Malaysian Herbal Monograph

Patawali Stem

Tinospora crispa (L.) Hook.f. & Thomson

Menispermaceae

Figure 1 : T. crispa. (a) Whole plant; (b)-(c) stems; (d) leaf. (Photos courtesy of Thiyagu, MARDI, 2014)

DEFINITION

Patawali stem consists of dried stem of T. crispa (L.) Hook.f. & Thomson (Menispermaceae)

SYNONYM

Tinospora rumphii Boerl, Tinospora tuberculata (Lamk) Beumee ex. K Heyne.

VERNACULAR NAMES

Heavenly elixir, heartleaf moonseed (English) [ 1 ], akar patawali, putarwali, petawali, daun akar wali, batang wali, akar seruntum (Malay), bo ye qing niu dan (Chinese) [ 2 ], kattukkodi (Tamil).

CHARACTER

ColourGreenish brown
OdourCharacteristic
TasteVery bitter

IDENTIFICATION

Plant Morphology

A woody climber up to 15 m long, older stems very prominently, tuberculate and producing very long filiform aerial roots. The stem contains an exceedingly bitter milky sap. Leaves broadly ovate to obircular, 7–14 cm x 6–12 cm. Inflorescence appearing when plant is leafless. Flowers usually come with 3 petals. Fruits are ellipsoidal, about 2 cm long and orange in colour [ 3 , 4 , 5 ].

Microscopy

Microscopic characters of T. crispa stem powder consist of cork cells in surface view, numerous starch granules with parenchyma cells, collenchymas cells; annular, spiral, reticulate, pitted and bordered pitted vessel cells, up to 160 µm in diameter; fragments of bast fibres with dentated wall and solitary crystals; fragments of lignified parenchyma and annular vessel fragments with parenchyma cells; parenchyma cell containing calcium oxalate crystals adjacent to bast fibres [ 6 ].

Figure 2 : Microscopic characters of T. crispa stem powder. (a-b) Cork cells; (c-d) starch granules; (e) collenchyma cells; (f-g) bordered pitted vessel cells; (h) fibre; (i) fibre and starch granules; (j-m) solitary crystals. [Scale bars: a,g=50 µm;b-f, h-k, m=20 µm; l=10 µm]

Colour Tests 

Observed colour of solution after treatment with various reagents:

NH4OH (25%)Yellow
FeCl3 (5%)Yellow

Thin Layer Chromatography (TLC)

Figure 3 : TLC profiles of syringin (S) and methanol extract of T. crispa dried stem observed under (a) UV at 254 nm and (b) UV at 366 nm before spraying; (c) visible light and (d) UV at 366 nm after spraying with 10% solution of sulfuric acid in ethanol.

Test Solutions Extract about 0.5 g of the T. crispa dried stem with 5 mL of methanol. Sonicate the solution for 15 min and filter.
Standard solution Dissolve 1 mg of syringin [CAS no.: 118-34-3] in 1 mL methanol to produce 1 mg/mL.
Stationary Phase HPTLC silica gel pre-coated plate 60 F254, 10 x 10 cm.
Mobile phase Chloroform : methanol : water ( 8:2:0.2 ) (v/v/v)
Application
  1. Syringin standard solution (S); 2 µL, as a band
  2. Methanol extracts of T. crispa dried stem; 6 µL as a band
Development distance Equilibrate the chamber with the mobile phase for 15 minutes, develop vertically for 7.0 cm.
Drying Air drying
Detection
  1. UV 254 nm and
  2. UV 366 nm before spraying;
  3. Visible light and d) UV 366 nm after spraying with with 10% solution of sulfuric acid in ethanol, and heat at 105°C until the colours of the zones become visible

High Performance Liquid Chromatography (HPLC)

Test solution Extract about 0.5 g of the T. crispa dried stem with 2 mL of methanol. Sonicate the solution for 2 hr and filter the solution through a 0.45 µm PTFE syringe filter and inject the filtrate into the HPLC column.
Standard solution Dissolve 1 mg of syringin [CAS no.: 118-34-3] in 1 mL methanol to produce 1 mg/mL. 
Chromatographic system

Detector: UV 220 nm 

Column: C18  (5 µm, 4.6 mm I.D. x 250 mm) (preferable Phenomenex-Luna C18)

Column oven temperature: Ambient         

Flow rate: 1 mL/min         

Injection volume: 15 µL

Mobile Phase (gradient mode)

Run Time

(min)

A – 0.1% aqueous formic acid
(%)

B – Acetonitrile (%)

0

95

5

20

60

40

40

50

50

50

15

85

55

95

5

System suitability requirement

Perform at least five replicate injections of the standard solutions (1 mg/mL). The requirements of the system suitability parameters are as follow:

  1. Symmetry factor (As) is not more than 1.5.
  2. Percentage of relative standard deviation (RSD) of the retention time (tr) for syringin is not more than 2.0%. 
Acceptance criteria
  1. Retention time (tr) of syringin in the test solution is similar to the tr of the standard solution.
    The ultraviolet (UV) spectrum of syringin in the test solution is similar to  the UV   spectrum of syringin in the standard solution (optional supportive data). 
figure4

Figure 4 : HPLC chromatogram of syringin standard solution (1 mg/ mL) at tr = 15.542 min.

figure5

Figure 5 : HPLC full chromatogram of methanol extract of T. crispa dried stem showing peak corresponding to syringin at tr = 15.449 min.

Figure 6 : UV spectrum of syringin (1 mg/mL) and methanol extract of T. crispa dried stem.

PURITY TESTS

Foreign Matter
Not more than 2%
Ash Contents
Total ash Not more than 5%
Acid-insoluble ash Not more than 1%
Loss on Drying
Not more than 11%
Extractive Values
Water-soluble extracts
Hot method Not less than 18%
Cold method Not less than 15%
Ethanol-soluble extracts
Hot method Not less than 8%
Cold method Not less than 8%

SAFETY TESTS

Heavy Metals
Arsenic Not more than 5.0 mg/kg
Mercury Not more than 0.5 mg/kg
Lead Not more than 10.0 mg/kg
Cadmium Not more than 0.3 mg/kg
Microbial Limits
Total bacterial count Not more than 105 cfu/g
Total yeast and mould count Not more than 104 cfu/g
Bile-tolerant gram negative Not more than 104 cfu/g
Specific Pathogens
Salmonella spp. Absent in 25 g
Escherichia coli Absent in 1 g
Staphylococcus aureus Absent in 1 g
Pseudomonas aeruginosa Absent in 1 g

CHEMICAL CONSTITUENTS

The stems contain N-trans-feruloyltyramine and N-cis-feruloyl tyramine in theethyl acetate fraction as well as a phenolicglucoside, namely, tinotuberide in thebutanol fraction [ 7 ]. Subsequently, a diterpeneglucoside, borapetoside A, and its aglycone,borapetol A were isolated from the stems, followed by the isolation ofborapetoside B, and its aglycone, borapetol B [ 8 , 9 ]. The presence of borapetoside C–H andtinotufolin A–F were also identified from thestems and leaves [ 10 , 11 , 12 , 13 ].

The cis clerodane-type furanoditerpenoids include, (2R,5R,6R,8S,9S,10S,12S)-15,16-epoxy-2-hydroxy-6-O-{β-D-glucopyranosyl-(1→6)-α-Dxylopyranosyl}-cleroda-3,13(16),14-trien-17,12-olid-18-oic acid methyl ester, (2R,5R,6R,8R,9S,10S,12S)-15,16-epoxy-2-hydroxy-6-O-(β-D-glucopyranosyl)-cleroda-3,13(16),14-trien 17,12-olid-18-oic acid methyl ester, (5R,6R,8S,9R,10R,12S)-15,16-epoxy-2-oxo-6-O-(β-D glucopyranosyl)-cleroda-3,13(16),14-trien-17,12-olid-18-oic acid methyl ester, methyl (2R,7S,8S)-8[(2S)-2-(3,4-dihydroxy-2,5-dimethoxytetrahydro-3-furanyl)-2-hydroxyethyl]-2,8-dimethyl-10-oxo-11-oxatricyclo[7.2.1.0]dodec-3-ene-3-carboxylate, (5R,6R,8S,9R,10S,12S)-15,16-epoxy-2-oxo-6-O-(β D-glucopyranosyl)-cleroda-3,13(16),14-trien-17,12-olid-18-oic acid methylester, (2R,5R,6S,9S,10S,12S)-15,16-epoxy-2-hydroxy-6-O-(β-D-glucopyranosyl)-cleroda-3,7,13(16),14-tetraen 17,12-olid-18-oic acid methyl ester, (5R,6S,9S,10S,12S)-15,16-epoxy-2-oxo-6-O-(β-D glucopyranosyl)-cleroda-3,7,13(16),14-tetraen-17,12-olid-18-oic acid methyl ester, (3R,4R,5R,6S,8R,9S,10S,12S)-15,16-epoxy-3,4-epoxy-6-O-(β-D-glucopyranosyl)-cleroda-3,13(16),14 trien-17,12-olid-18-oic acid methyl ester and (1R,4S,5R,8S,9R,10S,12S)-15,16-epoxy-4-O-(β-D glucopyranosyl)-cleroda-2,13(16),14-triene-17(12),18(1)-diolide [ 14 ].

The stems also contained five flavone glycosides identified as luteolin 4’-methyl ether 7-glucoside, genkwanin 7-glucoside, luteolin 4’-methyl ether 3’-glucoside, diosmetin and genkwanin [ 15 ].

The stems contained aporphine alkaloid, N-formylasimilobine 2-Oβ-D-glucopyranoside, N-formylasimilobine 2-Oβ-D-glucopyranosyl-(1 → 2)-β-D-glucopyranoside (tinoscorside A), N-formylanonaine, N-formyldehydroanonaine, N-formylnomuciferine, N-demethyl-N-formyldehydronornuciferine, magnoflorine, paprazine, N-transferuloyltyramine and cytidine
[ 16 , 17 ].

The dichloromethane extract showed presence of vanillin, syringin, N formylannonain, N-formylnornuciferin, borapetosides B, C and F, N-cis-feruloyltyramine, N-trans feruloyltyramine and secoisolariciresinol [ 18 ].

The chloroform extracts of the dried stems showed presence of cycloeucalenol and cycloeucalenone [ 19 ].

The n-butanol extract of the stems was found to contain salsolinol, adenosine, uridine, tyramine, higenamine, syringing, borapetoside A, B, D and E, adenine and litcubinine [ 20 ].

The ethanolic extract showed presence of 2-O-lactoylborapetoside B, 6’-O lactoylborapetoside B, tinocrispol A, borapetosides A-F, borapetols A and B and columbin [ 21 ].

MEDICINAL USES

Uses described in folk medicine, not supported by experimental or clinical data

In folk medicine, an infusion of stem T. crispa is useful as vermifuge. A decoction of the whole plant is used for cholera [ 22 , 23 ]. This plant is useful for indigestion, stomachache, jaundice [ 22 ], sore eyes (external application), itchiness, wounds, mosquito bites [ 23 ] and fever [ 22 , 23 ]. It is also used in treatment of syphilis (external application), small pox and rheumatism [ 23 ].

Biological and pharmacological activities supported by experimental data

Antibacterial activity
Ethanol (95%v/v) extract of T. crispa dried stem inhibited the growth of eight methicilin-resistant Staphylococcus aureus isolated strains with minimum inhibitory concentrations (MIC) of 0.40-0.78 mg/mL and minimum bacterial counts (MBC) of 0.78-1.56 mg/mL by using checkerboard assay [ 24 ].

Aqueous, ethanol, and chloroform extracts of T. crispa dried stem (25, 50, 75, and 100% concentration/strength) inhibited the growth of Clostridium diphtheria, Streptococcus pneumonia and Shigella flexneri in a dose-dependent manner by using in vitro disc diffusion assay. From the same assay, the growth of Escherichia coli was only inhibited by aqueous and chloroform extracts at concentrations of 50-100% while the growth of Staphylococcus aureus was inhibited by ethanol extract at concentrations of 75% and 100%
[ 25 ].

Antiplasmodial activity
Ethanol (95% v/v) extract of T. crispa stem administered intraperitoneally (20, 40, and 80 mg/kg/day) to female ICR mice (pre-inoculated with Plasmodium yoelii, aged 6 weeks) for 20 days exhibited antiplasmodial activity in a dose-dependent manner with lower percentage of P. yoelii (18.12 ±2.24%) compared to ethanol (30%v/v) control (45.45 ±3.67%) on day 8. A pre-inoculated mouse given 80 mg/kg/day extract was alive without P. yoelii anymore in its blood stream on day 20 [ 26 ].

Antifilarial activity
Aqueous extract of T. crispa (10 mg/mL) against Dirofilaria immitis with mean relative movability (RM) value of 30.15% after 24 hours treatment (p < 0.05) compared to DMSO control (100%) [ 27 ].

Aqueous extract of T. crispa stem (0.5, 1.0, 5.0, and 10.0 mg/mL) exhibited antifilarial activity against adults worms of Brugia malayi (harvested from 4-6 weeks old male cat infected with L3-stage larvae from Aedes togoi) with the mean relative movability (RM) values of adults worms ranging between 0-100, in a dose-dependent manner, after 24 hours incubation compared to RPMI1640 culture medium as control (mean RM value of 100) [ 28 ].

Antinociceptive activity
Ethanol extract of T. crispa dried stem administered intraperitoneally in a single dose (30, 100, and 300 mg/kg body weight) to male Balb-C mice (aged 5-7 weeks) significantly exhibited antinociceptive activity in a dose-dependent manner with writhing response values of 3-15 (p ≤ 0.05) compared to saline control (36 ±1.45) by using acetic acid-induced writhing test and latency of discomfort of 10.84-13.16 seconds (p ≤ 0.05) compared to saline control (8.12 ±0.71) at 240 minutes by using hot plate test [ 29 ].

Anti-inflammatory activity
Ethanol extract of T. crispa dried stem administered intraperitoneally in a single dose (30, 100, and 300 mg/kg body weight) to Sprague Dawley rats (aged 8-10 weeks) significantly exhibited anti-inflammatory activity in a dose-dependent manner with the means of paw edema ranging between 0.11-0.41 mL/h (p ≤ 0.05) compared to saline control (0.57 ±0.05) at 1 hour by using carrageenan-induced paw edema test [ 30 ].

Antihyperglycemic activity
Water-ethanol extract of T. crispa stem (400 µg/mL) significantly increased 2-deoxy-D-glucose uptake (214.49 ±9.00% above basal, p < 0.01) in isolated rat’s L6 skeletal muscle cells after 24 hours incubation compared to metformin (205.80 ±6.28% above basal). However, this extract-stimulated glucose uptake may adversely be inhibited by 3.5 µM cycloheximide (glucose uptake < 100%) compared to partially inhibited metformin-stimulated glucose uptake (< 150%). This extract also significantly increased AMPKα1 (2.10 ±0.38 fold above basal, p < 0.05) and PPARγ (6.73 ±0.88 fold above basal, p < 0.05) mRNA levels after 15 minutes treatment while significantly increased GLUT1 mRNA level (1.29 ±0.38, p < 0.05) after 4 hours compared to control [ 31 ].

Anti-oxidant activity
Aqueous extract of T. crispa stem (dose) scavenged DPPH free radicals (86.51 ±0.07%) and inhibited oxidation of ferric ions (0.89 ±0.07mmol/L, p < 0.05) compared to vitamin C (96.36 ±0.90%, 1.05 ±0.00mmol/L) and BHT (96.51 ±0.95%, 1.03 ±0.03mmol/L) controls. This extract also inhibited lipid peroxidation (39.2 ±5.14%) by using thiobarbituric acid assay compared to vitamin C (73.2 ±5.14%) and BHT (75.8 ±6.08%) controls. The extract contained flavonoids (1.58 µg/mg of catechin, 0.85 µg/mg of luteolin, 1.44 µg/mg of morin and 1.38 µg/mg rutin) and phenolic (0.29 ±0.01 mg gallic acid equivalent/100 g of fresh sample) [ 31 ].

Methanol, chloroform, and water extracts of T. crispa stem showed significant total phenolic content (79.00-255.33 mg gallic acid equivalents/g extract, p < 0.05) by using Folin-Ciocalteu method and flavonoid content (2.67-9.53 mg quercetin equivalents/g extract, p < 0.05) by using Dowd method. Only methanol extract inhibited the scavenging activity of 1,1-diphenyl-2-picrylhydrazyl (DPPH) free radicals with IC50 value of 12 µg/mL compared to vitamin C control by using DPPH free radical scavenging assay [ 32 ].

Aqueous extract of T. crispa dried stem was prepared through different temperatures with respective time length, i.e. 20°C/24h, 40°C/12h, 60°C/6h, 80°C/3h, and 100°C/15 min. The extract at 60°C/6h significantly inhibited DPPH free radicals with 85.95 ±0.52% inhibition (p<0.05) compared to 20°C/24h (66.86 ±0.55%) and also inhibited thiobarbituric acid (TBA) (39.20 ±2.97%) [ 32 ].

Antihypercholesterolemic activity
Aqueous extract of T. crispa stem (450 mg/kg/day body weight) given to male New Zealand white rabbits fed with high cholesterol diet for 10 weeks significantly increased the levels of glutathione peroxidase (494.32 ±96.72U/L) and high density lipoprotein (HDL) (9.23 ±1.36mmol/L) (p <0.05) compared to untreated high cholesterol diet rabbits (220.41 ±19.69U/L , 0.86 ±0.11mmol/L). This extract also significantly decreased the levels of triglycerides and low density lipoprotein (LDL) (8.38 ±0.99mmol/L) (p < 0.05) compared to untreated high cholesterol diet rabbits (16.46 ±1.60 mmol/L). No foam cell formation was observed in the aorta of rabbits [ 34 ].

Cardiotonic activity
Triterpenes (cycloeucalenol and cycloeucalenone) isolated from crude chloroform extract of T. crispa dried stem (5.6×10-5 M for each triterpene) exhibited mild cardiotonic effects on isolated rat’s right and left atria (male Wistar rats weighed 250-300 g) compared to ethanol (80%v/v) control [ 35 ].

Cytochrome inhibitory activity
Methanol and ethanol-soluble fractions from aqueous extract of T. crispa stem (1.65 mg/mL) respectively inhibited activity of drug metabolizing enzymes from human liver microsomes, i.e. CYP3A4 (>70% inhibition; <30% inhibition) compared to ketoconazole control and CYP2D6 (>70% inhibition; <30% inhibition) compared to quinidine control
[ 36 ].                                    

Methanol extract of T. crispa stem (0.5 mg/mL) inhibited the activity of drug metabolizing enzymes from human liver microsomes, i.e. CYP3A4 (>30% inhibition) compared to troleandomycin control (37.9% inhibition) and CYP2D6 (<30% inhibition) compared to paroxetine control (60.7% inhibition) [ 37 ].

Cytotoxic activity
Methanol and water crude extract of T. crispa dried stem (5-500 µg/mL) showed less cytotoxicity effect (IC50 value > 500 µg/mL) on HL-60, MCF-7 and HepG2 cancer cell lines by using MTT assay [ 38 ].

Antiproliferative activity
Methanol, chloroform, and water extracts of T. crispa stem inhibited the growth of MCF-7 (IC50 = 33.75-42.75 µg/mL), MDA-MB-231 (IC50 = 44.83-51.25 µg/mL), HeLa (IC50 = 46.13-53.83 µg/mL), and 3T3 (IC50 = 52.58-65.50 µg/mL) cancer cell lines, in a dose-dependent manner compared to tamoxifen control by using MTT assay [ 32 ].

Aqueous extract of T. crispa dried stem inhibited the growth of Caov-3 (IC50 = 80 µg/mL compared to cisplatin control), HepG2 (IC50 = 60 µg/mL compared to cisplatin control), MCF-7 (IC50 = 60 µg/mL compared to tamoxifen control), and HeLa (IC50 = 79 µg/mL compared to tamoxifen control) cancer cell lines on day 3 by using MTT assay [ 33 ].

Clinical studies

A randomized double-blind placebo-controlled crossover trial to study the hypoglycemic effect of T. crispa was conducted involving 36 patients who met the metabolic syndrome criteria by National Cholesterol Education Program Adult Treatment Panel III and did not consume any oral hypoglycemic drug. These patients were divided into two groups, i.e. a group given 250 mg T. crispa dry extract powder capsule (consists of 0.98% dry weight marker substance A) and another given placebo, at 30 minutes before meal twice daily for two months period. After two months, each group of patients received the other treatment. The fasting blood glucose (112.06 ± 13.98mg/dL) and triglyceride (135.78 ± 65.59mg/dL) levels in treatment group were significantly decreased (p < 0.05) and the HDL level (48.03 ±9.95mg/dL) was significantly increased (p < 0.05) while total cholesterol (189.31 ±33.91mg/dL) and LDL (114.11 ±30.82mg/dL) levels were not significantly changed (p>0.05), respectively compared to baseline. No significant difference in above measured parameters, calorie intakes, and body weights between treatmentand placebo groups during the treatment period. 91.6% of total patients adhered to T. crispa (assessed by capsule count method). The extract increased level of liver enzymes (AST and ALT) more than three times the baseline levels [ 41 ].

A randomized double blind placebo controlled trial to study the hypoglycemic effect of T. crispa was conducted involving 40 patients (included dropouts of six patients) with type 2 diabetes mellitus and glycosylated hemoglobin more than 8.5%, aged above 35 years old, who consumed adequate dose of oral hypoglycemic drug for at least two months, and refused insulin injection. These patients were divided evenly into two groups, i.e. a group given 1 g of T. crispa powder capsule and another given placebo, thrice daily for six months period. Fasting plasma glucose, glycosylated hemoglobin, and insulin levels in treatment group were not significantly different (p ≥ 0.0.5) compared to baseline and placebo. Body weight of treatment group decreased significantly but cholesterol increased significantly (p ≤ 0.05). Changes of liver enzymes profile were not observed [ 42 ].

SAFETY INFORMATION

Preclinical studies (Toxicology studies)

Acute toxicity
Oral single dose acute toxicity study using aqueous mixture of dried powder of T. crispa stem on female Sprague Dawley rats (aged between 8 and 12 weeks old) showed no toxic effect on the parameters observed which includes behaviors, body weight, food and water intakes. All rats were observed for 14 days prior to necropsy. No death was found throughout the study period. Necropsy revealed no significant abnormality. No-observed-adverse-effect level (NOAEL) is more than 2,000 mg/kg body weight[ 39 ].

Ethanol (95%v/v) extract of T. crispa stem (1, 2, and 4 g/kg body weight) given single dose orally to ICR mice (25±2 g body weight) showed no sign of toxicity after seven days observation [ 40 ].

Chronic toxicity
Ethanol (95%v/v) extract of T. crispa stem (0.02, 0.16, and 1.28 g/kg/day) given orally to Wistar rats (200-230 g body weight) for 180 days showed no effect on hematological parameters (hematocrit, white blood cells, platelet, neutrophil, eosinophil, lymphocyte, monocyte). The extract at the concentration of 1.28 g/kg/day significantly decreased creatinine and potassium levels, reduced rats’ body weight (p<0.05, both genders) and showed morphological changes (bile duct proliferation and focal liver cell hyperplasia) on male rats’ liver compared to control [ 41 ].

Others (Adverse reaction, contraindication, side effect, warning, precaution)

Adverse Reaction
Diabetic patient may experience increased appetite, flatulence and dizziness while receiving 250 mg T. crispa dry extract powder capsule (consists of 0.98% dry weight marker substance A) [ 41 ].

Precaution
Extract-thioacetamide treated rats showed significant increase in the levels of liver enzymes and liver body weight ratios. Histological examination revealed hepatocytes degeneration, centrilobular necrosis of hepatocytes, and inflammatory cell infiltration containing lymphocytes and mononuclear cells on liver of the rats [ 43 ].

250 mg T. crispa dry extract powder capsule (consists of 0.98% dry weight marker substance A) given to diabetic patients, at 30 minutes before meal twice daily for two months period, increased the level of liver enzymes (AST and ALT) more than three times the baseline levels. This extract may be a risk factor to elevated liver enzymes [ 41 ].

DOSAGE

Information and data have not been established

STORAGE

Store below 30°C. Protect from light and moisture.

REFERENCES

  1. StuartXchange. [Internet] Tinospora crispa; 2013 [cited on 9th September 2014]. Available from: http://www.stuartxchange.com/Makabuhay.html
  2. Tropicos. [Internet] Missouri: Missouri Botanical Garden; 2014 [cited on 9th September 2014]. Available from: http://tropicos.org/Name/50053822
  3. Yusuf UK, Horsten SFAJ, Lemmens RHMJ. Tinospora crispa (L.) Hook.f. Thomson. in: de Padua, L.S., Bunyapraphatsara, N. and Lemmens, R.H.M.J (editors). Plant resources of South-East Asia No. 12(1): medicinal and poisonous plants 1. Backhuys Publisher, Leiden, The Netherlands. 1999;p.483-484.
  4. Forman LL. A revision of Tinospora (Menispermaceae) in Asia to Australia and the Pacific. The Menispermaceae of Malesia and adjacent areas: XI. Kew Bulletin. 1981;36(2):375–421.
  5. Forman LL. Menispermaceae. In Flora Malesiana ser. I, vol. 10. 1986;p.188-200.
  6. Thai Pharmacopoeia Committee. Thai herbal pharmacopoeia 2009, vol. I. Thailand: Department of Medical Sciences, Ministry of Public Health. 2009;p.9.
  7. Fukuda N, Yonemitsu M, Kimura T. Studies on the constituents of the stems of Tinospora tuberculata Beumee. I. N-trans-and N-cis feruloyl tyramine, and a new phenolic glucoside, tinotuberide. Chemical & Pharmaceutical Bulletin. 1983;31(1):156–161.
  8. Fukuda N, Yonemitsu M, Kimura T. Studies on the constituents of the stems of Tinospora tuberculata Beumee, III. New diterpenoids, borapetoside B and borapetol B. Chemical & Pharmaceutical Bulletin, 1986;34(7):2868–2872.
  9. Fukuda N, Yonemitsu M, Kimura T. Studies on the constituents of the stems of Tinospora tuberculata Beumee. II. New diterpenoids, borapetoside A and borapetol A. Chemical & Pharmaceutical Bulletin. 1985;33(10):4438–4444.
  10. Fukuda N, Nakamura M, Yonemitsu M, Kimura T, Isobe R, Komori T. Studies on the constituents of the leaves of Tinospora tuberculata, I.– Isolation and structure elucidation of two new furanoid diterpenes, tinotufolin A and B. Liebigs Annalen der Chemie. 1993;1993(3):325–327.
  11. Fukuda N, Yonemitsu M, Kimura T. Studies on the constituents of the stems of Tinospora tuberculata, IV. Isolation and structure elucidation of the five new furanoid diterpene glycosides borapetoside C–G. Liebigs Annalen der Chemie. 1993;1993(5):491–495.
  12. Fukuda N, Yonemitsu M, Kimura T, Isobe R, Komori T. Studies on the constituents of the leaves of Tinospora tuberculata, II. Isolation and structure elucidation of four new furanoid diterpenes, tinotufolin C–F. Liebigs Annalen der Chemie. 1994;1994(7):755–757.
  13. Fukuda N, Yonemitsu M, Kimura T. Studies on the constituents of the stems of Tinospora tuberculata, V. Isolation and structure elucidation of the new furanoid diterpene glucoside borapetoside H. Liebigs Annalen, 1995;1995(9):1689–1691.
  14. Choudhary MI, Ismail M, Shaari K, Abbaskhan A, Sattar SA, Lajis NH. Cis-Clerodane-type furanoditerpenoids from Tinospora crispa. Journal of Natural Products. 2010;73(4):541–547.
  15. Umi Kalsom Y, Noor H. Flavone O-glycosides from Tinospora crispa. Fitoterapia. 1995;66(3):280.
  16. Imphanban K, Kongkathip N, Dhumma-upakorn P, Mesripong R, Kongkathip B. Synthesis of N-formylnornuciferine with cardiotonic activity. Natural Sciences. 2009;43(4):738–744.
  17. Choudhary MI, Ismail M, Ali Z, Shaari K, Lajis NH. Alkaloidal constituents of Tinospora crispa. Natural Product Communications. 2010;5(11):1747–1750.
  18. Cavin A, Hostettmann K, Dyatmyko W, Potterat O. Antioxidant and lipophilic constituents of Tinosproa crispa. Planta Medica. 1998;64(5):393–396.
  19. Kongkathip N, Dhumma-upakorn P, Kongkathip B, Chawananoraset K, Sangchomkaeo P, Hatthakitpanichakul S. Study on cardiac contractility of cycloeucalenol and cycloeucalenone isolated from Tinospora crispa. Journal of Ethnopharmacology. 2002;83(1-2):95–99.
  20. Praman S, Mulvany MJ, Williams DE, Andersen RJ, Jansakul C. Hypotensive and cardiochronotropic constituents of Tinospora crispa and mechanisms of action on the cardiovascular system in anesthetized rats. Journal of Ethnopharmacology. 2012;140:166–178.
  21. Lam SH, Ruan CT, Hsieh PH, Su MJ, Lee SS. Hypoglycemic diterpenoids from Tinospora crispa. Journal of Natural Products, 2012; 5(2);153–159.
  22. Perry LM. Medicinal plants of East and Southeast Asia. Massachusetts: MIT Press. 1980;p.268.
  23. Burkill I. A dictionary of the economic products of the Malay Peninsula. Kuala Lumpur: Ministry of Agriculture, Malaysia. 1935;p.2165.
  24. Al-Alusi NT, Kadir FA, Ismail S, Abdullah MA. In vitro Interaction of combined plants Tinospora crispa and Swietenia mahagoni against methicillin-resistant Staphylococcus aureus. African Journal of Microbiology Research. 2010;4(21):2309-2312.
  25. Zakaria ZA, Mat Jais AM, Somchit MN, Sulaiman MR, Faizal FO. The in vitro antibacterial activity of Tinospora crispa extracts. Journal of Biological Sciences. 2006;6(2):398-401.
  26. Rungruang T, Boonmars T. In vivo antiparasitic activity of the Thai traditional medicine plant Tinospora crispa against Plasmodium yoelli. Southeast Asian Journal of Tropical Medicine and Public Health. 2009;40(5):898-900.
  27. Merawin LT, Arifah AK, Sani RA, Somchit MN, Zuraini A, Ganabadi S, Zakaria ZA. Screening of microfilaricidal effects of plant extracts against Dirofilaria immitis. Research in Veterinary Science, 2010;88(1):142–147.
  28. Zaridah MZ, Idid SZ, Omar AW, Khozirah S. In vitro antifilarial effects of three plants species against adult worms of subperiodic Brugia malayi. Journal of Ethnopharmacology. 2001;78(1):79-84.
  29. Sulaiman MR, Zakaria ZA, Lihan R. Antinociceptive and anti-inflammatory activities of Tinospora crispa in various animal models. International Journal of Tropical Medicine. 2008;3(3):66-69.
  30. Noipha K, Ninla-aesong P. The activation of GLUT1, AMPKα and PPARγ by Tinospora crispa in L6 myotubes. Spatula DD. 2011;1(4):245–249.
  31. Zulkhairi A, Hasnah B, Sakinah I, Nur Amalina I, Zamree MS, Mohd Shahidan A. Nutritional composition, antioxidant ability and flavonoid content of Tinospora crispa stem. Advances in Natural and Applied Sciences, 2009;3(1):88–94.
  32. Ibrahim MJ, Wan-Nor I’zzah WMZ, Narimah AHH, Nurul Asyikin Z, Siti-Nur Shafinas SAR, Froemming GA. Anti-proliperative and antioxidant effects of Tinospora crispa (batawali). Biomedical Research. 2011;22(1):57-62.
  33. Zulkhairi A, Abdah MA, M Kamal NH, Nursakinah I, Moklas MAM, Hasnah B, Fazali F, Khairunnur FA, Kamilah KAK, Zamree MS, Shahidan MMA. Biological properties of Tinospora crispa (akar patawali) and its antiproliferative activities on selected human cancer cell lines. Malaysian Journal of Nutrition. 2008;14(2):173-187.
  34. Zulkhairi A, Hasnah B, Zamree MS, Shahidan MA, Nursakinah I, Fauziah O, Maznah I, Taufik Hidayat M, Moklas MA, Khairul Kamilah AK. Potential of Tinospora crispa as a hypocholesterolemic agent in rabbits. Malaysian Journal of Medicine and Health Sciences. 2009;5(2):1-10.
  35. Kongkathip N, Dhumma-upakorn P, Kongkathip B, Chawananoraset K, Sangchomkaeo P, Hatthakitpanichakul S. Study on cardicac contractility of cycloeucalenol and cycloeucalenone isolated from Tinospora crispa. Journal of Ethnopharmacology. 2002;83(1-2):95-99.
  36. Usia T, Iwata H, Hiratsuka A, Watabe T, Kadota S, Tezuka Y. CYP3A4 and CYP2D6 inhibitory activities of Indonesian medicinal plants. Phytomedicine. 2006;13(1-2):67–73.
  37. Subehan, Usia T, Iwata H, Kadota S, Tezuka Y. Mechanism-based inhibition of CYP3A4 and CYP2D6 o Indonesian medicinal plants. Journal of Ethnopharmacology. 2006;105(3):449–455.
  38. Tungpradit R, Sinchaikul S, Phutrakul S, Wongkham W, Chen ST. Anti-cancer compound screening and isolation: Coscinium fenestratum, Tinospora crispa and Tinospora cordifolia. Journal of the Science Faculty of Chiang Mai University. 2010;37(3):476-488.
  39. Teh BP, Hamzah NF, Rosli SNS, Yahaya MAF, Zakiah I, Murizal Z. Acute oral toxicity study of selected malaysian medicinal herbs on Sprague Dawley rats. Institute for Medical Research, Ministry of Health; 2012. Report No.: HMRC 11-045/01/TT/S/P.
  40. Pranee C, Aimmanus A, Anchalee C, Pranee C. Toxicological study of crude extract of Tinospora crispa Mier ex Hook F. & Thoms. Thai Journal of Pharmaceutical Sciences. 1997;21(4):199-210.
  41. Sriyapai C, Dhumma-upakorn R, Sangwatanaroj S, Kongkathip N, Krittiyanunt S. Hypoglycemic effect of Tinospora crispa dry powder in outpatients with metabolic syndrome at King Chulalongkorn Memorial Hospital. Journal of Health Research. 2009;23(3):125-133.
  42. Sangsuwan C, Udompanthurak S, Vannasaeng S, Thamlikitkul V. Randomized controlled trial of Tinospora crispa for additional therapy in patients with type 2 diabetes mellitus. Journal of the Medical Association of Thailand. 2004;87(5):543-546.
  43. Farkaad AK, Faizah O, Mahmmod AA, Farida H, Pouya H. Effect of Tinospora crispa on thioacetamide-induced liver cirrhosis in rats. Indian Journal of Pharmacology. 2011;43(1):64-68.