Zingiber officinale Roscoe

Scientific Name

Zingiber officinale Roscoe

Synonyms

Amomum angustifolium Salisb. [Illegitimate], Amomum zingiber L., Amomum zinziba Hill [Spelling variant], Zingiber aromaticum Noronha [Invalid], Zingiber cholmondeleyi (F.M.Bailey) K.Schum., Zingiber missionis Wall., Zingiber sichuanense Z.Y.Zhu, S.L.Zhang & S.X.Chen, Zingiber zingiber (L.) H.Karst. [Invalid] [1]

Vernacular Name

MalaysiaHalia [2], halia pedas, alia, halia bara, halia padi, halia hudung, halia nasi, halia cina [3]
EnglishGinger, common ginger [2], canton ginger, East Indian ginger, Queensland ginger, stem ginger [4]
ChinaGan jiang, kan chiang, sheng chiang, sheng jiang [4], chiang [5]
IndiaAdrak, aadaa [2], nagara, palu, inguru [5], ardhrakam [6]
IndonesiaJahe, aliah, jae [2], jae merah, sunti [3]
ThailandKinkh, khingdaen, khing klaeng, khing phueak, sa e [2], khing [3]
PhilippinesLuya, baseng, laya [7]
CambodiaKhnhei, khnhei phlung [7]
MyanmarGyin [4]
LaosKhi:ng [7]
BangladeshAda, adrak [4]
JapanJinja, shouga [2], ata-le [6]
FranceGingembre, gingembre commun, gingembre officinal, gingembre traditionnel [2]
Arabs Zanjabil urratab, zanjabil ee-e-tar,khenseing, gnji [5], zanjabeel [6], zenjabil, zingibil [4]
Congo Tangawissi [4]
GhanaKakaduro [4]
NigeriaAtale, cittaraho [4]
Sierra LeoneGinga, kijei, ta san, to sena [4]
TogoAfou [4].

Geographical Distributions

Zingiber officinale is probably originated from south Asia [3]. It is been thought native to Indian and brought to Europe and East Africa by Arabs traders [7]. This plant is distributed in tropical and subtropical Asia and Far East Asia [8].

Botanical Description

Z. officinale is a member of the family Zingiberaceae. It is a biennial or perennial reed-like herb, grows up to 1m and mostly grown for its bulbous rhizome [9].

The stem is non-woody, covered with leaf sheath, with a subterranean, digitately branched rhizome [10].

The sheathing leaves linear lanceolate, about 5-30 cm long and 8-20 mm wide, dark green, alternate, smooth on the upper surface and slightly rough on the underneath which is covered with very fine hairs; ligule light green in colour. [10]

The flower stems shorter than leaf stems, bearing a few flowers, each surrounded by a thin bract and situated in axils of large, greenish yellow obtuse bracts, closely arranged at end of flower stem in whorl forming collectively an ovate-oblong spike; each flower shows a superior tubular calyx, split part way down one side; corolla orange yellow, composed of a tube, divided into 3 separated blunt lobes, linear oblong, ca. 2-2.5 cm long, slightly longer than bractea; dorsal lobe broader, 12 mm by 10 mm; 6 staminodes in 2 rows, the outer row of 3 inserted at mouth of corolla; the posterior 2, small, horn-like; the anterior petaloid, purple and spotted and divided into 3 rounded lobes; an inferior, 3-celled ovary with tufted stigma. [10]

The fruit is a capsule with small arillate seeds [10][11].

The rhizome occurs in horizontal, laterally flattened, irregularly branching pieces, 3-16 cm long, 3-4 cm wide, up to 2 cm thick, sometimes split longitudinally, pale yellowish buff or light brown externally, longitudinally striated, somewhat fibrous; branches known as “fingers” arise obliquely from the rhizomes, flattish, obovate, short, about 1-3 cm long; fracture, short and starchy with projecting fibres. Internally, yellowish brown, showing a yellow endodermis separating the narrow cortex from the wide stele, and numerous scattered fibrovascular bundles; abundant scattered oleoresin cells with yellow contents and numerous larger greyish points; vascular bundles, scattered on the whole surface [10][12][13][14].

Cultivation

Soil Suitability and Climate Requirement

Z. officinale is adapted to tropical environment and is suitable for growing in many locations in Malaysia. The plant thrives well at 1500 m above the sea level. The main growing area of Z. officinale is at Bukit Tinggi, Bentong. The area is about 600-700 m above the sea level with minimum and maximum temperature of 16oC and 28oC, respectively. The total annual rainfall of 2000-3000 mm is required for good growth. Z. officinale can be planted on various soil types from peat to heavy clay soil. Soil with good drainage is favoured to avoid bacterial wilt disease. It is very susceptible to waterlogged condition. [15]

Field Preparation

Land Preparation

Good land preparation is a prerequisite to ensure successful production. For mineral soil, the land has to be ploughed and rotovated 1-2 times. The newly-developed peat soil with pH of 3.6 has to be limed with Ground Magnesium Limestone (GML) at the rate of 7.5 t/ha. On mineral soil normally no lime is required unless the pH is less than 5. Bed is prepared after soil preparation especially for soil with poor drainage. The bed is normally 60-120 cm wide and 25-30 cm high. The distance between each bed is 30-80 cm. [15]

Production of Planting Materials

Z. officinale is propagated using clean, mature rhizomes of more than 9 months in the field. About 1500-2000 kg of fresh rhizomes is required for one hectare of land. Rhizomes have to be kept at cool, wet place to encourage bud formation which takes place 2-3 weeks at storage. Rhizomes are then split cut 5 cm long with 2 buds. About 25-30 cuttings are obtained from 1 kg of rhizomes. The cut rhizome has to be treated with Bordeaux mixture (2 kg copper sulphate, 2 kg lime and 250 litres water) or 1% formalin solution for 5 minutes. The cuttings are dried before planting. [15]

Field Planting

Planting should be done at the onset of rain. The planting distance is 25 cm within a row and 45 cm between rows with 2-3 rows in one bed. The cut rhizome is sown 10 cm deep with the bud upright. Mulching using dried coconut leaves or rice straw should be done 2-4 weeks after planting when newly-developed leaves just appear. This is done until three months to avoid leaf scorch which could lead to leaf spot disease. [15][16]

Field maintenance

Fertilisation

Z. officinale need considerably high nutrient supply. High organic and inorganic fertilisers are needed to ensure good growth. Inorganic compound fertiliser NPK (12:12:17:2+TE) is applied in split of 4-5 times per growing season and the fertiliser is divided equally. Organic fertiliser using chicken manure is applied 3-5 days before planting. [15]

Weed Control

Weed control is done once in 2-3 months. The next weeding is normally done together with hilling. Hilling is actually rising up the soil at the plant base to cover the rhizome from being exposed to sunlight and to encourage growth. Mulching using rice straw or dried coconut leaves helps to control weeds, avoids bed slacken off, controls nutrient loss and maintains soil moisture. [15]

Water management

A sufficient water supply is needed for ‘Z. officinale cultivation. Irrigation is needed during the early growth stage unless it rains. [15]

Pest and Disease Control

Stem borer (Udapes falus) is the main pest of Z. officinale. The borer makes holes in the stem and subsequently leaves will turn yellowish and dried. Stem borer can be controlled using insecticide such as permethrin, acephate or methamidophos. Bacterial wilt is the major disease that could lead to great loss in Z. officinale production. The disease is caused by Pseudomonas solanacearum, which can remain in the soil for years. The symptom is the yellowing of lower leaves and spreads to the upper leaves. The leaves subsequently become brown, wet, soft and easily detached from the stem. Planting using treated planting materials and alternating the area with other crops can reduce this disease problem. [15]

Harvesting

Harvesting is done for green Z. officinale or mature Z. officinale at 4-5 months and 8-9 months respectively, after planting. The best time of harvest is when the plant is at 7-8 months of age. Rhizome harvested after 9 months is fibrous and this lowers the quality. Harvesting is done when the stem becomes brownish. Z. officinale has to be harvested carefully using a hoe to avoid bruising the rhizome. The cleaned rhizome has to be kept away from full sunlight to avoid moisture loss and change of colour. The yield of mature Z. officinale and green Z. officinale are 15-22 t/ha and 8-12 t/ha respectively. [15]

Postharvest handling

Rhizome has to be cleaned from dried leaves, stems and soils. Z. officinale is placed in bamboo or plastic basket for washing under running water. Care is needed to avoid bruising Z. officinale during cleaning. To control fungus, rhizome is soaked in Bordeaux mixture (500 ppm) for 15 minutes and left to dry. The cleaned rhizome is kept at room temperature for 4-5 days to allow a protection layer to be formed outside the rhizome. [15]

Estimated cost of production

The estimated cost of production per hectare of Z. officinale is RM24720.00. Based on 15-22 t/ha rhizome yield, the production cost is estimated to be RM1.12-RM1.65/kg. The production cost was estimated based on the cost of current inputs during writing of this article. [15]

Chemical Constituent

Aqueous extract of Z. officinale rhizome has been reported to contain hexanoic phosphate and gingerlanosterol. [17]

Ethanol extracts of Z. officinale rhizome has been reported to contain diarylheptanoids (e.g. (3S,5S)-3,5-diacetoxy-1,7-bis(4-hydroxy-3-methoxyphenyl)heptane, 3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane, (3R,5S)-3-acetoxy-5-hydroxy-1, 7-bis(4-hydroxy-3-methoxyphenyl)heptane, (3R,5S)-3,5-dihydroxy-1-(4-hydroxy-3,5-dimethoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane, (5S)-5-acetoxy-1,7-bis(4-hydroxy-3-methoxyphenyl)heptan-3-one, 5-hydroxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptan-3-one, 5-hydroxy-1-(4-hydroxy-3-methoxyphenyl)-7-(3,4-dihydroxy-5-methoxyphenyl)heptan-3-one, 1,5-epoxy-3-hydroxy-1-(4-hydroxy-3,5-dimethoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl) heptane and 1,5-epoxy-3-hydroxy-1-(3,4-hydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane), acyclic diarylheptanes (e.g. (3R,5S)-3,5-di-acetoxy-1,7-bis(4-hydroxy-3-methoxyphenyl)heptane, (3R,5S)-3,5-diacetoxy-1-(4-hydroxy-3,5-dimethoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane, (3S,5S)-3,5-dihydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl)heptane, (3R,5S)-3,5-dihydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl)heptane), diarylheptanones (e.g. 5-hydroxy-1-(4-hydroxy-3-methoxyphenyl)-7-(3,4-dihydroxyphenyl)heptan-3-one, 7-(3,4-dihydroxy-5-methoxyphenyl)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl)heptan-3-one, 5-hydroxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptan-3-one, hexahydrocurcumin, 1,7-bis(4-hydroxy-3-methoxyphenyl)heptane-3,5-dione), cyclic diarylheptanes (e.g. 3-acetoxy-1,5-epoxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane, 1,5-epoxy-3-hydroxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(3,4-dihydroxyphenyl)heptane), diarylheptenones (e.g. 1,7-bis(4-hydroxy-3-methoxyphenyl)hept-4-en-3-one, gingerenone C), paradols (e.g. [6]-paradol, [8]-paradol, zingerone), gingerdiols (e.g. (3S,5S)-[6]-gingerdiol, (3R,5S)-[6]-gingerdiol, (3R,5S)-3,5-diacetoxy-[6]-gingerdiol), gingerols (e.g. [4]-gingerol, [6]-gingerol, [8]-gingerol, [10]-gingerol, 5-acetoxy-[6]-gingerol, [6]-gingeroldiacetate), gingerdione (e.g. [10]-gingerdione), dehydrogingerdiones (e.g. 1-dehydro-[6]-gingerdione, 1-dehydro-[8]-gingerdione), shogaols (e.g. [6]-shogaol, [10]-shogaol, [6]-dehydroshogaol, 6-hydroxy-[6]-shogaol), steroids (e.g. ß-sitosterol, stigmasterol, 6ß-hydroxystigmast-4-en-3-one) and others (e.g. 1-(3-methoxy-4-hydroxy-phenyl)-propan-1,2-diol, 1,7-bis(4-hydroxy-3-methoxyphenyl)heptane-3,5-diol, 6-(hydroxymethyl)tetrahydro-2H-pyran-3,4-5-triol, 5-[4-hydroxy-6-(4-hydroxyphenethyl)tetrahydro-2H-pyran-2-yl]-3-methoxybenzene-1,2-diol, 1-(3,4-dimethoxyphenyl)-5-hydroxy-decan-3-one, 4a,5ß-dihydroxybisabola-2,10-diene, galanolactone, citrylidenmalonsaeure, shogasulfonic acid A, 6-gingesulfonic acid, (E)-geranylferulic acid, (Z)-geranylferulic acid, (2R,3R,4S,5S,6R)-2((1S,2S,4S)-4-hydroxy-2-methylcyclohexyloxy)-sodium 6-gingesulfonate, sodium (E)-7-hydroxy-1,7-bis(4-hydroxyphenyl)hept-5-ene-3S-sulfonate, sodium (E)-7-hydroxy-1,7-bis(4-hydroxyphenyl)hept-5-ene-3R-sulfonate, hydroxyl-cineole-10-O-ß-D-glucosidase, trans-ß-sesquiphellandrol, trans-sesquipiperitol). [18][19][20][21][22]

Methanol extract of Z. officinale rhizome has been reported to contain gingerols (e.g. [6]-gingerol, [8]-gingerol, [10]-gingerol), gingerdiones (e.g. dehydro-[6]-gingerdione, dehydro-[10]-gingerdione), shogaols (e.g. [6]-shogaol, [8]-shogaol, [10]-shogaol), zingerines (e.g. [6]-zingerine, [8]-zingerine, [10]-zingerine), [6]-gingediol and [6]-paradol [23][24][25]. Whereas the methanolic aqueous extract had [6]-gingerol, [8]-gingerol, [10]-gingerol, [6]-shogaol, [6]-paradol, [6]-gingeacetate, [4]-gingesulfonic acid, [6]-gingesulfonic acid and shogasulfonic acids A-D [26].

Chloroform extract of Z. officinale rhizome has been reported to contain [6]-gingerol, [10]-gingerol, [6]-shogaol, [10]-shogaol and hexahydrocurcumin. [27]

Dichloromethane extract of Z. officinale rhizome has been reported to contain diarylheptanoids (e.g. (Z)-1,7-bis-(4’-hydroxy-3’-methoxyphenyl)-4-hepten-3-one, (E)-1,7-bis-(4’-hydroxy-3’-methoxyphenyl)-4-hepten-3-one, 1,7-bis-(4’-hydroxy-3’-methoxyphenyl)-5-methoxyheptan-3-one, 1-(4’-hydroxy-3’,5’-dimethoxyphenyl)-7-(4’-hydroxy-3’-methoxyphenyl)-4-hepten-3-one, 1,7-bis-(4’-hydroxy-3’-methoxyphenyl)-3-hydroxy-5-acetoxyheptane, 1,7-bis-(4’-hydroxy-3’-methoxyphenyl)-3,5-heptadione, meso and (3S,5S)-3,5-diacetoxy-1,7-bis-(4’-hydroxy-3’-methoxyphenyl)heptane, 3-acetoxy-1,5-epoxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane, epimer of 3-acetoxy-1,5-epoxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane at C-3, 1,5-epoxy-3-hydroxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane, epimer of 1,5-epoxy-3-hydroxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane at C-3, 1,5-epoxy-3-hydroxy-1-(4-hydroxy-3,5-dimethoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane), paradols (e.g. [1]-paradol, [6]-paradol, [7]-paradol, [8]-paradol, [9]-paradol, [10]-paradol, [11]-paradol, [13]-paradol, methyl [6]-paradol, methyl [8]-paradol, dihydro-[6]-paradol, acetoxydihydro-[6]-paradol, methyl ether derivative of acetoxydihydro-[6]-paradol, 1-hydroxy-[6]-paradol), gingerols (e.g. [4]-gingerol, [5]-gingerol, [6]-gingerol, [7]-gingerol, [8]-gingerol, [10]-gingerol, methyl [4]-gingerol, methyl [6]-gingerol, [4]-isogingerol, methyl [6]-isogingerol, acetoxy-[4]-gingerol, acetoxy-[6]-gingerol, acetoxy-[8]-gingerol, acetoxy-[10]-gingerol, methyl acetoxy-[6]-gingerol, methoxy-[4]-gingerol, methoxy-[6]-gingerol, methoxy-[8]-gingerol, methoxy-[10]-gingerol, demethoxy-[6]-gingerol, 5-acetoxy-3-deoxy-[6]-gingerol, 1-dehydro-[6]-gingerol, 6-dehydro-[6]-gingerol), shogaols (e.g. (E)-[4]-shogaol, (E)-[5]-shogaol, (Z)-[6]-shogaol, (E)-[6]-shogaol, (Z)-[8]-shogaol, (E)-[8]-shogaol, (Z)-[10]-shogaol, (E)-[10]-shogaol, (E)-[12]-shogaol, methyl (E)-[4]-shogaol, methyl (E)-[6]-shogaol, methyl (E)-[8]-shogaol, [6]-isoshogaol, demethoxy-[6]-shogaol, 3-acetoxy-3-dihydrodemethoxy-[6]-shogaol, 6-hydroxy-[6]-shogaol, 6-hydroxy-[8]-shogaol, 6-hydroxy-[10]-shogaol, 3-dihydro-[6]-demethoxyshogaol), gingerdiones (e.g. [6]-gingerdione, [8]-gingerdione, [10]-gingerdione, [12]-gingerdione, 1-dehydro-[3]-gingerdione, 1-dehydro-[6]-gingerdione, 1-dehydro-[8]-gingerdione, 1-dehydro-[10]-gingerdione, 1-dehydro-3-dihydro-[10]-gingerdione), gingerdioles (e.g. [4]-gingerdiol, [6]-gingerdiol, [8]-gingerdiol, [10]-gingerdiol, 5-acetoxy-[4]-gingerdiol, 3-acetoxy-[4]-gingerdiol, 5-acetoxy-[6]-gingerdiol, stereoisomer of 5-acetoxy-[6]-gingerdiol, 5-acetoxy-[7]-gingerdiol, methyl 5-acetoxy-[4]-gingerdiol, methyl 5-acetoxy-[6]-gingerdiol, diacetoxy-[4]-gingerdiol, diacetoxy-[6]-gingerdiol, diacetoxy-[8]-gingerdiol, methyl diacetoxy-[4]-gingerdiol, methyl diacetoxy-[6]-gingerdiol, methyl diacetoxy-[8]-gingerdiol, methyl diacetoxy-[10]-gingerdiol, (2E)-geranial acetal of [4]-gingerdiol, (2Z)-neral acetal of [6]-gingerdiol, (2E)-geranial acetal of [6]-gingerdiol, acetaldehyde acetal of [6]-gingerdiol, cyclic methyl orthoester [6]-gingerdiol, cyclic methyl orthoester [10]-gingerdiol) and others (e.g. 6-(4’-hydroxy-3’-methoxyphenyl)-2-nonyl-2-hydroxytetrahydropyran, 1-(4’-hydroxy-3’-methoxyphenyl)-2-nonadecen-1-one, 1-(3’,4’-dimethoxyphenyl)-2-nonadecen-1-one, 1-(4’-hydroxy-3’-methoxyphenyl)-7-decen-3-one, 1-(4-hydroxy-3-methoxyphenyl)-2,4-dehydro-6-decanone, 1-(4’-hydroxy-3’-methoxyphenyl)-7-octen-3-one, 1-(4’-Hydroxy-3’-methoxyphenyl)-7-dodecen-3-one, 3-(4’-hydroxyphenyl)-1-propanal, 3-(4’-hydroxy-3’-methoxyphenyl)-1-propanal, 3-(3’,4’-dihydroxy-5’-methoxyphenyl)-1-propanal, 3-(4’-hydroxy-3’,5’-dimethoxyphenyl)-1-propanal, 3-(3’-hydroxy-4’,5’-dimethoxyphenyl)-1-propanal, 2-dehydro-3-(3’,4’-dimethoxyphenyl)-1-propanal, 2-dehydro-3-(3’,4’,5’-trimethoxyphenyl)-1-propanal, 4-(4’-hydroxyphenyl)-2-butanone, 4-(3’,4’-dihydroxy-5’-methoxyphenyl)-2-butanone, 4-(4’-hydroxy-3’,5’-dimethoxyphenyl)-2-butanone, 4-(3’-hydroxy-4’,5’-dimethoxyphenyl)-2-butanone, 4-(3’,4’,5’-trimethoxyphenyl)-2-butanone, 5-(4’-hydroxy-3’-methoxyphenyl)-pent-2-en-1-al, 5-(4’-hydroxy-3’-methoxyphenyl)-3-hydroxy-1-pentanal, 1,7-bis-(4’-hydroxy-3’-methoxyphenyl)-4-heptene-3-one, 1,7-bis-(4’-hydroxy-3’-methoxyphenyl)-3,5-heptadione, p-hydroxybenzaldehyde, vanillin, 4-vinylguaiacol, 4-vinylpyrogallol monomethyl ether, 4-vinylsyringol, acetovanillone, methyl vanillate, 2-(4’hydroxy-3’-methoxyphenyl)ethyl acetate, ferulic acid, zingerol, zingerone, zingerone methyl ether). [28][29][30]

Petroleum ether extract of Z. officinale rhizome has been reported to contain [4]-gingerol, [6]-dehydrogingerdione and [6]-dihydrogingerdione. [31]

Oleoresin of Z. officinale fresh rhizomes has been reported to contain trans-6-shogaol, trans-10-shogaol, α-zingiberene, 10-Gingerdione, cis-10-Shogaol, 8-Gingerdione, 6-Gingerdiol-diacetate, trans-8-Shogaol, cis-8-Shogaol, 6-Gingerol, 6-Paradol, β-Sesquiphellandrene, E,E-α-Farnesene, β-Bisabolene, ar-Curcumene. [32]

Essential oils of Z. officinale rhizome has been reported to contain allo aroma dendrene, a-bergamotene, ß-besabolene, endo borneol, endo bornyl acetate, ß-bourbonene, γ-cadinene, calamenene, calarene, camphene, camphor, trans-2-caren-4-ol, 1,8-ceneole, z-citral (neral), citral (geranial), citronella, ß-citronellol, citronellyl acetate, a-copaene, ß-cubebene, cuparene, a-curcumene, cyclosativen, 1-decanol, ß-elemene, γ-elemene, elemol, (-)-epiglobulol, ß-eudesmol, ß-farnasene, farnesene, (-)-farnesol, trans-geraniol, geranyl acetate, germacrene B, a-guaen, 2-heptanol, juniper camphor, ledol, linalool, 6-methyl-5-hepten-2-one, a-muurolene, ß-myrcene, myrtenal, nerolidol, nerol, 2-nonanone, cis-ocimene, a-phellandrene, ß-phellandrene, a-pinene, 2-ß-pinene, a-selinene, ß-selinene, epibicyclo sesquiphellandrene, ß-sesquiphellandrene, cis-ß-sesquiphellandrol, trans-ß-sesquiphellandrol, terpinene-4-ol, α-terpineol, α-terpinolene, torreyol, 2-undecanone, verbenone, viridiflorol and zingiberene. [33][34][35]

Essential oil Z. officinale fresh rhizomes has been reported to contain α-zingiberene, Valencene, 6-methyl-5-hepten-2-one + myrcene, β-phellandrene, 1,8-cineole, α-terpineol, citronellol, geraniol, bornyl acetate, ar-curcumene, (E,E)-α-farnesene, germacrene-D, β-bisabolen, (E)-nerolidol, and elemol. [32][36]

Z. officinale rhizome has been reported to contain to contain diarylheptanoids (e.g. 5-hydroxy-7-(4-hydroxyphenyl)-1-(4-hydroxy-3-methoxyphenyl)-3-heptanone, 3,5-diacetoxy-7-(3,4-dihydroxyphenyl)-1-(4-hydroxy-3-methoxyphenyl)heptane, 5-hydroxy-7-(4-hydroxy-3,5-dimethoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)-3-heptanone, 5-hydroxy-7-(4-hydroxy-3,5-dimethoxyphenyl)-1-(4-hydroxy-3-methoxyphenyl)-3-heptanone, (3R,5S)-3,5-dihydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl)heptane, (3S,5S)-3,5-diacetoxy-1,7-bis(3,4-dihydroxyphenyl)heptane, (4E,6E)-7-(3,4-dihydroxy-5-methoxyphenyl)-1-(4-hydroxy-3-methoxyphenyl)hepta-4,6-dien-3-one, 5-hydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl)heptan-3-one, 7-(3,4-dihydroxyphenyl)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl)heptan-3-one, (E)-1,7-bis(4-hydroxy-3-methoxyphenyl)hept-1-ene-3,5-dione, (E)-7-(4-hydroxy-3-methoxyphenyl)-1-(4-hydroxyphenyl)-hept-1-ene-3,5-dione, 1,5-epoxy-3-hydroxy-1-(3,4-dihydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl)heptane, (3S,5S)-3,5-diacetoxy-1,7-bis(3,4-dihydroxyphenyl)heptane, curcumin, tetrahydrocurcumin, hexahydrocurcumin), diarylheptenones (e.g. gingerenone A-C, isogingerenone B), gingerdiols (e.g. [6]-gingerdiol, (3R,5S)-5-acetoxy-3-hydroxy-1-(4-hydroxy-3-methoxyphenyl)decane, (3R,5S)-3-acetoxy-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl)decane, (3R,5S)-3,5-diacetoxy-1-(4-hydroxy-3-methoxyphenyl)decane and (3R,5S)-3,5-diacetoxy-1-(3,4-dimethoxyphenyl)decane) and [10]-dehydrogingerdione. [37][38][39][40][41]

Crude extract of Z. officinale rhizome has been reported to contain ginger proteases (GP-I and GP-II), 1,5-epoxy-3-hydroxy-1-(3-methoxy-4,5-dihydroxyphenyl)-7-(4-hydroxyphenyl)-heptane, 10-O-ß-D-glucopyranosyl-hydroxy cineole, 1-(4-O-ß-D-glucopyranosyl-3-methoxyphenyl)-3,5-dihydroxydecane, 5-O-ß-D-glucopyranosyl-3-hydroxy-1-(4-hydroxy-3-methoxyphenyl)decane, [6]-gingerol, [8]-gingerol, [10]-gingerol, [6]-shogaol, [8]-shogaol, [10]-shogaol, [6]-dehydroshogaol, [8]-dehydroshogaol, [10]-dehydroshogaol and [1]-dehydrogingerdione. [42][43][44][45][46][47]

Plant Part Used

Root/ Rhizome [48]

Traditional Use

Z. officinale is used not only as a dietary ingredient but also for its medicinal value; often for the same purpose. Z. officinale is widely known for its effect on the gastrointestinal system, and in African traditional medicine, this is no different. When used as a culinary herb, Z. officinale can be useful as a carminative, diuretic and antiemetic [6]. The dried rhizomes have been used as a primary ingredient for stomachics which are used to treat nausea, indigestion and flatulence [49]. In cases of abdominal pain, Zimbabwe people used the ground rhizome by steeped it in hot water and drunk. It also has been chewed raw and the juices swallowed for general abdominal pain [50][51]. Several pieces of the rhizome of Z. officinale have been used in a decoction along with the aerial parts of Ocimum americanum and Xylopia aethiopica in order to treat colic, constipation or irregularity [48].

Z. officinale has also been used as a popular respiratory aide. Decoctions of Z. officinale rhizome have been mixed with milk in order to suppress coughing [52]. In cases of persistent cough or bronchitis, the rhizome has been chewed raw. In order to ease the intensity of the rhizome, a sweetener is added [48].

Z. officinale rhizome has been used as a stimulant. An infusion of the rhizome is thought to stimulate the Central Nervous System or even relieve Amnesia [53]. The macerated rhizome is eaten twice daily as a general stimulant [54].

Preclinical Data

Pharmacology

Antidiarrheal activity

Methanol extract of Z. officinale rhizome (up to 100 µg/mL) inhibited Escherichia coli heat-labile enterotoxin (LT)-induced diarrhoea by blocking the binding of LTB to cell-surface receptor ganglioside GM1 with an IC50 value of 2.0 µg/mL using GM1-enzyme-linked immunosorbent assay. [55]

Antihyperlipidimic acitivity

Ethanol extract of Z. officinale rhizome (200 mg/kg) administered orally to cholesterol-fed rabbits for duration of 10 weeks showed a hypocholesterolemic effect similar to the drug gemfibrozil. The extract significantly (p<0.01) reduced the elevated serum and tissue cholesterol, serum triglycerides, serum lipoproteins and phospholipids in the rabbit with lower degree of atherosclerosis observed. [56]

Ethanol extract of Z. officinale rhizome (200 mg/kg/day) administered orally to streptozotocin-induced diabetic male and female Wistar rats for a duration of 20 days significantly (p<0.01) decreased serum total cholesterol, triglycerides, liver and pancreas thiobarbituric acid reactive substances values and significantly (p<0.01) increased the level of high-density lipoprotein. [57]

Aqueous extract of Z. officinale rhizome (500 mg/kg/day) administered orally and intraperitoneally to adult female Sprague Dawley rats for a duration of 4 weeks significantly (p<0.05) decreased cholesterol level with no changes in triglycerides level. [58]

Standardized ethanol extract of Z. officinale consumed as a dietary supplement significantly decreased the development of atherosclerotic lesions, and also was associated with a significant reduction in plasma and LDL cholesterol levels and a significant reduction in the LDL basal oxidative state, as well as their susceptibility to oxidation and aggregation. [59]

Thermogenic activity

Gingerols and shogaols isolated from Z. officinale have been reported to induce perfused rats hindlimb muscle to consume oxygen in association with increases in perfusion pressure and lactate production, leading to a thermogenic state. [60]

Antioxidant activity

Extract of Z. officinale (100 mg/kg/day) administered orally to lead acetate-induced apoptosis male Wistar rats for a duration of 10 weeks significantly (p<0.05) increased plasma superoxides dismutase activity, plasma catalase activity and decreased the plasma malondialdehyde. [61]

Geranial from the essential oil and oleoresins (ethanol, methanol, CCl4 and isooctane) of Z. officinale showed stronger antioxidant activity compared to butylated hydroxyanisole (BHT), which is an antioxidant commonly used in foods when evaluated against mustard oil by peroxide, anisidine, thiobarbituric acid (TBA), ferric thiocyanate (FTC) and 2,2′-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging methods. [62]

A glucoside of 6-gingerdiol (5-O-beta-D-glucopyranosyl-3-hydroxy-1-(4-hydroxy-3-methoxyphenyl)decane) isolated from fresh Z. officinale showed strong antioxidant activity by a linoleic acid model system and DPPH radical-scavenging ability compared to its aglycon, 6-gingerdiol. [44]

Z. officinale (1% w/w) fed to rats showed significant increase in blood glutathione content and decreased in lipid peroxidation by supporting the activities of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase, comparable to natural antioxidant ascorbic acid (100 mg/kg, body wt) [63]. The extract also attenuated the malathion induced lipid peroxidation and oxidative stress in rats by decreased the elevated malondialdehyde (MDA) levels in serum, activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) in erythrocytes and glutathione reductase (GR) and glutathione S-transferase (GST) in serum while increased the glutathione (GSH) level in whole blood [64].

Immunomodulatory activity

Volatile oil of Z. officinale (0.001-10 ng/mL) administered to mice in vitro showed that the essential oil could modulate immune function by significantly (p < 0.01) inhibiting T-lymphocyte proliferation, decreased the number of the total T lymphocytes and T helper cells (p < 0.01) in a concentration-dependent manner, but increased the percentage of T suppressor cells to the total T lymphocytes in the mice. It also inhibited IL-1alpha secretion by the mice peritoneal macrophages in a concentration-dependent manner. In vivo examination showed that oral administration of the Z. officinale volatile oil (0.125, 0.25 and 0.5 g/kg body weight) weakened the delayed type of hypersensitivity response to 2,4-dinitro-1-fluorobenzene in the immune-sensitised mice (p < 0.05). In turn, this could be beneficial in many clinical conditions such as autoimmune diseases. [65]

Anticancer activity

6-gingerol, a phenolic alkanone isolated from Z. officinale showed anticancer activity to human cell lines such as gastric cancer cell and breast cancer cell (MDA-MB-231). The compound enhanced the TRAIL-induced viability reduction of gastric cancer cells by inhibiting TRAIL-induced NF-kappaB activation. 6-gingerol showed metastasis effects of MDA-MB-231 by inhibit cell adhesion, invasion, motility, activity and the amount of Matrix Metalloproteinase 2 (MMP2) or Matrix Metalloproteinase 9 (MMP9). [66][67]

Antithrombotic activity

Aqueous extract of Z. officinale rhizome (500 mg/kg/day) administered orally to adult female Sprague Dawley rats for a duration of 4 weeks significantly (p<0.05) decreased (50%) the synthesis of thromboxane B. The extract administered intraperitoneally showed no reduction of thromboxane B synthesis. [58]

Profertility activity

Aqueous extract of Z. officinale rhizome (24 mg/mL) administered orally three times weekly to metiram induced-testicular damage of sexually mature male albino mice for a duration of 6 weeks significantly (p<0.05) increased the diameter of seminiferous tubules and epithelial height of testis, increased spermatogenic cells and decreased the percentage of Bax positive germ cells. [68]

Aqueous extract of Z. officinale rhizome (500 and 1000 mg/kg/day) administered orally to adult male rats for a duration of 14 and 28 days significantly (p<0.05) increased weight of testis and epididymis, sperm count and motility, testosterone level and significantly (p<0.05) decreased malonhydialdehyde level. [69]

Antidiabetic activity

Aqueous extract of Z. officinale root (500 mg/kg/day) administered orally to alloxan-induced diabetic albino rats for a duration of 6 weeks significantly (p<0.05) decreased serum glucose level. [70]

The juice of Z. officinale rhizome (4mL/kg/day) administered orally to streptozotocin-induced diabetic male Sprague Dawley rats for a duration of 6 weeks significantly (p<0.05) decreased fasting blood glucose level and significantly (p<0.05) increased insulin level. [71]

Two compounds ( 2-(4-hydroxy-3-methoxyphenyl)ethanol and 2-(4-hydroxy-3-methoxyphenyl)ethanoic acid) isolated from Z. officinale showed a good inhibitors of recombinant human aldose reductase with IC50 values of 19.2 ± 1.9 µM and 18.5 ± 1.1 µM, respectively. The compounds also significantly suppressed sorbitol accumulation in human erythrocytes and lens galactitol accumulation in 30% of galactose-fed cataract rat. The result suggested the dietary supplement of Z. officinale that contain aldose reductase inhibitors properties can contribute to the protection against or improvement of diabetic complications. [72]

A concurrent treatment of ginger extract (25 or 50 mg/kg) and glibenclamide (5 mg/kg) significantly reduced non-fasting blood glucose level by 26 and 25%, respectively, compared to 7.9% reduction when glibenclamide was used alone in rat model of streptozotocin (STZ)-induced diabetes [128]

In another study, a combination of ginger extract and a sub-optimal dose of glibenclamide (0.5 mg/kg) was found to exert effects similar to a full therapeutic dose of glibenclamide (1 mg/kg) in the STZ-induced diabetic model, highlighting the possibility of reduced side-effects of antidiabetics (due to the lower dose required) when used in combination with ginger extract [129]

Antimicrobial activity

Essential of Z. officinale rhizome showed antimicrobial action against yeast, filametous fungi, gram positive bacteria, and gram negative bacteria, when tested using the agar diffusion method among two other Zingiberaceae namely Aframomum danielli (Hook. f.) K. Schum and Curcuma longa L. [73] The high-intensity ultrasound-assisted (HI-US) solvent-extracted of Z. officinale Thai species also showed antimicrobial activity against Listeria monocytogenes and Salmonella Typhimurium DT 104 using an agar dilution assay [74].

Antibacterial

The green parts of Z. officinale also showed antibacterial activity by inhibit M. luteus in heat treatment using boiling water at 100oC within 20 min. [75]

Antifungal activity

Ethanol extract of Z. officinale rhizome (10 mg/mL) inhibited the growth of Candida albicans with MIC values of 2 mg/mL by using antifungal disk assay. [76]

Essential oil of Z. officinale fresh rhizome exhibited different strengths of antifungal activity against different species. Antifungal activity was shown to be 100% against Fusarium oxysporum while the oleoresin extract was 100% antifungal against Aspergillus niger. [32] Another study indicated that essential oil and CCl4 oleoresin of Z. officinale showed a100% inhibition against Fusarium moniliforme. [62]

Essential oil of Z. officinale was found to be least efficient antifungal action when tested against fluconazole-susceptible yeasts. The susceptibilities of fluconazole-resistant C. albicans, C. dubliniensis, and Candida non-albicans to ginger essential oils were higher than those of the fluconazole-susceptible yeasts (P<0.05). [77]

Essential oil of Z. officinale showed moderate antifungal activity when tested against food born pathogens Fusarium moniliforme, Aspergillus flavus and Aspergillus fumigatus using agar dilution technique with the dose recorded were between 800 and 2500 ppm (parts per million). [78]

Antiviral

Z. officinale dried rhizome showed antirhinoviral activity in vitro using plaque reduction test with IC50 value of 0.44 µM compared to rhinovirus IB. The most active compound was identified as β-sesquiphellandrene. [79]

Toxicity

Acute toxicity

A patented standardized ethanol extract of Z. officinale rhizome EV.EXT™ 33 (up to 100 mg/kg) administered orally to male Wistar rats for duration of 3 hours showed no changes in systolic blood glucose or heart rate. [80]

Oral single dose acute toxicity study on female Sprague Dawley rats (aged between 8 and 12 weeks old) using aqueous mixture of Z. officinale rhizome powder 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. LD50 > 2000 mg/kg. [81]

Z. officinale root (2500 mg/ kg) in saline administered orally as a single dose to adult male albino rats for a duration of 24 hours significantly (P<0.0001) decreased blood pressures and heart rate and induced prenecrotic changes in cardiac tissues with LD50 value of > 2500 mg/kg. [82]

Methanol and aqueous extract of Z. officinale root (up to 17.5 g/kg bw) administered orally as a single dose to male Swiss mice (8-10 weeks old) for a duration of 48 hours showed no toxic effect with LD50 value of 10.25 and 11.75 g/kg bw respectively. [83]

Sub-acute toxicity

Dried Z. officinale roots (500 mg/kg/day) in saline administered orally to adult male albino rats for a duration of 28 days showed hypotension and bradycardia effect with degenerative changes in cardiac myocyte fibers. [82]

Fine powder of Z. officinale roots (up to 2000 mg/kg/day) in gum arabic administered orally to male and female Sprague Dawley rats for a duration of 35 days showed no toxic effect in behavior, growth, and food and water consumption. [84]

Teratogenicity

Z. officinale rhizome tea (up to 50 g/L/day) administered orally to pregnant Sprague Dawley rats (8-12 weeks old) for a duration of 5 days from gestation day 6 to gestation day 15 showed no teratogenic effect with advanced skeletal development of fetuses and increased growth of surviving foetuses. [85]

Clinical Data

Clinical findings

Antiemetic activity

Outpatient surgery

A clinical trial to study the antiemetic property of Z. officinale root as compared to metoclopramide and placebo by monitoring the incidence of nausea and vomiting in outpatient surgery was carried out. It is a prospective, randomised double-blind controlled trial which involved 120 gynaecological patients of American Society of Anesthesiologists graded 1-3; aged over 16 years old, scheduled for elective laparoscopic surgery on a day stay basis. The patients were divided into 3 groups, they were given two capsules containing 10 mg metoclopramide, 1 g powdered Z. officinale rhizome, and 1 g lactose for three respective patient groups, at 1 hour before anaesthesia induction. The observations were made at discharge from the recovery ward, at hospital discharge and 24 hours postoperatively. Results showed a decreased in postoperative nausea or vomiting incidence. The requirement of postoperative antiemetic agents were also decreased as compared to the placebo treated patients. [86]

A double blind randomized controlled trial to study the efficacy of Z. officinale to prevent nausea and vomiting after outpatient gynecological laparoscopy has been carried out and compared to placebo among the 80 patients. The results recorded significant difference in the incidence of the nausea between Z. officinale treated [12 (30%)] and placebo treated [23 (57.50%)] group. The visual analogue nausea scores (VANS) was lower in Z. officinale group than placebo at 2 and 4 hours (p < 0.05), similar with the vomiting frequency incidence. It showed that Z. officinale is effective in prevention of nausea after outpatient gynecological laparoscopy. [87] Similar study has been carried out involving 120 patients who underwent major gynecologic surgery. The result also showed promising effect of Z. officinale as antiemetic to after surgery patients. [88]

A double-blind, randomised, placebo-controlled study showed that the effectiveness of Z. officinale root presurgically as an antiemetic agent was comparable with metoclopramide when studied in 60 women who had major gynaecological surgery. The incidences of nausea in the Z. officinale group showed significantly (p < 0.05) fewer compared to placebo. However the administration of antiemetic after operation was significantly (p < 0.05) greater in the placebo group compared to the other two groups. [89]

Chemotherapy effects

A Phase II clinical trial (randomized, double-blind, placebo-controlled trial) has been carried out to study the effect of encapsulated Z. officinale treatment to162 patients with cancer who were receiving chemotherapy and had experienced nausea and vomiting during at least one previous round of chemotherapy. This study was found that the Z. officinale was not effective in reducing the prevalence or severity of acute or delayed chemotherapy-induced nausea and vomiting. However it appeared well tolerated, with no difference in all adverse events and significantly less fatigue and miscellaneous adverse events in the ginger group. [90]

A small clinical study to 28 patients with cancer receiving chemotherapy for the first time found that a high protein meal given with Z. officinale may reduce the delayed nausea of chemotherapy and reduced use of antiemetic medications. [91]

A clinical studies has been carried out to patients undergoing photopheresis aged 80 years old and over administered with Z. officinale prior to ingestion of required drug 8-MOP. Z. officinale reduced the nausea side effect caused by 8-MOP therapy. [92]

Antimotion sickness

Z. officinale is best known for its ability to lessen the nausea and vomiting associated with motion sickness. In fact, studies have found that it may be more effective than drug alternatives for many conditions and situations that make the stomach feel unsettled. Several clinical studies have been published which support antiemetic activity of Z. officinale compared to drug or placebo therapy. 28 human volunteers were evaluated for antimotion sickness activity of Z. officinale root characterized its effects on gastric function. The volunteers made timed head movements in a rotating chair until they reached an endpoint of motion sickness short of vomiting (malaise III or M-III). The results showed that Z. officinale powder (whole root, 500 or 1,000 mg) or Z. offcinale fresh root (1000 mg) did not exhibited antimotion sickness but performed an average of 147.5 more head movements (p < 0.01) after administered orally with scopolamine (0.6 mg) than after placebo. The rate of gastric emptying was significantly (p < 0.05) slowed when tested immediately after M-III but was inhibited less when tested 15 min after M-III. The trials was concluded that Z. officinale did not possed antimotion sickness and significantly not alter gastric function during motion sickness. [93][94]

Another controlled, double-blind study was carried out to study the antimotion sickness mechanism by determine whether nystagmus response to optokinetic or vestibular stimuli might be altered by some agent contained in powdered Z. officinale root and being compared with placebo and with dimenhydrinate. The study showed that neither the vestibular nor the oculomotor system which were importance in the decisive occurrence of motion sickness were influenced by Z. officinale. The case of reduction of motion sickness might be due to the influence of Z. officinale on the gastric system. [95]

Antiseasickness

A clinical trial to study the effect of Z. officinale roots against seasickness has been carried out to 80 naval cadets and unaccustomed to sailing in heavy seas in a double-blind randomized placebo trial. The Z. officinale root significantly (p < 0.05) reduced the tendency to vomiting and cold sweating better than placebo with modified Protection Index (PI) of 72% was calculated regarding to vomiting.The ingestion of Z. officinale root also cause remarkably fewer symptoms of nausea and vertigo, but with statistically no significant difference. [96]

Nausea and vomiting of pregnancy

A clinical trial to compare Z. officinale and vitamin B6 for the treatment of nausea and vomiting in pregnancy was conducted. It is a randomised double-blind controlled trial which involved 70 pregnant women with nausea at or before 17 weeks gestation. The patients took 2 capsules containing powdered of Z. officinale root (1 g/day) or vitamin B6 (40 mg/day) after breakfast and dinner for 4 days. Z. officinale was found to be significantly more effective in alleviating the severity of nausea as compared to vitamin B6 group. Z. officinale also was equally effective with vitamin B6 group in reducing the number of vomiting episode during early pregnancy period. [97]

A single blind clinical trial study has been conducted to determine the effects of Z. officinale capsule in nausea and vomiting of pregnancy compared to placebo involving 67 pregnant women with the complaines. The experimental group received Z. officinale capsules (250 mg) for duration of 4 days, and placebo received the same prescription form. Administration of Z. officinale capsules significantly (p < 0.01) showed a higher rate of improvement (85%) compared to placebo (56%). It also significantly (p < 0.05) decreased the vomiting times compared to women who received placebo (50% and 9%, respectively). [98]

A double-blind randomized cross-over trial has been carried out among 30 women to study the efficacy of powdered Z. officinale root and placebo in the treatment of hyperemesis gravidarum, a condition in morning sickness where severe dehydration and electrolyte disturbances may occur through excessive vomiting. The results showed that Z. officinale (1 g, 250 mg each capsule) administered daily for duration of 4 days could removing the hyperemesis gravidarum symptoms better than placebo. [99] It has been reported that this effect may be due to part of Z. officinale could increase in gastric emptying and its thromboxane synthetase activity. [100]

A double blind randomized controlled trial of 170 pregnant women also found that Z. officinale was as effective as the over-the-counter (OTC) drug dimenhydrinate (Dramamine) in the treatment of nausea and vomiting during pregnancy with fewer side effects including drowsiness. [101]

A randomized, controlled equivalence trial involving 291 women less than 16 weeks pregnant found that women using Z. officinale in early pregnancy will reduce their nausea, dry retching, and vomiting symptoms, which is equivalent to pyridoxine hydrochloride (vitamin B6). [102][103]

A randomized controlled trial in a double-masked design involving pregnant women with nausea and vomiting who first attended an antenatal clinic at or before 17 weeks gestation was carried out to study the effect of Z. officinale to relieve their severity of nausea and vomiting. The result showed that the use of Z. officinale for treatment of nausea in pregnancy effectively decreased the number of events as well as lessening the severity of nausea. [104]

Effect on gastric emptying

A randomized double blind study of 24 healthy volunteers concluded that Z. officinale capsules (total 1200 mg) accelerated gastric emptying and stimulates antral contractions in healthy volunteers compared to placebo. [105]

A randomised, placebo-controlled, double-blind crossover clinical trial has been carried out to 16 healthy volunteers to investigate the effect of Z. officinale (1 g) on the gastric emptying rate. The oral paracetamol absorption model has been used for measurement and the ingestion of Z. officinale did not showed any effect on gastric emptying. Thus the result concluded that antiemetic activity of Z. officinale was not associated with gastric emptying effect. [106]

Z. officinale rhizome extract (2 x 100 mg) taken orally improved gastroduodenal motility in the fasting state and after a standard test meal in 12 healthy volunteers involved in the study to determine the effect of Z. officinale on fasting and postprandial gastroduodenal motility with stationary manometry. [107]

However, there has been a report of dried Z. officinale root having no value in decreasing postoperative nausea, but product quality and lack of standardization may have contributed to this negative finding. [108]

Antiarthitis

Z. officinale extract (Zintona EC)has been studied to 29 patients (6 men and 23 women) in the age range of 42 to 85 years with symptomatic gonarthritis (ACR criteria). A randomized double blind, crossover study was compared with placebo as a control for duration of 6 months. The results showed that there was highly statistically significant (p<0.001) difference between the visual analog scale (VAS) of pain and handicap of the two groups by the end of 24 weeks. However, at crossover both groups showed a statistically significant decrease in VAS of pain on movement and of handicap, but the differences between the groups did not reach statistical significance. It was concluded that the Z. officinale extract showed a significant superiority over the placebo group 3 months after crossover study. [109]

A study involving 56 patients with rheumatoid arthritis (28), osteoarthritis (18), and muscular discomfort (10) cosumed Z. officinale for a duration from 3 months to 2.5 years showed it was effective in more than 75% of the arthritis patients while all the patients with muscular discomfort experienced relief in pain. The Z. officinale consumption also did not show any adverse effect to the patients. [110]

A randomized, double-blind, placebo-controlled, multicenter, and parallel-group study lasting 6-weeks has been carried out to 247 patients to evaluate the safety and efficacy of 2 ginger species (Z. officinale and Alpinia galanga) in osteoarthritis (OA) of the knee. The Z. officinale extract group had greater response in the primary endpoint of reduced knee pain upon standing as well as all the secondary endpoints evaluated. Less rescue medication (acetaminophen) was used by the Z. officinale group. More gastrointestinal adverse effects, most of them mild were experienced by the Z. officinale group. It is important to note that the change in the quality of life was equal between the Z. officinale and placebo group. [111]

Another study found no significant advantage of using Z. officinale root over conventional anti-inflammatory agents such as ibuprofen when administered to patients with osteoarthritis of the hip or knee in a controlled, double blind, double dummy, cross-over study with a wash-out period of one week followed by three treatment periods in a randomized sequence, each of three weeks duration. [112]

Knee Pain

A small study used aromatic essential oil massage among elderly patients with moderate to severe knee pain. The aromatherapy oil contained 1% Z. officinale oil and 5% Citrus sinesis (sweet orange) oil while the placebo oil only contained olive oil. After 3 weeks of massage, it was found that both groups had a decrease in stiffness and pain intensity. However, at the one week mark the aromatherapy group had less pain and improved physical function that was not seen in the placebo group. [113]

Platelet aggregation

Dried Z. officinale (5 g) included in dietary supplementation of 100 g butter by 20 healthy volunteers for duration of 7 days showed that it significantly (p < 0.001) inhibited the platelet aggregation induced by ADP (adenosine diphosphate) and epinephrine caused by the butter consumpption in their meals. Meanwhile there were no significant alteration in platelet aggregation in the placebo group and both group did not showed any change of serum lipids. [114]

Antihyperlipidaemic activity

A double blind controlled clinical study has been carried out to patients with hyperlipidemia in 2 cardiac clinics Cardiac Disease Clinic, Babol, north of Iran to study the effect of fine powder of Z. officinale on their lipid level. It was found that a significant reduce in triglyceride, cholesterol, low density lipoprotein (LDL), very low density lipoprotein (VLDL) after the treatment with encapsulated ginger. [115]

Precautions

Z. officinale rhizome is not recommended for children less than 6 years of age [116]. Contact dermatitis on the fingers has been found in sensitive patients [117].

Based on pharmacology of Z. officinale products, caution should be used in patients with bleeding disorders. [118]

If you are planning to have any type of surgery or dental work, stop using this dietary supplement for at least 14 days prior to the procedure. [119]

Side effects

Z. officinale is generally considered safe for use. There is little risk of toxicity in normal doses, but some individuals may experience gastrointestinal upset. [120]

Z. officinale also may cause contact dermatitis in sensitive individuals. [121]

Z. officinale was reported to cause severe hyperkalemia in a patient with cirrhosis. Use ginger with caution in those with liver disorders or pre-disposed to liver diseases. [122]

Pregnancy/Breast Feeding

If pregnant or nursing, consult a physician before use. A laboratory animal study reported that when a proprietary extract of Z. officinale was administered to pregnant rats during the period of organogenesis, with neither maternal nor developmental toxicity at daily doses of up to 1000 mg/kg body weight seen. [123]

Age limitation

No documentation.

Adverse reaction

No documentation.

Interaction & Depletion

Interaction with drug

As constituents in Z. officinale may inhibit platelet aggregation, it may alter the effects of medications used for this purpose. Z. officinale may have interaction with anticoagulant medications, antiplatelet medications and Nifedipine (Procardia). It was reported in an in vitro study that it could increase the antiplatelet activity of nifedipine. [124][125][126]

Interaction with other Herbs

No documentation.

Contraindications

No documentation.

Case Report

No documentation.

Poisonous Management

No documentation.

Line Drawing

Figure 1. The line drawing of Zingiber officinale Roscoe. [7]

References

  1. The Plant List. Ver1.1. Zingiber officinale Roscoe.[homepage on the Internet] .c2013 [updated 2012 Mar 26; cited 2014 Oct 14] Available from: http://www.theplantlist.org/tpl1.1/record/kew-273361
  2. Philippine Medicinal Plants. Luya. Zingiber officinale Roscoe. [homepage on the Internet]. c2013 [updated 2014 April; cited 2014 Oct 9]. Available from: http://www.stuartxchange.com/Luya.html
  3. Herbal Medicine Research Centre, Institute for Medical Research. Compendium of Medicinal Plants Used in Malaysia. Volume 2. Kuala Lumpur: HMRC IMR, 2002; p. 454.
  4. Quattrocchi U. CRC world dictionary of medicinal and poisonous plants: Common names, scientific names, eponyms, synonyms and etymology. Volume V R-Z. Boca Raton, Florida: CRC Press, 2012. p. 837-838.
  5. Kapoor LD. CRC Handbook of Ayurvedic Medicinal Plants. Boca Raton, Florida: CRC Press; 1990. p. 341.
  6. Maurice M. Handbook of African Medicinal plants. 2nd ed. Boca Raton, Florida: CRC Press, 1993; p. 263.
  7. Sutarno H, Hadad EA, Brink M. Zingiber officinale Roscoe. In: de Guzman CC, Siemonsma JS, editors. Plant Resources of South-East Asia No. 13: Spices. Leiden, Netherlands: Backhuys Publisher, 1999; p. 238-244.
  8. Nair KPP. The agronomy and economy of turmeric and ginger: The invaluable medicinal spice crops. London: Elsevier, 2013; p.232.
  9. Magness JR, Markle GM, Compton CC. Food and Feed Crops of the United States. New Jersey Interregional Research Project IR-4, IR Bul. 1 (Bul. 828 New Jersey Agr. Expt. Sta.). 1971.
  10. Standard of Asean herbal medicine. Volume 1. Jakarta, Indonesia: ASEAN countries. 1993.
  11. Keys JD. Chinese herbs, their botany, chemistry and pharmacodynamics. Rutland, VT, CE Tuttle. 1976.
  12. Youngken HW. Textbook of pharmacognosy. 6th ed. Philadelphia: Blakiston Publication Company; 1950.
  13. British pharmacopoeia. London: General Medical Council, 1993; p. 305
  14. African pharmacopoeia. Volume 1. 1st ed. Lagos: Organization of African Unity, Scientific, Technical & Research Commission. 1985.
  15. Mansor P, Muhamad Ghawas M, Sentoor K. Halia (Zingiber officinale Ross). In: Musa Y, Muhammad Ghawas M, Mansor P, editors. Penanaman tumbuhan ubatan & beraroma. Serdang: MARDI, 2005; p. 29 -35.
  16. Leong AC, Mansor P. Halia: Pengeluaran dan pengunaan. In: Tan SL, et al, editors. Prosiding bengkel tanaman yang kurang dieksploitasi. 18 May 1993. Kuala Lumpur: Vinlin Press Sdn. Bhd., 1993; p. 75-86.
  17. Chung IM, Ali M, Ahmad A. Isolation and characterization of water-soluble new gingerlanosterol from Zingiber officinale rhizomes. Asian J Chem. 2005;17(3):1915-1920.
  18. Imm J, Zhang G, Chan L-K, Nitteranon V, Parkin KL. [6]-Dehydroshogaol, a minor component in ginger rhizome, exhibits quinone reductase inducing and anti-inflammatory activities that rival those of curcumin. Food Res Int. 2010;43:2208-2213.
  19. Ma J, Jin X, Yang L, Liu Z-L. Diarylheptanoids from the rhizomes of Zingiber officinale. Phytochemistry. 2004;65:1137-1143.
  20. Peng F, Tao Q, Wu X, et al. Cytotoxic, cytoprotective and antioxidant effects of isolated phenolic compounds from fresh ginger. Fitoterapia. 2012;83:568-585.
  21. Li F, Wang Y, Parkin KL, et al. Isolation of quinone reductase (QR) inducing agents from ginger rhizome and their in vitro anti-inflammatory activity. Food Res Int. 2011;44:1597-1603.
  22. Hong SS, Oh JS. Phenylpropanoid ester from Zingiber officinale and their inhibitory effects on the production of nitric oxide. Arch Pharm Res. 2012;35(2):315-320.
  23. He XG, Bernart MW, Lian LZ, Lin LZ. High-performance liquid chromatography–electrospray mass spectrometric analysis of pungent constituents of ginger. J Chromatogr A. 1998;796:327-334.
  24. Araya JJ, Zhang H, Prisinzano TE, Mitscher LA, Timmermann BN. Identification of unprecedented purine-containing compounds, the zingerines, from ginger rhizomes (Zingiber officinale Roscoe) using a phase-trafficking approach. Phytochemistry. 2011;72:935-941.
  25. Lee SW, Lim JH, Kim MS, et al. Phenolic compounds isolated from Zingiber officinale roots inhibit cell adhesion. Food Chem. 2011;128:778-782.
  26. Hori Y, Miura T, Hirai Y, et al. Pharmacognostic studies on ginger and related drugs -part 1: five sulfonated compounds from Zingiberis rhizome (Shokyo). Phytochemistry. 2003;62:613-617.
  27. Lin RJ, Chen CY, Chung LY, Yen CM. Larvicidal activities of ginger (Zingiber officinale) against Angiostrongylus cantonensis. Acta Tropica. 2010;115:69-76.
  28. Kikuzaki H, Nakatan N. Cyclic diarylheptanoids from rhizomes of Zingiber officinale. Phytochemistry. 1996;43(1):273-277.
  29. Jolad SD, Lantz RC, Chen GJ, Bates RB, Timmermann BN. Commercially processed dry ginger (Zingiber officinale): Composition and effects on LPS-stimulated PGE2 production. Phytochemistry. 2005;66:1614-1635.
  30. Jolad SD, Lantz RC, Solyom AM, Chen GJ, Bates RB, Timmermann BN. Fresh organically grown ginger (Zingiber officinale): Composition and effects on LPS-induced PGE2 production. Phytochemistry. 2004;65:1937-1954.
  31. Rahuman AA, Gopalakrishnan G, Venkatesan P, Geetha K, Bagavan A. Mosquito larvicidal activity of isolated compounds from the rhizome of Zingiber officinale. Phytother Res. 2008;22(8):1035-1039.
  32. Singh G. Studies on essential oils, Part 42: Chemical, antifungal, antioxidant and sprout suppressant studies on ginger essential oil and its oleoresin. Flav Frag J. 2004;20(1):1-6.
  33. Kizhakkayil J, Sasikumar B. Characterization of ginger (Zingiber officinale Rosc.) germplasm based on volatile and non-volatile components. Afr J Biotechnol. 2012;11(4):777-786.
  34. Bednarczyk AA, Galetto WG, Kramer A. cis- and trans-ß-sesquiphellandrol. Two new sesquiterpene alcohols from oil of ginger (Zingiber officinale Roscoe). J Agric Food Chem. 1975;23(3):499-501.
  35. Smith RM, Robinson JM. The essential oil of ginger from Fiji. Phytochemistry. 1981;20(2):203-206.
  36. Wohlmuth H. Essential oil composition of diploid and tetraploid clones of ginger (Zingiber officinale Roscoe) grown in Australia. J Agric Food Chem. 2006;54(4):1414-1419.
  37. Kikuzaki H, Kobayashi M, Nakatani N. Diarylheptanoids from rhizomes of Zingiber officinale. Phytochemistry. 1991;30(11):3647-3651.
  38. Kikuzaki H, Tsai SM, Nakatani N. Gingerdiol related compounds from the rhizomes of Zingiber officinale. Phytochemistry. 1992;31(5):1783-1786.
  39. Endo K, Kanno E, Oshima Y. Structures of antifungal diarylheptenones, gingerenones A, B, C and isogingerenone B, isolated from the rhizomes of Zingiber officinale. Phytochemistry. 1990;29(3):797-799.
  40. Li N, Wang L, Zu L, Wang K, Di L, Wang Z. Antioxidant and cytotoxic diarylheptanoids Isolated from Zingiber officinale rhizomes. Chin J Chem. 2012;30(6):1351-1355.
  41. Choi SY, Park GS, Lee SY, Kim JY, Kim YK. The conformation and CETP inhibitory activity of [10]-dehydrogingerdione isolated from Zingiber officinale. Arch Pharm Res. 2011;34(5):727-731.
  42. Choi KH, Laursen RA. Amino-acid sequence and glycan structures of cysteine proteases with proline specificity from ginger rhizome Zingiber officinale. Eur J Biochem. 2000;267:1516-1526.
  43. Zhao Y, Tao QF, Zhang RP, et al. Two new compounds from Zingiber officinale. Chin Chem Lett. 2007;18:1247-1249.
  44. Sekiwa Y, Kubota K, Kobayashi A. Isolation of novel glucosides related to gingerdiol from ginger and their antioxidative activities. J Agric Chem. 2000:48(2):373–377.
  45. Kizhakkayil J, Sasikumar B. Characterization of ginger (Zingiber officinale Rosc.) germplasm based on volatile and non-volatile components. Afr J Biotechnol. 2012;11(4):777-786.
  46. Wu TS, Wu YC, Wu PL, Chern CY, Leu YL, Chan YY. Structure and synthesis of [n]-dehydroshogaols from Zingiber officinale. Phytochemistry. 1998;48(5):889-891.
  47. Charles R, Garg SN, Kumar S. New gingerdione from the rhizomes of Zingiber officinale. Fitoterapia. 2000;71:716-718.
  48. Neuwinger HD. African traditional medicine: A dictionary of plant use and applications with supplement: search system for diseases. Birkenwaldstr, Germany: Medpharm Scientific Publisher; 2000.
  49. Van Wyk BE, Van Oudtshoorn B, Gericke N. Medicinal plants of South Africa. Pretoria, South Africa: Briza Publications, 2009; p. 320.
  50. Chinemana F, Drummond R, Mavi S, De Zoysa I. Indigenous plant remedies in Zimbabwe. J Ethnopharmacol. 1985;14(2):159-172.
  51. Gelfand M, Mavi S, Drummond RB, Ndemera B. The traditional medical practitioner in Zimbabwe. Gweru, Zimbabwe: Mambo Press; 1985.
  52. El-Kamali HH, El-Khalid SA. The most common herbal remedies in Dangola Provence, Norther Sudan. Fitoterapia. 1998;69:118-121.
  53. Nwosu MO. Herbs for mental disorders. Fitoterapia. 1999;70:58-63.
  54. Nuomi E, Amvan Zollo P, Lontsi D. Aphrodisiac plants used in Cameroon. Fitoterapia. 1998; 69:125-134.
  55. Chen JC, Huang LJ, Wu SL, Kuo SC, Ho TY, Hsiang CY. Ginger and its bioactive component inhibit enterotoxigenic Escherichia coli heat-labile enterotoxin-induced diarrhea in mice. J Agric Food Chem. 2007;55:8390-8397.
  56. Bhandari U, Sharma JN, Zafar R. The protective action of ethanolic ginger (Zingiber officinale) extract in cholesterol fed rabbits. J Ethnopharmacol. 1998;61:167-171.
  57. Bhandari U, Kanojia B, Pillai KK. Effect of ethanolic extract of Zingiber officinale on dyslipidaemia in diabetic rats. J Ethnopharmacol. 2005;97:227-230.
  58. Thomson M, Al-Qattan KK, Al-Sawan SM, Alnaqeeb MA, Khan I, Ali M. The use of ginger (Zingiber officinale Rosc.) as a potential anti-inflammatory and antithrombotic agent. Prostaglandins Leukot Essent Fatty Acids. 2002;67(6):475-478.
  59. Fuhrman B, Rosenblat M, Hayek T, et al. Ginger extract consumption reduces plasma cholesterol, inhibits LDL oxidation and attenuates development of atherosclerosis in stherosclerotic, spolipoprotein E-deficient mice. J Nutr. 2000;130(5):1124-1131.
  60. Eldershaw TP, Colguhoun EQ, Dora KA, et al. Pungent principles of ginger (Zingiber officinale) are thermogenic in the perfused rat hindlimb. Int J Obes Relat Metab Disord. 1992;16(10):755-763.
  61. Khaki AA, Khaki A. Antioxidant effect of ginger to prevents lead-induced liver tissue apoptosis in rat. J Med Plant Res. 2010;4(14):1492-1495.
  62. Singh G, Kapoor IP, Singh P, de Heluani CS, de Lampasona MP, Catalan CA. Chemistry, antioxidant and antimicrobial investigations on essential oil and oleoresins of Zingiber officinale. Food Chem Toxicol. 2008;46(10):3295-3302.
  63. Ahmed RS, Seth V, Banerjee BD. Influence of dietary ginger (Zingiber officinales Rosc) on antioxidant defense system in rat: Comparison with ascorbic acid. Indian J Exp Biol. 2000;38(6):604-606.
  64. Ahmed RS, Seth V, Pasha ST, et al. Influence of dietary gnger (Zingiber officinales Rosc) on Oxidative Stress Induced by Malathion in Rats. Food Chem Toxicol. 2000;38(5):443-450.
  65. Zhou HL. The modulatory effects of the volatile oil of ginger on the cellular immune response in vitro and in vivo in mice. J Ethnopharmacol. 2006;105(1-2):301-305.
  66. Ishiguro K, Ando T, Maeda O, et al. Ginger ingredients reduce viability of gastric cancer cells via distinct mechanisms. Biochem Biophys Res Commun. 2007;362(1):218-223. Epub 2007 Aug 10.
  67. Lee HS, Seo EY, Kang NE, Kim WK. [6]-Gingerol inhibits metastasis of MDA-MB-231 human breast cancer cells. J Nutr Biochem. 2008;19(5):313-319. Epub 2007 Aug 1.
  68. Sakr SA, Badawy GM. Effect of ginger (Zingiber officinale R.) on metiram-inhibited spermatogenesis and induced apoptosis in albino mice. J Applied Pharm Sci. 2011;1(4):131-136.
  69. Morakinyo AO, Adeniyi OS, Arikawe AP. Effects of Zingiber officinale on reproductive functions in the male rat. Afr J Biomed Res. 2008;11:329-334.
  70. Jafri SA, Abass S, Qasim M. Hypoglycemic effect of ginger (Zingiber officinale) in alloxan induced diabetic rats (Rattus norvegicus). Pak Vet J. 2011;31(2):160-162.
  71. Akhani S, Vishwakarma S, Goyal R. Antidiabetic activity of Zingiber officinale in streptozotocin-induced type I diabetic rats. J Pharm Pharmacol. 2004;56:101-105.
  72. Kato A, Higuchi Y, Goto H, et al. Inhibitory effects of Zingiber officinale Roscoe derived components on aldose reductase activity in vitro and in vivo. J Agric Food Chem. 2006;54(18):6640-6644.
  73. Martins AP, Salgueiro L, Gonçalves MJ, et al. Essential oil composition and antimicrobial activity of three Zingiberaceae from S.Tomé e Príncipe. Planta Med. 2001;67(6):580-584.
  74. Thongson C, Davidson PM, Mahakarnchanakul W, Weiss J. Antimicrobial activity of ultrasound-assisted solvent-extracted spices. Lett Appl Microbiol. 2004;39(5):401-406.
  75. Chen HC, Chang MD, Chang TJ. Antibacterial properties of some spice plants before and after heat treatment. [Article in Chinese]. Zhonghua Min Guo Wei Sheng Wu Ji Mian Yi Xue Za Zhi. 1985;18(3):190-195.
  76. Atai Z, Atapour M, Mohseni M. Inhibitory effect of ginger extract on Candida albicans. Am J Appl Sci. 2009;6(6):1067-1069.
  77. Pozzatti P, Scheid LA, Spader TB, Atayde ML, Santurio JM, Alves SH. In vitro activity of essential oils extracted from plants used as spices against fluconazole-resistant and fluconazole-susceptible Candida spp. Can J Microbiol. 2008;54(11):950-956.
  78. Nguefack J, Leth V, Amvam Zollo PH, Mathur SB. Evaluation of five essential oils from aromatic plants of Cameroon for controlling food spoilage and mycotoxin producing fungi. Int J Food Microbiol. 2004;94(3):329-334.
  79. Denyer CV, Jackson P, Loakes DM, et al. Isolation of Antirhinoviral Sesquiterpenes from Ginger (Zingiber officinale). J Nat Prod. 1994;57(5):658-662.
  80. Weidner MS, Sigwart K. The safety of a ginger extract in the rat. J Ethnopharmacol. 2000;73:513–520.
  81. 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. Report No.: HMRC 11-045/01/ZO/RH/K. Kuala Lumpur: Institute for Medical Research, Ministry of Health; 2012.
  82. Iman A. Elkhishin, Ibrahim A. Awwad. A study of the cardiovascular toxic effects of Zingiber officinale (ginger) in adult male albino rats and its possible mechanisms of action. Mansoura J Forensic Med Clin Toxicol. 2009;17:109-125.
  83. Shalaby MA, Hamowieh AR. Safety and efficacy of Zingiber officinale roots on fertility of male diabetic rats. Food Chem Toxicol. 2010;48:2920–2924.
  84. Rong X, Peng G, Suzuki T, Yang Q, Yamahara J, Li Y. A 35-day gavage safety assessment of ginger in rats. Regul Toxicol Pharmacol. 2009;54:118–123.
  85. Wilkinson JM. Effect of ginger tea on the fetal development of Sprague-Dawley rats. Reprod Toxicol. 2000;14:507–512.
  86. Phillips S, Ruggier R, Hutchinson SE. Zingiber officinale (ginger)-an antiemetic for day case surgery. Anaesthesia. 1993;48(8):715-717.
  87. Pongrojpaw D, Chiamchanya C. The efficacy of ginger in prevention of post-operative nausea and vomiting after outpatient gynecological laparoscopy. J Med Assoc Thai. 2003;86(3):244-250.
  88. Nanthakomon T, Pongrojpaw D. The efficacy of ginger in prevention of postoperative nausea and vomiting after major gynecologic surgery. J Med Assoc Thai. 2006;89 Suppl 4:S130-136.
  89. Bone ME, Wilkinson DJ, Young JR, McNeil J, Charlton S. Ginger root–A new antiemetic. The effect of ginger root on postoperative nausea and vomiting after major gynaecological surgery. Anaesthesia. 1990;45(8):669-671.
  90. Zick SM, Ruffin MT, Lee J, et al. Phase II trial of encapsulated ginger as a treatment for chemotherapy-induced nausea and vomiting. Support Care Cancer. 2009;17(5):563-572. Epub 2008 Nov 13.
  91. Levine ME, Gillis MG, Koch SY, Voss AC, Stern RM, Koch KL. Protein and ginger for the treatment of chemotherapy-induced delayed nausea. J Altern Complement Med. 2008;14(5):545-551.
  92. Meyer K, Schwartz J, Crater D, Keyes B. Zingiber officinale (ginger) used to prevent 8-Mop associated nausea. Dermatol Nurs. 1995;7(4):242-244.
  93. Mowrey DB, Clayson DE. Motion sickness, ginger, and psychophysics. Lancet. 1982;1(8273):655-667.
  94. Stewart JJ, Wood MJ, Wood CD, Mims ME. Effects of ginger on motion sickness susceptibility and gastric function. Pharmacology. 1991;42(2):111-120.
  95. Holtmann S, Clarke AH, Scherer H, Höhn M. The Anti-motion sickness mechanism of ginger. A comparative study with placebo and dimenhydrinate. Acta Otolaryngol. 1989;108(3-4):168-174.
  96. Grontved A, Brask T, Kambskard J, Hentzer E. Ginger root against sea sickness: A controlled trial on the open sea. Acta Otolaryngol. 1988;105:45-49.
  97. Jenabi E, Mohammad-Alizadeh CS. Comparing ginger and vitamin B6 for the treatment of nausea and vomiting in pregnancy: A randomized controlled trial. Midwifery. 2009;25:649–653.
  98. Ozgoli G, Goli M, Simbar M. Effects of ginger capsules on pregnancy, nausea, and vomiting. J Altern Complement Med. 2009;15(3):243-246.
  99. Fischer-Rasmussen W, Kjaer SK, Dahl C, Asping U. Ginger Treatment of Hyperemesis Gravidarum. Eur J Obstet Gynecol Reprod Biol. 1991;38(1):19-24.
  100. Backon J. Ginger in Preventing Nausea and Vomiting of Pregnancy; A Caveat Due to Its Thromboxane Synthetase Activity and Effect on Testosterone Binding. Eur J Obstet Gynecol Reprod Biol. 1991;42(2):163-164.
  101. Pongrojpaw D, Somprasit C, Chanthasenanont A. A randomized comparison of ginger and dimenhydrinate in the treatment of nausea and vomiting in pregnancy. J Med Assoc Thai. 2007;90(9):1703-1709.
  102. Smith C, Crowther C, Willson K, Hotham N, McMillian V. A randomized controlled trial of ginger to treat nausea and vomiting in pregnancy. Obstet Gynecol. 2004;103(4):639-645.
  103. Sripramote M, Lekhyananda N. A randomized comparison of ginger and vitamin B6 in the treatment of nausea and vomiting of pregnancy. J Med Assoc Thai. 2003;86(9):846-853.
  104. Vutyavanich T, Kraisarin T, Ruangsri R. Ginger for nausea and vomiting in pregnancy: randomized, double-masked, placebo-controlled trial. Obstet Gynecol. 2001;97(4):577-582.
  105. Wu KL, Rayner CK, Chuah SK, et al. Effects of ginger on gastric emptying and motility in healthy humans. Eur J Gastroenterol Hepatol. 2008;20(5):436-440.
  106. Phillips S, Hutchinson S, Ruggier R. Zingiber officinale does not affect gastric emptying rate. A randomised, placebo-controlled, crossover trial.Anaesthesia. 1993;48(5):393-395.
  107. Micklefield GH, Redeker Y, Meister V, Jung O, Greving I, May B. Effects of ginger on gastroduodenal motility. Int J Clin Pharmacol Ther. 1999;37(7):341-346.
  108. Arfeen Z, Owen H, Plummer JL, Ilsley AH, Sorby-Adams RA, Doecke CJ. A double-blind randomized controlled trial of ginger for the prevention of postoperative nausea and vomiting. Anaesth Intensive Care. 1995;23(4):449-452.
  109. Wigler I, Grotto I, Caspi D, Yaron M. The effects of Zintona EC (a ginger extract) on symptomatic gonarthritis. Osteoarthritis Cartilage. 2003;11(11):783-789.
  110. Srivastava KC, Mustafa T. Ginger (Zingiber officinale) in rheumatism and musculoskeletal Disorders. Med Hypotheses. 1992;39(4):342-348.
  111. Altman RD, Marcussen KC. Effects of a ginger extract on knee pain in patients with osteoarthritis. Arthritis Rheum. 2001;44(11):2531-2538.
  112. Bliddal H, Rosetzsky A, Schlichting P, et al. A randomized, placebo-controlled, cross-over study of ginger extracts and ibuprofen in osteoarthritis. Osteoarthritis Cartilage. 2000;8(1):9-12.
  113. Yip YB, Tam ACY. An experimental study on the effectiveness of massage with aromatic ginger and orange essential oil for moderate-to-severe knee pain among the elderly in Hong Kong. Complement Ther Med. 2008;16(3):131-138.
  114. Verma SK, Singh J, Khamesra R, Bordia A. Effect of ginger on platelet aggregation in man. Indian J Med Res. 1993;98:240-242.
  115. Alizadeh-Navaei R, Roozbeh F, Saravi M, Pouramir M, Jalali F, Moghadamnia AA. Investigation of the effect of ginger on the lipid levels. A double blind controlled clinical trial. Saudi Med J. 2008;29(9):1280-1284.
  116. World Health Organization. WHO monographs on selected medicinal plants. Volume 1. Geneva: WHO, 1999; p.277-287.
  117. Seetharam KA, Pasricha JS. Condiments and contact dermatitis of the finger tips. Indian J Dermatol Venereol Leprol. 1987;53:325–328.
  118. Heck AM, DeWitt BA, Lukes AL. Potential interactions between alternative therapies and warfarin. Am J Health Syst Pharm. 2000;57(13): 1221-1227.
  119. Pribitkin ED, Boger G. Herbal therapy: What every facial plastic surgeon must know. Arch Facial Plast Surg. 2001;3(2):127-132.
  120. Desai HG, Kalro RH, Choksi AP. Effect of ginger and garlic on DNA content of gastric aspirate. Ind J Med Res. 1990;92:139-141.
  121. Minamoto K, Harada K, Wei QJ, Wei CN, Omori S, Ueda A. Occupational allergic contact dermatitis from mioga (Zingiber miogarosc.) in greenhouse cultivators. Int J Immunopathol Pharmacol. 2007;20(2 Suppl 2):31-34.
  122. Rivero Fernández M, Moreira Vicente V, Rodríguez AL, Ruiz del Arbol Olmos L. Severe hyperkalaemia caused by ginger in cirrhotic patient. [Article in Spanish]. Med Clin (Barc). 2007;129(10):398-399.
  123. Weidner MS, Sigwart K. Investigation of the teratogenic potential of a Zingiber officinale extract in the rat. Reprod Toxicol. 2001;15(1):75-80.
  124. Guh JH, Ko FN, Jong TT, Teng CM. Antiplatelet effect of gingerol isolated from Zingiber officinale. J Pharm Pharmacol. 1995;47(4):329-332.
  125. Suekawa M, Ishige A, Yuasa K, Sudo K, Aburada M, Hosoya E. Pharmacological studies on ginger, I. Pharmacological actions of pungent constitutents, (6)-gingerol and (6)-shogaol. J Pharmacobiodyn. 1984;7(11):836-848.
  126. Young HY, Liao JC, Chang YS, Luo YL, Lu MC, Peng WH. Synergistic effect of ginger and nifedipine on human platelet aggregation: A study in hypertensive patients and normal volunteers. Am J Chin Med. 2006;34(4):545-551.
  127. Blumenthal M, Busse WR, Goldberg A, et al. The complete German commission e monographs, therapeutic guide to herbal medicines (Zingiberis rhizoma). Austin, Texas: American Botanical Council, 1998; p.135-136.
  128. Al-Omaria IL, Afifib FU, Salhaba AS. Therapeutic effect and possible herb drug interactions of ginger (Zingiber officinale Roscoe, Zingiberaceae) crude extract with glibenclamide and insulin. world. 2012;9(11).
  129. Rafieian-Kopaei M, Nasri H. The ameliorative effect of Zingiber officinale in diabetic nephropathy. Iranian Red Crescent Medical Journal. 2014;16(5)

in this scope
Malaysian Herbal Monograph​
Medicinal Herbs & Plants Monographs​
Traditional Chinese Medicine Herbs (Professional Data)
Herbal Medicines Compendium (HMC) - U.S​