Academia.eduAcademia.edu
International Journal of Biological Chemistry 9 (6): 318-331, 2015 ISSN 1819-155X / DOI: 10.3923/ijbc.2015.318.331 © 2015 Academic Journals Inc. A Review of the Medicinal Plants of Genus Orthosiphon (Lamiaceae) 1 Mukesh K. Singh, 1Bina Gidwani, 1Anshita Gupta, 1 Chanchal Deep Kaur, 2Pranita P. Kashyap and 3D.K. Tripathi 1 Hemant Dhongade, 1 Shri Rawatpura Sarkar Institute of Pharmacy, Kumhari, Durg, Chhattisgarh, India Maharashtra Institute of Pharmacy, Bramhapuri, Chandrapur, Maharashtra 3 Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh, India 2 Corresponding Author: Mukesh Kumar Singh, Shri Rawatpura Sarkar Institute of Pharmacy, Kumhari, Durg, Chhattisgarh, India Tel: +91-9691699320 ABSTRACT In the genus Orthosiphon (Lamiaceae), Orthosiphon aristatus, Orthosiphon pallidus, Orthosiphon thymiflorus, Orthosiphon stamineus are widely used in traditional medicine to prevent different diseases such as diabetes, kidney stone, edema, rheumatism, hepatitis, hypertensive and jaundice. A different variety of phytoconstituents has been isolated from the Orthosiphon species which include monoterpenes, diterpenes, triterpenes, saponins, organic acid and flavonoids compound. Antidiabetic, anti-inflammatory, antioxidant, hepatoprotective, analgesic and nephroprotective activities have been reported in the plant extract and phytoconstituents. Hence, the purpose of this review is to provide a comprehensive report about the Orthosiphon genus based on its toxicity in order to identify its therapeutic potential and future prospects for betterment of research. Key words: Orthosiphon, phytochemistry, pharmacological activities, toxicity studies INTRODUCTION The genus Orthosiphon was coined from two Latin words, iorthos and siphon. The words referred to straight while siphon meant tube like or cylindrical. These two words actually referred to the straight tube like flowers that were produced by the Orthosiphon species and this was considered as one of the main characteristics of the Labiatae or Lamiaceae family (Keng and Siong, 2006). The genus Orthosiphon benth in tribe ocimeae comprises 40 species and was recorded from the old world: in tropical and subtropical Asia including Southern Africa and Madagascar. The species usually occurs in grassland, woodland or forest margins (Sadashiva et al., 2013). Some of these species are important medicinal plants that are used in herbolism and thought to have medicinal properties. Up to date, the genus provided a large number of chemical compounds of which some indicated dynamic pharmacological activity (Sundarammal et al., 2012). Orthosiphon aristatus has long history of medicinal use in Indonesia, Malaysia, Southeast Asia, this plant was initially recorded as a treatment for diabetes, kidney stone and hypertension (Matsubara et al., 1999; Ohashi et al., 2000; Masuda et al., 1992; Shibuya, 1999). Orthosiphon pallidus is herbaceous shrub native to South East Asia and India has been used to treat urinary lithiasis, edema, fever, influenza, rheumatism, hepatitis and jaundice (Kiruthika and Meenakshi, 2011). Orthosiphon thymiflorus used in India to treat cytotoxic, diabetic, 318 Int. J. Biol. Chem., 9 (6): 318-331, 2015 anti-inflammatory and hypertensive (Sundarammal et al., 2012; Sini et al., 2012; Kavimani et al., 1997). Orthosiphon stamineus is used to treat diabetes, hypertension, oedema, epilepsy, fever, influenza and jaundice (Arafat et al., 2008; Akowuah et al., 2005; Ho et al., 2010; Awale et al., 2003a). The traditional indigenous uses and pharmacology of ethnobotanic herbs provides basic knowledge for further development of medicinal plants and a useful approach for drug discovery (Heinrich and Gibbons, 2001). The genus Orthosiphon comprises an impressive number of species some of which have been used in traditional medicine. Hence the purpose of this review is to provide a comprehensive report about the genus based on its toxicity in order to identify its therapeutic potential and future prospects for betterment of research. This will be possible through analysis of collected data related to botany, local and traditional uses, pharmacology and toxicology of Orthosiphon species. BOTANICAL DESCRIPTION Orthosiphon plants are herbaceous shrubs which grow to a height of 1.5 m. Orthosiphon is a popular garden plant with whitish flower having unique identification and bluish filaments resembling a cast’s whiskers. Orthosiphon pallidus Royle ex Benth, O. aristatus, O. thymiflorus and O. stamineus are commonly used in traditional medicines. The morphology characteristics of O. pallidus are as follows: perennial herb with a woody root stock not aromatic. Stems are diffusely branched ascending erect 10-35 cm, slender, quadrangular, velvety or almost hairless. Leaves are ovate, 1-3.5×1.2, palegreen, slightly fleshy, nearly entire to saw-toothed, gland-dotted, stalked, velvety to almost hairless. Flower stalks are 2 mm in flower and up to 6 mm in fruit, velvety in lower part, upper lobe ovate-circular. Orthosiphon aristatus is slender, smooth or hairy undershrub 30-60 cm high. Leaves in distant pair, narrowed in to the stalk ovate, 5-10 cm long, pointed at bath ends, coarsely toothed margins. The flowers are borne with extreme lax racemes. The calyxes of flowers have naked throat and bell shape with two slender lower teeth. Corolla is purple-white in color with 2.5 cm long and smooth. Nutlets are oblong and compressed. Orthosiphon thymiflorus straggling, somewhat shrubby perennial herb up to 1.5 m tall not or hardly aromatic. Stems several ascending to erect to 4-angled, normally well branched, retrorsely pubescent along the angles and sometimes with dense hairs. Leaves are usually oval or elliptic with 1.4-4.5 cm long but larger in well shaded plants, glandular punctuate and hairless/pubescent mostly along the veins beneath, margin scalloped or toothed, petiole up to 25 mm long. Orthosiphon stamineus is a perennial herb. It attains 0.3-1.5 m high and 4 angle stem. Leaves are simple, opposite, ovate, oblong lanceolate, elliptic orrhamboid, which have 2-4 cm wide and 4-7 cm long. The flowers are white, blue or violet. TRADITIONAL USES OF SELECTED SPECIES The plants of genus Orthosiphon have been used by the various parts of Asia and Africa. Traditional uses of selected Orthosiphon species (Table 1) point to their importance especially in the treatment of diabetes, kidney stone, influenza, hepatitis and jaundice. In order to cover all published botanical names, a list of synonyms based on the relevant taxonomic literature is provided (Table 2). The list encompasses representatives of the genus that have ethanomedicinal relevance according to the present comprehensive literature review. 319 Table 1: Traditional uses, pharmacological and biological activities of selected Orthosiphon species Species Region Plant part Traditional uses Pharmacological activity Orthosiphon Indonesia Dried leaves and Used in hypertension Antibacterial activity aristatus and Malaysia tops of stem and diabetes Indonesia Leaves Used as a diuretics Diuretic effects and Malaysia Indonesia Dried leaves Used in hypertension Antihypertensive Used in diabetes Southeast Asia and Australia Dried leaves Aerial part 320 Baluchistan, Arabia, Whole plant India (Kashmir, Punjab, (coarse powder) West Bihar and Southwards to Travancore) Orthosiphon Aliyar foot hills of valparal, Fresh leaves thymiflorus Coimbatore, Tamilnadu Treatment of renal inflammation used in Kidney stone Used in dysuria Used to treat urinary lithiasis, edema, influenza, rheumatism, hepatitis and jaundice Treatment of neurasthenia, general tonic and aphrodisiac Antioxidant Reference Chen et al. (1989) Aqueous extract Chen et al. (1999) Water decoction Matsubara et al. (1999), Ohashi et al. (2000), Masuda et al. (1992) and Shibuya et al. (1999) Di et al. (2013) and Hsu et al. (2010) Antioxidant and anti-inflammatory Methanol, ethanol and water extract Antioxidant and anti-inflammatory Anticancer (51.74% cytotoxicity) Hexane extract Lower the blood pressure and inhibition of heart of pithed frog Absolute alcohol and Basu and Singh Antioxidant, cytotoxic and vasodialative Absolute alcohol Di et al. (2013) and Hsu et al. (2010) Ashokan and Muthuraman (2011) Basu and Sing (1956) (1956) Hydrodistilation, Sundarammal et al. Clevenger and (2012) apparatus Attapady palakkad and Powdered plant Anticancer Cytotoxic activity, anti Imbbibition, Sini et al. (2012) Kerala material diabetic, antihepatotoxic, meceration and antibacterial and hypertensive percolation in chloroform Tirunelveli and Tamilnadu Whole plant Aquaretic Diuretic activity and Meceration in boiling Kavimani et al. (1997) anti-inflammetory water Maruthamalal hills Dried leaves Larvacidal Larvacidal activity Hexane, chloroform, Kovendan et al. (2012) Coimbatore and Tamilnadu ethyl acetate, acetone and methanol Orthosiphon Malaysia, Indonesia and Leaf part (fresh) Treating stone diseases Bladder inflammation, food preservative, Methanol, Chloroform, Awale et al. (2003a, b), stamineus Japan and gout, Java tea and inhibitory effect in growth of calcium Ethyl acetate and Hossain and Ismail decocted leaves as crystal, diabetes, hypertension, Acetone (2013), Akowuah et al. diuretics rheumatism, tonsillitis, menstrual (2005), Ho et al. (2010), disorder, urinary lithiasis, biliary Arafat et al. (2008), litiasis, epilepsy, oedema, eruptive fever, Akowuah et al. (2005) hepatitis, jaundice, influenza, gonorrhoea, and Hossain and Ismail syphilills, renal calculus, gallstone, (2013) diuretics, inhibitory activity of nitric oxide and body detoxification Myanmar Leaf part (dried) Antidiabetics to treat Diabetes, urinary tract and renal Methanol Awale et al. urinary tract and renal diseases (2003a, b, 2004) and diseases Han et al. (2008) China, Indonesia and Arial part Urinary lithiasis, edema, eruptive fever, Methanol Awale et al. Veitnam influenza, hepatitis and jaundice (2003b, 2004) and Paton et al. (2004) Int. J. Biol. Chem., 9 (6): 318-331, 2015 Southeast Asia and Australia Orthosiphon Africa and pallidus South-East Asia Dried leaves Active extract Aqueous extract Int. J. Biol. Chem., 9 (6): 318-331, 2015 Table 2: Representatives of genus Orthosiphon used in traditional medicine and their synonyms Orthosiphon species Synonyms Orthosiphon adenocaulis Orthosiphon adornatus, Orthosiphon affinis Benth, Orthosiphon adscendence and Orthosiphon albiflorus Orthosiphon allenii Orthosiphon amabilis, Orthosiphon ambiguous Bolus and Orthosiphon angolensis Orthosiphon aristatus var. aristatus Orthosiphon asperus, Orthosiphon atacorensis, Orthosiphon australis and Orthosiphon bartsioides Orthosiphon biflorus Orthosiphon bodinieri, Orthosiphon bolusii, Orthosiphon bracteatus, Orthosiphon brevicaulis, Orthosiphon buchananii and Orthosiphon bracteotus Orthosiphon bullosus Orthosiphon buryi, Orthosiphon calaminthoides, Orthosiphon cameronii, Orthosiphon canescens and Orthosiphon capitatus Orthosiphon cladotrichos Orthosiphon cleistocalyx, Orthosiphon colouratus, Orthosiphon comosus Wight and Orthosiphon comosus Baker Orthosiphon cuanzae Orthosiphon debilis, Orthosiphon decipiens, Orthosiphon degasparisianum and Orthosiphon diffuses Orthosiphon discolor Orthosiphon dissitifolius, Orthosiphon ehrenbergii, Orthosiphon ellenbecki and Orthosiphon elliottii Orthosiphon ellipticus Orthosiphon emirnensis and Orthosiphon engleri Perkins Orthosiphon ferruginous Orthosiphon foliosus Orthosiphon fruticosus Orthosiphon gerrardii, Orthosiphon glabratus Benth, Orthosiphon glabratus var. Palviflorus (Benth) and Orthosiphon glabrascene Orthosiphon glandulosus Orthosiphon glutinosus Chiov., Orthosiphon gofensis S. Moore and Orthosiphon grandiflorus Bold. Orthosiphon hanningtonii Orthosiphon helenae Buscal, Orthosiphon heterochrous Briq, Orthosiphon heterophyllus Gurke, Orthosiphon hildebrandtii Vatke, Orthosiphon hildebrandtii Baker, Orthosiphon hispidus Benth., Orthosiphon hockii, Orthosiphon holubii and Orthosiphon homblei Orthosiphon humbertii Orthosiphon humilis, Orthosiphon incisus and Orthosiphon inconcinnus Orthosiphon incurvus Orthosiphon inodorus, Orthosiphon iodocalyx Briq, Orthosiphon johnstonii Baker, Orthosiphon kelleri Briq, Orthosiphon kirkii Baker and Orthosiphon labiatuss Orthosiphon lanatus Doan Orthosiphon lanceolatus Gurke, Orthosiphon lanceolatus, Orthosiphon latidens, Orthosiphon laurentii, Orthosiphon liebrechtsiauum, Orthosiphon linraris Benth, Orthosiphon longipes Baker, Orthosiphon macranthus, Orthosiphon macrocheilus, Orthosiphon macrophyllus, Orthosiphon mairei, Orthosiphon malosanus Baker, Orthosiphon marmoritis, Orthosiphon marquesii Briq., Orthosiphon menthifolius Briq and Orthosiphon massinensis Orthosiphon miserabilis Orthosiphon molis Baker, Orthosiphon mombasicus, Orthosiphon mossianus, Orthosiphon muddii, Orthosiphon natalensis and Orthosiphon neglectus Orthosiphon nigripunctatus Orthosiphon nyasicus, Orthosiphon obbiadensis, Orthosiphon oblongifolius, Orthosiphon obscurus and Orthosiphon omatus Orthosiphon parvifolius Orthosiphon pascuensis, Orthosiphon persimilis, Orthosiphon petiolaris, Orthosiphon petrensis, Orthosiphon physocalycinus and Orthosiphon pretoriae Orthosiphon pseudoaristatus Orthosiphon pseudomatus, Orthosiphon pseudorubicundus, Orthosiphon pseudoserratus, Orthosiphon rabaiensis, Orthosiphon reflexus, Orthosiphon rehmannii, Orthosiphon retinervis and Orthosiphon rhodesianus Orthosiphon robustus Orthosiphon rogersii and Orthosiphon roseus Orthosiphon rubicundus Benth Orthosiphon rubicundus var. canescene Orthosiphon rubicundus var. Orthosiphon rubicundus var. hohenackeri, Orthosiphon rubicundus var. macrocarpus, hainanensis Orthosiphon rubicundus var. mollissimus and Orthosiphon rubicundus var. rigidus Orthosiphon rubicundus var. Orthosiphon rufinervis and Orthosiphon salagensis rubicundus Orthosiphon sarmentotus Orthosiphon scabridus Orthosiphon schimperi Orthosiphon schinzianus, Orthosiphon secundiflorus, Orthosiphon serratus, Orthosiphon shirensis, Orthosiphon silvicola, Orthosiphon sinensis, Orthosiphon somalensis, Orthosiphon spicatus Baker, Orthosiphon spicatus Benth, Orthosiphon spiralis, Orthosiphon stamineus, Orthosiphon stenophyllus, Orthosiphon stuhlmannii, Orthosiphon subvelutinus, Orthosiphon suffrutescene, Orthosiphon tagawae Orthosiphon tenuiflorus, Orthosiphon tenuifrons, Orthosiphon teucriifolius, Orthosiphon teucriifolius var. galpinianus, Orthosiphon teucriifolius var. teucriifolius and Orthosiphon thorncroftii Orthosiphon thymiflorus Orthosiphon thymiflorus var. viscosus, Orthosiphon tomentosus Benth, Orthosiphon tomentosus De wild, Orthosiphon tomentosus var. glabratus, Orthosiphon tomentotus var. parviflorus, Orthosiphon tomentosus var. rubiginosus, Orthosiphon tomentotus var viscosus, Orthosiphon transvaalensis and Orthosiphon tristis Benth Orthosiphon truncates Orthosiphon tuberosus, Orthosiphon tubiformis, Orthosiphon tubulascene, Orthosiphon unyikensis Orthosiphon usambarensis, Orthosiphon varians and Orthosiphon velteri Orthosiphon vernalis Orthosiphon viatorum and Orthosiphon villosus Orthosiphon violaceus Orthosiphon virgatus, Orthosiphon viscosus and Orthosiphon welkefieldii Orthosiphon wattii Orthosiphon welwitschii, Orthosiphon wilmsii gurke, Orthosiphon wilmsii var. komghensis, Orthosiphon wilmsii var. wilomsii, Orthosiphon woodii and Orthosiphon xylorrhizus 321 Int. J. Biol. Chem., 9 (6): 318-331, 2015 PHYTOCHEMISTRY These plants generally reported to contain monoterpenes, diterpenes, triterpenes, saponins, flavonoids, organic acids and etc. Considering the similarity of the chemical constituents of plants in the same genus. We summarized the phytochemical studies of five investigated plants, including O. stamineus, O. ariatatus, O. pallidus, O. thymiflorus and O. diffuses. This summary allows an understanding of the general and phytochemical constituents that has been discovered. It should also aid in further utilization of the plant resources in this genus. Selected chemical structure identified in Orthosiphon plants are depicted in Fig. 1. (a) H3C O O O O H3C Bzo Bzo CH3 AcO H CH3 O HO OH CH3 HO OAc AcO H3C CH3 CH3 H HO H3C H CH3 HO H H CH3 HO H3C H CH3 CH3 HO H3C CH3 CH3 H3C O O CH3 CH3 OH CH3 H H CH3 CH3 H3C O H CH3 H OAc (d) CH3 H3C H3C CH3 H CH3 H3C CH 3 H3C O OH H CH3 H3C CH2 CH3 OH Bzo Bzo CH3 O OH H CH3 O (c) (b) CH2 CH3 OH CH3 OH CH3 HO H3C CH3 (e) OH CH3 HO H3C CH3 CH3 (f) CH3 O H3C O Bzo Bzo CH3 CH2 H CH3 OH AcO O CH3 H AcO O OH HO AcO OAc AcO CH3 CH2 CH3 OH Bzo Bzo CH3 H HO OH O OH H CH3 HO CH2 H CH3 CH3 OH OH O OAc (h) OH O H CH3 (g) OH O H CH3 Bzo Bzo CH3 CH2 H CH3 OAc H CH3 Bzo Bzo CH3 CH2 O OAc Bzo CH3 CH3 O Bzo CH3 OH O OH OAc Bzo Bzo CH3 CH3 AcO H CH2 CH3 OH O OH OAc H H CH3 CH3 OH (i) O H CH3 H HO H3C CH3 (j) CH3 CH2 CH3 OH C H 3 CH2 OH CH3 H CH3 H3C O O CH3 CH3 O O CH3 O O H3C O O OH O O CH3 CH3 CH3 CH3 HO CH3 H3C CH2 (k) H3C OH H2C O CH3 CH3 Fig. 1: Continue 322 O O CH3 Int. J. Biol. Chem., 9 (6): 318-331, 2015 OH (l) H3C CH3 O O OH O O O H3C O O O OH CH3 CH3 O OH H2C (m) CH3 CH3 (n) CH3 HO O H CH3 CH3 O O O O CH3 CH3 CH3 O O CH3 CH3 O O CH3 O CH3 CH3 (o) CH2 H CH2 H3C CH3 H H3C CH3 CH3 CH3 H3C O (p) CH3 OH OH HO CH3 H H3C CH3 + N CH3 H3C CH3 CH3 + N CH3 CH3 (r) O (q) H3C CH3 OH H CH2 H3C H3C H3C CH3 OH (s) CH3 CH3 CH3 CH3 H3C CH3 CH3 O H3C CH3 CH3 H3C CH3 CH2 Fig. 1(a-s): Chemical Structures of typical and bioactive constituents isolated from Orthosiphon species, (a) 3’-hydroxy-5, 6, 7, 4’-tetramethoxyflavoneNeoorthosiphol A, (b) Neoorthosiphol Bα-amyrin, (c) -amyrinMaslinic acid, (d) Urosolic Acid Oleanolicv Acid, (e) Orthosiphonone A Orthosiphonone B, (f) Orthosiphol A Orthosiphol B, (g) Myo-inositol, (h) Neoorthosiphol A Neoorthosiphol B, (i) Betulinic acid -Elemene, (j) -Caryophyllene Caffeic acid, (k) Sinensetin Tetra-methyl scutellarein, (l) Eupatorin Cirsimaritin, (m) Acetovanillochromene Orthochromene A, (n) Methylripario Chromene Agermacrene-D, (o) -Selinen -Cadinol, (p) Choline Betaine, (q) O-cyamenea-terpineol, (r) LyrolValencene and (s) Nephthalin Camphor -elemene Moreover, previous research have detected 116 chemical compound have been isolated from the O. stamineus. They were 3-hdroxy 5, 6, 7, 4 tetramethoxy flavones, 2-O-deacetyl Orthosiphol J, 4' hydroxyl-5,6,7-trimethoxy flavone, α-cadinol, α-humulene, β-bourbonene, β-caryophyllene, β-elemene, β-pinene, aurantiamide acetate, caffeic acid depside A-C, cismaritin, eugenol, eupatorin, ladanein, methylripario chromene A, neoorthosiphol A-B, neoOrthosiphon A, norstaminol A-C, Othosiphol A-Z, Orthosiphon A-D, pillion, quercetin, rosamarinic acid salvigenin, secoorthosiphol A-C, siphonol A-E, staminol A-D, ursolic acid, betulinic acid, vomifoliol beta amyrin, α-amyrin, maslinic acid, oleanolicy acid and other minor constituents (Adnyana et al., 2013; Ameer et al., 2012; Guerin et al., 1989). 323 Int. J. Biol. Chem., 9 (6): 318-331, 2015 In the case of O. aristatus, the major constituents were sequiterpenes including β-elemene, β-caryophyllene, orthochromene A, acetovanillochromene, sinensetin, tetramethyl scutellarein, eupatorin, neoOrthosiphons A and B, Orthosiphones A and B with some minor constituents (Shibuya et al., 1999; Schut and Zwaving, 1986; Bombardelli, 1972; Lyckander and Malterud, 1992). In the case of O. pallidus is rich in gemacrene D, β-selinene, α-cadinol, choline, betainine, Orthosiphonone A and B, Orthosiphol A and B with some minor other constituents (Basu and Sing, 1956; Basu and Singh, 1956). Moreover O. thymiflorus and diffuses leaves were identified 33 and 25 compound. Most of the compounds are terpenoids. Orthosiphon thymiflorus content camphor, o-cymene, α-terpineal, nephthaline, lyrol, α-elemene and valencene etc. Other major compound of Orthosiphon diffuses were t-caryophyllene, octocosane, n-eicosane, limonene, β-ocimene and kauran-18-al and minor compounds were farnesol, calarene, octanol, β-selive, β-bisebolene, α- terpinolene and methylioso stearate etc. (Sadashiva et al., 2013). PHARMACOLOGICAL PROPERTIES Anti-inflammatory activity: Mostly 60-75% of the medicinal species of Orthosiphon reported in Table 1 have been traditionally used for treatment of inflammation and diseases like arthritis, bronchitis and rheumatoid. The pharmacological activity of the species of genus Orthosiphon provides primarily in vivo information for anti-inflammatory effects. In different studies on O. stamineus methanolic extract on various amount model suggested that oral administration of methanolic extract of O. stamineus exerted significant anti-inflammatory activity from 250-1000 mg kgG1 of dose (Ameer et al., 2012). The activity of chloroform extract was studied on various models like anti-peritoneal capillary permeability, carrageenan-induced rat paw edema along with in vitro radical scavenging activity. It was found that oral administration of chloroform extract at 500-1000 mg kgG1 reduced edema and no dye leakage to the peritoneal cavity (Yam et al., 2010). Masuda et al. (1992) investigated that isolation of Orthosiphol A and B showed strong inhibitory activity against the inflammation induced by a tumor promoter on the ears gene targated mice (Masuda et al., 1992). Antioxidant activity: Several Orthosiphon species traditionally used for expectorant and rheumatism indicated antioxidant activity. In different studies of O. stamineus for different extract (50% hydroalcoholic, distilled water, 50-70% hydroacetone and chloroform extract) was investigated that for free radical scavenging activity using different model like DPPH, superoxides and xanthin oxidase that O. stamineus extract showed potential antioxidant activity. The highest activity was found in hydroacetone extract. Other study found that all the extract had potential antioxidants comparable to that of some standard antioxidants BHA and quercetin (Adnyana et al., 2013). Hepatoprotective activity: Yam et al. (2009) reported that pretreatment with methanolic extract of O. stamineus to hepatoprotective activity in CCL4- induced liver damage in rats. It was investigated that hepatoprotective effects caused by antioxidants properties (Yam et al., 2007). Another study Maheswari et al. (2008) investigated that methanol extract of O. stamineus showed hepatoprotective activity on paracetamol-induced rats. Further, they proposed that there quality of medicinal plant due to ability to prevent the depletion of the tissue GSH (Maheswari et al., 2008). 324 Int. J. Biol. Chem., 9 (6): 318-331, 2015 Anticancer activity: Stampoulis et al. (1999) proposed cytotoxic activity of methanolic extract of O. stamineus against liver methanolic clon 26-LS carcinoma cells. The isolated compound stamina lactones A and B and norstaminal a showed mild cytotoxic activity against high malignant live metal stalic clone carcinoma cells (Stampoulis et al., 1999). Another study Awale et al. (2003a) investigated the possible cytotoxic activity a compound isolated from japans O. stamineus against highly malignant liver metastatices murine colon 26-LS carcinoma and human HT-1080 fibrosarcoma cell line (Adnyana et al., 2013). Antihypertensive activity: The antihypertensive activity of aqueous extract of leaves and active constituent isolated from O. stamineus benth was examined. Methylripariochromene A (from aqueous extract of leaves), Orthochromene A, Orthosiphonone A and B and neoorthosiphol A and B (from CHcl3 fraction of leaves), tetramethylscutell are in posses diuretic action. These constituents led to decrease in blood pressure and cardiac output. Subcutaneous administration of aqueous decoction of leaves led to decrease in systolic blood pressure conscious SHRSP. Does dependent decrease in urinary volume was observed ofter oral administration of isolated constituents of Orthosiphon stamineus benth urinary excretion of electrolytes was increased 2-3 times. These results confirmed that flavonoids and isopimarane-type compounds contributes significant antihypertensive activity (Adnyana et al., 213; Ameer et al., 2012). Koay and Amir (2012) investigated antihypertensive activity of O. stamineus benth in combination with folic acid, coenzyme-Q, policosanol which indicated effective control of high blood pressure in patients with metabolic syndrome (Koay and Amir, 2012). Gastro protective activity: Methanolic extract of leaves of O. stamineus benth posses significant effects for treatment gastric ailments. Fifty percentage of methanolic extract led to decrease in ulcer index, gastric mucosa mucosal damage, lipid peroxidation with an increase in mucus secretion. The antiulcerogenic activity was investigated in male Sprague Dawley rats against ethanolinduced ulcers. The traces of histological changes, mucosal secretion, Ulcer index and lipid. Peroxidation level was estimated using both in vitro and ex vivo models. The results showed significant does dependent gastro protective responces (125-1000 mg kgG1) (Yam et al., 2009). Antisebum activity: Sebum is an oily waxy matter secreted by exocrine sebaceous gland. Antisebum activity is observed in plants with phenolic and flavonoidal, terpennidal contents. O. stamineus benth exhibit prominent antisebum activity. The leaf extracts of O. stamineus decrese the activity of enzyme 5 α-reductase. The enzyme triggers the secretion of sebum. The extract of O. stamineus inhibits the synthesis of squaline (30 carbon natural orgaic compound) importent sebum constituents and help in skin glow there by reducing the oily appearance. Two percentage of leaf extract of O. stamineus reduces the oily appearance of skin and significantly reduces the pore size leading to improved skin complexion (Vogelgesang et al., 2011). Hyperlipidemic acivity: The aqueous extract of O. stamineus benth showed significant hyperlipidemic activity in diabetic rats. Mariam et al. (1996) investigated the oral admnistration of aqueous extract of O. stamineus benth on lipid profile in normal and Streptozotocic induced diabetic male wistar rats (Mariam et al., 1996). 325 Int. J. Biol. Chem., 9 (6): 318-331, 2015 Nephroprotective activity: Adnyana et al. (2013) investigated the potential of hydroalcoholic O. stamneus. The study revealed that the plant posses nephroprotective activity significantly at a dose of 50 mg kgG1. When compared to standard drug hydrochlorothiazide (10 mg kgG1). Similarly when the methanolic extract of the plant was investigated gentamycin-induced nephrotic model, A does dependent nephroprotective effect was observed (100-200 mg kgG1) with a steep decrease in decreased serum creatinine and blood urea level (Adnyana et al., 2013). Antipyretic activity: Antipyretic study of O. stamineus hydrochloric extract executed a profound effect from a dose range of 50-1000 mg kgG1 b.wt. The yeast induced pyrexia model was employed to investigate the effect. Similarly the effect was observed in 50% methanolic extract of O. stamineus in yeast-induced pyrexia in Sprague Dawley rats was investigated. The study showed that oral administration of the extract in the range from 450-1000 mg kgG1 led to no reduction in body temperature, but a significant alleviation of the pyrexia induced by yeasts was observed (Yam et al., 2008). Antiangiogenic activity: Plant O. stamineus possess significant anti-angiogenic activity. Ethanolic extract of O. stamineus showed retarding effect on the colorectal tumor and human umbical vein endothetical cell formation. Ethanolic extract of the plant at a concentration of (211±0.26 pg mLG1) inhibited VEGF in vitro and in vivo (53-54) (Sahib et al., 2009; Goodwin, 2007). Antibacterial activity: The studies on O. stamineus extract showed antibacterial activity on serotypes c and d of Streptococcus mutans (MIC = 7.8-23.4 mg mLG1). The potency decreased about one-half for type d but no change was found in type c, with the presence of 5% sucrose (Chen et al., 1989). Orthosiphon stamineus methanolic extract at concentration of 50% inhibited Bacillus subtilis, Bacillus cereus, Litseria monocytogenes, Staphylococcus aureus, Escherichia coli, Vibri parahaemolyticus, Salmonella enteritidis, Salmonella typhimurium and Klebsiella pneumoniae. This antibacterial activities of O. stamineus may be due to the high concentration of rosmarinic acid (Hossain et al., 2008). Whole O. stamineus plant (powdered) methanolic extract demonstrated inhibitory activity against vibrio parahaemolyticus in vitro. The inhibition showed with O. stamineus extracts was comparable to the inhibition seen with that of 5% lactic acid; this may be likely due to high concentration of rosmarinic acid found in the O. stamineus extracts (Ho et al., 2010). Antidiabetic activity: In oral glucose tolerance test, the water extract at doses of 0.2-1.0 g kgG1 significantly decreased plasma glucose concentration in dose-dependent manner for both normal and diabetic rats. At a dose of 1.0 g kgG1 showed similar effect with glibenclamide (5 mg kgG1). In diabetic rats, after they were given the extract orally (0.5 g kgG1) for 14 days, plasma glucose concentrations were reduced significantly. In addition, plasma triglyceride concentration was also lower in the extract-treated diabetic rats than that of untreated group. Furthermore, plasma HDL-cholesterol concentration was significantly increased in diabetic rats treated with the extract. In perfused rat pancreas, 100 μg mLG1 extract potentiated the glucose-induced insulin secretion (Sriplang et al., 2007). Antidiabetic effects of the chloroform, methanol, petroleum ether and water extracts of Orthosiphon stamineus was studied. Chloroform extract at a dose of 1 g kgG1 b.wt., significantly 326 Int. J. Biol. Chem., 9 (6): 318-331, 2015 reduced blood glucose level. Further, this extract was fractionated and finally one subfraction showed similar antidiabetic effect with metformin (Mohamed et al., 2011a). Diuretic activity: Diuretic activity of O. stamineus hydroalcohol extract from aerial parts was reported. At a dose of 50 mg kgG1, this extract showed similar effectivity with hydrochlorothiazide at a dose of 10 mg kgG1 (Beaux et al., 1999). Other studies reported that a water extract and tincture of leaves enhanced ion excretion of rats which were not due to the potassium content of the starting material (Englert and Harnischfeger, 1992). Arafat et al. (2008) studied the diuretic and hypouricemic activity of different O. stamineus methanol extracts by Sprague, Dawley rats model. A single dose infusion (2 g kgG1) of methanol and methanol: Water (1:1) extracts possesses significant diuretic action, where the effect was quantitatively similar to the control, hydrochlorothiazide. Repeated dose of 0.5 g kgG1 of methanol: water (1:1) extracts showed an increase in diuresis from the third day of treatment. Oral administration of 0.5, 1.0 and 2.0 g kgG1 of methanol: water (1:1) extracts significantly reduced serum urate level of hyperuricemic rats at hour 6, whereby the decrease in the uric acid level was also observed for the standard, allopurinol at hour 6 (Arafat et al., 2008). Adam et al. (2009) investigated the diuretic effects of Orthosiphon stamineus aqueous extract. Orally at doses of 5 and 10 mg kgG1 to Sprague, Dawley rats and was compare with furosemide or hydrochlorothiazide at 10 mg kgG1. Urine pH, urine volume, urine density and urine electrolytes were determined every hour for 4 h. Blood was assayed for albumin, glucose, Blood Urea Nitrogen (BUN) and creatinine. Orthosiphon stamineus extract exhibited dose-dependent diuretic activity. However, Na+ and Cl!excretion was not markedly elevated but urinary excretion of K+ was significantly increased. Orthosiphon stamineus extracts increased the serum BUN, creatinine and blood glucose level slightly (Adam et al., 2009). The diuretic, saluretic and uricosuric actions of 50 and 70% ethanol extracts of O. stamineus (700 mg kgG1) in rats revealed that the diuretic effect of the 50% ethanolic extract was higher than that of the 70% ethanolic extract or furosemide. It was characterized by higher absolute excretion of sodium and lower potassium wasting. Furthermore, the same 50% ethanol extract showed a relatively higher uricosuric effect. As the hydrophilicity of the extract increases, its diuretic and uricosuric effects also increase. This may be attributed to the abundance of polyphenols (Olah et al., 2003). TOXICITY STUDY The only toxicity literature and reports on members of the Orthosiphon genus were concerning O. stamineus. Different studies proved that the possible acute toxicity effects of orally administered Orthosiphon stamineus plant extract in rats. Acute toxicity was evaluated by LD50 method. No toxicity was found at a dose of 2 g kgG1 (Padilla et al., 1996). Another study Mohamed et al. (2011b) proved that standardized 50% ethanol plant extract at a dose 5 g kgG1 given orally to Sprague Dawley rats did not show an changes in macroscopic and microscopic. These results were proved that subchronic toxicity. Different concentration of plant extract (1250-5000 mg kgG1) on male and female Sprague Dawley rats for 4 weeks, showed no significant changes with control group. The parameters were hematological, organ weight, biochemical value, macroscopic and microscopic observation of the heart, brain, liver, kidney, spleen, tests, uterus and stomach (Mohamed et al., 2011a). 327 Int. J. Biol. Chem., 9 (6): 318-331, 2015 Recently Muhammad et al. (2011) investigated genotoxicity of O. stamineus using salmonellal microsome mutation and mouse bone marrow micronucleus assays method. The results were concluded that use of Orthosiphon stamineus in traditional medicine poses no genotoxic risk (Muhammad et al., 2011). SUMMARY AND CONCLUSION In the present review, summarized to congregate traditional use of medicinal plants in the genus Orthosiphon and research on its phytochemical, pharmacological and toxicological information on O. aristatus, O. pallidus, O. thymiflorus and O. stamineus, medicinal herbs used in the India and all over the world. Survey of literature data provided a practical base for further scientific research on this genus. In another equally very important to understand if the pharmacological studies on this genus are available to validate their traditional uses. Preliminary report in experimental studies says that it is significantly effective in diseases related to gastrointestinal, lungs and liver. Hence the purpose of this review is to provide comprehensive report about the genus based on its toxicity in order to identify its therapeutic potential and further prospects for betterment of research and provides basic knowledge for development of medicinal plants and useful approach for drug discovery. REFERENCES Adam, Y., M.N. Somchit, M.R. Sulaiman, A.A. Nasaruddin, A. Zuraini, A.A. Bustamam and Z.A. Zakaria, 2009. Diuretic properties of Orthosiphon stamineus Benth. J. Ethnopharmacol., 124: 154-158. Adnyana, I.K., F. Setiawan and M. Insanu, 2013. From ethnopharmacology to clinical study of Orthosiphon stamineus Benth. Int. J. Pharm. Pharmaceut Sci., 5: 66-73. Akowuah, G.A., Z. Ismail, I. Norhayati and A. Sadikun, 2005. The effects of different extraction solvents of varying polarities on polyphenols of Orthosiphon stamineus and evaluation of the free radical-scavenging activity. Food Chem., 93: 311-317. Ameer, O.Z., I.M. Salman, M.Z. Asmawi, Z.O. Ibraheem and M.F. Yam, 2012. Orthosiphon stamineus: Traditional uses, phytochemistry, pharmacology and toxicology. J. Med. Food, 15: 678-690. Arafat, O.M., S.Y. Tham, A. Sadikun, I. Zhari, P.J. Haughton and M.Z. Asmawi, 2008. Studies on diuretic and hypouricemic effects of Orthosiphon stamineus methanol extracts in rats. J. Ethnopharmacol., 118: 354-360. Ashokan, K. and M.S. Muthuraman, 2011. Anticancer studies on Orthosiphon pallidusm royle. and Peristrophe bicalyculata nees. J. Pharm. Res., 4: 2654-2656. Awale, S., Y. Tezuka, A.H. Banskota and S. Kadota, 2003a. Siphonols A-E: Novel nitric oxide inhibitors from Orthosiphon stamineus of Indonesia. Bioorgan. Med. Chem. Lett., 13: 31-35. Awale, S., Y. Tezuka, A.H. Banskota, I.K. Adnyana and S. Kadota, 2003b. Highly-oxygenated isopimarane-type diterpenes from Orthosiphon stamineus of Indonesia and their nitric oxide inhibitory activity. Chem. Pharm. Bull., 51: 268-275. Awale, S., Y. Tezuka, M. Kobayashi, J.Y. Ueda and S. Kadota, 2004. NeoOrthosiphonone a; A nitric oxide (NO) inhibitory diterpene with new carbon skeleton from Orthosiphon stamineus. Tetrahedron Lett., 45: 1359-1362. Basu, N.K. and H. Sing, 1956. Investigation of Orthosiphon pallidus royle I. Preliminary chemical study and isolation of Orthosiphonol. J. Am. Pharmaceut. Assoc., 9: 595-598. 328 Int. J. Biol. Chem., 9 (6): 318-331, 2015 Basu, N.K. and H. Singh, 1956. Investigation of Orthosiphon pallidus Royle II. Isolation of choline and betaine. J. Am. Pharmaceut. Assoc., 9: 598-601. Beaux, D., J. Fleurentin and F. Mortier, 1999. Effect of extracts of Orthosiphon stamineus benth, Hieracium pilosella L., Sambucus nigra L. and arctostaphylos uva-ursi (L.) spreng. in rats. Phytother. Res., 13: 222-225. Bombardelli, E., 1972. Flavonoid constituents of Orthosiphon stamineus. Fitoterapia, 43: 35-38. Chen, C.P., C.C. Lin and N. Tsuneo, 1989. Screening of Taiwanese crude drugs for antibacterial activity against Streptococcus mutans. J. Ethnopharmacol., 27: 285-295. Chen, H., F. Chen, Y.L. Zhang and J.Y. Song, 1999. Production of rosmarinic acid and lithospermic acid B in Ti transformed Salvia miltiorrhiza cell suspension cultures. Process Biochem., 34: 777-784. Di, X.X., S.Q. Wang, X.L. Zhang, B. Wang, H.X. Lou and X.N. Wang, 2013. Diterpenoids from the aerial parts of Orthosiphon aristatus var. aristatus. Phytochemistry, 6: 412-417. Englert, J. and G. Harnischfeger, 1992. Diuretic action of aqueous Orthosiphon extract in rats. Planta Med., 58: 237-238. Goodwin, A.M., 2007. In vitro assays of angiogenesis for assessment of angiogenic and anti-angiogenic agents. Microvascular Res., 74: 172-183. Guerin, J.C., H.P. Reveillere, P. Ducrey and L. Toupet, 1989. Orthosiphon stamineus as a potent source of methylripariochromene A. J. Nat. Prod., 52: 171-173. Han, C.J., A.H. Hussin and S. Ismail, 2008. Toxicity study of Orthosiphon stamineus benth (Misai Kucing) on sprague dawley rats. Trop. Biomed., 25: 9-16. Heinrich, M. and S. Gibbons, 2001. Ethnopharmacology in drug discovery: An analysis of its role and potential contribution. J. Pharm. Pharmacol., 53: 425-432. Ho, C.H., I. Noryati, S.F. Sulaiman and A. Rosma, 2010. In vitro antibacterial and antioxidant activities of Orthosiphon stamineus Benth. extracts against food-borne bacteria. Food Chem., 122: 1168-1172. Hossain, M.A. and Z. Ismail, 2013. Isolation and characterization of triterpenes from the leaves of Orthosiphon stamineus. Arabian J. Chem., 6: 295-298. Hossain, M.A., Z. Ismail, A. Rahman and S.C. Kang, 2008. Chemical composition and anti-fungal properties of the essential oils and crude extracts of Orthosiphon stamineus Benth. Ind. Crops Prod., 27: 328-334. Hsu, C.L., B.H. Hong, Y.S. Yu and G.C. Yen, 2010. Antioxidant and anti-inflammatory effects of Orthosiphon aristatus and its bioactive compounds. J. Agric. Food Chem., 58: 2150-2156. Kavimani, S., R. Ilango, J. Thangadurai, B. Jaykar, U. Majumdar and M. Gupta, 1997. Diuretic activity of aqueous extract of Orthosiphon thymiflorus in rats. Indian J. Pharmaceut. Sci., 59: 96-98. Keng, C.L. and L.P. Siong, 2006. Morphological similarities and differences between the two varieties of cat’s whiskers (Orthosiphon stamineus Benth.) grown in Malaysia. Int. J. Bot., 2: 1-6. Kiruthika, A. and S.M. Meenakshi, 2011. Anticancer studies on Orthosiphon pallidus royle. and Peristrophe bicalyculata nees. J. Pharm. Res., 4: 2654-2656. Koay, Y.C. and F. Amir, 2012. A survey of the chemical constituents and Biological activities of Orthosiphon stamineus. Sci. Int., 24: 133-138. 329 Int. J. Biol. Chem., 9 (6): 318-331, 2015 Kovendan, K., K. Murugan, S. Vincent and D.R. Barnard, 2012. Mosquito larvicidal properties of Orthosiphon thymiflorus (Roth) Sleesen. (Family: Labiatae) against mosquito vectors, Anopheles stephensi, Culex quinquefasciatus and Aedes aegypti (Diptera: Culicidae). Asian Pac. J. Trop. Biomed., 5: 299-305. Lyckander, I.M. and K.E. Malterud, 1992. Lipophilic flavonoids from Orthosiphon spicatus as inhibitors of 15-lipoxygenase. Acta Pharmaceutica Nordica, 4: 159-166. Maheswari, C., R. Maryammal and R. Venkatanarayanan, 2008. Hepatoprotective activity of Orthosiphon stamineus on liver damage caused by paracetamol in rats. Jordan J. Biol. Sci., 1: 105-108. Mariam, A., M.Z. Asmawi and A. Sadikun, 1996. Hypoglycaemic activity of the aqueous extract of Orthosiphon stamineus. Fitoterapia, 67: 465-468. Masuda, T., K. Masuda, S. Shiragami, A. Jitoe and N. Nakatani, 1992. Orthosiphol A and B, novel diterpenoid inhibitors of TPA (12-O-tetradecanoylphorbol-13-acetate)-induced inflammation, from Orthosiphon stamineus. Tetrahedron, 48: 6787-6792. Matsubara, T., T. Bohgaki, M. Watarai, H. Suzuki, K. Ohashi and H. Shibuya, 1999. Antihypertensive actions of methylripariochromene A from Orthosiphon aristatus, an Indonesian traditional medicinal plant. Biol. Pharm. Bull., 22: 1083-1088. Mohamed, E.A.H., A.J. Mohamed, M.Z. Asmawi, A. Sadikun, O.S. Ebrika and M.F. Yam, 2011a. Antihyperglycemic effect of Orthosiphon stamineus Benth leaves extract and its bioassayguided fractions. Molecules, 16: 3787-3801. Mohamed, E.A.H., C.P. Lim, O.S. Ebrika, M.Z. Asmawi, A. Sadikun and M.F. Yam, 2011b. Toxicity evaluation of a standardised 50% ethanol extract of Orthosiphon stamineus. J. Ethnopharmacol., 133: 358-363. Muhammad, H., M.R. Gomes-Carneiro, K.S. Poca, A.C.A.X. De-Oliveira and A. Afzan et al., 2011. Evaluation of the genotoxicity of Orthosiphon stamineus aqueous extract. J. Ethnopharmacol., 133: 647-653. Ohashi, K., T. Bohgaki, T. Matsubara and H. Shibuya, 2000. Indonesian medicinal plants. XXIII. Chemical structures of two new migrated pimarane-type diterpenes, neoorthosiphols A and B and suppressive effects on rat thoracic aorta of chemical constituents isolated from the leaves of Orthosiphon aristatus (Lamiaceae). Chem. Pharm. Bull., 48: 433-435. Olah, N.K., L. Radu, C. Mogosan, D. Hanganu and S. Gocan, 2003. Phytochemical and pharmacological studies on Orthosiphon stamineus Benth. (Lamiaceae) hydroalcoholic extracts. J. Pharmaceut. Biomed. Anal., 33: 117-123. Padilla, M.C.L., J.T. Capo, A.H. Rodriguez, J.L.C. Freixas and L.S.C. Alvarez, 1996. Efecto diuretico y toxicidad aguda del Orthosiphon aristatus blume (Te de Rinon). Rev. Cubana Plant Med., 1: 26-30. Paton, A.J., D. Springate, S. Suddee, D. Otieno and R.J. Grayer et al., 2004. Phylogeny and evolution of basils and allies (Ocimeae, Labiatae) based on three plastid DNA regions. Mol. Phylogenet. Evol., 31: 277-299. Sadashiva, C.T., P. Sharanappa, Y. Naidoo, C.T. Sulaimon and I. Balachandran, 2013. Chemical composition of essential oil from Orthosiphon diffuses Benth. J. Med. Plants Res., 7: 170-172. Sahib, H.B., Z. Ismail, N.H. Othman and A.M.S. Abdul-Majid, 2009. Orthosiphon stamineus benth. methanolic extract enhances the anti-proliferative effects of tamoxifen on human hormone dependent breast cancer. Int. J. Pharmacol., 5: 273-276. 330 Int. J. Biol. Chem., 9 (6): 318-331, 2015 Schut, G.A. and J.H. Zwaving, 1986. Content and composition of the essential oil of Orthosiphon aristatus. Planta Med., 52: 240-241. Shibuya, H., 1999. Chemical structures of two new isopimarane- type diterpenes, Orthosiphonones A and B and a new benzochromene, orthochroinene A from the leaves of Orthosiphon aristatus (Lamiaceae). Chem. Pharmaceut. Bull., 47: 695-698. Shibuya, H., T. Bohgaki, T. Matsubara, M. Watarai, K. Ohashi and I. Kitagawa, 1999. Chemical structures of two new isopimarane-type diterpenes, Orthosiphonones A and B and a new benzochromene, orthochromene a from the leaves of Orthosiphon aristatus (Lamiaceae). Chem. Pharm. Bull., 47: 695-698. Sini, K.R., Y. Haribabu, M.S. Sajith and K.S. Sreekumar, 2012. In-vitro cytotoxic activity of Orthosiphon thymiflorus Roth.) sleensen leaf extract against dalton lymphoma ascites cell line. J. Chem. Pharmaceut. Res., 4: 917-921. Sriplang, K., S. Adisakwattana, A. Rungsipipat and S. Yibchok-Anun, 2007. Effects of Orthosiphon stamineus aqueous extract on plasma glucose concentration and lipid profile in normal and streptozotocin-induced diabetic rats. J. Ethnopharmacol., 109: 510-514. Stampoulis, P., Y. Tezuka, A.H. Banskota, K.Q. Tran, I. Saiki and S. Kadota, 1999. Staminolactones A and B and norstaminol A: Three highly oxygenated staminane-type diterpenes from Orthosiphon stamineus. Org Lett., 1: 1367-1370. Sundarammal, S., R. Thirugnanasampandan and M.T. Selvi, 2012. Chemical composition analysis and antioxidant activity evaluation of essential oil from Orthosiphon thymiflorus (Roth) Sleesen. Asian Pac. J. Trop. Biomed., 2: S112-S115. Vogelgesang, B., N. Abdul-Malak, C. Reymermier, C. Altobelli and J. Saget, 2011. On the effects of a plant extract of Orthosiphon stamineus on sebum-related skin imperfections. Int. J. Cosmetic Sci., 33: 44-52. Yam, M.F., R. Basir, M.Z. Asmawi and Z. Ismail, 2007. Antioxidant and hepatoprotective effects of Orthosiphon stamineus Benth. standardized extract. Am. J. Chinese Med., 35: 115-126. Yam, M.F., M.Z. Asmawi and R. Basir, 2008. An investigation of the anti-inflammatory and analgesic effects of Orthosiphon stamineus leaf extract. J. Med. Food., 11: 362-368. Yam, M.F., L.F. Ang, I.M. Salman, O.Z. Ameer and V. Lim et al., 2009. Orthosiphon stamineus leaf extract protects against ethanol-induced gastropathy in rats. J. Med. Food., 12: 1089-1097. Yam, M.F., V. Lim, I.M. Salman, O.Z. Ameer and L.F. Ang et al., 2010. HPLC and anti-inflammatory studies of the flavonoid rich chloroform extract fraction of Orthosiphon stamineus leaves. Molecules, 15: 4452-4466. 331