) and total bilirubin have confirmed hepatic damage after CCl4 administration. C. epithymum prevented the toxic effect in both anticipatory and curative models, which may be due to the presence of various bioactive moieties, including phenolics, flavonoids, and alkaloids (185). Many investigators have studied the curative effect of C. reflexa against liver damage induced by cisplatin, paracetamol, carbon tetrachloride, ethanol, isoniazid, and rifampicin. Various biochemical measurements were observed including ALT, AST, ALP, and total bilirubin before and after the administration of C. reflexa extract. It improved liver function by significantly reducing the serum ALT, AST, and ALP levels in affected rats comparable to standard. Histopathological examination of liver section supports the results (32, 242, 243). Ethanolic extract of C. australis also appeared as liver protector against acetaminophen intoxication in an animal model. Two groups of rats were intoxicated on day eight after receiving doses of C. australis seed and stem extract separately for seven days. In untreated rats, severe periportal hepatic necrosis, considerably raised serum liver damage markers, noticeably augmented lipid peroxidation and suppressed liver antioxidant enzymes activities were witnessed. Comparative evaluation of seed and stem extract proves that stem is a more potent hepatoprotective counterpart than seed (70). Seeds of C. chinensis are commonly employed to nourish and improve hepatic disorders in China and various other Asian countries. Oxidative stress can stimulate the development of acetaminophen-induced hepatotoxicity. Liver protecting and antioxidant activities of ethanolic and aqueous extracts of C. chinensis on acetaminophen-induced hepatotoxicity in rats. Ethanolic extract showed a significant hepatoprotective effect at an oral dose of 125 and 250 mg/kg confirmed by the measurement of various Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. parameters and observation of liver histopathology. Comparatively same doses of the aqueous extract were found ineffective rather; it resulted in further hepatic deterioration (33). C. chinensis nanoparticles were found more effective in this regard (198, 239). Thus, from the above findings it can be observed that many Cuscuta species are promising hepatoprotective agents supporting the claims of traditional healers. Further investigations on chemical components are needed to pinpoint the findings. Antidiabetic Diabetes mellitus is becoming a growing threat for a vast population in almost all countries of the world due to a sluggish lifestyle leading to reduced physical activity and increase in obesity (244). Methanolic and chloroform extracts of C. reflexa whole plant exhibited significant hypoglycemic activity at doses of 50, 100, and 200 mg/kg body weight. Oral glucose tolerance test was used to estimate the effect in glucose-loaded Long Evans rats (50). Administration of methanolic extract C. reflexa to glucose-loaded mice led to notable reductions in blood glucose and improved metabolic alterations, thereby justifying its traditional folkloric claims (89, 245). Antidiabetic activity of C. chinensis was evaluated in dexamethasone-induced insulin-resistant human liver carcinoma (HepG2) cells (246). C. chinensis polysaccharides can reduce blood sugar level in type2 diabetes. Efficacy was tested on alloxan-induced diabetes in a mice model. Orally administrated doses of 300 and 600 mg/kg remarkably decreased the elevated fasting blood glucose (247-248). In a similar study, oral administration of 200 and 400 mg/kg polysaccharides significantly lessened blood glucose along with glycosylate serum protein (249). A Chinese herbal prescription, Zhujing pill, having more than 50 % C. chinensis protected retina of diabetic rats, possibly through its antioxidation and anti-inflammatory effects (250). Recently mechanism of hypoglycemic activity of C. chinesis on type 1 diabetic disease was investigated using a rat model. Daily administration of C. chinesis extract returned fasting serum insulin and fasting blood glucose to normal value by upregulating the gene expression of hepatic and pancreas genes (251). It is crucial to continue the exploration of hypoglycaemic effect of more plants as these are blessed with similar chemical profile. Anti-inflammatory Inflammatory reactions play a decisive role in different phases of pathogenesis of cancer. So, there may be an assumption that anti-inflammatory drugs can induce apoptosis in cancerous cells and may be equally beneficial as preventive measure and therapy (252). Aqueous and alcoholic extracts of stem of C. reflexa and its ethyl acetate fraction showed remarkable anti-inflammatory activity in in vitro and in vivo tests. Inflammation was induced by various chemicals like histamine and lipopolysaccharide. It was observed that extracts inhibited inflammatory responses that can be related to the presence of flavonoids, phenols, and polyphenols in this plant (43-44, 253). C. reflexa significantly suppressed inflammation by reducing 1239 Noureen et al. edema volume up to 80 % in rats as compared to standard 96.36 % (254). C. campestris markedly inhibited carrageenaninduced edema in rats by oral pretreatment with 100 mg/kg extract (47). C. chinensis, by suppressing the inflammatory responses showed the potential for treatment of brain inflammation (255). Moreover, λ-carrageenan-induced paw edema treatment by using the methanolic extract of C. chinensis seed in mice, also confirmed its anti-inflammatory effect (256). C. pedicelleta and C. arvensis were found effective against inflammation (168, 257). Further studies must be conducted to clarify the mechanism and to figure out the active principle behind the activity. Antibacterial, antifungal, and antiviral Continuous and urgent exploration is required for new antimicrobial agents with new compositions and diverse mechanisms of action to overcome antimicrobial modifications (9). Methanolic extract of C. reflexa was found significantly active against a broad spectrum of bacterial species including S. aureus, P. aeruginosa, S. dysenteriae, S. boydii, and E. coli with impressive zone of inhibition (27, 258-260). Xanthomonas campestris (XC) is a widely spread infectious agent causing a huge loss in food crops with visible symptoms and leave shedding. Aqueous decoction and infusion extract of C. pedicellata were evaluated for antibacterial activity against diverse pathovars of XC using in vitro well diffusion method. Inhibition zone diameter was observed from 1.0 to 5.0 cm (74). The methanolic extract also showed promising high antimicrobial activity (168). C. australis is another species having notable antibacterial effect. The 50 % methanolic extract was fractionated by hexane, ethyl acetate, and butanol with various polarities. All fractions were tested against fungal, yeast and various Gram-positive and Gram-negative bacteria. All extracts except n-hexane were found effective against different species (261). Additionally, methanolic extract of C. epithymum was also significantly active against Bordetella bronchiseptica demonstrating zone of inhibition from 10–14 mm (262). C. europaea was active against Staphylococcus aureus even higher than standard drug Amoxicillin. These results lead toward the concept that this plant can be used as a safer option against this microbe (263). Recently essential oils and carotenoids separated from C. mitraeformis were found antibacterial (78). In addition to many other species of genus Cuscuta, C. racemose offers flavonoids as chief metabolites. Slightly positive antimicrobial activity of this plant was observed against S. aureus using dilution in a liquid medium method. Minimum inhibiting concentration was 2.0 mg/ml. Phenolic compounds are documented as antimicrobial substances. So, the activity can be ascribed to the flavonoids and tannins in the plant (52). Several secondary metabolites like flavans, flavones, and quinic acid derivatives have been found active against HIV infection. Crude aqueous extracts of C. reflexa exhibited anti-HIV activity. Virus inhibition may be attributed to the combinatory effects of nine closely related compounds (49). An antiviral protein with significantly high inhibiting property was isolated 1240 An overview of the genus Cuscuta from the aqueous extract of C. reflexa (219). Methanolic extract of C. campestris showed weak anti-HIV activity (264). A number of species have been found effective against microbes. It is recommended that further studies with isolated components instead of extracts may be more useful to identify the active compounds. Antitumor effect Some species of the genus Cuscuta afford alkaloids with indolic nuclei that are considered potential antitumor substances. C. chinensis is a popular antitumor prescription in the Unani medicine system. Oral administration of the plant extract at a dose of 1 g/kg noticeably delayed the appearance and growth of skin papilloma and reduced the chances of carcinoma (30). Anticancer activity of C. chinensis has been evaluated by several pharmacological studies using a variety of cell lines. Results prove that it can act as an integrative approach to encounter ever-growing disease management (22, 31, 265- 267). In vivo anticancer potential of C. reflexa was determined by using murine models. Alcoholic extract and its chloroform fraction were found more potent. It showed highest toxicity against human breast cancer cell lines. Similarly, chloroform part of extract of alcohol showed considerable tumor growth inhibition, which reveals that these extracts interfere in cell proliferation to inhibit cancer (15). It can induce apoptosis in Hep3B cells (253). Phenolic components isolated from C. reflexa were also assessed in HCT116 colorectal cells amongst which 1-O-p-hydroxycinnamoylglucose could show considerable anticancer activity (10). The seed extract of C. kotschyana induced apoptosis in breast cancer cell line (MCF7) (77). As the major active phytoconstituents of C. kotschyana are flavonols, quercetin, and kaempferol (231) and quercetin has been found to reduce cell viability of quite a lot of cancer cell lines in vitro (268-269). Therefore, these facts are consistent with results that the exposure of MCF7 cells to C. kotschyana considerably reduced viability (77). C. campestris also has anticancer agents (270). Detection and evaluation of phytochemicals suggested that eugenol epoxide, lutein epoxide, and lupeol epoxide formed the most active fractions and exhibited the cytotoxic effects against breast cancer cells (271). In a recent effort, efficacy of a Korean herbal formula Ga Gam Nai Go Hyan containing C. japonica against benign prostatic hyperplasia was evaluated. This herbal prescription significantly decreases prostate weight by regulating inflammatory responses and apoptosis (92). There is need to develop new technologies such as nanoparticles to improve the therapeutic effect of compounds isolated from these plants. Further efforts may be used to design sustained and targeted drug release systems to improve avoiding side effects. Immunological effects Ethanolic extract of C. chinensis showed considerable adjuvant potentials towards cellular and humoral immune responses in mice models and can be used as vaccine adjuvants. Extract enhanced specific antibodies (IgG, IgG1, and IgG2b) to a noticeably high level by affecting Th1 and Th2 cell functions (272). Dendritic cells play a key role in regulating immune responses Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta and are a major target to develop immune modulators. n-butanol and methanol extracts exhibited the immunosuppressive effect on dendritic cells. Kaempferol was identified as the main flavonoid of methanol fraction. Results suggest that kaempferol has potential to treat chronic inflammatory and autoimmune diseases (273). Furthermore, aqueous extract of C. chinensis also improved the immune responses (274). C. chinensis can protect against tertiary butyl hydroperoxide induced murine osteoblastic MC3T3-E1cell injury. Aqueous extract of seeds protected cells in a dose-dependent manner by modulating the oxidative stress-induced apoptosis probably owing to its antioxidant potential (240). C. australis may act as an immunopotentiator for mammals by increasing the percentage of phagocytosis (275). C. australis hyperoside can decrease T or B lymphocyte proliferation and phagocytic activity of the peritoneal M and mediate immune regulation (276). Effect on the neuronal system C. chinensis can act as a neuroactive agent and improves memory by inducing cell differentiation. Glycoside of the plant induced neuronal differentiation in rat pheochromocytoma PC12 cells (277). In another experiment, C. chinensis improved memory and inhibited acetylcholinesterase activity in scopolamine-induced dysmnesia mice (278). Oral administration of its aqueous extract recovered the ischemia-induced lethal damage of neurons and prevented learning disability (51). A traditional Chinese formula Wu-Zi-Yan-Zong containing C. chinensis suppresses neuroinflammatory responses and can act as an effective therapeutic agent to prevent and treat neuroinflammatory defects (279). Anti-aging activities C. chinensis is an important antiaging prescription of the Chinese herbal medicinal system. Various experimental efforts have been employed to test the certainty of the claim. Polysaccharides of C. chinensis can exhibit anti-aging effects by scavenging free radicals and opposing lipid peroxidation (280). Ethanolic extract of C. chinensis significantly suppressed the non-enzymatic glycosylation of D-galactose-induced rat aging model (281). Various research reports obviously show that it can regulate immune responses, prolong cell cycle, positively affect body metabolism, improve physiology of internal body organs, and stress management, which proves its anti-aging effects (282). Antihypertensive Ethanolic extract of C. reflexa decreased arterial blood pressure and heartbeat rate in Pentothal anesthetized rats. Experimental data indicated that it is a non-specific depressant on all the isolated tissues tested (283). In the course of experiments, ethyl acetate fraction of C. japonica exhibited distinctive angiotensin-converting enzyme (ACE) inhibition at a dose of 400 mg/ml. Four caffeoylquinic acid derivatives were isolated from the active fraction having inhibitory effects on ACE activity. Presence of these metabolites, at least in part is responsible for the antihypertensive activity extract (229). Anti-osteoporotic activity C. chinensis effectively boasted tissue regeneration Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. of damaged bones by promoting the formation of osteoblasts from their precursor cells (284). It has been demonstrated in an experimental report that aqueous extract of C. chinensis significantly stimulated the differentiation and proliferation of osteoblasts in rat bone cells, but the osteoclasts activities were inhibited (285-286). Antagonistically antiosteoporotic effect of C. chinensis was also observed. Five flavonoids were isolated from which kaempferol and hyperoside were found osteogenic in nature (22). Renoprotective effects Aqueous and alcoholic extract of C. reflexa exhibited substantial diuretic activity in Wister rats. Total urine volume and Na +, K + and Cl− concentration was estimated after a dose of 300 mg/kg extract. There was a marked rise in Na + and K + excretion (45). C. chinensis has been used as a kidney tonic since ancient times. Effect of seed extract on renal function parameters in the rat model having ischemia/reperfusion-induced acute renal failure was studied. Results indicate that C. chinensis extract ameliorates renal functions and regulates urine concentration (287). Effect on the reproductive system C. reflexa has an antifertility effect. Methanolic extract arrested the normal estrus cycle and decreased ovarian and uterus weight in adult female mice. Flavonoids are reported as antifertility agents, and C. reflexa is rich in flavonoids, so results can be attributed to the presence of such compounds (42). C. chinensis extract, and its isolations can improve reproductive systems of both males and females. Ethanolic extract of C. chinensis induces a relaxing effect on cavernous penile tissue and may improve erectile dysfunction conditions (288). Many formulations of C. chinensis with other herbal prescriptions enhanced penile erection, improved erectile dysfunction, infantile uteruses, and motility of sperm (154, 289-291). An herbal formula, KH-204 containing C. chinensis, ameliorates erectile dysfunction by its antioxidant and lipid profile improving property (292). Effect of various flavonoids from C. chinensis on sex hormones, and prevention of induced and threatened abortion were evaluated by measuring different parameters in a mice model (293-297). Anti-mutagenic activity Mutations elicit an innate metabolic defect in regular cellular systems and lead to morbidity and mortality in mutated organisms. Therefore, exploration for novel bioactive phytocompounds to encounter promutagenic and carcinogenic effects is a subject of keen interest (298). Preliminary evaluation of methanolic extract of C. chinensis suppressed 90 % of mutagenic effect against Trp-P-1 in the Ames test, suggesting it as a potential antimutagenic agent (299). Mutagenic and antimutagenic effects of C. reflexa were also studied by the Ames test against well-known positive mutagens including 2-aminofluorine, 4-nitroo-phenylenediamine, and sodium azide in Salmonella typhimurium (TA 98 and TA 100) bacterial strains. The extract revealed noteworthy antimutagenic activity against 4-nitro-o-phenylenediamine and sodium azide 1241 Noureen et al. for S. typhimurium strains (122). Cardiovascular activities The aging process is accompanied by so many diseases like diabetes, cancer, dementia, and cardiovascular diseases. Heart diseases, leading causes of mortality are due to cardiomyocyte apoptosis which play a key role in myocardial damage and heart failure (300-302). In an experiment, effect of polysaccharide of C. chinensis was investigated on D-galactose induced apoptosis of cardiomyocytes in an aging rate model. Apoptosis parameter evaluation indicated that polysaccharide extract decreased the apoptosis of cardiomyocytes (303). C. chinensis extract can increase coronary blood flow and decrease myocardial oxygen consumption (304). CNS depressant activities and anti-depressant activities Central nervous system (CNS) disorders comprise 12 % of deaths worldwide and are still a hugely challenging endeavor for health care systems. Plenty of Convolvulaceae species, including Cuscuta members, are used to treat CNS related diseases traditionally and might be used as alternatives (184). C. campestris affects the CNS action and decreases motor activity of mice sited on a rotarod. Various tests applied indicated the CNS-depressant activity of the extract, which probably seems due to an anesthetizing effect (8, 47). In another experimental trial, methanolic extract of C. reflexa served as a good anxiolytic agent in mice at a dose of 400 mg/kg (305). C. chinensis methanolic extract considerably reduced immobility times estimated by FST forced swimming test, which reveals its antidepressant activity (306). While its aqueous extract shows CNS-depressant activity in mice by reducing motor activity and the tonic/clonic phases of electrically-induced seizures in rats (157). Recently a Chinese herbal medicine, Tiansi liquid, containing C. chinensis was evaluated for its antidepressant activity, and possible mechanism of action was predicted by in silico study (307). Capsules of C. planiflora (500 mg) prepared by a pharmacist were found effective for major depression patients. In a study period of eight weeks depression was measured before and after by Beck Depression Inventory and Hamilton Depression Inventory (308). Effect on melanin production C. chinensis can promote melanogenesis of amelanotic melanocytes and improved the tyrosinase activities (247-248). Furthermore, it significantly enhanced skin melanin and tyrosinase production. It also positively affected vitiligo treatment in guinea pigs (309). Moreover, there is another report on melanogenesis effect of C. chinensis seeds aqueous and ethanolic extracts both in vitro and in vivo. The aqueous extract showed inhibitory effect on tyrosinase, while the ethanolic extract displayed the opposite effect in tyrosinase activity (160). In a similar study aqueous and ethanolic extracts of C. chinensis seeds significantly influenced the melanogenesis by regulating the activity of tyrosinase (310). Consumption of the C. chinensis extract with milk reduced the melatonin synthesis and thus ameliorated 1242 An overview of the genus Cuscuta the elimination of melasma (311). C. japonica has an inhibitory effect on mushroom tyrosinase activity (312). It can also be used to improve hyperpigmentation. It was ascertained by the treatment of alpha-melanocyte-stimulating hormoneinduced melanogenesis with aqueous extract in mouse melanoma cells (69). Anti hair fall and anthelmintic activities Hair loss is a feared side effect of chemotherapy and creates a psychologically distressing condition among millions of men and women due to the deprivation of their major esthetic display feature. Plants as hair growth promotors have found their use in almost all traditional medicinal systems. C. reflexa extract is useful in the treatment of alopecia by promoting hair growth (40, 313). Methanolic extract of C. chinensis was used as an anthelmintic drug against Dactylogyrus intermedius in goldfish (314). Analgesic and psychopharmacological C. campestris has analgesic properties. The whole plant grown on Nerium indicum was studied. Acetic acid induced writhing test and heat conduction method were used to study the described activity in an animal model. A dose of 400 mg/kg methanolic extract gave significant results as compared to standard Diclofenac sodium (46). In a similar experiment, protecting response against p-benzoquinone-induced writhing was studied by giving a dose of 100 mg/kg to mice, which suggested the analgesic activity of the extract (47). C. chinensis also has a pain-relieving ability which was examined by using acetic acid-induced writhing response and formalin-induced paw licking method (256). Petroleum ether extract of C. reflexa noticeably decreased the spontaneous activity and behavior profile of Swiss albino mice. Steroids, the major constituents of the extract may be responsible for such changes (39). C. Japonica treatment improved the cognitive function of mice in a dose-dependent manner. Novel object recognition and passive avoidance test proved that it might improve learning and memory (315). Antipyretic and antiulcer Antipyretics agents lessened the body temperature in fever. Efficacy of C. reflexa as an antipyretic agent was confirmed in yeast induced pyrexia in rats. Aqueous and ethanolic extracts were both found active and started rectal temperature decline after three hours of dose. A dose of 400 mg/kg weight reduced the elevated temperature approximately 83.8 % (ethanolic) and 79 % (aqueous) as compared to the standard drug (96.5 %, Paracetamol) after six hours of treatment (48). C. campestris markedly lowered the body temperature of hyperthermic and normothermic mice (47). Lyophilized raw extract of C. racemosa possesses antiulcer activity, which was ascertained by a test showing 44.22 % rate of activity, and 37.05 % rate of cure against acute and sub-chronic models of ulcers, respectively (52). Anticonvulsant and anti-obesity C. epithymum have effective anticonvulsant constituents and delayed the onset of seizure (316). Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta Methanolic extract of C. reflexa stem demonstrated preventive effects against convulsion created by chemical agents in mice. Catecholamines levels augmented considerably. After a six-week treatment, γ-aminobutyric acid (GABA) involved in seizure activity was noticeably increased in the brains of mice (317). Ethanolic extract of C. reflexa significantly reduced convulsions by delaying onset and duration of seizures in an albino mice model. A dose of 400 mg/kg showed maximum delay in pentylenetetrazole induced convulsions (238). C. pedicellata is widely used for management of obesity. Ethanolic extract of C. pedicellata has significantly reduced the bodyweight along with serum lipid profile in high-fat diet-fed rats (26). Recently, polyphenols are reported to possess anti-obesity activity (318). Cytotoxicity, insecticidal, antiarthritic, and wound healing activity The ethanolic extract C. reflexa, parasitizing Nerium oleander, exhibited promising cytotoxic activity (208). Lectin-like glycoproteins isolated from C. europaea demonstrated the cytotoxic effects of LLP and LLP on C127 and B-16 cells (319). Various extracts of the plant have larvicidal potential against mosquitoes (320). C. reflexa protects against arthritis and nephrotoxicity. A dose at 600 mg/kg considerably reduced paw edema and joint swelling up to 71.22 % (321). Aqueous and ethanolic extracts of C. reflexa stem at 200 mg/kg and 400 mg/kg were able to heal wounds in a rat model (322). Conclusion Cuscuta genus is a rich and diverse source of many valuable chemical components. It is loaded with flavonoids, alkaloids, lignans, polysaccharides, steroids, volatile oils, and resin glycosides. Medicinal importance of its various species is part of prehistoric texts. Traditionally it is considered a miracle genus equipped with broad spectrum of remedial values. Decoctions, extracts, paste, powder, juice, and infusions of different parts of the plants are important herbal prescriptions in traditional medicinal systems. A lot of experimentation has been employed to verify its phytotherapy as claimed by traditional healers and local inhabitants. C. reflexa, C. chinensis, C. pedicellata, C. approximate, C. monogyna, C. campestris, and C. mitraeformis have shown impressive antioxidant activity. C. chinensis, C. australis, C. reflexa, and C. epithymum are significantly hepatoprotective in nature. Some species of Cuscuta including C. reflexa, C. chinensis, C. campestris, C. japonica, and C. kotschyana have been reported potentially antitumor against various cancer cell lines. Moreover crude extracts and compounds from the various parts possessed antibacterial, antiosteoporotic, anti-inflammatory, antihypertensive, analgesic, anti hair fall, analgesic, and antiestrogenic properties. Rich and unrivaled medicinal history demands verification with modern scientific methodologies. Only a few of the species are thoroughly investigated up till now, especially C. reflexa and C. chinensis out of nearly 170, while the rest of the members are partially or fully undiscovered in terms of phytochemistry and pharmacology. Most of the efforts are limited to in Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. vitro and in vivo animal models or cell line level. Very few clinical studies are reported in humans. Although a good deal of secondary metabolites with multitudinous pharmacological attributes have been isolated, identified, and characterized but most of the pharmacological investigations are extract-based. Further studies must be conducted to clarify the mechanism and to figure out the active principle behind the activity to use these compounds as leads and template in development of new drugs. References 1. Sermakkani M, Thangapandian V. GC-MS analysis of Cassia italica leaf methanol extract. Asian J Pharm Clin Res 2012; 5:90-94. 2. Gulfraz M, Sadiq A, Tariq H, Imran M, Qureshi R, Zeenat A. Phytochemical analysis and antibacterial activity of Eruca sativa seed. Pak J Bot 2011; 43:1351-1359. 3. Ramasamy S, Manoharan AC. Antibacterial effect of volatile components of selected medicinal plants against human pathogens. Asian J Microbiol Biotechnol Environ Sci 2004; 6:209-210. 4. Hoareau L, DaSilva EJ. Medicinal plants: a re-emerging health aid. Electron J Biotechnol 1999; 2:3-4. 5. Gurib-Fakim A. Medicinal plants: traditions of yesterday and drugs of tomorrow. Mol Aspects Med 2006; 27:1-93. 6. Rates SM. Plants as source of drugs. Toxicon 2001; 39:603613. 7. Balandrin MF, Klocke JA. Medicinal, aromatic, and industrial materials from plants. In Medicinal and Aromatic Plants I. Berlin. Heidelberg: Springer 1988. p.3-36. 8. Prajapati ND, Purohit SS. Agro S Colour Atlas of Medicinal Plants. Agrobios (India); 2003. 9. Rojas R, Bustamante B, Bauer J, Fernández I, Albán J, Lock O. Antimicrobial activity of selected Peruvian medicinal plants. J Ethnopharmacol 2003; 88:199-204. 10. Riaz M, Bilal A, Ali MS, Fatima I, Faisal A, Sherkheli MA, et al. Natural products from Cuscuta reflexa Roxb. with antiproliferation activities in HCT116 colorectal cell lines. Nat Prod Res 2017; 315:583-587. 11. Kaul K, Jaitak V, Kaul VK. Review on pharmceutical properties and conservation measures of Potentilla fulgens Wall. ex Hook.-a medicinal endangered herb of higher Himalaya. Indian J Nat Prod Resour 2011; 2:298-306. 12. Benkeblia N. Antimicrobial activity of essential oil extracts of various onions (Allium cepa) and garlic (Allium sativum). Food Sci Technol 2004; 37:263-268. 13. Anjum F, Bukhari SA, Shahid M, Anwar S, Afzal M and Akhter N. Comparative evaluation of antioxidant potential of parasitic plant collected from different hosts. J Food Process Technol 2013; 4:1-6. 14. Jafari E, Bahmanzadegan A, Ghanbarian G, Rowshan V. Antioxidant activity and total phenolic content from aerial parts of three Cuscuta species. Anal Chem Lett 2015; 5:377384. 15. Bhagat M, Arora JS, Saxena AK. In vitro and in vivo antiproliferative potential of Cuscuta reflexa Roxb. J Pharm Res 2013; 6:690-695. 16. Rao VS, Dasaradhan P, Krishnaiah KS. Antifertility effect of some indigenous plants. Indian J Med Res 1979; 70:517-520. 17. Costa-Lotufo LV, Khan MT, Ather A, Wilke DV, Jimenez PC, Pessoa C, et al. Studies of the anticancer potential of plants used in bangladeshi folk medicine. J Ethnopharmacol 2005; 99:21-30. 1243 Noureen et al. 18. Begum HA, Hamayun M, Zaman K, Hussain A, Ruaf M. Phytochemical evaluation of ethnobotanically selected medicinal plants of mardan, pakistan. J Adv Bot Zool 2015; 3:1-5. 19. Qureshi R, Bhatti GR. Ethnobotany of plants used by the thari people of nara desert, pakistan. Fitoterapia 2008; 79:468-473. 20. Sharma H, Kumar A. Ethnobotanical studies on medicinal plants of rajasthan (india): a review. J Med Plants Res 2011; 5:1107-1112. 21. Malhotra SP, Dutta BK, Gupta R, Gaur YD. Medicinal plants of the indian arid zone. J Agric Tradit Bot Appl 1966; 13:247-288. 22. Yang L, Chen Q, Wang F, Zhang G. Antiosteoporotic compounds from seeds of Cuscuta chinensis. J ethnopharmacol 2011; 135:553-560. 23. Schmelzer GH, Gurib-Fakim A. (2013). Plant resources of tropical Africa 11 (2): medicinal plants 2. Plant resources of tropical Africa 11: Medicinal Plants 2 P. 101-105 24. Sharma L, Khandelwal S. Weeds of rajasthan and their ethno-botanical importance. Stud Ethno-Med 2010; 4:75-79. 25. Jang IM. Treatise on asian herbal medicines. Seoul: Haksulpyunsu-kwan in Research Institute of Natural Products of Seoul National University. 2003. 26. Zekry SH, Abo-elmatty DM, Zayed RA, Radwan MM, ElSohly MA, Hassanean HA, et al. Effect of metabolites isolated from Cuscuta pedicellata on high fat diet-fed rats. Med Chem Res 2015; 24:1964-1973. 27. Raza MA, Mukhtar F, Danish M. Cuscuta reflexa and Carthamus Oxyacantha: potent sources of alternative and complimentary drug. SpringerPlus. 2015; 4:76-82. 28. Inamdar FB, Oswal RJ, Chorage TV, Garje K. In vitro antimicrobial activity of Cuscuta reflexa ROXB. Int Res J Pharm 2011; 2:214-216. 29. Kalita D, Saikia J. Ethnomedicinal, Antibacterial and antifungal potentiality of Centella asiatica, Nerium indicum and Cuscuta reflexa -widely used in tiwa tribe of morigaon district of assam, india. Int J Phytomed 2012; 4:380-385. 30. Nisa M, Akbar S, Tariq M, Hussain Z. Effect of Cuscuta chinensis water extract on 7, 12-dimethylbenz [a] anthraceneinduced skin papillomas and carcinomas in mice. J Ethnopharmacol 1986; 18:21-31. 31. Umehara K, Nemoto K, Ohkubo T, Miyase T, Degawa M, Noguchi H. Isolation of a new 15-membered macrocyclic glycolipid lactone, Cuscutic Resinoside a from the seeds of Cuscuta chinensis: a stimulator of breast cancer cell proliferation. Planta Med 2004; 70:299-304. 32. Balakrishnan BR, Sangameswaran B, Bhaskar VH. Effect of methanol extract of Cuscuta reflexa aerial parts on hepatotoxicity induced by antitubercular drugs in rats. Int J Appl Res Nat Prod 2010; 3:18-22. 33. Yen FL, Wu TH, Lin LT, Lin CC. Hepatoprotective and antioxidant effects of Cuscuta chinensis against acetaminopheninduced hepatotoxicity in rats. J Ethnopharmacol 2007; 111:123-128. 34. Borole SP, Oswal RJ, Antre RV, Kshirsagar SS, Bagul YR. Evaluation of anti-epileptic activity of Cuscuta reflexa Roxb. Res J Pharm Biol Chem Sci 2011; 2:657-663. 35. Amol P, Vikas P, Kundan C, Vijay P, Rajesh C. In vitro free radicals scavenging activity of stems of Cuscuta reflexa. J Pharm Res 2009; 2:58-61. 36. Bao X, Wang Z, Fang J, Li X. Structural features of an immunostimulating and antioxidant acidic polysaccharide from the seeds of Cuscuta chinensis. Planta Med 2002; 68:237243. 37. Noureen S, Noreen S, Ghumman SA, Batool F, Arshad M, 1244 An overview of the genus Cuscuta Noreen F, et al. Seeds of giant dodder (Cuscuta reflexa) as a function of extract procedure and solvent nature. Not Bot Hort Agrobot Cluj 2018; 46:653-662. 38. Anis E, Anis I, Ahmed S, Mustafa G, Malik A, Afza N, et al. α-glucosidase inhibitory constituents from Cuscuta reflexa. Chem Pharm Bull 2002; 50:112-114. 39. Pal DI, Panda CH, Sinhababu SA, Dutta AR, Bhattacharya SH. Evaluation of psychopharmacol effects of petroleum ether extract of Cuscuta reflexa Roxb stem in mice. Acta Pol Pharm 2003; 60:481-486. 40. Pandit S, Chauhan NS, Dixit VK. Effect of Cuscuta reflexa Roxb on androgen-induced alopecia. J Cosmet Dermatol 2008; 7:199-204. 41. Roy RK, Thakur M, Dixit VK. Development and evaluation of polyherbal formulation for hair growth–promoting activity. J Cosmet Dermatol 2007; 6:108-112. 42. Gupta M, Mazumder UK, Pal DK, Bhattacharya S. Antisteroidogenic activity of methanolic extract of Cuscuta reflexa Roxb. stem and Corchorus olitorius Linn. seed in mouse ovary. Indian J Exp Biol 2003; 41:641-644. 43. Katiyar NS, Rao NV, Gangwar AK. Evaluation of antiinflammatory activity of stem extracts of Cuscuta reflexa Roxb in rats. Int J Res Pharm Biomed Sci 2012; 3:1805-1808. 44. Udavant PB, Satyanarayana SV, Upasani CD. Preliminary screening of Cuscuta reflexa stems for anti inflammatory and cytotoxic activity. Asian Pac J Trop Biom 2012; 2:13031307. 45. Sharma S, Hullatti KK, Prasanna SM, Kuppast IJ, Sharma P. Comparative study of Cuscuta reflexa and Cassytha filiformis for diuretic activity. Pharmacogn Res 2009; 1:327-330. 46. Ghule RS, Venkatanarayan R, Thakare SP, Jain H, Ghule PR. Analgesic activity of Cuscuta campestris Yuncker a parasitic plant grown on Nerium indicum Mill. J Adv PharmTechnol Res 2011; 1:45-51. 47. Agha AM, Sattar EA, Galal A. Pharmacological study of Cuscuta campestris Yuncker. Phytother Res 1996; 10:117-120. 48. Bhattacharya S, Roy B. Preliminary investigation on antipyretic activity of Cuscuta reflexa in rats. J Adv Pharm Technol Res 2010; 1:83-87. 49. Mahmood N, Piacente S, Burke A, Khan AL, Pizza C. Constituents of Cuscuta reflexa are anti-HIV agents. Antivir Chem Chemother 1997; 8:70-74. 50. Rahmatullah M, Sultan S, Toma T, Lucky S, Chowdhury M, Haque W, et al. Effect of Cuscuta reflex stem and Calotropis procera leaf extracts on glucose tolerance in glucose-induced hyperglycemic rats and mice. Afri J Tradit Complementary Altern Med 2010; 7:109-112. 51. Chung TW, Koo BS, Choi EG, Kim MG, Lee IS, Kim CH. Neuroprotective effect of a chuk-me-sun-dan on neurons from ischemic damage and neuronal cell toxicity. Neurochem res 2006; 31:1-9. 52. Ferraz HO, Silva MG, Kato ETM, Barros S, Bacchi EM. Antiulcer and antioxidant activities and acute toxicity of extracts of Cuscuta racemosa Mart (Convolvulaceae). Lat Am Jo Pharm 2011; 30:1090-1097. 53. Teware K. Pytochemical extraction and TLC estimation of extract of Cuscuta reflexa . World J Pharm Pharm Sci 2016; 5:378-384. 54. Kuijt J. The biology of parasitic flowering plants. University of California Press, Berkeley; 1969. 55. Liao GI, Chen MY, Kuoh CS. Cuscuta L. (Convolvulaceae) in Taiwan. Taiwania 2000; 45:226-34. 56. Parker C, Riches CR. Parasitic weeds of the world: biology and control. CAB International; 1993. 57. Yuncker TG. The genus Cuscuta. Mem Torrey Bot Club Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta 1932; 18:109-331. 58. Chrtek J, Osbornová J. Notes on the synanthropic plants of Egypt 3. Grammica campestris and other species of family Cuscutaceae. Folia Geobot Phytotax 1991; 26:287-314. 59. Cronquist A, Takhtadzhian AL. An integrated system of classification of flowering plants. Columbia University Press; 1981. 60. Dahlgren G. The last Dahlgrenogram. System of Classification of the Dicotyledons. The Davis and Hedge Festschrift. 1989. p. 249-260. 61. Dawson JH, Musselman LJ, Wolswinkel PI, Dörr I. Biology and control of Cuscuta. Rev Weed Sci 1994; 6:265-317. 62. Fang R, Musselman L, Plitmann U, Cuscuta In P. Raven and C.Y. Wu (eds.) Flora Chin 1995; 16:322-325. 63. Gwo-Ing LI, Ming-Yih CH, Chang-Sheng KU. Pollen morphology of Cuscuta (Convolvulaceae) in Taiwan. Bot Bull Acad Sinica 2005; 46:75-81. 64. Hadaĉ E, Chrtek J. Notes on the taxonomy of Cuscutaceae. Folia Geobot 1970; 5:443-445. 65. Täckholm V, Boulos L. Supplementary notes to Student’s flora of Egypt. Cairo Univ. Herbarium; 1974. 66. Takhtajan A. Flowering Plants: Origin and Dispersal, Oliver and Boyd, Edinburgh; 1969. 67. Patel S, Sharma V, Chauhan NS, Dixit VK. An updated review on the parasitic herb of Cuscuta reflexa Roxb. Jo Chin Integr Med 2012; 10:249-255. 68. Donnapee S, Li J, Yang X, Ge AH, Donkor PO, Gao XM, Chang YX. Cuscuta chinensis Lam.: a systematic review on ethnopharmacology, phytochemistry and pharmacology of an important traditional herbal medicine. J ethnopharmacol 2014; 157:292-308. 69. Jang JY, Kim HN, Kim YR, Choi YH, Kim BW, Shin HK, et al. Aqueous fraction from Cuscuta japonica seed suppresses melanin synthesis through inhibition of the p38 mitogenactivated protein kinase signaling pathway in B16F10 cells. J ethnopharmacol 2012; 141:338-344. 70. Folarin RO, Omirinde JO, Bejide R, Isola TO, Usende LI, Basiru A. Comparative hepatoprotective activity of ethanolic extracts of Cuscuta australis against acetaminophen intoxication in wistar rats. Int Sch Res Notices 2014; 2014:1-6. 71. Dangwal LR, Rana CS, Sharma A. Ethno-medicinal plants from transitional zone of Nanda evi Biosphere Reserve, District Chamoli, Uttarakhand. India 2011; 2:116-120. 72. Haq F. The ethno botanical uses of medicinal plants of Allai Valley, Western Himalaya Pakistan. Int J Plant Res 2012; 2:2134. 73. Meena AK, Rao MM. Folk herbal medicines used by the Meena community in Rajasthan. Asian J Tradit Med 2010; 5:19-31. 74. Ali A, Haider MS, Hanif S, Akhtar N. Assessment of the antibacterial activity of Cuscuta pedicellata Ledeb. Afri J Biotechnol 2014; 13:430-433. 75. Lakhdari W, Dehliz A, Acheuk F, Mlik R, Hammi H, Doumandji-mitiche B, et al. Ethnobotanical study of some plants used in traditional medicine in the region of Oued Righ (Algerian Sahara). J Med Plants Stud 2016; 4:6-10. 76. Njoroge GN, Bussmann RW. Traditional management of ear, nose and throat (ENT) diseases in Central Kenya. J Ethnobiol Ethnomed 2006; 2:54-63. 77. Sepehr MF, Jameie SB, Hajijafari B. The Cuscuta kotschyana effects on breast cancer cells line MCF7. J Med Plants Res 2011; 5:6344-6351. 78. Villa N, Pacheco Y, Rubio E, Cruz R, Lozoya E. Essential oil composition, carotenoid profile, antioxidant and antimicrobial activities of the parasitic plant Cuscuta mitraeformis. Bol Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. latinoam Caribe plantas med aromát 2017; 16:463-470. 79. Weimann C, Heinrich M. Indigenous medicinal plants in Mexico: the example of the Nahua (Sierra de Zongolica). Bot Acta 1997; 110:62-72. 80. Ballabh B, Chaurasia OP, Ahmed Z, Singh SB. Traditional medicinal plants of cold desert Ladakh—used against kidney and urinary disorders. J Ethnopharmacol 2008; 118:331-339. 81. Holm LG, Holm L, Holm E, Pancho JV, Herberger JP. World weeds: natural histories and distribution. John Wiley & Sons; 1997. 82. Furuhashi T, Furuhashi K, Weckwerth W. The parasitic mechanism of the holostemparasitic plant Cuscuta. J Plant Interact 2011; 6:207-219. 83. Kaiser B, Vogg G, Fürst UB, Albert M. Parasitic plants of the genus Cuscuta and their interaction with susceptible and resistant host plants. Front Plant Sci 2015; 6:45-54. 84. Shen HW, Ye W, Hong L, Huang H, Wang Z, Deng X, et al. Progress in parasitic plant biology: host selection and nutrient transfer. Plant Biol 2006; 8:175-185. 85. Kelly CK, Horning K. Acquisition order and resource value in Cuscuta attenuata. Proc Nat Acad Sci 1999; 96:1321913222. 86. Nwokocha MI, Aigbokhan EI. Host range and preference of Cuscuta campestris (Yunck). among common weeds in Benin city, Nigeria. Niger J Bot 2013; 26:1-29. 87. Prather LA. Biology of Cuscuta Attenuata Waterfall. Proc Oklahoma Acad Sci1990; 73:7-13. 88. Diggs GM, Lipscomb BL, O’Kennon RJ, Mahler WF, Shinners LH. Shinners’ and Mahler’s illustrated flora of North Central Texas. Bot Res Inst Texas 1999. 89. Akter MH, Akter MH, Rahmatullah M. Synergistic antihyperglycemic activity of Coccinia grandis leaves and Cuscuta reflexa stems. J Pharm Pharm Sci 2016; 5:236-243. 90. Vijikumar S. Cuscuta reflexa Roxb.–a wonderful miracle plant in ethnomedicine. Indian J Nat Sci 2011; 11:676-683. 91. Mavlonov GT, Ubaidullaeva KA, Kadryaeva GV, Kuznetsova NN. Cytotoxic components of Cuscuta. Chem Nat Compd 2008; 44:409-410. 92. Shin SJ, Lee KH, Chung KS, Cheon SY, An HJ. The traditional Korean herbal medicine Ga-Gam-Nai-Go-Hyan suppresses testosterone-induced benign prostatic hyperplasia by regulating inflammatory responses and apoptosis. Exp Ther Med 2017; 13:1025-1031. 93. Talha J, Priyanka M, Akanksha A. Hypertension and herbal plants. Int Res J Pharm 2011; 2:26-30. 94. Kuo CS, Liao GI. Flower initiation and development in Cuscuta australis R. Br.(Convolvulaceae). Taiwania 1993; 38:99-108. 95. Quattrocchi U. CRC world dictionary of plant names: common names, scientific names, eponyms, synonyms, and etymology: CRC Press. 2000. 96. Weinberg T, Lalazar A, Rubin B. Effects of bleaching herbicides on field dodder (Cuscuta campestris). Weed sci 2003; 51:663-670. 97. Joshi SK, Sanjay G. Cuscuta europaea Linn. (Dodder plant): an emerging threat to plant diversity of Valley of Flowers. Curr Sci 2003; 84:1285-1286. 98. Papuc C, Crivineanu M, Nicorescu V, Predescu C. Reactive oxygen species scavenging activity and hepatoprotective effects of a polyphenolic extract obtained from Cuscuta Europaea. Rev Chim (Bucharest) 2012; 9:869-873. 99. Jafari EF, Assadi MO, Ghanbarian GA. A revision of Cuscutaceae family in Iran. Iran J Bot 2016; 22:23-29. 100. Costea M, Stefanović S. Evolutionary history and taxonomy of the Cuscuta umbellata complex (Convolvulaceae): evidence 1245 Noureen et al. of extensive hybridization from discordant nuclear and plastid phylogenies. Taxon 2010; 59:1783-1800. 101. Hashem A, Patabendige D, Roberts C. Biology and management of red dodder-a new threat to the grains industry. In15th Australian Weeds Conference, Papers and Proceedings, Adelaide, South Australia, 24-28 September 2006: Managing weeds in a changing climate Weed Management Society of South Australia. 2006 p. 163-166. 102. Orr GL, Haidar MA, Orr DA. Smallseed dodder (Cuscuta planiflora) gravitropism in red light and in red plus far-red. Weed sci 1996; 44:795-796. 103. Farah AF, Al-Abdulsalam MA. Effect of field dodder (Cuscuta campestris Yuncker) on some legume crops. Sci J King Faisal Univ (Basic Appl Sci) 2004; 5:103-113. 104. Peng WH, Chen YW, Lee MS, Chang WT, Tsai JC, Lin YC, et al. Hepatoprotective effect of Cuscuta campestris Yunck. whole plant on carbon tetrachloride induced chronic liver injury in mice. Int J Mol Sci 2016; 17:2056-2067. 105. Youssef SA. Medicinal and non-medicinal uses of some plants found in the middle region of Saudi Arabia. J Med Plants Res 2013; 7: 2501-2517. 106. Hillman FH. Dodder In Relation To Farm Seeds. US Department of Agriculture. 1907. 107. Mukhtar I, Atiq M, Hanan A, Iqbal Z. Antifungal activity of Cuscuta reflexa Roxb. Pakistan J Phytopathol 2012; 24:163166. 108. Quattrocchi U. CRC world dictionary of medicinal and poisonous plants: common names, scientific names, eponyms, synonyms, and etymology: CRC Press. 2012. 109. Shahid M, Rao NK. New records of three Convolvulaceae species to the flora of the United Arab Emirates. J New Biol Sci 2016; 5:114-121. 110. Doyle GJ. Cuscuta epithymum (L.) L. (Convolvulaceae), its hosts and associated vegetation in a limestone pavement habitat in the Burren lowlands in county Clare (H9), Western Ireland. In Biology and Environment: Proceedings of the Royal Irish Academy; 1993. p.61-67. 111. Hussain F, Leghari IH, Naveed S. Vegetation in sindh: an analytical and literary study. Karoonjhar 2015; 7:11-28. 112. Piwowarczyk R, Guzikowski S, Góralski G, DenysenkoBennett M, Kwolek D, Joachimiak AJ. First report of dodder (Cuscuta epithymum) parasitizing hemiparasitic species of santalaceae (thesium) and orobanchaceae (euphrasia, melampyrum, odontites, orthantha, and rhinanthus) in Poland. Plant Dis 2018; 102:456-460. 113. Shimi P, Rezapanah MR. A study of Smicronyx robustus faust (Curculionidae) as a biological control agent of eastern dodder (Cuscuta monogyna Vahl.) Iran J Agric Sci 1995; 1:4351. 114. Anac E, Kaya I, Tepe I. Determination of alfalfa dodder (Cuscuta approximata Bab.) damage on alfalfa (Medicago sativa L.) grown in Van, Turkey. In Proceedings of Joint Workshop of the EWRS Working Groups Weed Management in Arid and Semi-arid Climate and Weed Management Systems in Vegetables 2011. p. 4-8. 115. Tepe I, Celebi SZ, Kaya I, Ozkan RY. Control of smoothseed alfalfa dodder (Cuscuta approximata) in alfalfa (Medicago sativa). Int J Agric Biol 2017; 19:199-203. 116. Bhadrecha P, Kumar V, Kumar M. Medicinal plant growing under sub-optimal conditions in trans-himalaya region at high altitude. Def Life Sci J 2017; 2:37-45. 117. Bibi T, Ahmad M, Tareen RB, Tareen NM, Jabeen R, Rehman SU, et al. Ethnobotany of medicinal plants in district Mastung of Balochistan province-Pakistan. J Ethnopharm 2014; 157:79-89. 1246 An overview of the genus Cuscuta 118. Petrovska BB. Historical review of medicinal plants’ usage. Pharmacogn Rev 2012; 6:1-5. 119. Ogbulie JN, Ogueke CC, Okorondu S. Antibacterial properties of A. cordifolia, M. flurum, U. chamae, B. pinnatum, C. albidum and A. ciliata on some hospital isolates. Niger J Microbiol 2004; 18:249-255. 120. Chopra RN, Nayar L, Chopra IC. Glossary of Indian medicinal plants. New Delhi. C SIR. 1956. 121. Chopra R, Chopra I Handa K, Kapur L. Indigenous drugs of India UN Dhur and Sons. Pvt. Ltd., Calcutta. 1958. p. 358. 122. Dokuparthi SK, Banerjee N, Kumar A, Singamaneni V, Giri AK, Mukhopadhyay S. Phytochemical investigation and evaluation of antimutagenic activity of the extract of Cuscuta reflexa Roxb by Ames Test. Int J Pharm Sci Res 2014; 5:34303434. 123. Saini P, Mithal R, Menghani E. A parasitic medicinal plant Cuscuta reflexa : an overview. Int J Sci Eng Res 2015; 6:951959. 124. Singh S, Sharma A. Studies on ethnomedicinal Plant of Baghicha Jashpur Chattisgarh. J Sci Lett 2017; 2:48-55. 125. Basak S, Banerjee A, Manna CK. Role of some ethno medicines used by the Santal tribal people, of the district Bankura, WB, India, for abortifacient purposes. J Med Plants Stud 2016; 4:125-129. 126. Singh RS, Shahi SK. Diversity of medicinal plants of Ratanpur region of Bilaspur district (Chhattisgarh). J Med Plants 2017; 5:276-281. 127. Singh S. Ethnobotanical study of some climbers of Parsa district forest of Nepal. J Med Plants 2016; 4:6-10. 128. Mohapatra SS, Sarma J, Roy RK, Panigrahi S, Ganguly S. Ethnomedicinal plants used in balasore district of Odisha: a comprehensive report. Int J Cur Microbiol App Sci 2018; 7:1959-1963. 129. Kirtikar KR, Basum BD. Indian medicinal plants. Vol 1. Delhi: Periodical Experts Book Agency; 1984. 130. Darias V, Bravo L, Rabanal R, Mateo CS, Luis RG, Perez AH. New contribution to the ethnopharmacological study of the Canary Islands. J Ethnopharmacol 1989 1; 25:77-92. 131. Chowdhury M, Das AP. Folk medicines used by the Rabha tribe in Coochbehar district of West Bengal: a preliminary report. Adv Ethnobot 2007:289-296. 132. Rai Y, Kumar D. Survey on medicinal climbers in meerut district, Uttar Pradesh, India. Imperial J Interdisciplinary Res 2016; 2:603-610. 133. Patel JN, Patel NK. Study of parasite hosts of the genus Cuscuta and its traditional uses in Planpur Taluka, Gujarat, India. Ethnobot Leaf 2010; 14:126-135. 134. Dutta ML. Plants used as ethnomedicine by the Thengal Kacharies of Assam, India. Asian J Plant Sci Res 2017; 7:7-8. 135. Khalid M, Bilal M, Hassani D, Zaman S, Huang D. Characterization of ethno-medicinal plant resources of karamar valley Swabi, Pakistan. J Radiat Res Appl Sci 2017; 10:152-163. 136. Khattak NS, Nouroz F, Rahman IU, Noreen S. Ethno veterinary uses of medicinal plants of district Karak, Pakistan. J ethnopharmacol 2015; 171:273-279. 137. Kumar S, Singh BS, Singh RB. Ethnomedicinal plants uses to cure different human diseases by rural and tribal peoples of Hathras district of Uttar Pradesh. J Pharmacogn Phytochem 2017; 6:346-348. 138. Azam MN, Mannan MA, Ahmed MN. Medicinal plants used by the traditional medical practitioners of Barendra and Shamatat (Rajshahi & Khulna Division) region in Bangladesh for treatment of cardiovascular disorders. J Med Plants 2014; 2:9-14. Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta 139. Khanday ZH, Singh S. Ethnomedicinal Plants used for curing various skin diseases in Shopian district of Jammu and Kashmir. J Phytology 2017; 9:5-6. 140. Senthilkumar S, Vijayakumari K. A review-pharmacology of medicinal plants. Int J Univers Pharm Bio Sci 2016; 5:37-59. 141. Shahidullah M, Al-Mujahidee M, Uddin SN, Hossan MS, Hanif A, Bari S, et al. Medicinal plants of the Santal tribe residing in Rajshahi district, Bangladesh. Am Eur J Sustain Agric 2009; 3:220-226. 142. Singh EA, Kamble SY, Bipinraj NK, Jagtap SD. Medicinal plants used by the Thakar tribes of Raigad district, Maharashtra for the treatment of snake-bite and scorpionbite. Int J Phytother Res 2012; 2:26-35. 143. Hossan MS, Hanif A, Khan M, Bari S, Jahan R, Rahmatullah M. Ethnobotanical survey of the Tripura tribe of Bangladesh. Am Eur J Sustain Agric 2009; 3:253-261. 144. Patel H, Patel N. Sacred and medicinal plant diversity of patan sacres grove of Patan District (NG). Life Sci Leaf 2017; 92:50-60. 145. Siwakoti M, Siwakoti S. Ethnomedicinal uses of plants among the satar tribe of Nepal. J Econ Taxon Bot 2000; 24:323333. 146. Saheb TS, Rao BR, Venkateswarlu M, Swamulu M. Medicinal plants used for jaundice by the tribal people of nallamalais in Andhra Pradesh. J Pharmacogn Phytochem 2018; 7:528-531. 147. Divakara BN, Prasad S. Ethnomedicinal importance of invasive alien flora of latehar and hazaribagh districts: Jharkhand. Indian For 2015; 141:1172-1175. 148. Mahmud MR, Parvin A, Anny IP, Akter F, Tarannom SR, Moury SI, et al. Home remedies of village people in six villages of Dinajpur and Rangpur districts, Bangladesh. World J Pharm Pharm Sci 2015; 4:63-73. 149. Rahmatullah M, Khatun Z, Hasan A, Parvin W, Moniruzzaman M, Khatun A, et al. Survey and scientific evaluation of medicinal plants used by the Pahan and Teli tribal communities of Natore district, Bangladesh. Afr J Tradit Complementary Altern Med 2012; 9:366-373. 150. Seliya AR, Patel NK. Ethnomedicinal uses of climbers from Saraswati river region of Patan district, North Gujarat. Ethnobot leaf 2009; 13:865-872. 151. Qureshi R, Bhatti GR, Memon RA. Ethnomedicinal uses of herbs from northern part of Nara desert, Pakistan. Pak J Bot 2010; 42:839-851. 152. Patil JU, Biradar SD. Folkloric medicinal plants of Hingoli district, Maharashtra. 2011; 2:97-101. 153. Van Sam H, Baas P, Kebler PJ. Traditional medicinal plants in Ben En national park, Vietnam. Blumea Biodivers Evol Biogeogr Plants 2008; 53:569-601. 154. Sohn DW, Kim HY, Kim SD, Lee EJ, Kim HS, Kim JK, et al. Elevation of intracavernous pressure and NO-cGMP activity by a new herbal formula in penile tissues of spontaneous hypertensive male rats. J ethnopharmacol 2008; 120:176-180. 155. Deepakkumar R, Sabari E, Karthick M, Raysad KS. Traditionally used ethno-medicinal plants of the Kurumba communities surrounded in Thalamalai hills, Namakkal district, Tamil Nadu. South Indian J Biol Sci 2017; 3:15-26. 156. Patil SJ, Patil HM. Ethnomedicinal herbal recipes from satpura hill ranges of shirpur tahsil, dhule, maharashtra. India Res J Recent Sci 2012; 1:333-336. 157. Akbar S, Nisa M, Tariq M. CNS depressant activity of Cuscuta chinensis Lam. Int J Crude Drug Res 1985; 23:91-94. 158. Fahmy GM. Qatar biodiversity newsletter. Ostrich 2008; 2:1-5. 159. Rizk AM, El-Ghazaly GA. Medicinal and poisonous plants Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. of Qatar. University of Qatar; 1995. 160. Wang TJ, An J, Chen XH, Deng QD, Yang L. Assessment of Cuscuta chinensis seeds, effect on melanogenesis: comparison of water and ethanol fractions in vitro and in vivo. J Ethnopharmacol 2014; 154:240-248. 161. Shubhangi P, Patil DA. Herbal haircare as revealed by people in Jalgaon district, Maharashtra, India. J Exp Sci 2012; 3:32-34. 162. Ghayoumi A, Mashayekhi A. Scleroderma treatment in iranian traditional medicine: a case report. Adv Herb Med 2016; 2:1-4. 163. Tavili A, Farajollahi A, Pouzesh H, Bandak E. Treatment induced germination improvement in medicinal species of Foeniculum vulgare Miller and Cuscuta epithymum (L.) L. Mod Appl Sci 2010; 4:163-169. 164. Haq F, Ahmad H, Alam M. Traditional uses of medicinal plants of Nandiar Khuwarr catchment (District Battagram). Pakistan J Med Plants Res 2011; 5:39-48. 165. Dangwal LR, Sharma A, Rana CS. Ethnomedicinal plants of the Garhwal Himalaya used to cure various diseases: a case study. N Y Sci J 2010; 3:28-31. 166. Nedelcheva A, Pavlova D, Krasteva I, Nikolov S. Medicinal plants biodiversity and their resources of one serpentine site in the Rhodope MTS (Bulgaria). Nat Montenegr 2010; 9:373387. 167. Uniyal B, Shiva V. Traditional knowledge on medicinal plants among rural women of the Garhwal Himalaya, Uttaranchal. Indian J Tradit Knowl 2005; 4:259-266. 168. Naz R, Ayub H, Nawaz S, Islam ZU, Yasmin T, Bano A, et al. Antimicrobial activity, toxicity and anti-inflammatory potential of methanolic extracts of four ethnomedicinal plant species from Punjab, Pakistan. BMC Complement Altern Med 2017; 17:302-315. 169. Nita RD, Haresh DL. Ethno-botanical survey of some medicinal plants in jatasankar region of girnar forest, gujarat, india. Glob J Res Med Plants Indig Med 2013; 2:830-841. 170. Paudel N, Adhikari DC, Das BD. Some medicinal plants uses in ethnical group from batnagar, eastern, Nepal. Am Sci Res J Eng Tech Sci 2018; 41:233-239. 171. Ahirwar RK. Diversity of ethnomedicinal plants in Boridand forest of district Korea, Chhattisgarh, India. Am J Plant Sci 2015; 6:413-425. 172. Yaseen G, Ahmad M, Potter D, Zafar M, Sultana S, Mir S. Ethnobotany of medicinal plants for livelihood and community health in deserts of Sindh-Pakistan. In Plant and Human Health, Volume 1. Springer, Cham. 2018. p. 767-792. 173. Kala CP. Ethnomedicinal botany of the Apatani in the Eastern Himalayan region of India. J Ethnobiol Ethnomed 2005; 1:1-8. 174. Akter MH, Akter MH, Prodhan MT, Akter S, Akter N, Sultana J, et al. Documentation of plant-based remedies of a folk herbalist of Comilla district, Bangladesh. World J Pharm Pharm Sci 2017; 6:1-11. 175. Khatun MM, Rahma M. Medicinal plants used by the village Pania under Baghmara District, Bangladesh. Discovery 2018; 54:60-71. 176. Azam MN, Ahmed MN, Rahman MM, Rahmatullah M. Ethnomedicines used by the Oraon and Gor tribes of Sylhet district, Bangladesh. Am-Eurasian J Sustain Agric 2013; 7:391402. 177. Verma N, Yadav RK. Cuscuta reflexa: a paracitic medicinal plant. Plant Arch 2018; 18:1938-1942. 178. Kumar S, Sharma SD, Kumar N. Ethnobatanical study of some common plants from district hamirpur of Himachal Pradesh (India). Int J Adv Res 2015; 3:492-496. 1247 Noureen et al. 179. Shipa A, Koli S, Akter K, Shahriar SS, Rahmatullah M. Phytotherapeutic practices of a folk medicinal practitioner in Kishoreganj district, Bangladesh. J Med Plants 2018; 6:240242. 180. Khan W, Khan SM, Ahmad H. Ethno-ecology, Human Health and Plants of the Thandiani Sub Forest Division, Abbottabad, KP, Pakistan. In Plant and Human Health, Volume 1. Springer, Cham. 2018. p. 547-567. 181. Chen GT, Lu Y, Yang M, Li JL, Fan BY. Medicinal uses, pharmacology, and phytochemistry of convolvulaceae plants with central nervous system efficacies: a systematic review. Phytother Res 2018; 32:823-864. 182. Ganapaty SE, Ramaiah MA, Yasaswini KA, Kumar CR. Determination of total phenolic, flavonoid, alkaloidal contents and in vitro screening for hepatoprotective activity of Cuscuta epithymum (L) whole plant against CCl4 induced liver damage animal model. Int J Pharm Pharm Sci 2013; 5:738-742. 183. Sahranavard S, Ghafari S, Mosaddegh M. Medicinal plants used in Iranian traditional medicine to treat epilepsy. Seizure 2014; 23:328-332. 184. Farnsworth NR, Morris RW. Higher plants-the sleeping giant of drug development. Am J Pharm Sci Support Public Health 1976; 148:46-52. 185. Dillard CJ, German JB. Phytochemicals: nutraceuticals and human health. J Sci Food Agric 2000; 80:1744-1756. 186. Ahmad M, Khan MA, Zafar M, Sultana S. Ethnomedicinal demography and ecological diversification of some important weeds from district attock Pakistan. Pak J Weed Sci Res 2006; 12:37-46. 187. Dhalwal K, Shinde VM, Mahadik KR, Namdeo AG. Rapid densitometric method for simultaneous analysis of umbelliferone, psoralen, and eugenol in herbal raw materials using HPTLC. J Sep Sci 2007; 30:2053-2058. 188. Mir MA, Sawhney SS, Jassal MM. Qualitative and quantitative analysis of phytochemicals of Taraxacum officinale. Wudpecker J Pharm Pharmacol 2013; 2:1-5. 189. Almodaifer S, Alsibaie N, Alhoumendan G, Alammari G, Kavita MS. Role of phytochemicals in health and nutrition. BAO J Nutr 2017; 3:28-34. 190. Savithramma N, Rao ML, Suhrulatha D. Screening of medicinal plants for secondary metabolites. Middle East J Sci Res 2011; 8:579-584. 191. Yahara S, Domoto H, Sugimura C, Nohara T, Niiho Y, Nakajima Y, et al. An alkaloid and two lignans from Cuscuta chinensis. Phytochem 1994; 37:1755-1757. 192. Garcia MR, Erazo GS, Pena RC. Flavonoids and alkaloids from Cuscuta (Cuscutaceae). Biochem Syst Ecol 1995; 23:571572. 193. Miyahara K, Du XM, Watamab M, Sugimura C, Yahara S, Nohara T. Resin glycosides. XXIII. Two novel acylated trisaccharides related to resin glycoside from the seeds of Cuscuta chinensis. Chem Pharm bull 1996; 44:481-485. 194. Du XM, Kohinata K, Kawasaki T, Guo YT, Miyahara K. Components of the ether-insoluble resin glycoside-like fraction from Cuscuta chinensis. Phytochem 1998; 48:843-850. 195. Yen FL, Wu TH, Lin LT, Cham TM, Lin CC. Concordance between antioxidant activities and flavonol contents in different extracts and fractions of Cuscuta chinensis. Food Chem 2008; 108:455-462. 196. He XH, Yang WZ, Meng AH, He WN, Guo DA, Ye M. Two new lignan glycosides from the seeds of Cuscuta chinensis. J Asian Nat Prod Res 2010; 12:934-939. 197. Fan BY, Luo JG, Gu YC, Kong LY. Unusual ether-type resin glycoside dimers from the seeds of Cuscuta chinensis. Tetrahedron 2014; 70:2003-2014. 1248 An overview of the genus Cuscuta 198. Wang J, Tan D, Wei G, Guo Y, Chen C, Zhu H, et al. Studies on the Chemical Constituents of Cuscuta chinensis. Chem Nat Compd 2016; 52:1133-1136. 199. Ibrahim M, Rehman K, Hussain I, Farooq T, Ali B, Majeed I, et al. Ethnopharmacological investigations of phytochemical constituents isolated from the genus cuscuta. Crit Rev Eukaryot Gene Expr 2017; 27:113-150. 200. Löffler C, Czygan FC, Proksch P. Phenolic constituents as taxonomic markers in the genus Cuscuta (Cuscutaceae). Biochem Syst Ecol 1997; 25:297-303. 201. Wink M, Witte L. Quinolizidine alkaloids in Genista acanthoclada and its holoparasite, Cuscuta palaestina. J Chem Ecol 1993; 19:441-448. 202. Ye M, Li Y, Yan Y, Liu H, Ji X. Determination of flavonoids in Semen Cuscutae by RP-HPLC. J Pharm Biomed Anal 2002; 28:621-628. 203. Siddiqui MS, Memon AA, Memon S, Baloch SG. Cuscuta reflexa as a rich source of bioactive phenolic compounds. J Herbs Spices Med Plants 2017; 23:157-168. 204. Ramya B, Natrajan E, Vijaykumar S, John Vasanth MS, Muthuramsanjivani VK. Isolation and characterization of bioactive metabolites in Cuscuta reflexa Roxb. Indian J Nat Sci 2010; 1:134-139. 205. Uddin SJ, Shilpi JA, Middleton M, Byres M, Shoeb M, Nahar L, et al. Swarnalin and cis-swarnalin, two new tetrahydrofuran derivatives with free radical scavenging activity, from the aerial parts of Cuscuta reflexa. Nat Prod Res 2007; 21:663-668. 206. Tripathi VJ, Yadav SB, Upadhyay AK. A new flavanone, reflexin, from Cuscuta reflexa and its selective sensing of nitric oxide. Appl Biochem Biotechnol 2005; 127:63-67. 207. Chatterjee DP, Sahu RK. Chemical characterization of the flavonoid constituents of Cuscuta reflexa. UK J Pharm Bio Sci 2014; 2:13-16. 208. Awasthi LP. The purification and nature of an antiviral protein from Cuscuta reflexa plants. Arch Virol 1981; 70:215223. 209. Shekarchi M, Kondori BM, Hajimehdipoor H, Abdi L, Naseri M, Pourfarzib M, et al. Finger printing and quantitative analysis of Cuscuta chinensis flavonoid contents from different hosts by RP-HPLC. Food Nutr Sci 2014; 5:914-922. 210. Zhan W, Zhisheng H. Studies on the chemical constituents of the seed of chinese dodder (Cuscuta chinensis). Chin Tradit Herb drugs 1998; 9:115-117. 211. Ye M, Yan Y, Guo DA. Characterization of phenolic compounds in the Chinese herbal drug Tu-Si-Zi by liquid chromatography coupled to electrospray ionization mass spectrometry. Rapid Commun Mass Spectrom 2005; 19:14691484. 212. Tsai YC, Lai WC, Du YC, Wu SF, El-Shazly M, Lee CL, et al. Lignan and flavonoid phytoestrogens from the seeds of Cuscuta chinensis. J Nat Prod 2012; 75:1424-1431. 213. Du XM, Sun NY, Nishi M, Kawasaki T, Guo YT, Miyahara K. Components of the ether-insoluble resin glycoside fraction from the seed of Cuscuta australis. J Nat Prod 1999; 62:722725. 214. Ferraz HO, Silva MG, Carvalho R, Suffredini IB, Kato E, Arakaki F, et al. Phytochemical study and evaluation of the antimicrobial activity and cytotoxicity of Cuscuta racemosa. Rev Bras Farm 2011; 21:41-46. 215. Shailajan S, Joshi H. Optimized separation and quantification of pharmacologically active markers quercetin, kaempferol, ß-sitosterol and lupeol from Cuscuta reflexa Roxb. J Pharm Res 2011; 4:1851-1853. 216. Versiani MA, Kanwal A, Faizi S, Farooq AD. Cytotoxic cardiac glycoside from the parasitic plant Cuscuta reflexa. Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta Chem Nat Compd 2017; 53:915-922. 217. Jahan IA, Akbar PN, Enayetullah M, Ahmmad N, Nuruddin M, Ahmed MR. Elemental and fatty acid content of four medicinal plants: Kaiempferia rotunda, Cuscuta reflexa, Centella asiatica and Asparagus racemosus. European J Med Plants 2015;1-10. 218. Bais N, Kakkar A. Comparative phytochemical analysis of Cuscuta reflexa parasite grown on Cassia fistula and Ficus benghlensis by GC-MS. Int J Pharm Pharm Sci 2013; 5:350-355. 219. Rath D, Panigrahi SK, Kar DM, Maharana L. Identification of bioactive constituents from different fractions of stems of Cuscuta reflexa Roxb. Using GC-MS. Nat Prod Res 2017; 32:1977-1981. 220. Mukherjee R, Bordoloi J, Goswami A, Goswami BC. Carotenoids of dodder (Cuscuta reflexa ) grown on hedge, Clerodendrum enermy. Adv Nat Appl Sci 2008; 2:99-103. 221. Ye M, Yan Y, Ni X, Qiao L. Studies on the chemical constituents of the herba of Cuscuta chinensis. J Chinese Med Mat 2001; 24:339-341. 222. Cheng PP, Shi J, Du P, Liu DH, Cao X, Wen X. Fatty acid in the Cuscuta chinensis lam by capillary gas chromatography. Acad Periodical Farm Prod Process 2013; 8:116-118. 223. Kwon Y, Chang B, Kim C. Antioxidative constituents from the seeds of Cuscuta chinensis. Nat Prod Sci 2000; 6:135-138. 224. Xiang SX, He ZS, Ye Y. Furofuran lignans from Cuscuta chinensis. Chin J Chem 2001; 19:282-285. 225. Lin Q, Jia LY, Sun QS. Chemical constituents of the seeds of Cuscuta chinensis Lam.[J]. J Shenyang Pharm Univ 2009; 12:110. 226. Oh H, Kang DG, Lee S, Lee HS. Angiotensin converting enzyme inhibitors from Cuscuta japonica Choisy. J Ethnopharmacol 2002; 83:105-108. 227. Baccarini A, Bertossi F, Bagni N. Carotenoid pigments in the stem of Cuscuta australis. Phytochemistry 1965; 4:349351. 228. Hongzhu G, Jiashi L. Study on constituents of the seed from Cuscuta Australis. J Beijing Univ Tradit Chin Med 2000; 23:20-23. 229. Guo H, Li J. Study on flavonoids of Cuscuta australis R. Br. China J Chin Materia Med 1997; 22:38-39. 230. Sousa AL, Sales QS, Braz-Filho R, de Oliveira RR. Lignans and flavonoids isolated from Cuscuta racemosa Mart. & Humb (Convolvulaceae) by droplet counter-current chromatography. J Liq Chromatogr R T 2012; 35:2294-2303. 231. Bacchi EM. Flavonoids from Cuscuta racemosa. Planta Medi 1993; 59:605-606. 232. Abdallah WE, Elsayed WM, Abdelshafeek KA. Chemical constituents and radical scavenging activity of Cuscuta pedicellata seed extracts. Int J ChemTech Res 2016; 9:580-587. 233. Szymańska R, Kruk J. Tocopherol content and isomers’ composition in selected plant species. Plant Physiol Biochem 2008; 46:29-33. 234. Prior RL, Cao G. Antioxidant phytochemicals in fruits and vegetables: diet and health implications. Hort Sci 2000; 35:588-592. 235. Rice-Evans C. Flavonoids and isoflavones: absorption, metabolism, and bioactivity. Free Radic Biol Med 2004; 7:827828. 236. Chanda S, Dave R, Kaneria M, Nagani K. Seaweeds: A Novel, Untapped Source of Drugs From Sea to Combat Infectious Diseases. Current research, Technology And Education Topics In Applied Microbiology Microbial Biotechnology 2010. p. 473-480. 237. Tanruean K, Poolprasert P, Kumla J, Suwannarach N, Lumyong S. Bioactive compounds content and their biological Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. properties of acetone extract of Cuscuta reflexa Roxb. grown on various host plants. Nat Prod Res 2017; 3:1-4. 238. Hussain SA, Farheen S, Sultana T, Tabassum A, Hussain SI, Khan R. Evaluation of anticovulsant and anioxidant activity of selected medicinal plants. World J Pharm Pharm Sci 2017; 6:1899-1914. 239. Yen FL, Wu TH, Lin LT, Cham TM, Lin CC. Nanoparticles formulation of Cuscuta chinensis prevents acetaminopheninduced hepatotoxicity in rats. Food Chem toxicol 2008; 46:1771-1777. 240. Gao JM, Li R, Zhang L, Jia LL, Ying XX, Dou DQ, et al. Cuscuta chinensis seeds water extraction protecting murine osteoblastic MC3T3-E1 cells against tertiary butyl hydroperoxide induced injury. J Ethnopharmacol 2013; 148:587-595. 241. Zhen GH, Jiang B, Bao YM, Li DX, An LJ. The protective effect of flavonoids from Cuscuta chinensis in PC12 cells from damage induced by H2O2. J Chin Med Mater 2006; 29:10511055. 242. Amaresh P, Seemanchala R, Debashis P, Arpan M, Bijan G, Kumar BN. hepatoprotective activity of whole part of the plant Cuscuta reflexa Roxb.(Convolvulaceae) in chloroform, ethanol and paracetamol induced hepatotoxic rat models. Int J Pharm Clin Res 2014; 6:127-132. 243. Taghizadieh M, Issabeagloo E, Valiloo MR, Afshari F, Asadi J. Hepatoprotective and antioxidant activity of ethanolic extract of aerial parts of Cuscuta reflexa Robx. on liver damage due to cisplantin in rats. Baltica 2014; 27:274-279. 244. Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract 2010; 87:4-14. 245. Rath D, Kar DM, Panigrahi SK, Maharana L. Antidiabetic effects of Cuscuta reflexa Roxb. in streptozotocin induced diabetic rats. J Ethnopharmacol 2016; 192:442-449. 246. Ma JZ, Yang LX, Shen XL, Qin JH, Deng LL, Ahmed S, et al. Effects of traditional Chinese medicinal plants on anti-insulin resistance bioactivity of DXMS-induced insulin resistant HepG2 cells. Nat Prod Bioprospect 2014; 4:197-206. 247. Li DZ, Peng DY, Zhang R, Xu XX. Effects of Cuscuta chinensis polysaccharide on diabetic mice by alloxan. Anhui Med Pharm J 2008; 12:900-911. 248. Li XJ, You HY, Yang J, Liu BM, You G, Song Y. Aqueous extracts of Cuscuta chinensis Lam induces differentiat ion of amelanotic melanocytes of human hair follicles. Chin J Dermatovenereol 2008; 22:13-15. 249. Xu XX, Li DZ, Peng DY, Zhang R. Effects of Cuscuta chinensis Polysaccharide on Glucose-lipid Metabolism in Diabetic Rats. Chin J Exp Tradit Med Formula 2011; 17:232-234. 250. Lei X, He J, Ren C, Zhou Y, Chen X, Dou J. Protective effects of the Chinese herbal medicine prescription Zhujing pill on retina of streptozotocin-induced diabetic rats. Biomed Pharmacother 2018; 98:643-650. 251. Al-Sultany, Fadia H, Al-Saadi A H, Al-Husainy IM. Evaluated the Up–regulation in gene expression of hepatic insulin gene and hepatic insulin receptor gene in type 1 diabetic rats treated with Cuscuta chinesis Lam. J Babylon Univ 2018; 26:75-93. 252. Trinchieri G. Cancer and inflammation: an old intuition with rapidly evolving new concepts. Annu Rev Immunol 2012; 30:677-706. 253. Suresh V, Sruthi V, Padmaja B, Asha VV. In vitro antiinflammatory and anti-cancer activities of Cuscuta reflexa Roxb. J Ethnopharmacol 2011; 134:872-877. 254. Katiyar NS, Singh AP, Gangwar AK, Rao NV. Evaluation of carrageenan induced antiinflammatory activity of stem extracts of Cuscuta reflexa (Roxb) in rats. Int J Res Pharm Chem 1249 Noureen et al. 2015; 5:322-326. 255. Kang SY, Jung HW, Lee MY, Lee HW, Chae SW, Park YK. Effect of the semen extract of Cuscuta chinensis on inflammatory responses in LPS-stimulated BV-2 microglia. Chin J Nat Med 2014; 12:573-581. 256. Liao JC, Chang WT, Lee MS, Chiu YJ, Chao WK, Lin YC, et al. Antinociceptive and anti-inflammatory activities of Cuscuta chinensis seeds in mice. The Am J Chin Med 2014; 42:223-242. 257. Koca U, Küpeli-Akkol E, Sekeroglu N. Evaluation of in vivo and in vitro biological activities of different extracts of Cuscuta arvensis. Nat Prod Commun 2011; 6:1433-1436. 258. Islam R, Rahman MS, Rahman SM. GC-MS analysis and antibacterial activity of Cuscuta reflexa against bacterial pathogens. Asian Pac J Trop Dis 2015; 5:399-403. 259. Pal DK, Mandal M, Senthilkumar GP, Padhiari A. Antibacterial activity of Cuscuta reflexa stem and Corchorus olitorius seed. Fitoterapia 2006; 77:589-591. 260. Bibi Y, Naeem J, Zahara K, Arshad M, Qayyum A. In Vitro antimicrobial assessment of selected plant extracts from pakistan. Iran J Sci Technol A 2018; 42:267-272. 261. Okiei W, Ogunlesi M, Ademoye MA. An assessment of the antimicrobial properties of extracts of various polarities from Chasmanthera dependens, Emilia coccinea and Cuscuta australis, herbal medications for eye diseases. J Appl Sci 2009; 9:4076-4080. 262. Bonjar S. Evaluation of antibacterial properties of some medicinal plants used in iran. J Ethnopharmacol 2004; 94:301305. 263. Abdullah JA, Hammadi AA, Hakem R, Hatef Z, Hussein N. Study effect of plant extraction for Cuscuta europaea (Dodder) against two species of bacteria Staphylococcus aureus and Escherichia coli. J Contemp Med Sci 2016; 2:133-137. 264. Etedali P, Behbahani M, Rahiminejad RM, Rad SJ. Effect of crude extracts and fractions of Cuscuta campestris and two different hosts on peripheral blood mononuclear cells and HIV replication. Int J Biosci 2014; 4:83-89. 265. Ahmed HM, Yeh JY, Tang YC, Cheng WT, Ou BR. Molecular screening of chinese medicinal plants for progestogenic and anti-progestogenic activity. J Biosci 2014; 39:453-461. 266. Alaoui-Jamali M, editor. Alternative and complementary therapies for cancer: Integrative approaches and discovery of conventional drugs. Springer Science Business Media; New York, USA, 2010. p.63. 267. Zeraati F, Zamani A, Goodarzi MT, Hashjin SM, Razzaghi K. In vitro cytotoxic effects of Cuscuta chinensis whole extract on human acute lymphoblastic leukemia cell line. Iran J Med Sci 2015; 35:310-314. 268. Choi EJ, Kim GH, Kim T. Equol induced the apoptosis via cell cycle arrest in MDA-MB-453 but not in MCF-7 cells. Faseb J 2008; 22:265-265. 269. Magee PJ, Raschke M, Steiner C, Duffin JG, Pool-Zobel BL, Jokela T, et al. Equol: a comparison of the effects of the racemic compound with that of the purified S-enantiomer on the growth, invasion, and DNA integrity of breast and prostate cells in vitro. Nutr Cancer 2006; 54:232-242. 270. Selvi EK, Turumtay H, Demir A, Turumtay EA. Phytochemical profiling and evaluation of the hepatoprotective effect of Cuscuta campestris by high-performance liquid chromatography with diode array detection. Anal Lett 2018; 51:1464-1478. 271. Behbahani M. Evaluation of in vitro anticancer activity of Ocimum basilicum, Alhagi maurorum, Calendula officinalis and their parasite Cuscuta campestris. PloS one 2014; 9:1-13. 272. Pan HJ, Sun HX, Pan YJ. Adjuvant effect of ethanol extract of semen cuscutae on the immune responses to ovalbumin in 1250 An overview of the genus Cuscuta mice. J Ethnopharmacol 2005; 99:99-103. 273. Lin MK, Yu YL, Chen KC, Chang WT, Lee MS, Yang MJ, et al. Kaempferol from semen cuscutae attenuates the immune function of dendritic cells. Immunobiology 2011; 216:11031109. 274. Lin HB, Lin JQ, Lu N, Yi XY. Comparative study on immune enhancement effects of four kinds of dodder seeds in shandong province. J Chin Integr Med 2003; 1:51-53. 275. Xiao J, Cui F, Ning T, Zhao W. Effects of alcohol extract from Polygonatum odoratum and Cuscuta australis on immunological function of mice injured by burns. Chin J Chin Mater Med 1990; 15:557-559. 276. Gu LG, Ye M, Yan YN, Jia L, Zhao JQ. Study of Cuscuta australis hyperoside on immunological function of mice in vivo and in vitro. Chin J Tradit Chin Med Inf 2001; 8:42-44. 277. Jian-Hui L, Bo J, Yong-Ming B, Li-Jia A. Effect of Cuscuta chinensis glycoside on the neuronal differentiation of rat pheochromocytoma PC12 cells. Int J Dev Neurosci 2003; 21:277-281. 278. Liu ZY, Yang YG, Zheng B. Effect of improving memory and inhibiting acetylcholinesterase activity by invigorating-qi and warming-yang recipe. Chin J Integr Tradit West Med 1993; 13:675-676. 279. Yu Q, Song FJ, Chen JF, Dong X, Jiang Y, Zeng KW, et al. antineuroinflammatory effects of modified wu-zi-yan-zong prescription in β-amyloid-stimulated BV2 microglia via the NF-κB and ERK/p38 MAPK signaling pathways. J Evid Based Complementary Altern Med 2017; 2017:1-10. 280. Cai XG, Xu AX, Ge B, Gao X, Yang SH. Effects of a polysaccharide from CCL on inhibiting oxygen free radical threshold of senile mice model. Acta Acad Med Mil Tertiae 2005; 27:1326-1328. 281. Li CS, Deng HB, Li DD, Li ZH. Advances and challenges in screening traditional chinese anti-aging materia medica. Chin J Integr Med 2013; 19:243-252. 282. Yang FY, Huang J. “Tai Ping Sheng Hui Fang” in the antiaging effects medical research. J Guiyang Coll of Tradit Chin Med 1998; 2:7-8. 283. Gilani AU, Aftab K. Pharmacological actions of Cuscuta reflexa. Int J Pharma 1992; 30:296-302. 284. Yao CH, Tsai CC, Chen YS, Chang CJ, Liu BS, Lin CC, et al. Fabrication and evaluation of a new composite composed of tricalcium phosphate, gelatin and Chi-Li-Saan as a bone substitute. Am J Chin Med 2002; 30:471-482. 285. Yang HM, Shin HK, Kang YH, Kim JK. Cuscuta chinensis extract promotes osteoblast differentiation and mineralization in human osteoblast-like MG-63 cells. J Med Food 2009; 12:8592. 286. Yang M, Sun J, Lu Z, Chen G, Guan S, Liu X, et al. Phytochemical analysis of traditional chinese med using liquid chromatography coupled with mass spectrometry. J Chromatogr A 2009; 1216:2045-2062. 287. Shin S, Lee YJ, Kim EJ, Lee AS, Kang DG, Lee HS. Effect of Cuscuta chinensis on renal function in ischemia/reperfusioninduced acute renal failure rats. Am J Chin Med 2011; 39:889902. 288. Sun K, Zhao C, Chen XF, Kim HK, Choi BR, Huang YR, et al. Ex vivo relaxation effect of Cuscuta chinensis extract on rabbit corpus cavernosum. Asian J Androl 2013; 15:134-137. 289. Peng SJ, Lu RK, Yu LH. Effect of semen cuacutae, rhizoma curculiginis, radix morindae, officinalis on human spermatozoa’s motility and membrane function in vitro. Chin J West Med 1997; 17:145-147. 290. Shah GR, Chaudhari MV, Patankar SB, Pensalwar SV, Sabale VP, Sonawane NA. Evaluation of a multi-herb supplement for Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 An overview of the genus Cuscuta erectile dysfunction: a randomized double-blind, placebocontrolled study. BMC Complementary Altern Med 2012; 12:155-163. 291. Linmao Y. Integrating chinese and western medicine to treat infantile uterus. Herb J Tradit Chin Med 1992; 14:40-54. 292. Jang H, Bae WJ, Kim SJ, Cho HJ, Yuk SM, Han DS, et al. The herbal formula KH-204 is protective against erectile dysfunction by minimizing oxidative stress and improving lipid profiles in a rat model of erectile dysfunction induced by hypercholesterolaemia. BMC Complementary Altern Med 2017; 17:129-140. 293. Yang J, Wang Y, Bao Y, Guo J. The total flavones from semen cuscutae reverse the reduction of testosterone level and the expression of androgen receptor gene in kidney-yang deficient mice. J Ethnopharmacol 2008; 119:166-171. 294. Wang J, Wang M, Ou Y, Wu Q. Effects of flavonoids from semen cuscutae on changes of beta-EP in hypothalamuses and FSH and LH in anterior pituitaries in female rats exposed to psychologic stress. J Chin Med Mater 2002; 25:886-888. 295. Ma HX, You ZL, Wang RG. Effect of total flavones from Cuscuta chinensis on expression of Th type-1/Th type-2 cytokines, serum P and PR in abortion rats model. J Chin Med Mater 2008; 31:1201-1204. 296. Ma HX, You ZL, Wang XY. Effect of total flavones from Cuscuta chinensis on expression of Fas/FasL, PCNA and HBEGF in SD rats model with bromocriptine-induced abortion. J Chin Med Mater 2008; 31:1706-1709. 297. Zhu JF, She YC, Zhou CH. Experimental and clinical studies on the effect of Shou Tai Wan and additives on threatened abortion. J Integr Tradit Western Med 1987; 7:407-409. 298. Aqil F, Zahin M, Ahmad I. Antimutagenic activity of methanolic extracts of four ayurvedic medicinal plants. Indian J Exp Biol 2008; 46:668-672. 299. Nakahara K, Trakoontivakorn G, Alzoreky NS, Ono H, Onishi-Kameyama M, Yoshida M. Antimutagenicity of some edible Thai plants, and a bioactive carbazole alkaloid, mahanine, isolated from Micromelum minutum. J Agric Food Chem 2002; 50:4796-4802. 300. Zweier JL, Talukder MH. The role of oxidants and free radicals in reperfusion injury. Cardiovasc Res 2006; 70:181190. 301. Veinot JP, Gattinger DA, Fliss H. Early apoptosis in human myocardial infarcts. Hu Pathol 1997; 28:485-492. 302. Rabkin SW. Apoptosis in human acute myocardial infarction: the rationale for clinical trials of apoptosis inhibition in acute myocardial infarction. Sch Res Exch 2009; 2009:1-10. 303. Sun SL, Guo L, Ren YC, Wang B, Li RH, Qi YS, et al. Antiapoptosis effect of polysaccharide isolated from the seeds of Cuscuta chinensis lam on cardiomyocytes in aging rats. Mol Biol Rep 2014; 41:6117-6124. 304. Zhongrong L, Pengtie L, Tiejun F, Yuanqiao J, Ruozhu W. The effect of three extraction technique of Chinese dodder seed on cardiovascular activity. Nat Prod Res Develop 2004; 16:532-533. 305. Thomas S, Shrikumar S, Velmurugan C, Kumar BA. Evaluation of anxiolytic effect of whole plant of Cuscuta reflexa. World J Pharm Sci 2015; 4:1245-1253. 306. Mokhtarifar N, Sharif B, Naderi N, Mosaddegh M, Faizi M. Evaluation of anti-depressant effects of Cuscuta chinensis in experimental models. Res Pharm Sci 2012; 7:826-827. 307. Cheng D, Murtaza G, Ma S, Li L, Li X, Tian F, et al. In silico prediction of the anti-depression mechanism of a herbal formula (tiansi liquid) containing Morinda officinalis and Cuscuta chinensis. Molecules 2017; 22:1614-1630. Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019 Noureen et al. 308. Firoozabadi A, Zarshenas MM, Salehi A, Jahanbin S, Mohagheghzadeh A. Effectiveness of Cuscuta planiflora ten and Nepeta menthoides Boiss & Buhse in major depression: a triple-blind randomized controlled trial study. J Evi based Complementary Altern Med 2015; 20:94-97. 309. Shen L, Huang YY, Wang XN, Du J, Wang YY, Zhang DQ. Pharmacological effect of cuscutae Semen by external useon experimental vitiligo in guinea pigs. Chin J Exp Tradit Med Formulae 2012; 18:199-202. 310. Wang TJ, An J, Chen XH, Deng QD, Yang L. Assessment of Cuscuta chinensis seeds, effect on melanogenesis: comparison of water and ethanol fractions in vitro and in vivo. J Ethnopharmacol 2014; 154:240-248. 311. Mojtabaee M, Mokaberinejad R, Hamzeloo-Moghadam M, Nasab MR, Adhami S, Farshi S, et al. The effect of the traditional medicine product” Milk-Cuscuta” on skin hyperpigmentation in patients with melasma. Middle East J Family Med 2018; 7:204-211. 312. Suk KD, Lee SJ, Bae JM. Inhibitory effects of Cuscuta japonica extract and C. australis extract on mushroom tyrosinase activity. Korean J Pharma 2004; 35:380-383. 313. Patel S, Sharma V, Chauhan NS, Dixit VK. A study on the extracts of Cuscuta reflexa Roxb in treatment of cyclophosphamide induced alopecia. Daru J Pharm Sci 2014; 22:27-34. 314. Huang AG, Yi YL, Ling F, Lu L, Zhang QZ, Wang GX. Screening of plant extracts for anthelmintic activity against Dactylogyrus intermedius (Monogenea) in goldfish (Carassius auratus). Parasitol Res 2013; 112:4065-4072. 315. Moon M, Jeong HU, Choi JG, Jeon SG, Song EJ, Hong SP, et al. Memory-enhancing effects of Cuscuta japonica Choisy via enhancement of adult hippocampal neurogenesis in mice. Behav Brain Res 2016; 311:173-182. 316. Mehrabani M, Modirian E, Ebrahimabadi AR, Vafazadeh J. Study of the effects of hydro-methanol extracts of Lavandula vera DC and Cuscuta epithymum Murr on the seizure induced by pentylentetranzol in mice. J Kerman Univ Med Sci 2014; 14:25-32. 317. Gupta MA, Mazumder UK, Pal D, Bhattacharya S, Chakrabarty SU. Studies on brain biogenic amines in methanolic extract of Cuscuta reflexa Roxb and Corchorus olitorius Linn seed treated mice. Acta Pol Pharma 2003; 60:207-210. 318. Ohta Y, Sami M, Kanda T, Saito K, Osada K, Kato H. Gene expression analysis of the anti-obesity effect by apple polyphenols in rats fed a high fat diet or a normal diet. J Oleo Sci 2006; 55:305-314. 319. Kakhorova KA, Khashimova ZS, Terenteva EO. Studies on cytotoxicity and antioxidant activities of lectin-like proteins from phytoparasites (Cuscuta eurospaea). Asian Pharm Pharmacol 2018; 4:265-270. 320. Bhan S, Mohan L, Srivastava CN. Efficacy of Cuscuta reflexa extract and its synergistic activity with Temephos against mosquito larvae. Int J Mosquito Res 2015; 2:34-41. 321. Alamgeer, Niazi SG, Uttra AM, Qaiser MN, Ahsan H. Appraisal of anti-arthritic and nephroprotective potential of Cuscuta reflexa. Pharm Biol 2017; 55:792-798. 322. Gautam T, Thakur V, Gupta V. Phytochemical Screening and Wound Healing Potential of Cuscuta reflexa Roxb. Int J Pharm Life Sci 2018; 9:21-21. 323. Sakib MH, Hossain MS, Hossain MS, Al Mahmood A, Sarkar MY, Rahman S, Shill LK. In-vitro cytotoxicity and antioxidant property evaluation from methanolic extract of Cuscuta Reflexa flowers. Asian J Med Biol Res 2015; 1:285-291. 324. Manirujjaman M, Suchana S, Collet T, Nawshin LN, Chowdhury MA. Antimicrobial effects of ethanolic extracts 1251 Noureen et al. from Cuscuta reflexa Roxb (Convolvulaceae). Int J Pharmacogn Phytochem Res 2016; 8:930-932. 325. Praseeja RJ, Sreejith PS, Asha VV. Studies on the apoptosis inducing and cell cycle regulatory effect of Cuscuta reflexa Roxb chloroform extract on human hepatocellular carcinoma cell line, Hep 3B. Int J Appl Res Nat Prod 2015; 8:37-47. 326. Roohina Ali S, Haque S, Versiani MA, Faizi S, Farooq 1252 An overview of the genus Cuscuta AD. Cytotoxicity and chromosomal aberrations induced by methanolic extract of Cuscuta reflexa and its pure compounds on meristematic cells of Allium species. Pak J Pharm sci 2017; 30:521-529. 327. Mala FA, Sofi MA. Evaluation of antihistaminic Activity of herbal drug isolated from Cuscuta reflexa Roxb. Ann Plant Sci 2017; 6:1807-1810. Iran J Basic Med Sci, Vol. 22, No. 11, Nov 2019