ArticlePDF Available

Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature

Authors:
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
1
Original Article
Pharmacogn J. 2019; 11(6)Suppl:
A Multifaceted Journal in the eld of Natural Products and Pharmacognosy
www.phcogj.com
Cite this article: Mans DRA, Djotaroeno M, Friperson P, Pawirodihardjo J. Phytochemical
and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A
Review of the Literature. Pharmacog J. 2019;11(6)Suppl:
Phcogj.com
Phytochemical and Pharmacological Support for the Traditional
Uses of Zingiberacea Species in Suriname - A Review of the
Literature
Dennis RA Mans*, Meryll Djotaroeno, Priscilla Friperson, Jennifer Pawirodihardjo
INTRODUCTION
e plant family Zingiberaceae or ginger family
includes about 50 genera and roughly 1,600 known
species of owering perennial plants that are
distributed throughout tropical Africa, Asia, and the
Americas.1 e highest diversity is encountered in
south-eastern Asia, and the greatest concentration
in the north-eastern region of India with 19 genera
and 88 species.1 e majority of the Zingiberacea
members grows in humid and shady places.1 Most
are small to large herbaceous plants with distichous
leaves having basal sheaths that overlap to form a
pseudostem.1 ey are characterized by aromatic
and creeping horizontal or tuberous rhizomes,
hermaphroditic owers consisting of a single
functional stamen (the pollen-producing part of
the ower) that runs through the pistil (the ovule-
producing part of the ower), and petals that are
sterile stamina called staminodes.1
Many members of the Zingiberaceae are
economically important as spices, ornamental
plants, cosmetics, traditional medicines, and/or
ingredients of religious rituals. e seeds from
the genera Amomum Roxb. and Elettaria Maton
give the spice cardamom, the world’s third-most
expensive spice in price per weight aer vanilla and
saron.2 e rhizomes from the ai ginger Alpinia
galanga (L.) Willd., the turmeric Curcuma longa L.,
and the common ginger Zingiber ocinale Roscoe
are well-known spices, condiments, and avoring
compounds.3 And the leaves and owers from C.
longa, the grains of paradise Aframomum melegueta
K. Schum., the white ginger Hedychium coronarium
J. Koenig, and the east-Indian galangal Kaempferia
galanga L. are consumed as vegetables and used as
key ingredients of spicy savory dishes and/or herbal
teas.3
Various species in the genera Alpinia Roxb., 1810,
Curcuma L., Globba L., Hedychium J. Koenig,
Kaempferia L., and Renealmia L.f. have visually
attractive owers and inorescences and are
cultivated as ornamentals.3 e rhizomes from many
species in the genera Alpinia Roxb., 1810, Curcuma
L., Hedychium J. Koenig, and Zingiber Mill., 1754
contain essential oils for producing soaps, cosmetics,
and perfumes.3 In addition, a number of ginger
species are medicinally used in various traditional
systems throughout the world. Examples are A.
melegueta, C. longa, and Z. ocinale, preparations
of which are used for treating many diseases
ranging from infectious diseases and inammatory
conditions to hypertension and diabetes mellitus.4
Parts from A. melegueta, C. longa, and R. alpinia are
also used in religious ceremonies.3
is paper rst presents some general information
about the Zingiberacea family, subsequently provides
some background on Suriname, then extensively
addresses the traditional uses of one representative
species of the seven Zingiberacea genera in the
ABSTRACT
The Zingiberacea or ginger family is a family of flowering plants comprising roughly 1,600
species of aromatic perennial herbs with creeping horizontal or tuberous rhizomes divided
into about 50 genera. The Zingiberaceae are distributed throughout tropical Africa, Asia, and
the Americas. Many members are economically important as spices, ornamentals, cosmetics,
traditional medicines, and/or ingredients of religious rituals. One of the most prominent
characteristics of this plant family is the presence of essential oils in particularly the rhizomes
but in some cases also the leaves and other parts of the plant. The essential oils are in general
made up of a variety of, among others, terpenoid and phenolic compounds with important
biological activities. The Republic of Suriname (South America) is well-known for its ethnic and
cultural diversity as well as its extensive ethnopharmacological knowledge and unique plant
biodiversity. This paper first presents some general information on the Zingiberacea family,
subsequently provides some background about Suriname and the Zingiberacea species in the
country, then extensively addresses the traditional uses of one representative of the seven
genera in the country and provides the phytochemical and pharmacological support for these
uses, and concludes with a critical appraisal of the medicinal values of these plants.
Key words: Zingiberaceae, Suriname, Traditional uses, Rationale, Phytochemical composition,
Pharmacological activity.
Dennis RA Mans*, Meryll
Djotaroeno, Priscilla Friperson,
Jennifer Pawirodihardjo
Department of Pharmacology, Faculty of
Medical Sciences, Anton de Kom University of
Suriname, Paramaribo, SURINAME.
Correspondence
Dennis RA Mans
Department of Pharmacology, Faculty
of Medical Sciences, Anton de Kom
University of Suriname, Kernkampweg 6,
Paramaribo, SURINAME.
Phone no: 597 441071;
E-mail: dennismans16@gmail.com
History
Submission Date: 01-10-2019;
Review completed: 13-10-2019;
Accepted Date: 14-10-2019.
DOI : 10.5530/pj.2019.11.
Article Available online
http://www.phcogj.com/v11/i6s
Copyright
© 2019 Phcogj.Com. This is an open-
access article distributed under the terms
of the Creative Commons Attribution 4.0
International license.
2
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
country as well as the phytochemical and pharmacological support for
these uses, and concludes with a critical appraisal of the medicinal value
of these plants.
BACKGROUND
General aspects of Suriname
e Republic of Suriname is located on the north-east coast of South
America between French Guiana and Guyana and borders the Atlantic
Ocean to the north and Brazil to the south. It is the smallest and least
populated country in South America, occupying a land area of roughly
165,000 km2 and harboring an estimated 590,000 inhabitants.5 Roughly
80% of the population lives in the relatively narrow northern coastal
zone while the remaining 20% populates the savannas and hinterlands
in the interior of the country.5 Suriname’s most important economic
means of support are crude oil drilling, gold mining, agriculture,
sheries, forestry, and ecotourism.6 ese activities have substantially
contributed to the gross domestic income in 2017 of USD 2,996 billion
and the average per capita income in that year of USD 5,150. 6 is
positions Suriname on the World Bank’s list of upper-middle income
economies.7
Despite its relatively small population, Suriname is one of the ethnically,
religiously, culturally, and linguistically most diverse countries in
the world.5 In addition to the original inhabitants, the Indigenous
Amerindians, the country is home to the descendants from enslaved
Africans from various African countries called Maroons, those of
mixed black and white origin called Creoles; the descendants from
indentured laborers from China, India (called Hindustanis), and the
Indonesian island of Java (called Javanese), as well as those from several
European and Middle Eastern countries.5 More recently, individuals
from a number of Latin American and Caribbean countries including
Brazil, Guyana, French Guiana, the Netherlands Antilles, Haiti, Cuba,
and Venezuela have settled in Suriname.5
Traditional medicine in Suriname
e various ethnic groups in Suriname have largely preserved their
culture and identity, still practicing their original religion and speaking
their original language in addition to Dutch (the ocial language) and
Surinamese or Sranan Tongo.5,8 e same holds true for their specic
perceptions of health and disease, ethnopharmacological traditions,
and traditional medical concepts.8 is was probably partially due to
the divide-and-conquer policy of the colonial government which kept
the several groups isolated, and partially to the desire of each ethnic
group to adhere to its particular customs as a means of strengthening
the own identity in the new and unfamiliar environment of Suriname.8
Important factors which helped preserve the various traditional
medical systems were the extensive botanical knowledge of the
newcomers and their previous acquaintance with useful plants. Indeed,
the newcomers readily recognized many edible and medicinal plants
in Suriname because these plants, along with numerous commodities,
people, animals, and diseases had been transferred from the Old World
(Europe, Asia, and Africa) to the New World (the Americas) and vice
versa during the Columbian Exchange in the 15th and 16th centuries.9
A few of many examples of such plants were maize (Zea mays L.
(Poaceae)), cassava (Manihot esculenta Crantz (Euphorbiaceae)),
okra (Abelmoschus esculentus (L.) Moench (Malvaceae)), and sesame
(Sesamum indicum L. (Pedaliaceae)).9 Z. mays and M. esculenta were
native to South America and had been introduced in Africa in the
16th century, while A. esculentus and S. indicum had been brought to
Suriname from Africa.9
Furthermore, the enslaved African and Asian indentured laborers
cultivated several food crops and medicinal plants from leovers
of the meals they were given during the trans-Atlantic journey.8 A
well-established example is the African rice Oryza glaberrima Steud.
(Poaceae) that has probably been introduced in Suriname by enslaved
African women who prepared meals on the slave ships and intentionally
collected rice seeds.10 e new arrivals also grew useful plants from plant
parts they had brought along from their home country to Suriname
to prepare their specic foods, traditional medicines, cosmetics, and
ritual artefacts.8 A few examples are the tamarind Tamarindus indica
L. (Fabaceae), the milkvetch or huáng qí Astragalus propinquus
Schischkin (Fabaceae), the neem plant Azadirachta indica A. Juss.,
1830 (Meliaceae), and the cat’s whiskers or kumis kutying Orthosiphon
stamineus Bold. (Lamiaceae).11 ese plants are important in African
traditional medicine, Chinese traditional medicine, Indian Ayurveda,
and Javanese Jamu, respectively.11
Zingiberaceae species in Suriname
In addition to the above-mentioned plant species and many others,
several members of the Zingiberaceae family have been introduced
into Suriname.12-15 ey are commonly used for preparing condiments
and so drinks, as spices in food, for medicinal purposes, and in ritual
practices.12-15 A. melegueta and H. coronarium, for instance, originate
from Africa where they had considerable economic and medicinal
signicance, and have probably been brought to Suriname and
popularized in the country by enslaved Africans and their descendants.
16 C. longa and H. coronarium are important plants in Indian Ayurveda
and are associated with Hindustani indentured workers and their
children17, and A. alpinia as well as K. galanga presumably originate
from Indonesia, where they have been for centuries important plants
in Jawa traditional medical and cultural practices.14 Not surprisingly,
particularly the Surinamese Javanese abundantly use Zingiberacea
species in health supplements known as jamus.14
Only 7 of the 50 genera of the Zingiberaceae plant family are present
in Suriname. ese include the genera Aframomum, Alpinia, Curcuma,
Hedychium, Kaempferia, Renealmia, and Zingiber. Table 1 shows
a representative species of each genus that is commonly used in the
country. Hereunder, the various applications of these plants including
their traditional medical uses are in detail addressed, and the scientic
support for these uses is provided on the basis of available phytochemical
and pharmacological information.
Aframomum melegueta K. Schum.
e grains of paradise or Guinea pepper A. melegueta is native to the
western African coast but is now cultivated in most African countries as
well as parts of South America and the Caribbean including Suriname.
e plant bears trumpet-shaped, purple owers which develop into
pods of 5 to 7 centimeters long (Figure 1) that contain many small,
reddish-brown seeds. Between the 16th and the 19th century, the sharp-
tasting seeds were in high demand in Europe as an alternative for the
relatively expensive black pepper Piper nigrum L. (Piperaceae) from
Asia. e thriving trade of A. melegueta seeds in that period is reected
by the name ‘Pepper Coast’ or ‘Grain Coast’ given to the coastal area
of western Africa that was then one of the centers of A. melegueta
cultivation and trade, and currently comprises the Republic of Liberia.
A. melegueta seeds are commonly used in western and northern
African cuisines as a spicy seasoning for sausages and meats and as a
avoring for hot and cold beverages, ice cream, candy, and bread. In
various other parts of the world, the seeds are used in gourmet cuisine
as a replacement for pepper; to avor certain cra beers, gins, and
the Scandinavian alcoholic beverage akvavit; and as a condiment in
exquisite dishes such as exclusive okra stews and apple pie recipes. A.
melegueta seeds have also been used to provide a ctitious strength to
alcoholic beverages, but this practice has been declared illegal and has
been banned.
3Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Plant species
(vernacular names in
English; Surinamese/
Javanese/Hindustani/
Dutch)
Traditional medicinal uses Phytochemical composition Pharmacologcial activities
Aframomum melegueta
(Roscoe) K. Schum.
(grains of paradise;
nengrekondre pepre)
Respiratory tract infections, gastrointestinal
problems, snakebites and scorpion stings,
cancer, infertility, hypertension, diabetes
mellitus8,14,15
Arylalkanoids such as paradols, shogaols,
and gingerols, labdane diterpenoids such
as zerumin A, sesquiterpenes such as
humulene and caryophyllene, avonoids
such as quercetin and kaempferol16,17
Antimicrobial,19-22 antiparasitic,24,25
antiinammatory,23 anticancer and
chemopreventive activity,28-31 blood
pressure-lowering,36 hypoglycemic
activity;37 (male) fertility booster;32,33
abortifacient35
Alpinia galanga (L.) Willd.
(ai ginger; laos)
Microbial infections, HIV infection,
rheumatic disorders, gastrointestinal
ailments, headache and body aches7,39,40,41
Phenylpropanoids such as 1’S’-1’-
acetoxychavicol acetate, terpenes/
terpenoids such as β-pinene, avonoids
such as galangin and alpinin43,44
Antimicrobial,46-50 anti-HIV,51
antiparasitic,52,53 antiinammatory,54-57
antiulcer,58,59 hepatoprotective activity60
Curcuma longa L.
(turmeric; haldi/kunyit)
Inammations, precancerous conditions
and cancer, microbial infections, brain
disorders such as depression and
Alzheimer's disease7, 8,13,63,64
Diarylheptanoids including curcuminoids
such as curcumin, diarylpentanoids,
sesquiterpenes, monoterpenes, diterpenes,
and triterpenoids65-67
Antinammatory,68-83 chemopreventive and
anticancer,85-92 antimicrobial,62,93-95
Antiparasitic,96 anti-Alzheimer activity97-99
Hedychium coronarium J.
König
(white buttery ginger;
gember lelie)
Microbial infections, parasitic infections,
inammatory complaints, cancer104,105
Labdane diterpenes such as, coronarins,
farnesane sesquiterpenes such as nerolidol
and hedychiols, monoterpenes such as
1,8-cineole and α- and β-pinene107-109
Antimicrobial,110-115 antiparasitic,112,115,116
antiinammatory and analgesic,117-120
anticancer and chemopreventive
activity111,121-124
Kaempferia galanga (L.)
Willd.
(East-Indian galangal;
kentyur)
Microbial infections, inammatory
conditions, parasitic infections, headaches,
mouth ulcers, toothache, dermatological
problems, anxiety and depression7,126-128
Phenylpropanoids such as ethyl p-methoxy
cinnamate; avonols such as kaempferol;
terpenoids such as 1,8-cineole129,130
Antimicrobial,131-143 antiparasitic,136-143
antiinammatory, antinociceptive, and
sedative activity144-150
Renealmia alpinia (Rottb.)
Maas
(ink plant; bigi masusa)
Snake bites and scorpion stings,
bacterial infections, gastrointestinal
problems, fungal infections, convulsions
and seizures,8,151-154,172 anxiety,170
neurodegenerative disturbances171
Monoterpenes such as β-pinene; labdane
diterpenoids; diarylheptanoids; phenolic
compounds such as coumarins and
pinostrobin; desmethoxyyangonin155-158
Antivenom,152,159,160,162 analgesic,
antinociceptive, and anti-inammatory,161
antimicrobial,163-165 antiparasitic,116,165 MAO-
inhibitory activity169
Zingiber ocinale Roscoe
(common ginger; dyindya)
Nausea and vomiting, microbial infections,
parasitic infections, inammatory
conditions176-178
Sequiterpene hydrocarbons such as
zingiberol;179 phenolic compounds such
as gingerols, shogaols, paradols, and
zingerone179,180
Antiemetic,182-185,188,189 antimicrobial,191-193
antiparasitic,194-197 antiinammatory and
analgesic activity198-202
Table 1: Representative Zingiberacea species in Suriname, their traditional medical uses, and the phytochemical and pharmacological support for
these uses.
Figure 1: Seed pods of Aframomum melegueta (Roscoe) K. Schum.
(from: https://images.app.goo.gl/7i3LE4owB5vwjDJx7).
A. melegueta has a long traditional medical use in Africa and
Afro-American communities including those of the Surinamese
Maroons where it is known as ‘nengrekondre pepre’ (‘pepper from
the homeland of the Africans’). Seed preparations are used, among
others, against infections and inammations of the respiratory tract
and the gastrointestinal system; to repel pests of stored grains; to ght
cancer; for treating infertility; and against hypertension and diabetes
mellitus.15,18,19 A hot-water infusion of the seeds would also help against
stuttering when drunk from the larynx of a howler monkey or a large
snail shell12, and an alcoholic extraction would serve as an aphrodisiac.15
A. melegueta seeds are, furthermore, essential components of Maroon
rituals and herbal baths to exorcise evil spirits and neutralize witchcra,
but also in practices to attract good fortune.15
Important phytochemicals in A. melegueta seeds, seed extracts, and
the seed essential oil are arylalkanoids such as paradols, shogaols, and
gingerols, the aromatic ketones responsible for the strong aromatic
avor and pungent, peppery taste associated with the plant.20 Other
phytochemicals in the seeds are labdane diterpenoids such as zerumin A
and (E)-labda 8(17),12-diene-15,16-dial; sesquiterpene hydrocarbons
such as humulene and caryophyllene; and avonoids such as quercetin
and kaempferol and their derivatives.20,21 Of note, the relative abundance
and diversity of avonoids, diterpenoids, and sesquiterpenoids may
represent a chemotaxonomic marker of the genus Aframomum,
distinguishing it from other genera in the Zingiberaceae.22
4
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
e traditional use of A. melegueta against respiratory tract infections
is supported by the meaningful antibacterial and antifungal activity
(including activity against methicillin-resistant Staphylococus aureus
(MRSA)) of preparations and constituents of the seeds and rhizomes.23,24
ese eects were also achieved with various avonoids, terpenoids,
as well as 6-paradol and related compounds isolated from these parts
of the plant.24,25 In addition, the seed essential oil inhibited the in
vitro proliferation of Bacillus cereus, an important causative agent of
foodborne illness.26 An ethanolic seed extract also reduced paw edema
in laboratory rats in a process involving inhibition of cyclooxygenase-2
(COX-2) activity,27 and 6-paradol and 6-shogaol stimulated the
expression of proinammatory genes in an assay for proinammatory
gene expression.27 ese ndings are in accordance with the traditional
use of A. melegueta against inammatory conditions.
Indications for the usefulness of A. melegueta seed extracts as well as
6-gingerol and 6-shogaol against pests of stored foods are provided by
the repellent activity of both the seed extract and (S)-2-heptanol, (S)-
2-heptyl acetate, and (R)-linalool against the maize weevil Sitophilus
zeamais, an infamous agricultural pest.28 ese substances also elicited
antifeedant activity towards the subterranean termite Reticulitermes
speratus that is considered an urban pest of wooden constructions
in Japan.29 ese observations provide a tentative explanation for the
preference of wild western lowland gorillas to eat this plant and use it
to make the nests where they sleep at night.30 is behavior presumably
protects them from a bacterial or a viral infection that would cause
brosing cardiomyopathy, a common cause of heart failure and/or
sudden death in these animals.30
Indications for potential antitumor activity of A. melegueta were
provided by the inhibitory eects of 6-paradol as well as organic extracts
of the rhizome on the proliferation of various human tumor cell lines
(see, for instance31). e cytotoxic eects were accompanied by signs
of apoptosis31, possibly through a caspase-3-dependent pathway31.
Evidence for chemopreventive properties of A. melegueta came from the
inhibitory eects of 6-paradol and/or some of its synthetic derivatives
on the promotion of skin carcinogenesis and ear edema in ICR mice
induced by the laboratory tumor promoter phorbol 12-myristate
13-acetate (PMA).32 Furthermore, these compounds led to a reduction
in frequency and number of skin tumors caused by the laboratory
carcinogen 7,12-dimethylbenz[a]anthracene and promoted by PMA as
well as the induction of PMA-induced ornithine decarboxylase activity
in the animals.32 6-Paradol and its derivatives also led to a decrease in
DNA damage in cultured cancer cells.32
e use of A. melegueta seed preparations for improving fertility in
humans is supported by the stimulatory eect of an aqueous seed
extract on mating behavior, sexual arousal, and reproductive function
parameters in male Wistar rats 33, and the increased testosterone levels
in the animals following intaperitoneal injection of the seed oil.34 On
the other hand, administration of a seed extract to Sprague Dawley
rats led to termination of rst trimester pregnancy,35 suggesting that A.
melegueta seed also has abortifacient properties.
Other reported pharmacological eects of A. melegueta include blood
pressure-lowering activity of a seed preparation in both normotensive
and hypertensive individuals,36 and the hypoglycemic activity of an
aqueous leaf extract in alloxan-induced diabetic and non-diabetic
rats through the stimulation of insulin secretion from remnant or
regenerated pancreatic β cells.37 ese observations give some credit
to the traditional use of A. melegueta seed preparations against
hypertension and diabetes mellitus.
Alpinia galanga (L.) Willd.
e ai ginger, java galangal, greater galangal, or laos Alpinia galanga
(L.) Willd is one of four plants known as galangals or blue gingers. It
forms light-red or pale yellow rhizomes from which large clumps of
stalks arise which have small greenish-white owers (Figure 2) that
develop into orange-red fruits. A. galanga is probably native to Indonesia
and southern China but is now cultivated for its rhizome in various
other south-eastern Asian countries as well as Suriname. e rhizome
has an aromatic odor and a pungent and spicy taste comparable to that
of the common ginger Z. ocinale, and is used - either fresh or dried,
or powdered or sliced - as a avoring in many Indonesian, Chinese,
ai, Indian, and Surinamese dishes. e owers, ower buds, fruits,
and young shoots have a avor reminiscent of cardamom and are also
edible. e rhizome essential oil is used to confer an aroma of pine
needles to liqueurs such as Chartreuse and Angostura and certain so
drinks, and for preparing Essence d'Amali that is widely used in the
perfume industry.38
Preparations from fresh or dried A. galanga rhizomes have a wide range
of traditional applications. ese substances are used, among others,
against microbial infections including opportunistic (fungal) infections
such as those occurring in AIDS patients, HIV infection, parasitic
infections, rheumatic disorders, and gastrointestinal ailments.14,39,40
Furthermore, the dried rhizome is the most important raw material
in the worldwide renowned topical over-the-counter Chinese pain
reliever Tiger balm as well as other traditional Chinese remedies.41 And
in African-American hoodoo folk magic, A. galanga rhizome is known
as ‘Little John to chew’, ‘Chewing John’, and the ‘lucky court case root’,
as it would bring luck in court cases and legal matters when it is chewed
on and the juice is spit on the oor of the courthouse.42
Some of A. galanga’s traditional uses may be attributed to the
presence in the rhizome of various phytochemicals with meaningful
pharmacological activities including phenylpropanoids such as
1’S’-1’-acetoxychavicol acetate, 1’S-1’-acetoxyeugenol acetate, and
p-hydroxycinnamaldehyde; terpenes and terpenoids such as β-pinene,
camphor, and eugenol; as well as avonoids such as galangin and
alpinin.43,44 1’S’-1’-acetoxychavicol acetate is one of the pungent
Figure 2: Flower of Alpinia galanga (L.) Willd. (from: https://images.
app.goo.gl/XVNmeSWm7jxx6Ls8A).
5Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
ingredients of the rhizome38 while some of the terpenoids are
ingredients of the volatile rhizome essential oil and contribute to the
taste of the rhizome.45
e use of A. galanga against microbial infections is supported by
the activity of the rhizome essential oil and several organic rhizome
extracts against a variety of bacterial species including common food
borne bacteria46,47 as well as a number of fungi.46,48 ese eects have
been ascribed to 1’S’-1’-acetoxychavicol acetate in these preparations.49
is compound also inhibited R-plasmid transfer in various multidrug-
resistant bacteria49 and the active removal of drugs from drug-resistant
Mycobacterium spp.50 For these reasons, 1’S’-1’-acetoxychavicol acetate
may be pursued as a lead compound for developing more ecacious
antibacterial antibiotics.
at A. galanga also may be of benet against HIV infection is based
on the blockage of HIV-1 replication in cultured peripheral blood
mononuclear cells by 1’S-1’-acetoxychavicol acetate isolated from
the rhizomes.51 is compound inhibited HIV mRNA translocation
to the cytoplasm for translation into viral proteins, and acted
synergistically with the reverse transcriptase inhibitor didanosine in
halting HIV-1 replication in these cells.51 e supposed antiparasitic
properties of the plant are supported by the benecial eects of various
phenylpropanoids, terpenoids, and avonoids in rhizome extracts
against malaria in laboratory mice52 as well as their substantial in vitro
activity against promastigotes of Leishmania (Leishmania) donovani53,
the causative agent of visceral leishmaniasis.
Support for the traditional use of A. galanga against rheumatic
disorders came from the antiinammatory activities of methanolic
and ethanolic rhizome extracts in carrageenan-induced paw edema
and pleurisy in laboratory rats.54,55 ese eects have been attributed to
1’S-1’-acetoxychavicol acetate and 1’S-1’-acetoxyeugenol acetate which
inhibited the release of β-hexosaminidase (a marker of IgE-mediated
degranulation) in cultured RBL-2H3 peripheral blood cells, reduced
the production of IgE-mediated tumor necrosis factor (TNF)-α)
and interleukin (IL)-4 by these cells, and inhibited local anaphylaxis
in laboratory mice.56 p-Hydroxycinnamaldehyde may contribute to
the antiinammatory activity of A. galanga, since an acetone extract
enriched with this compound inhibited the release of hyaluronan,
sulfated glycosaminoglycans, and metalloproteinase-2 from primary
human chondrocytes challenged with the proinammatory cytokine
IL-1β.57
1'S-1'-acetoxychavicol acetate and 1'S-1'-acetoxyeugenol also reduced
the damage in the gastric mucosa of laboratory rats induced by HCl or
aspirin58 and were probably also responsible for the inhibitory eects
of rhizome and seed extracts on the development of gastric ulcers and
gastric mucosal damage produced in the animals by pyloric ligation
and hypothermic restraint stress.59 ese observations may justify
the traditional use of A. galanga against gastrointestinal conditions.
Notably, a crude rhizome extract substantially decreased the number
of necrotic cells in the liver of Sprague-Dawley rats treated with
paracetamol60, suggesting that A. galanga rhizome preparations also
possess hepatoprotective proporties.
Curcuma longa L.
e turmeric or yellow ginger Curcuma longa L. (1753) presumably
arose by selection and vegetative propagation of a hybrid between the
wild turmeric Curcuma aromatica Salisb. that is native to India, Sri
Lanka, and the eastern Himalayas, and other closely related species.
As a result, C. longa is not found in the wild and is only known as a
domesticated plant. e plant is abundantly cultivated in India (where
it is known as ‘haldi’) and Indonesia (where it is known as ‘kunyit’),
as well as many other tropical and subtropical regions throughout the
world. It is sterile but readily produces new sprouts from branches of its
pulpy orange-yellow tuberous underground rhizomes (Figure 3). e
maternal plant gives rise to yellow-white owers which do not produce
viable seed.
For human use, the rhizomes are boiled for several hours and then
dried in hot ovens, aer which they are ground to a deep orange-yellow
powder that has a bitter, slightly acrid yet sweet taste. e powder is
an essential ingredient of curry, an indispensable spice in many hot,
savory, and/or sweet south-eastern Asian and Surinamese-Hindustani
dishes. It is also widely used as a coloring agent in cheeses, butters, and
mustards, manufactured food products such as canned beverages, dairy
products, orange juice, popcorn, sweets, cake icings, cereals, sauces,
and gelatins.61 e essential oil from the fresh rhizome produced by
steam distillation is called turmerol and is incorporated in perfumes to
confer a spicy, fresh, and sweet fragrance61.
C. longa plays an important role in various social and religious Hindu
rituals symbolizing inner purity and pride as well as fertility and
prosperity. us, the rhizomes are oen given as a present to pregnant
women.17 A paste prepared from fresh rhizomes in coconut oil is
applied on the face of the bride and the groom on the the day before
the wedding ceremony as part of a purication ritual.17 Such a paste is
also rubbed on the forehead of newborn babies to protect them from
demons and the evil eye and promote their well-being.17 As this practice
may ward o harmful bacteria62, C. longa rhizome resin and essential
oil have been incorporated in several sunscreens and facial creams
including anti-acne creams.17
C. longa rhizome is also extensively used for preparing traditional
medicines, particularly in Indian Ayurveda and Unani as well as
Indonesian Jawa. ese medications are used against a variety of
inammatory conditions such as rheumatoid arthritis, inammations
of the gastrointestinal tract and liver, and eye inammations, but also
for treating precancerous conditions and cancer, various microbial
infections, as well as brain disorders such as depression and Alzheimer's
disease.15,17,63,64 Furthermore, C. longa rhizome is a key ingredient of
jamus to promote health and tness and to enhance mental functioning
and well-being.14
Important bioactive constituents of C. longa rhizome are polyphenolic
compounds such as diarylheptanoids (including the yellow-colored
curcuminoids such as curcumin which are responsible for the
characteristic color and flavor of the rhizome) and diarylpentanoids,
as well as terpenoids such as sesquiterpenes, monoterpenes,
diterpenes, and triterpenoids.65-67 e most common curcuminoid
in the plant is curcumin that makes up approximately 90% of the
Figure 3: Rhizome of Curcuma longa L. (from: https://images.app.goo.gl/
ZnQgCAFiXVrJHgt8A).
6
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
curcuminoid content, as well as its derivatives demethoxycurcumin
and bisdemethoxycurcumin.65,66 e sesquiterpenes are the main
constituents of the rhizome essential oil, while the monoterpenes
dominate the essential oils from the leaves and the owers.66,67 e
major volatile principles of the rhizome oil are α- and β-turmerone and
aromatic -turmerone.66,67
A host of pharmacological studies support the traditional use of C.
longa preparations as well as curcumin against arthritic conditions (see,
for instance,68,69). Furthermore, administration of a rhizome extract, the
rhizome volatile oil, curcumin itself, natural analogues of curcumin, or
semi-synthetic curcumin analogues led to a decrease in carrageenin-
or formaline-induced rat paw edema as well as cotton pellet-induced
granuloma in laboratory rats.70-73 e use of curcumin also resulted in
a substantial reduction in the inammatory swelling in rats suering
from arthritis induced by treatment with Freuds adjuvant.72 e
antiinammatory eects probably occurred through interference at
dierent levels of the arachidonic acid inammatory cascade and
inhibition of proinammatory compounds such as prostaglandins,
leukotrienes, and COX-2.68,69 Of note, a C. longa rhizome extract even
protected collagen-induced arthritic Sprague-Dawley rats from the
degenerative changes in the bone and ankle joints to a comparable
extent as betamethasone.74 Importantly, clinical trials with curcumin
produced encouraging results in patients with rheumatoid arthritis75
and postoperative inammation.76
e usefulness of C. longa in inammatory gastrointestinal conditions
is sustained by the inhibitory eects of curcumin on the gastric mucosal
damage caused by indomethacin in laboratory animals77 and on the
production of inammatory cytokines, intercellular adhesion molecule
1, and TNF-α in the animals.77 Furthermore, curcumin substantially
improved the prole of inammatory markers, severity of diarrhea,
and colonic architecture in laboratory mice with colitis induced by the
laboratory model compound trinitrobenzenesulfonic acid.78 Clinical
trials indeed showed benecial eects of curcumin or a standardized C.
longa rhizome extract in patients with peptic ulcers79 or inammatory
bowel disease.80 In fact, a Cochrane analysis revealed that curcumin
may be a safe and eective therapy for the maintenance of remission
in quiescent ulcerative colitis.81 In addition, there is some support for
the traditional use of C. longa against ocular inammations besides
arthritic disease and gastrointestinal inammations. is is based on
the encouraging results of oral curcumin in (a relatively small group of)
patients with eye inammations such as anterior uveitis and idiopathic
orbital inammatory pseudotumors while not producing serious side-
eects.82,83
Chronic inammation is considered an important driving factor
for malignant transformation and cancer progression84, providing
a rationale for using C. longa and its curcuminoid constituents as
chemopreventive and anticancer compounds. Indeed, comparably to
the antiinammatory eects, the antineoplastic eects would occur
through the modulation of critical intracellular signaling pathways
such as the NF-κB pathway.85 e potential chemopreventive eects
of curcumin have been observed in several preclinical models86-88 and
a few human studies.89-91 In addition to reducing the inammatory
cancer microenvironment, these eects might be due to the promotion
of apoptosis, inhibition of survival signals, and scavenging of reactive
oxidative species.92
Other potentially interesting pharmacological activities of C. longa
and its constituents that give credence to the traditional claims
are antimicrobial, antiparasitic, and anti-Alzheimer eects. e
antimicrobial properties have been observed in various standard
bacterial strains62,93,94 including common periopathogens95 and may
account for the inclusion of C. longa rhizome in jamus for treating
inamed gums, abscesses, menstrual pains, and skin rash7 as well
as the application of a C. longa-based Javanese ointment called
bobok for alleviating the discomfort of, among others, toothache.7
Furthermore, the broad antiparasitic activity of curcumin96 may explain
the Surinamese-Javanese custom of including C. longa rhizome in
preparations for treating pinworm infections in children.7
e potential usefulness of C. longa preparations against
neurodegenerative disorders is supported by their capacity to reduce
the deposition of plaques similar to those of Alzheimer’s disease in the
brains of aged mice and the oxidative damage and amyloid pathology
in transgenic mouse models of Alzheimer's disease97, as well as that of
curcumin and dimethoxycurcumin to lessen lead-induced memory
decits in rats.98 Although the evidence supporting the ecacy of
curcumin in Alzheimer’s disease is currently insucient, the data
thus far available are suciently encouraging to justify further eorts
to optimize absorption, bioavailability, and the timing and length of
intervention of the treatment.99
Hedychium coronarium J. König
e white ginger, buttery ginger lily, or white buttery ginger
Hedychium coronarium J. König, in Suriname also known as ‘gember
lelie’ (Dutch for ‘ginger lily’), is native to southern China, Taiwan,
Myanmar, northeast India, and Nepal. It may have been brought to
South America and the Caribbean by enslaved Africans who used the
leaves of this plant as mattresses during their trans-Atlantic journey.100
e vernacular name ‘(white) buttery ginger’ refers to the shape
of the owers which resembles a ying buttery (Figure 4). is is
also reected by its vernacular name ‘or de mariposa’ (Spanish for
‘buttery ower’) in Cuba, where it has become the national ower.
However, H. coronarium has been declared an invasive herb in several
parts of Africa and the Americas.101 Its rapid vegetative reproduction
through underground spread of the rhizomes makes it dicult to
control its expansion.101 Notwithstanding, the considerable demand of
this plant for preparing Ayurvedic medicines has led to such an extent
of overharvesting that it has become an endangered species in certain
parts of India.102
Fortunately, H. coronarium is cultivated in many tropical and
subtropical countries as an ornamental garden plant and as a source
for ower garlands and cut owers. e essential oil of the owers has
a scent reminiscent of jasmine and is oen incorporated in commercial
cosmetic preparations such as perfumes, skin conditioners, and facial
masks. e juice from the mature seeds is used as a hair and skin
treatment by native Hawaiians.100 Both the owers and the rhizomes are
consumed as vegetables in parts of south-eastern Asia. e dried stem
Figure 4: Flower of Hedychium coronarium J. König (from: https://
images.app.goo.gl/xKuFp1A6ZnpZYVFS6).
7Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
contains 43 to 48% cellulose, making it a useful source of raw material
for making paper.103
H. coronarium is also medicinally used in various traditional systems
throughout the world. Preparations from virtually all its parts are used
for treating, among others, microbial infections, parasitic infections,
inammatory complaints such as sti and sore joints along with pain
from rheumatism and arthritis, as well as cancer.104,105 e typical scent
of the ower is for an important part determined by the terpenes
β-transocimenone, α-farnesene, linalool, 1,8-cineole, and α-terpineol
in the essential oil.106 Important phytochemicals in the (essential oils
from the) rhizomes and leaves are the large array of biologically active
labdane diterpenes such as coronains, coronarins, hedychilactones,
and hedychenones, 107 farnesane sesquiterpenes such as nerolidol
and hedychiols,108 as well as the monoterpenes 1,8-cineole, β-pinene,
myrcene, limonene, and benzoyl eugenol.109
Pharmacological evidence for antimicrobial activity of H. coronarium
came from the broad antibacterial and antifungal eects of the rhizome
essential oil, extracts from the rhizomes, preparations from the leaves,
and decoctions from the owers.110-113 e antimicrobial activities
have been ascribed to various terpenoids in the essential oils110,112 Of
note, the sesquiterpenoid coronarin D isolated from the rhizomes also
showed considerable antibacterial and antifungal eects.113-115 In some
cases, the antifungal activity was comparable to that of standard drugs
such as nystatin and griseofulvin.113,114
Furthermore, the rhizome essential oil displayed remarkable activity
against earthworms and tapeworms115; an ethanolic rhizome extract
was active against cultured amastigotes from L. (L.) amazonensis
(Trypanosomatidae)116 and a chloroquine-resistant strain of Plasmodium
falciparum116; and leaf and rhizome essential oils exerted mosquito
larvicidal activities.112 ese eects have partially been attributed to
α-pinene, β-pinene, and 1,8-cineol112, and support the traditional use of
H. coronarium against parasitic infections.
Indications for antiinammatory and analgesic activity of H.
coronarium were provided by the inhibitory eects of rhizome
(essential oil) preparations on the activities of 5-lipoxygenase and
proinammatory cytokines in vitro117, as well as on carrageenan-induced
paw edema, heat- or acetic acid-induced writhing, and elongation
of tail ick time in laboratory mice.118,119 Furthermore, treatment of
laboratory mice with a methanolic rhizome extract led to suppression
of their motor activity and exploratory behavior, suggesting that central
mechanisms of pain perception had been depressed.120 ese eects
have been associated with labdabe diterpenes and farnesane-type
sesquiterpenes in the preparations.108,117,119.
Potential anticancer activity of H. coronarium is supported by the
cytotoxicity of a partially puried rhizome extract against brine shrimp
nauplii111, as well as that of several coronarins and their derivatives
isolated from the rhizome against cultured human and animal cancer
cell lines.121-123 Interestingly, some of the diterpenes substantially
inhibited the growth of cultured human umbilical vein endothelial
cells, suggesting that they may possess meaningful anti-angiogenic
properties.122 e cell growth inhibitory eects were in some cases
accompanied by cell cycle arrest at the G1 phase and signs of apoptosis.123
In addition, several coronarin labdane diterpenes as well as benzoyl
eugenol showed meaningful cancer chemopreventive activity in in vitro
assays, inhibiting NF-κB, COX-1 and COX-2 activities, the induction of
antioxidant response element, and cell proliferation.124
Kaempferia galanga (L.) Willd
e east-Indian galangal, sand ginger, aromatic ginger, or kentyur
Kaempferia galanga (L.) Willd. is a small, stemless herb that grows from
a rhizomatous rootstock, and is characterized by the thick, rounded
leaves that lay at in a rosette on the ground. e plant develops beautiful
white owers with an amethyst heart which have some resemblance to
orchids (Figure 5) and is widely grown as an ornamental in tropical
gardens. K. galanga is probably native to India but may have originated
from Myanmar. It is cultivated for its rhizome in southern China, India,
Bangladesh, ailand, Cambodia, Vietnam, as well as Suriname, but is
in general gathered from the wild for local use as a food and medicine.
As a result, K. galanga has become a highly priced but endangered
species in India.125
e rhizome has a slightly pungent fragrance with a spicy aroma
that resembles that of ginger and is a vital spice in oriental cuisine
that contributes to the unique taste and avor of many Indonesian,
Malaysian, ai, and Surinamese-Javanese dishes. e rhizome is also
used as a condiment and, when dried, as a substitute for turmeric in
curry powder. e young rhizomes and the young leaves are eaten raw,
steamed, in curries or cooked with chilli paste and served as a side dish
with rice. e crushed aromatic leaves are also used as a perfume in
washing hair.126,127 e essential oil extracted from the rhizome is used
in perfumery.126,127 and the powdered root is added to body powders
and cosmetics126,127 but also to repel moths in wardrobes.126,127
In addition, K. galanga has many traditional medical uses.
Preparations from particularly the rhizome are used against, among
others, colds, sore throat, coughing, bronchitis, asthma, rheumatism
and several other microbial infections and inammatory conditions;
various parasitic infections including helminthiasis and malaria;
headaches, mouth ulcers, and toothaches; skin problems such as
dandru, leprosy, and psoriasis; as well as restlessness, stress, anxiety,
and depression.126-128 Interestingly, Surinamese Javanese apply a K.
galanga-based preparation on the skin of babies to remove excessive
body hair; however, at a later age the child also does not develop hair
on arms and legs.14
e main pharmacologically active ingredients of K. galanga
probably are phenylpropanoids such as ethyl p-methoxy cinnamate,
p-methoxycinnamic acid, ethyl cinnamate, and cinnamaldehyde;
avonols such as kaempferol and kaempferide; as well as a number
of terpenoids such as 1,8-cineole, g-careen, and borneole.129,130ese
compounds are constituents of the rhizome essential oil and have been
associated with various pharmacological activities, supporting some of
the traditional uses.127,128 Leaves and owers of the plant also contain a
number of avonoids with biological activity.130
at K. galanga may possess antimicrobial activity is supported by
the inhibitory eects of extracts from its rhizome and leaves on the
growth of a variety of pathogenic bacterial and fungal species.131-133
Figure 5: Flower of Kaempferia galanga (L.) willd. (from: https://images.
app.goo.gl/o9A3tx2YGBDJoxji9).
8
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
including that of Lactobacillus acidophilus, the bacterium responsible
for dental caries.134 e antimicrobial activity might be attributed to
ethyl p-methoxy cinnamate that showed meaningful antibacterial
activity against the skin bacteria Staphylococcus aureus, Staphylococcus
epidermidis, and Cutibacterium acnes without causing allergic
irritation.135 ese ndings may account for the above-mentioned
traditional use of K. galanga preparations against mouth ulcers and skin
conditions.
Several lines of evidence support pesticidal, larvicidal, and mosquito-
repellent activities of K. galanga. Methanolic leaf and rhizome extracts
as well as the rhizome essential oil elicited considerable repellent and
larvicidal activity against harmful mosquito vectors such as Aedes
spp., Anopheles spp., Armigeres subalbatus, Culex spp., and Mansonia
uniformis,136-138 including those resistant to the household insecticide
pyrethroid.139 ese mosquito strains are the carriers and transmitters
of serious diseases ranging from yellow fever and chikungunya to
malaria and lariasis. Furthermore, the rhizome essential oil displayed
strong contact toxicity against the booklouse Liposcelis bostrychophila
,140 and a rhizome extract showed anthelmintic activity against the
Bangladeshi earthworm Pheretima posthuma and insecticidal activity
against the rice weevil Sitophilus oryzae.141 Notably, neither a crude K.
galanga rhizome extract nor the rhizome essential oil produced signs of
dermal irritation,142 providing further support for the usefulness of this
plant against dermatological problems. e larvicidal and mosquito-
repellent properties of K. galanga may particularly be attributable to
ethyl p-methoxycinnamate, but also to other phenylpropanoids and/or
terpenoids and/or kaempferol 139,140,142,143
Indications for antinociceptive and antiinammatory activities of K.
galanga came from the substantial inhibitory eects of an aqueous leaf
extract and an alcoholic rhizome extract in abdominal constriction,
tail ick, hot plate, and/or formalin assays as well as carrageenan and/
or cotton pellet granuloma models.144-146 Markedly, in a double-blind
randomized clinical trial with patients suering from osteoarthritis of
the knee, the rhizome extract exerted the same eect on pain, stiness,
and physical interference as the nonsteroidal antiinammatory
drug meloxicam.147 e analgesic eects probably occurred through
both central components related to opioid receptors and peripheral
components associated with the COX pathway.145 e antiinammatory
eects probably involved the suppression of IL-1, TNF-α, as well as
angiogenesis-related events.130,147 Both eects were probably caused,
at least partially, by ethyl-p-methoxycinnamate.148 Together, these
observations support the traditional use of K. galanga leaves and
rhizomes for treating mouth ulcers, headaches, sore throat, swellings,
stomach ache, toothache, and rheumatism.
Importantly, K. galanga may also possess meaningful sedative
properties. is can be derived from the considerable decrease in
locomotor activity, onset and duration of thiopental sodium-induced
sleeping time, and/or exploratory activities of Swiss mice which had
received an extract of the rhizome and/or leaf by inhalation or per
os.149,150 e apparent central nervous system-depressant properties of
the plant have been attributed to ethyl trans-p-methoxycinamate and
ethyl-cinnamate.149 ese ndings support the traditional use of the
plant against anxiety and depression, as well as its use in aromatherapy
in Japan against sleeplessness and stress and as an ingredient of pain
relief Ayurvedic massage blends.149
Renealmia alpinia (Rottb.) Maas
e inkplant, bigi masusa, or blaka masusa Renealmia alpinia (Rottb.)
Maas, also known as Renealmia exaltata L. l.), bears fruits that are red
when immature and turn purple-black when mature (Figure 6), and
then contain numerous seeds embedded in a yellow pulp. It grows from
red, aromatic rhizomes and oen forms large colonies. e vernacular
name ‘inkplant’ reects the previous use of the dark-colored sap of the
peels from the ripe fruits as an ink for writing and applying skin tattoos151
Reddish-colored ink was obtained by adding lime juice to the sap.151 R.
alpinia is native to tropical America and can be found from Mexico to
Brazil and in several Caribbean islands. In Suriname, it grows wild in
secondary forests and on river banks, but it is cultivated for the red-
brown oil from the pulp around the seeds that is used for preparing the
widely appreciated tasty Creole rice dish ‘masusa moksi aleysi’ (literally
‘masusa mixed rice’). Peasants from the interior of Brazil oen drink
a juice prepared from the leaves as a cooling beverage in hot weather.
And in Mexico, tamales and empanadas are given a special spicy taste
by steaming them while wrapped in fresh R. alpinia leaves.
R. alpinia is also an important plant in various traditional medical
systems. Preparations from the rhizomes and leaves are used against
the symptoms of snake bites and scorpion stings in humans and
(hunting) dogs; bacterial infections causing fever, gastrointestinal
problems, or heart problems manifesting as shortness of breath or chest
colds; fungal infections including those causing dandru and vaginitis;
and convulsions during, for instance, epileptic seizures.15,152-154 In
Suriname, particularly Maroons use preparations from R. alpinia leaves
or rhizomes - either alone or together with parts from other plants -
as a blood-purifying agent, against infertility in women, to speed up
delivery, and to prevent uterine inammation aer childbirth and to
quickly obtain a slim gure aer pregnancy.15 Parts of the plant are
also included into herbal baths for strengthening the nerves, to convey
spiritual strength and self-condence, to remove sadness, despair, and
depression, for protection from the evil eye, and to dispel the spirit of a
deceased person.13 Curiously, according to ancient Amerindian belief,
women should avoid holding R. alpinia fruits too long in their hands as
this would accelerate aging.15
e characteristic avor and fragrance of R. alpinia seed oil is
attributable to monoterpenes such as β-pinene, limonene, and
β-phellandrene, as well as β-carotene, labdane diterpenoids, and
diarylheptanoids.155 Other important phytochemicals in the rhizomes
and the leaves are biologically active phenolic compounds such as
coumarins and the avanone pinostrobin, several labdane diterpenes,
as well as the kavalactone desmethoxyyangonin.155-158
e coumarins and pinostrobin have been associated with the
meaningful antivenom properties of R. Alpinia.159,160 is has been
inferred from the substantial analgesic, antiedematous, antihemorrhagic,
antidebrinating, anticoagulant, and neutralizing eects of rhizome
and leaf extracts containing these compounds in laboratory mice
poisoned with B. atrox venom.152,159,160 Furthermore, pinostrobin (the
Figure 6: Fruits from Renealmia alpinia (Rottb.) Maas (from: https://
images.app.goo.gl/qZF2bk1icRpYip3R8).
9Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
main ingredient of a dichloromethane leaf extract) elicited strong in
vitro analgesic, antinociceptive, and antiinammatory activities while
inhibiting the local tissue damage caused by the hemorrhagic eects
of the viper venom.161 Pinostrobin may neutralize the eects of snake
bites by opposing the anticoagulant and membrane-damaging eects of
phospholipase A2 activity in the venom.162
e use of R. alpinia against microbial infections is supported by the
bactericidal activity of leaf and stem extracts against Bacillus subtilis163
and the fungicidal activity of rhizome extracts against several species
of Candida, the dermatophytic fungus Trichophyton rubrum, and two
varieties of the encapsulated yeast Cryptococcus neoformans. 164,165
ese activities have been attributed to several labdane diterpenoids in
the extracts.163-165 ese compounds have also been implicated in the
antileishmanial activity against cultured amastigotes of Leishmania
(Leishmania) chagasi and L. (L.) amazonensis;116,165 the antiprotozoal
activity against epimastigotes of Trypanosoma cruzi, the causative agent
of Chagas’ disease;165 the antimalarial activity against a chloroquine-
resistant strain of P. falciparum;116 and the cytotoxic eects against
several cancer cell lines.156,166
Desmethoxyyangonin was previously isolated from the kava-kava plant
Piper methysticum167 and reversibly inhibited monoamine oxidase
(MAO) B in the central nervous system, thereby increasing serotonin
and dopamine levels.168 Desmethoxyyangonin puried from the
dichloromethane extract of R. alpinia leaves also potently inhibited
recombinant human MAOs - particularly MAO B - in an in vitro study169
us, this compound may not only suppress anxiety, stimulate feelings
of well-being, and promote attention,170 but also counter neurological
diseases associated with errors in MAOs such as neurodegenerative
disturbances171 and seizures,172 supporting the traditional use of
R. alpinia preparations for treating seizures in children and other
childhood conditions.151
Zingiber ocinale Rosco e.
e common ginger or dyindya Z. ocinale probably originates from
south-eastern Asia where it was presumably domesticated (it does not
exist anymore in its wild state), and subsequently spread throughout the
rest of the continent and many other parts of the world. It may have been
introduced in Suriname by enslaved Africans and Javanese indentured
laborers via Western Africa and south-eastern Asia, respectively.10,14
e inorescences directly arise from the rhizome on separate shoots
(Figure 7) and bear clusters of white and pink ower buds that bloom
into yellow owers. Because of its esthetic appeal, the plant is oen used
as landscaping around tropical and subtropical homes. However, Z.
ocinale is mainly cultivated for its rhizome which was one of the rst
spices exported from Asia to Europe during the spice trade, i.e., before
the beginning of the Christian era, and was already highly appreciated
by the ancient Greeks and Romans.173
Z. ocinale has remained an economically valuable crop. In 2018,
approximately 3.3 million tonnes were produced worldwide,
representing a market revenue of U$ 5.3 billion.174 China dominated the
exports in that year, accounting for 390 kilotonnes or more than two-
thirds of total exports, distantly followed by ailand (54 kilotonnes),
Peru (21 kilotonnes), India (21 kilotonnes), Brazil (15 kilotonnes), and
e Netherlands (13 kilotonnes).174
e fresh or dried rhizomes are widely used as a hot, fragrant kitchen
spice in many South Asian, Latin American, and Caribbean cuisines
for avoring seafood, meat, and vegetarian dishes and for making
curries and other spicy dishes. Both the fresh and the dried rhizome
are used for these purposes, but the latter is about twice as pungent as
the former. e young rhizomes are juicy and eshy and have a mild
taste and are also pickled in vinegar or sherry as a snack, and included
in gingerbread, ginger cake, cookies, and speculaas, a spiced shortcrust
biscuit that is traditionally baked in e Netherlands and Belgium for
consumption around St. Nicholas’ Day (5 and 6 December, respectively)
and in Germany around Christmas.
In addition, the rhizomes can be immersed in boiling water to make
ginger herb tea which is sweetened with honey, and they can be made
into ginger wine, ginger ale, and ginger beer. e rhizome also contains
an essential oil that is used to avor essences as well as in perfumery.175
e young inorescences can be eaten raw, and the young, slightly
spicy leaves and young shoots can be eaten as a vegetable or pureed
and added to sauces and dips. e leaves can also be used to wrap food
while it is cooked, adding extra avor to the food.
Z. ocinale has a myriad of applications in various traditional
medical systems. In India, it is regarded as a universal medicine176
and preparations from the rhizome are ingredients of numerous
prescriptions in Ayur vedic and traditional Chinese medicine.176,177 ese
products are internally used to control nausea and vomiting including
those caused by morning and motion sickness; microbial infections
of the upper respiratory tract; parasitic infections such as lariasis;
inammatory conditions such as asthma as well as rheumatoid arthritis
and osteoarthritis; problems with the peripheral circulation including
hypertension; and externally to treat spasmodic pain, rheumatism,
pain in the muscles and joints of the lower back, menstrual cramps,
and sprains.176-178
e distinctive odor and avor of Z. ocinale rhizome mostly
result from volatile oils but also from some nonvolatile phenolic
compounds.179 e volatile oils account for 1 to 3% of the weight of
fresh rhizome and mainly consist of sequiterpene hydrocarbons,
predominantly zingiberol.179 e nonvolatile phenolic phytochemicals
consist of gingerols, shogaols, paradols, and zingerone, as well as
various other gingerol-related compounds179,180 e major pungent
compound and the best studied phytochemical in Z. ocinale is
6-gingerol.179,180 Zingerone is produced from gingerols during drying
and is less pungent.179-181 Shagoals are about twice more pungent when
compared to the gingerols and and are not found in raw ginger but are
the dehydrated products of gingerols that are formed during drying,
heating, or prolonged storage of the rhizome.179-181
Support for the traditional use Z. officinale against nausea and
vomiting is provided by initial animal studies suggesting that
preparations from the rhizome had antiemetic activity in nausea
caused by cyclophosphamide or cisplatin.182-184 These effects
would occur peripherally, within the gastrointestinal tract, by
Figure 7: Rhizome of Zingiber ocinale Roscoe (from: https://images.
app.goo.gl/5DDiHrm1zRP5NzyF7).
10
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
increasing gastric tone and motility through anticholinenergic and
antiserotonergic mechanisms.185 However, human studies have not led
to conclusive results, some speaking in favor of antiemetic properties
of Z. ocinale (see, for instance,186) but others contradicting these
actions (see, for instance, 187). On the other hand, various gingerols,
6-shogaol, and zingerone were shown to antagonize the activation
of cholinergic and serotonin receptors in laboratory models,188,189 the
main targets of emetogenic chemotherapeutic drugs. Furthermore, a
few reviews and meta-analyses concluded that Z. ocinale was better
than placebo in pregnancy-related nausea and vomiting as well as
nausea and vomiting induced by chemotherapy or motion sickness but
that this should be veried in suciently large clinical studies (see, for
instance,190).
Indications for the usefulness of Z. ocinale against microbial
infections came from the broad in vitro antibacterial and antifungal
activity of organic extracts from the rhizome.191,192 ese eects might
be associated with the avonoid fractions of the extracts.193 Various lines
of evidence also support the usefulness of the plant against parasitic
infections. us, 6-gingerol, 10-gingerol, 6-shogaol, 10-shogaol, and/
or hexahydrocurcumin isolated from the rhizome elicited larvicidal
activity against the yellow fever mosquito Aedes aegypti that carries
and spreads yellow fever, dengue fever, chikungunya, and Zika fever
viruses, as well as the southern house mosquito Culex quinquefasciatus,
the primary vector of the round worm Wuchereria bancroi that causes
lymphatic lariasis.194
Furthermore, methanol and aqueous extracts of Z. ocinale rhizome
displayed substantial anthelmintic activity against the barber's
pole worm Haemonchus contortus that causes anemia, edema, and
eventually death of infected sheep and goats195, as well as the Indian
earthworm Pheretima posthuma that has been used as a model for
intestinal roundworm parasites of humans.196 ese extracts also elicited
meaningful activity against larvae of the rat long worm Angiostrongylus
cantonensis as well as those of the parasitic sh nematode Anisakis
simplex.197 A. cantonensis produces angiostrongyliasis, the most
common cause of eosinophilic meningitis or meningoencephalitis in
south-eastern Asia and the tropical Pacic islands, and A. simplex is
associated with anisakiasis, a gastrointestinal infection characterized by
severe abdominal cramps.
e use of Z. ocinale as an antiinammatory and analgesic compound
is supported by the notable inhibitory eects of a rhizome extract on
edema, stretching, as well as jumping and hind paw-licking of laboratory
rodents subjected to the carrageenan-induced rat paw oedema test,
the acetic acid-induced writhing assay, and the hot plate test.198-200
Comparable results were found with several in vitro antiinammatory
assays activity such as protein denaturation inhibition, membrane
stabilization, protease inhibition, and anti-lipoxygenase assays.201 e
in vivo antiinammatory activity was mainly ssociated with certain
paradols, shogaols, and gingerols, and might involve, among others,
inhibition of LPS-induced PGE2 production202 and decreases in pro-
inammatory cytokines and chemokines resulting in inhibition of
the activation and migration of monocytes and leukocytes.200 ese
compounds also strongly inhibited COX-2 activity in intact cells.201
CONCLUDING REMARKS
In the current paper, phytochemical and pharmacological evidence have
been compiled to support the traditional medical uses of seven common
Zingiberacea species in Suriname. e data obtained are summarized
in Table 1 and indicate that there is for all plants scientic evidence to
support some of the traditional uses. erefore, there is at least some
merit to the broad use of these plants as traditional and alternative
medicines and as nutraceuticals, i.e., foods or food constituents that
provide health benets in addition to nutritional value. In addition,
drug regulatory authorities such as the FDA generally regard many if
not all Zingiberacea species as safe.203
Furthermore, almost all the plants possessed antimicrobial and
antiparasitic properties which may support their use against (certain)
infectious diseases. is is not unexpected when considering
the repellent properties of the pungent pharmacologically active
constituents in their essential oils. However, the apparent antimicrobial
and antiparasitic eects have in general been observed in a handful of
preclinical studies. Indeed, clinical proof that any of these compounds
elicit these eects is absent, particularly against more serious infectious
diseases such as, among others, malaria, chikungunya, laria, and
leishmaniasis. us, despite the availability of supporting data, claims
of antimicrobial and antiparasitic activities of Zingiberacea species
should be taken with caution to avoid the risk of using inecacious
substances and delay in seeking professional help in these cases.
Many of the plants also displayed antiinammatory and analgesic
activity, but again, mostly in studies with laboratory animals. us,
these data should also be regarded with caution. Still, the results from
clinical studies evaluating the ecacy of curcumin from C. longa
against rheumatoid arthritis75 and that of the rhizome extract from
K. galanga against osteoarthritis of the knee147 were encouraging. e
same holds true for clinical studies on the capacity of curcumin from C.
longa to improve neurodegenerative diseases such Alzheimer’s disease99
and that of Z. ocinale rhizome preparations to control pregnancy-
related, chemotherapy-associated, and motion sickness-related nausea
and vomiting.190
However, the relatively small size of the patient populations enrolled
in the studies as well as other aws in the study designs have led to
serious doubts about the reliability of the outcomes of the trials (see,
for instance75,76,190). Still, the many encouraging preclinical and clinical
data warrant re-evaluation of (some of) these substances in suciently
large and better designed clinical studies. ese studies should also
take into account that many pharmacologically active compounds of
Zingiberacea species are ingredients of volatile oils and thus poorly
water-soluble. is may create major pharmcodynamic challenges
when administered to humans. For instance, the high lipophilicity of
curcumin has led to too low oral bioavailability, chemical stability, and
intracellular concentrations to make the results from clinical studies
evaluating its anticancer activity reliable.204
is has led to attempts to increase the overall anticancer activity of
curcumin by introducing structural modications in the molecule
that would improve its selectivity towards cancer cells as well as its
bioavailability and/or stability, or to use delivery systems that would
improve its physicochemical properties.117 e active ingredients from
other Zingiberacea species may conceivably also pose such problems
which may be solved through comparable strategies. An example is
desmethoxyyangonin from R. alpinia that may be modied in order
to improve its delivery to the brain for treating seizures in children.131
ese studies may help denitely establish the therapeutic importance
of the Zingiberaceae.
REFERENCES
1. Kress WJ, Prince LM, Williams KJ. The phylogeny and a new classification
of the gingers (Zingiberaceae): evidence from molecular data. Am J Bot.
2002;89:1682-96.
2. George M, Cherian E. Emergent global marketing challenges for Kerala
cardamom producers vis-à-vis role of the spice board of India. Int J Comm
Develop Manag Stud. 2017;1:39-62.
3. Silva ABWR, Herath H, Senanayake SP, Swarnathilaka DBR. Phenetic and
genetic characterization of selected economically important species in the
family Zingiberaceae. Sri Lankan J Biol. 2018;3:34-43.
4. Wohlmuth H. Phytochemistry and pharmacology of plants from the ginger
family, Zingiberaceae. PhD thesis, Southern Cross University, Lismore, New
South Wales, Australia.
5. General Bureau of Statistics. Demographic data 2013-2016. 2018 https://www.
statistics-suriname.org/wp-content/uploads/2019/02/DEMOGRAFISCHE-
DATA-2013-2016.pdf.
11 Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
6. General Bureau of Statistics. Gross domestic product 2014-2018. 2018
https://statistics-suriname.org/wp-content/uploads/2019/09/SURINAME-BBP-
SCHATTINGEN-2014-2018-Version-2-GDP-ESTIMATES-2014-2018-Version-2.
pdf.
7. World Bank Group. Suriname - World Bank: Data 2019 https://data.worldbank.
org/country/suriname.
8. Mans DRA, Ganga D, Kartopawiro J. Meeting of the minds: traditional herbal
medicine in multiethnic Suriname. Chapter 6. In: El-Shemy H (ed). Aromatic
and Medicinal Plants - Back to Nature. InTech, Rijeka, Croatia. 2017.
9. Crosby AW. The Columbian exchange: biological and cultural consequences of
1492. Greenwood Press, Westport (CT), USA.1972.
10. Carney JA. Rice and memory in the age of enslavement: Atlantic passages to
Suriname. Slavery Abolition 2005;26:325-47.
11. Mans DRA. Nature, green in leaf and stem. Research on plants with medicinal
properties in Suriname. Clin Med Invest 2016;2:1-10.
12. May AF. Sranan oso dresi. Surinaams kruidenboek (Surinamese folk medicine.
A collection of Surinamese medicinal herbs). De Walburg Pers, Amsterdam,
The Netherlands. 1982.
13. Heyde H. Surinaamse medicijnplanten (Surinamese medicinal plants), 2nd ed.
Westfort, Paramaribo, Suriname. 1987.
14. Tjong Ayong G.Het gebruik van medicinale planten door de Javaanse
bevolkingsgroep in Suriname (The use of medicinal plants by the Javanese in
Suriname). Instituut voor de Opleiding van Leraren, Paramaribo, Suriname.1989.
15. Van Andel TR, Ruysschaert S. Medicinale en rituele planten van Suriname
(Medicinal and ritual plants of Suriname). KIT Publishers, Amsterdam, The
Netherlands. 2012.
16. Moret ES. Trans-Atlantic diaspora ethnobotany: legacies of West African and
Iberian Mediterranean migration in Central Cuba. Pp. 217-245. In: Voeks R,
Rashford J (eds). African Ethnobotany in the Americas. Springer, New York
(NY), USA. 2013.
1 7. Prasad S, Aggarwal BB. Turmeric, the golden spice: from traditional medicine
to modern medicine. Chapter 13. In: Benzie IFF, Wachtel-Galor S (eds). Herbal
Medicine: Biomolecular and Clinical Aspects, 2nd edition. CRC Press/Taylor &
Francis, Boca Raton (FL), USA. 2011.
18. Ogbole OO, Ajaiyeoba EO. Traditional management of tuberculosis in
Ogun State of Nigeria: the practice and ethnobotanical survey. Afr J Tradit
Complement Altern Med. 2009;7:79-84.
19. Gbolade A. Ethnobotanical study of plants used in treating hypertension in Edo
State of Nigeria. J. Ethnopharmacol. 2012;144:1-10.
20. Amadi SW, Zhang Y, Wu G. Research progress in phytochemistry and biology of
Aframomum species. Pharm Biol. 2016;54:2761-70.
21. Agim CS, Nosiri CI, Chukwuma A, Okechukwu A.Evaluation of the phytochemical
and GC-MS analysis of the aqueous seed extract of Aframomum melegueta. J
Pharmacogn Phytochem. 2017;6:101-4.
22. Wabo HK, Tane P, Connolly JD. Diterpenoids and sesquiterpenoids from
Aframomum arundinaceum. Biochem Syst Ecol. 2006;34:603-5.
23. Okigbo RN, Ogbonnanya O U. Antifungal effects of two tropical plants extracts
Ocimum gratissimum and Afromaomum melegueta on post-harvest yam
Discorea spp rot. Afr J Biotechnol. 2006;5:727-31.
24. Djeussi DE, Noumedem JA, Seukep JA, Fankam AG, Voukeng IK. Antibacterial
activities of selected edible plants extracts against multidrug-resistant Gram-
negative bacteria. BMC Complement Altern Med. 2013;13:164.
25. Ngwoke KG, Chevallier O, Wirkom VK, Stevenson P, Elliott CT. In vitro bactericidal
activity of diterpenoids isolated from Aframomum melegueta K.Schum against
strains of Escherichia coli, Listeria monocytogenes and St aphylococcus aureus.
J Ethnopharmacol. 2014;151:1147-54.
26. Uzeh RE, Oguntosin DO. Efficacy of essential oils from some African spices
against two strains of Bacillus cereus isolated from vegetable salad. J Food
Res. 2013;2:48-54.
27. Ilic NM, Dey M, Poulev AA, Logendra S, Kuhn PE, Raskin I. Antiinflammatory
activity of grains of paradise (Aframomum melegueta Schum) extract. J Agric
Food Chem. 2014;62:10452-7.
28. Ukeh DA, Birkett MA, Pickett JA, Bowman AS, Mordue AJ. Repellent activity
of alligator pepper, Aframomum melegueta, and ginger, Zingiber officinale,
against the maize weevil, Sitophilus zeamais. Phytochemistr y. 2009;70:751-58.
29. Escoubas P, Lajide L, Mizutani J. Termite antifeedant activity in Aframomum
melegueta. Phytochemistry. 1995;40:1097-9.
30. Dybas CL, Raskin I. Out of Africa: a tale of gorillas, heart disease and a swamp
plant. BioScience. 2007;57:392-7.
31. Keum YS, Kim J, Lee KH, Park KK, Surh YJ. Induction of apoptosis and caspase-3
activation by chemopreventive [6]-paradol and structurally related compounds
in KB cells. Cancer Lett. 2002;177:41-7.
32. Chung WY, Jung YJ, Surh YJ, Lee SS, Park KK. Antioxidative and antitumor
promoting effects of [6]-paradol and its homologs. Mutat Res. 2001;496:199-
06.
33. Mbongue G, Kamtchouing P, Dimo T. Effects of the aqueous extract of dry
seeds of Aframomum melegueta on some parameters of the reproductive
function of mature male rats. Andrologia. 2012;44:53-8.
34. Akpanabiatu MI, Ekpo ND, Ufot UF, Udoh NM, Akpan EJ, Etuk EU. Acute
toxicity, biochemical and haematological study of Aframomum melegueta seed
oil in male Wistar albino rats. J Ethnopharmacol. 2013;150: 590-4.
35. Inegbenebor U, Ebomoyi MI, Onyia KA, Amadi K, Aigbiremolen AE. Effect
of alligator pepper (Zingiberaceae Aframomum melegueta) on first trimester
pregnancy in Sprague Dawley rats. Niger J Physiol Sci. 2009;24:161-4.
36. Lawal B, Aderibigbe A, Essiet G, Essien A. Hypotensive and antihypertensive
effects of Aframomum melegueta seeds in humans. Int J Pharmacol.
2007;3:311-8 .
37. Adefegha SA, Oboh G. Inhibition of key enzymes linked to type 2 diabetes
and sodium nitroprusside-induced lipid peroxidation in rat pancreas by
water extractable phytochemicals from some tropical spices. Pharm Biol.
2012;50:857-65.
38. Yang X, Eilerman RG. Pungent principal of Alpinia galanga (L.) Swartz and its
applications. J Agric Food Chem. 1999;47:1657-62.
39. Verma RK, Mishra G, Singh P, Jha KK, Khosa RL. Alpinia galanga - an important
medicinal plant: a review. Der Pharmacia Sinica. 2011;2:142-54.
40. Shukla D, Jawaid T, Srivastava S. Alpinia galanga: an overview and herbal
interactions. Med Res Chron. 2017;4:301-5.
41. Wang S, Zhao Y, Zhang J, Huang X, Wang Y. Antidiarrheal effect of Alpinia
oxyphylla Miq. (Zingiberaceae) in experimental mice and its possible mechanism
of action. J Ethnopharmacol. 2015;168:182-90.
42. Yronwode C. Hoodoo herb and root magic. The Lucky Mojo Curio Co.,
Forestville (CA), USA. 2002.
43. Ma X-N, Xie C-L, Miao Z, Yang Q, Yang X-W. An overview of chemical constituents
from Alpinia species in the last six decades. RSC Adv. 2007;7:14114-44.
44. Chouni A, Santanu P. A review on phytochemical and pharmacological potential
of Alpinia galanga. Pharmacogn J. 2018;10:9-15.
45. De Pooter HL, Omar MN, Coolsaet BA, Schamp NM. The essential oil of greater
galanga (Alpinia galanga) from Malaysia. Phytochemistry. 1985;24:93-6.
46. Thuy Quynh VT, Duszkiewicz-Reinhard W. Antimicrobial activity of essential
oils from fresh and dried Alpinia galanga rhizomes. J Essent Oil Bear Plants.
2004;7:165-70.
47. Rao K, Ch B, Narasu LM, Giri A. Antibacterial activity of Alpinia galanga (L) Willd
crude extracts. Appl Biochem Biotechnol. 2010;162:871-84.
48. Janssen AM, Scheffer JJ. Acetoxychavicol acetate, an antifungal component of
Alpinia galanga L. Planta Med. 1985;51:507-11.
49. Latha C, Shriram VD, Jahagirdar SS, Dhakephalkar PK, Rojatkar SR. Antiplasmid
activity of 1’S’-1’-acetoxychavicol acetate from Alpinia galanga against
multidrug resistant bacteria. J Ethnopharmacol. 2009;123:522-5.
50. Roy SK, Pahwa S, Nandanwar H, Jachak SM. Phenylpropanoids of Alpinia
galanga as efflux pump inhibitors in Mycobacterium smegmatis mc2 155.
Fitoterapia. 2012;83:1248-55.
51. Ye Y, Li B. 1’S-1’-Acetoxychavicol acetate isolated from Alpinia galanga inhibits
human immunodeficiency virus type 1 replication by blocking Rev transport. J
Gen Virol. 2016;87:2047-53.
52. Al-Adhroey AH, Nor ZM, Al-Mekhlafi HM, Mahmud R. Median lethal dose,
antimalarial activity, phytochemical screening and radical scavenging of
methanolic Languas galanga rhizome extract. Molecules. 2010;15:8366-76.
53. Kaur A, Singh R, Dey CS, Sharma SS, Bhutani KK, Singh IP. Antileishmanial
phenylpropanoids from Alpinia galanga (Linn.) Willd. Indian J Exp Biol.
2010;48:314-7.
54. Unnisa A, Parveen TD. Anti-inflammatory and acute toxicity studies of the
extracts from the rhizomes of Alpinia galanga Willd. Der Pharm Sin. 2011;2:361-
7.
55. Subash KR, Prakash GB, Reddy KV, Manjunath K, Rao KU. Anti-inflammatory
activity of ethanolic extract of Alpinia galanga in carrageenan induced pleurisy
rats. Natl J Physiol Pharm Pharmacol. 2016;6:468-70.
56. Matsuda H, Morikawa T, Managi H, Yoshikawa M. Antiallergic principles from
Alpinia galanga: structural requirements of phenylpropanoids for inhibition of
degranulation and release of TNF-α and IL-4 in RBL-2H3 cells. Bioorg Med
Chem Lett. 2003;13:3197-02.
57. Phitak T, Choocheep K, Pothacharoen P, Pompimon W, Premanode B. The
effects of p-hydroxycinnamaldehyde from Alpinia galanga extracts on human
chondrocytes. Phytochemistry. 2009;70:237-43.
58. Matsuda H, Pongpiriyadacha Y, Morikawa T, Ochi M, Yoshikawa M.
Gastroprotective effects of phenylpropanoids from the rhizomes of Alpinia
galanga in rats: structural requirements and mode of action. Eur J Pharm.
2003;471:59-67.
59. Al-Yahya MA, Rafatullah S, Mossa JS, Ageel AM, AlSaid MS, Tariq M. Gastric
antisecretory, antiulcer and cytoprotective properties of ethanolic extract of
Alpinia galanga willd in rats. Phyther Res. 1990;4:112-4.
12
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
60. Hemabarathy B, Budin SB, Feizal V. Paracetamol hepatotoxicity in rats treated
with crude extract of Alpinia galanga. J Biol Sci. 2009;9:57-62.
61. Sharma RA, Gescher AJ, Steward WP. Curcumin: the story so far. Eur J Cancer.
2005;41:1955-68.
62. Moghadamtousi SZ, Kadir HA, Hassandarvish P, Tajik H, Abubakar S. A review
on antibacterial, antiviral, and antifungal activity of curcumin. Biomed Res Int.
2014:186864.
63. Gupta SC, Sung B, Kim JH, Prasad S, Li S, Aggarwal BB. Multitargeting
by turmeric, the golden spice: from kitchen to clinic. Mol Nutr Food Res.
2013;57:1510-28.
64. Kunnumakkara B, Bordoloi D, Padmavathi G, Monisha J, Roy NK. Curcumin,
the golden nutraceutical: multitargeting for multiple chronic diseases. Br J
Pharmacol. 2017;174:1325-48.
65. Li S, Yuan W, Deng G, Wang P, Yang P. Chemical composition and product quality
control of turmeric (Curcuma longa L.). Pharm Crops. 2011;2:28-54.
66. Sabale P, Modi A, Sabale V. Curcuma longa Linn. A phytochemical and
phytopharmacological review. Res J Pharmacogn Phytochem. 2013;5:59-68.
67. Stanojević JS, Stanojević LP, Cvetković DJ, Danilović BR. Chemical composition,
antioxidant and antimicrobial activity of the turmeric essential oil (Curcuma
longa L.). Advan Technol. 2015;4:19-25.
68. Jurenka JS. Anti-inflammatory properties of curcumin, a major constituent of
Curcuma longa: a review of preclinical and clinical research. Altern Med Rev.
2009;14:141-53.
69. Tabrizi R, Vakili S, Akbari M, Mirhosseini N, Lankarani KB. The eects of
curcumin-containing supplements on biomarkers of inflammation and oxidative
stress: a systematic review and meta-analysis of randomized controlled trials.
Phytother Res. 2018;33:253-62.
70. Arora RB, Basu N, Kapoor V, Jain AP. Anti-inflammatory studies on Curcuma
longa (turmeric). Indian J Med Res. 1971;59:1289-95.
71. Chandra D, Gupta SS. Anti-inflammatory and antiarthritic activity of volatile oil
of Curcuma longa (haldi). Indian J Med Res. 1972;60:138-42.
72. Srimal RC, Dhawan BN. Pharmacology of diferuloyl methane (curcumin), a non-
steroidal anti-inflammatory agent. J Pharm Pharmacol. 1973;25:447-52.
73. Rao TS, Basu N, Siddiqui HH. Anti-inflammatory activity of curcumin analogues.
Indian J Med Res. 1982;75:574-8.
74. Kamarudin TA, Othman F, Mohd Ramli ES, Md Isa N, Das S. Protective effect
of curcumin on experimentally induced arthritic rats: detailed histopathological
study of the joints and white blood cell count. EXCLI J. 2011;11:226-36.
75. Deodhar SD, Sethi R, Srimal RC. Preliminary study on antirheumatic activity of
curcumin (diferuloyl methane). Indian J Med Res. 1980;71:632-43.
76. Satoskar RR, Shah SJ, Shenoy SG. Evaluation of anti-inflammatory property
of curcumin in patients with post operative inflammation. Int J Clin Pharmacol
Ther Toxicol. 1986;24:651-4.
77. Thong-Ngam D, Choochuai S, Patumraj S, Chayanupatkul M, Klaikeaw N.
Curcumin prevents indomethacin-induced gastropathy in rats. World J
Gastroenterol. 2012;18:1479-84.
78. Ukil A, Maity S, Karmakar S, Datt a N, Vedasiromoni JR, Das PK. Curcumin,
the major component of food flavour turmeric, reduces mucosal injury in
trinitrobenzene sulphonic acidinduced colitis. Br J Pharmacol. 2003;139:209-
18.
79. Prucksunand C, Indrasukhsri B, Leethochawalit M, Hungspreugs K. Phase II
clinical trial on effect of the long turmeric (Curcuma longa Linn) on healing of
peptic ulcer. Southeast Asian J Trop Med Public Health. 2011;32:208-15.
80. Hanai H, Iida T, Takeuchi K, Watanabe F, Maruyama Y. Curcumin maintenance
therapy for ulcerative colitis: randomized, multicenter, double-blind, placebo-
controlled trial. Clin Gastroenterol Hepatol. 2006;4:1502-6.
81. Kumar S, Ahuja V, Sankar MJ, Kumar A, Moss AC. Curcumin for maintenance of
remission in ulcerative colitis Cochrane Database Syst Rev. 2012;17:CD008424.
82. Lal B, Kapoor AK, Asthana OP, Agrawal PK, Prasad R. Efficacy of curcumin in
the management of chronic anterior uveitis. Phytother Res. 1999;13:318-22.
83. 831. Lal B, Kapoor AK, Agrawal PK, Asthana OP, Srimal RC. Role of curcumin in
idiopathic inflammatory orbital pseudotumours. Phytother Res. 2000;14:443-
4 7.
84. Hanahn D, Weinberg RA. Hallmarks of cancer: the next generation. Cell.
2011;144:646-74.
85. Marquardt JU, Gomez-Quiroz L, Arreguin Camacho LO, Pinna F, Lee YH.
Curcumin eectively inhibits oncogenic NF-κB signaling and restrains
stemness features in liver cancer. J Hepatol. 2015;63:661-9.
86. Ushida J, Sugie S, Kawabata K, Pham QV, Tanaka T. Chemopreventive effect of
curcumin on N-nitrosomethylbenzylamine-induced esophageal carcinogenesis
in rats. Jpn J Cancer Res. 2000;91:893-8.
87. Perkins S, Verschoyle RD, Hill K, Parveen I, Threadgill MD. Chemopreventive
efficacy and pharmacokinetics of curcumin in the min/+ mouse, a model
of familial adenomatous polyposis. Cancer Epidemiol Biomarkers Prev.
2002;11:535-40.
88. Bower M, Aiyer H, Li Y, Martin R. Chemoprotective effects of curcumin in
esophageal epithelial cells exposed to bile acids. World J Gastroenterol.
201;16:4152-8.
89. Kuttan R, Sudheeran PC, Josph C. Turmeric and curcumin as topical agents in
cancer therapy. Tumori. 1987;73:29-31.
90. Cruz-Correa M, Shoskes DA, Sanchez P, Zhao R, Hylind LM. Combination
treatment with curcumin and quercetin of adenomas in familial adenomatous
polyposis. Clin Gastroenterol Hepatol. 2006;4:1035-8.
91. Horie S. Chemoprevention of prostate cancer: soy isoflavones and curcumin.
Korean J Urol. 2012;53:665-72.
92. Park W, Amin AR, Chen ZG, Shin DM. New perspectives of curcumin in cancer
prevention. Cancer Prev Res. 2013;6:387-400.
93. Gunes H, Gulen D, Mutlu R. Antibacterial effects of curcumin: an in vitro
minimum inhibitory concentration study. Toxicol Ind Health. 2016;32:246-50.
94. Teow SY, Liew K, Ali SA, Khoo AS, Peh SC. Antibacterial action of curcumin
against Staphylococcus aureus: a brief review. J Trop Med. 2016;2016:2853045.
95. Bomdya RS, Shah MU, Doshi YS, Shah VA, Khirade SP. Antibacterial activity of
curcumin (turmeric) against periopathogens - An in vitro evaluation. J Adv Clin
Res Insights. 2017;4:175-80.
96. Cheraghipour K, Marzban A, Ezatpour B, Khanizadeh S, Koshki J. Antiparasitic
properties of curcumin: a review. AIMS Agriculture and Food. 2019;4:1-18.
97. Rao R, Descamps O, John V, Bredesen DE. Ayurvedic medicinal plants for
Alzheimer’s disease: a review. Alzheimer’s Res Ther. 2012;4(3):22.
98. Dairam A, Limson JL, Walkins GM, Antunes E, Daya S. Curcuminoids, curcumin,
and demethoxycurcumin reduce lead-induced memory deficits in male Wistar
rats. J Agric Food Chem. 2007;55:1039-44.
99. Goozee KG, Shah TM, Sohrabi HR, Rainey-Smith SR, Brown B. Examining
the potential clinical value of curcumin in the prevention and diagnosis of
Alzheimer’s disease. Br J Nutr. 2016;115:449-65.
100. De Souza JA, Correia MCR. Biologia floral de Hedychium coronarium
Koen. (Zingiberaceae). (Floral biology of Hedychium coronarium Koen.
(Zingiberaceae)). (Rev Bras Horticult Ornament. 2007;13:21-30.
101. Meyer JY. Preliminary review of the invasive plants in the Pacific islands
(SPREP Member Countries). Pp. 85-114. In Sherley G (ed). Invasive species
in the Pacific: a technical review and draft regional strategy. South Pacific
Regional Environment Programme, Apia, Samoa. 2012.
102. Manish M. Current status of endangered medicinal plant Hedychium
coronarium and causes of population decline in the natural forests of Anuppur
and Dindori districts of Madhya Pradesh, India. Int Res J Bio Sci. 2013;2:1-6.
103. Bisht NS, Bhandari S. In vitro plant regeneration from seedling explants of
Hedychium coronarium J. Koenig. J Med Plants Res. 2012;6:5546-51.
104. Pachurekar P, Dixit AK. A review on pharmacognostical, phytochemical
and ethnomedicinal properties of Hedychium coronarium J. Koenig an
endangered medicine. Int J Chin Med. 2017;1:49-61.
105. Ray S, Sheikh M, Mishra S. Ethnomedicinal plants used by tribal of East
Nimar region, Madhya Pradesh. Indian J Trad Knowl. 2011;10:367-71.
106. Báez D, Pino JA, Morales D. Floral scent composition in Hedychium
coronarium J. Koenig analyzed by SPME. J Essent Oil Res. 2011;23:64-7.
107. Chimnoi N, Pisutjaroenpong S, Ngiwsara L, Dechtrirut D, Chokchaichamnankit
D. Labdane diterpenes from the rhizomes of Hedychium coronarium. Nat
Prod Res. 2008;22:1249-56.
108. Morikawa T, Matsuda H, Sakamoto Y, Ueda K, Yoshikawa M. New farnesane-
type sesquiterpenes, hedychiols a and b 8,9-diacetate, and inhibitors of
degranulation in RBL-2H3 cells from the rhizome of Hedychium coronarium.
Chem Pharm Bull (Tokyo). 2002;50:1045-9.
109. Dos Santos BCB, Barata LES, Marques FA, Baroni ACM, Karnos BAC.
Composition of leaf and rhizome essential oils of Hedychium coronarium
Koen. from Brazil. J Essent Oil Res. 2010;22:305-6.
110. Joy B, Rajan A, Abraham E. Antimicrobial activity and chemical composition
of essential oil from Hedychium coronarium. Phytother Res. 2007;21:439-43.
111. Aziz MA, Habib MR, Karim MR. Antibacterial and cytotoxic activities of
Hedychium coronarium J. Koenig. Res J Agric Biol Sci. 2009;5:969-72.
112. Ho JC. Antimicrobial, mosquito larvicidal and antioxidant properties of the
leaf and rhizome of Hedychium coronarium. J Chin Chem Soc. 2011;58:563-7.
113. Pandya CV, Jadeja AJ, Golakiya BA. Antifungal activity of crude extracts of
Hedychium coronarium. Int J Res Phytochem Pharmacol. 2014;4:4-6.
114. Kaomongkolgit R, Jamdee K, Wongnoi S, Chimnoi N, Techasakul S. Antifungal
activity of coronarin D against Candida albicans. Oral Surg Oral Med Oral
Pathol Oral Radiol. 2012;114:61-6.
115. Reuk-ngam N, Chimnoi N, Khunnawutmanotham N, Techasakul S.
Antimicrobial activity of coronarin D and its synergistic potential with
antibiotics. Biomed Res Int. 2014;581985.
116. Valadeau C, Pabon A, Deharo E, Albán-Castillo J, Estevez Y. Medicinal plants
from the Yanesha (Peru): evaluation of the leishmanicidal and antimalarial
activity of selected extracts. J Ethnopharmacol. 2019;123:413-22.
13 Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
11 7. Kiem PV, Kim Thuy NT, Tuan Anh HL, Nhiem NX, Minh CV. Chemical
constituents of the rhizomes of Hedychium coronarium and their inhibitory
effect on the pro-inflammatory cytokines production LPS-stimulated in bone
marrow-derived dendritic cells. Bioorg Med Chem Lett. 2011;21:7460-5.
118. Shrotriya S, Ali MS, Saha A, Bachar SC, Islam MS. Anti-inflammatory
and analgesic effects of Hedychium coronarium Koen. Pak J Pharm Sci.
2007;20:47-51.
119. Lu Y, Zhong CX, Wang L, Lu C, Li XL. Anti-inflammation activity and chemical
composition of flower essential oil from Hedychium coronarium. Afr J
Biotechnol. 2009;8:5373-7.
120. Dash PR, Nasrin M, Saha MR. Evaluation of analgesic and
neuropharmacological activities of methanolic rhizome extract of Hedychium
coronarium. Int J Pharma Sci Res. 2011;2(4):979984.
121. Suresh G, Reddy PP, Babu KS, Shaik TB, Kalivendi SV. Two new cytotoxic
labdane diterpenes from the rhizomes of Hedychium coronarium. Bioorg
Med Chem Lett. 2010;20:7544-8.
122. Zhan ZJ, Wen YT, Ren FY, Rao GW, Shan WG. Diterpenoids and a
diarylheptanoid from Hedychium coronarium with significant anti-angiogenic
and cytotoxic activities. Chem Biodivers. 2012;9:2754-60.
123. Ray A, Jena S, Dash B, Sahoo A, Kar B. Hedychium coronarium extract arrests
cell cycle progression, induces apoptosis, and impairs migration and invasion
in HeLa cervical cancer cells. Cancer Manag Res. 2019;11: 483-500.
124. Endringer DC, Taveira FSN, Kondratyuk TP, Pezzuto JM, Braga FC. Cancer
chemoprevention activity of labdane diterpenes from rhizomes of Hedychium
coronarium. Rev Bras Farmacogn. 2014;24:408-12.
125. Shankar D, Ved DK, Tandon V, Ramesh SR, Karim A, Singh P. Conserving
national resource - Need for national policy and national programme on
medicinal plant conservation, In: Pushpangadan P, Nair KSS (eds). Pp. 103-117.
Biodiversity in tropical forests: the Kerala scenario, Kerala State Committee
on Science, Technology and Environment, Kerala, India. 1997.
126. Dash PR, Raihan SZ, Ali MS. Ethnopharmacological investigation of the spice
Kaempferia galanga, 1st ed. Lambert Academic Publishing, Saarbrücken,
Germany. 2013.
127. Preetha TS, Hemanthakumar AS, Krishnan PN. A comprehensive review
of Kaempferia galanga L. (Zingiberaceae): a high sought medicinal plant in
tropical Asia. J Med Plants Stud. 2016;4:270-6.
128. Shetu HJ, Trisha KT, Sikta SA, Anwar R, Rashed SSB. Pharmacological
importance of Kaempferia galanga (Zingiberaceae): a mini review. Int J Res
Pharm Pharm Sci. 2018;3:32-9.
129. Tewtrakul S, Yuenyongsawad S, Kummee S, Latthya Atsawajaruwan L.
Chemical components and biological activities of volatile oil of Kaempferia
galanga Linn. Songklanakarin J Sci Technol. 2005;27:503-7.
130. Umar MI, Asmawi MZB, Sadikun A, Altaf R, Iqbal MA. Phytochemistry and
medicinal properties of Kaempferia galanga L. (Zingiberaceae) extracts. Afr J
Pharm Pharmacol. 2011;5:1638-47.
131. Parvez MAK, Mahboob HK, Zahurul I, Shek MH. Antimicrobial activities of
the petroleum ether, methanol and acetone extracts of Kaempferia galanga
L. rhizomes. J Life Earth Sci. 2005;1:25-9.
132. Kochuthressia KP, John Britto S, Jaseentha M, Raphae R. In vitro antimicrobial
evaluation of Kaempferia galanga L. rhizome extract. Am J Biotechnol Mol
Sci. 2011;2:1-5.
133. Dash PR, Nasrin M, Ali M. In vivo cytotoxic and in vitro antibacterial activities
of Kaempferia galanga. J Pharmacogn Phytochem. 2014;3:172-7.
134. Saraswati J, Septalita A, Bovita NA. Antibacterial effect of Kaempferia galanga
L extract on Lactobacillus acidophilusin vitro. Indon J Infect Dis. 2013;1:22-
8.
135. Elya B, Kusuma IM, Jufri M, Handayani R. Antibacterial tests against acne
in vitro, the physical stability and patch test using cream containing ethyl
pmethoxycinnamate extracted from Kaempferia galanga L. rhizome. Res J
Med Plants. 2016;10:426-34.
136. Yang YC, Park IK, Kim EH, Lee HS, Ahn YJ. Larvicidal activity of medicinal
plant extracts against Aedes aegypti, Ochlerotatus togoi, and Culex pipiens
pallens (Diptera: Culicidae). J Asia Pac Entomol. 2014;7:227-32.
137. Choochote W, Chaithong U, Kamsuk K, Jitpakdi A, Tippawangkosol P.
Repellent activity of selected essential oils against Aedes aegypti. Fitoterapia.
2007;78:359-64.
138. Dhandapani A, Kumar S, Kadarkarai M. Larvicidal, pupicidal and smoke toxicity
effect of Kaempferia galanga to the malarial vector, Anopheles stephensi.
Bioscan. 2011;6:329-33.
139. Sutthanont N, Choochote W, Tuetun B, Junkum A, Jitpakdi A. Chemical
composition and larvicidal activity of edible plant-derived essential oils
against the pyrethroid-susceptible and -resistant strains of Aedesaegypti
(Diptera: Culicidae). J Vector Ecol. 2011;35:106-15.
140. Liu XC, Liang Y, Shi WP, Liu QZ, Ligang Z. Repellent and insecticidal effects of
the essential oil of Kaempferia galanga rhizomes to Liposcelis bostrychophila
(Psocoptera: Liposcelidae). J Econ Entomol. 2014;107:1706-712.
141. Dash PR, Mou KM, Erina IN, Ripa FA, Al Masud KN. Study of anthelmintic and
insecticidal activities of different extracts of Kaempferia galanga. Int J Pharm
Sci Res. 2017;8:729-33.
142. Kanjanapothi D, Panthong A, Lertprasertsuke N, Taesotikul T, Rujjanawate
C. Toxicity of crude rhizome extract of Kaempferia galanga L. (Proh Hom). J
Ethnopharmacol. 2004;90:359-65.
143. Kim NJ, Byun SG, Cho JE, Chung K, Ahn YJ. Larvicidal activity of Kaempferia
galanga rhizome phenylpropanoids towards three mosquito species. Pest
Manag Sci. 2008;64:857-62.
144. Sulaiman MR, Zakaria ZA, Duad IA, Hidayat MT. Antinociceptive and anti-
inflammatory activities of the aqueous extract of Kaempferia galanga leaves
in animal models. J Nat Med. 2008;62:221-7.
145. Ridtitid W, Sae-Wong C, Reanmongkol W, Wongnawa M. Antinociceptive
activity of the methanolic extract of Kaempferia galanga Linn. in experimental
animals. J Ethnopharmacol. 2008;118:225-30.
146. Vittalrao AM, Shanbhag T, Kumari M, Bairy KL, Shenoy S. Evaluation of anti-
inflammatory and analgesic activities of alcoholic extract of Kaempferia
galanga in rats. Indian J Physiol Pharmacol. 2011;55:13-24.
147. Taslim NA, Djide MN, Rifai Y, Syahruddin AN, Rampo YR. Double-blind
randomized clinical trial of Kaempferia galanga L. extract as an anti-
inflammation (prostaglandin E2 and tumor necrosis factor-alpha) on
osteoarthritis. Asian J Pharm Clin Res. 2019;12:63-6.
148. Umar MI, Asmawi MZ, Sadikun A, Majid AM, Al-Suede FS. Ethyl-p-
methoxycinnamate isolated from Kaempferia galanga inhibits inflammation
by suppressing interleukin-1, tumor necrosis factor-α, and angiogenesis by
blocking endothelial functions. Clinics (Sao Paulo). 2014;69:134-44.
149. Huang L, Yagura T, Chen S. Sedative activity of hexane extract of Keampferia
galanga L. and its active compounds. J Ethnopharmacol. 2008;120:123-5.
150. Ali MS, Dash PR, Nasrin M. Study of sedative activity of different extracts
of Kaempferia galanga in Swiss albino mice. BMC Complement Altern Med.
2015;15:158.
151. Maas PJM. Renealmia (Zingiberaceae-Zingiberoideae), Costoideae (Additions)
(Zingiberaceae). Flora Neotropica. 1977;18:1-218.
152. Otero R, Núñez V, Jiménez SL, Fonnegra R, Osorio RG. Snakebites and
ethnobotany in the northwest region of Colombia: Part II: neutralization of
lethal and enzymatic effects of Bothrops atrox venom. J Ethnopharmacol.
2000;71:505-11.
153. Lans C, Harper T, Georges K, Bridgewater E. Medicinal and ethnoveterinary
remedies of hunters in Trinidad. BMC Complement Altern Med. 2011;1:10.
154. Ruysschaert S, Van Andel T, Van de Putte K, Van Damme P. Bathe the baby
to make it strong and healthy: plant use and child care among Saramaccan
Maroons in Suriname. J Ethnopharmacol. 2009;121:148-70.
155. Lognay G, Marlier M, Severin M, Haugruge E, Gibon V. On the characterization
of some terpenes from Renealmia alpinia Rottb. (Mass) oleoresin. Flavour
Fragr J. 1991;6:87-91.
156. Zhou BN, Baj NJ, Glass TE, Malone S, Werkhoven MC. Bioactive labdane
diterpenoids from Renealmia alpinia collected in the Suriname rainforest. J
Nat Prod. 1997;60:1287-93.
157. Yang SW, Zhou BN, Malone S, Werkhoven MC, van Troon F. A new labdane
diterpenoid from Renealmia alpinia collected in the Suriname rainforest. J Nat
Prod. 1999;62:1173-74.
158. Gilli C, Orlowska E, Kaiser D, Steyrer J, Rathgeb A. Diarylheptanoids,
flavonoids and other constituents from two neotropical Renealmia species
(Zingiberaceae). Biochem System Ecol. 2014;56:178-84.
159. Núñez V, Otero R, Barona J, Saldarriaga M, Osorio RG. Neutralization of the
edema-forming, defibrinating and coagulant effects of Bothrops asper venom
by extracts of plants used by healers in Colombia. Braz J Med Biol Res.
2014;37:969-77.
160. Patiño AC, Benjumea DM, Pereañez JA. Inhibition of venom serine proteinase
and metalloproteinase activities by Renealmia alpinia (Zingiberaceae) extracts:
comparison of wild and in vitro propagated plants. J Ethnopharmacol.
2013;149:590-6.
161. Gómez-Betancur I, Benjumea D, Patiño A, Jiménez N, Osorio E. Inhibition of
the toxic effects of Bothrops asper venom by pinostrobin, a flavanone isolated
from Renealmia alpinia (Rottb.) Maas. J Ethnopharmacol. 2014;155:1609-15.
162. Gómez-Betancur I, Pereañez JA, Patiño AC, Benjumea D. Inhibitory effect of
pinostrobin from Renealmia alpinia, on the enzymatic and biological activities
of a PLA2. Int J Biol Macromol. 2015;89:35-42.
163. Verpoorte R, Tsoi AT, van Doorne H, Svendsen AB. Medicinal plants
of Suriname. I. Antimicrobial activity of some medicinal plants. J
Ethnopharmacol. 1982;5:221-6.
164. Melo e Silva F, de Paula JE, Espindola LS. Evaluation of the antifungal potential
of Brazilian Cerrado medicinal plants. Mycoses. 2009;52:511-7.
14
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
165. Rangel ET. Atividade antiprotozoária, antifúngica e citotóxica de extratos de
plantas do bioma Cerrado, com ênfase em Leishmania (Leishmania) chagasi
(Antiprotozoal, antifungal, and cytotoxic activities of extracts from plants from
the Cerrado biome, with emphasis on Leishmania (Leishmania) chagasi). PhD
thesis, University of Brasilia, Brasília, Brazil. 2017.
166. De Mesquita ML, de Paula JE, Pessoa C, de Moraes MO, Costa Lotufo LV.
Cytotoxic activity of Brazilian Cerrado plants used in traditional medicine
against cancer cell lines. J Ethnopharmacol. 2009;123:439-45.
167. Uebelhack R, Franke L, Schewe HL. Inhibition of platelet MAO-B by kava
pyrone-enriched extract from Piper methysticum Forster (kava-kava).
Pharmacopsychiatry. 1998;31:187-92.
168. Baum SS, Hill R, Rommelspacher H. Effect of kava extract and individual
kavapyrones on neurotransmitter levels in the nucleus accumbens of rats.
Prog Neuropsychopharmacol Biol Psychiatry. 1998;22:1105-20.
169. Chaurasiya ND, León F, Ding Y, Gómez-Betancur I, Benjumea D. Interactions
of desmethoxyyangonin, a secondary metabolite from Renealmia alpinia,
with human monoamine oxidase-A and oxidase-B. Evid Based Complement
Alternat Med. 2017:4018724.
170. Tzeng YM, Lee MJ. Neuroprotective properties of kavalactones. Neural
Regen Res. 2015;10:875-7.
171. Bortolato M, Chen K, Shih JC. Monoamine oxidase inactivation: from
pathophysiology to therapeutics. Adv Drug Deliv Rev. 2008;60:1527-33.
172. Svob Strac D, Pivac N, Smolders IJ, Fogel WA, De Deurwaerdere P.
Monoaminergic mechanisms in epilepsy may offer innovative therapeutic
opportunity for monoaminergic multi-target drugs. Front Neurosci.
2016;10:492.
173. Langner E, Greifenberg S, Gruenwald J. Ginger: history and use. Adv Ther.
1998;15:25-44.
174. IndexBox. Global Ginger Market 2019 - U.S. Imports Increases Robustly,
Turning The Country Into The Most Promising Market. 2019. https://www.
globaltrademag.com/global-trade-daily/global-ginger-market-2019-u-s-
imports-increases-robustly-turning-the-country-into-the-most-promising-
market/.
175. Lawrence BM. Progress in essential oils. Perfumer and Flavorist. 1997;22:71-
83.
176. Sandeep S.Commentary on therapeutic role of ginger (Zingiber officinale) as
medicine for the whole world. Int J Pharmacogn Chinese Med. 2017;1:000102.
177. Imtiyaz S, Rahman K, Sultana A, Tariq M, Chaudhary SS. Zingiber officinale
Rosc.: a traditional herb with medicinal properties. TANG. 2013;3:e263.
178. Mekuriya W, Mekibib B. Review on the medicinal values of ginger for human
and animal ailments. J Vet Sci Technol. 2010;9:519.
179. Bao L, Deng A, Li Z, Du G, Qin H. Chemical constituents of rhizomes of
Zingiber officinale. Zhongguo Zhong Yao Za Zhi. 2010;35:598-601.
180. Jiang H, Solyom AM, Timmermann BN, Gang DR. Characterization of
gingerol-related compounds in ginger rhizome (Zingiber oficinale Rosc.)
by high-performance liquid chromatography/electrospray ionization mass
spectrometry. Rapid Commun Mass Spectrom. 2005;19:2957-64.
181. An K, Zhao D, Wang Z, Wu J, Xu Y, Xiao G. Comparison of different drying
methods on Chinese ginger (Zingiber officinale Roscoe): changes in volatiles,
chemical profile, antioxidant properties, and microstructure. Food Chem.
2016;197:1292-300.
182. Yamahara J, Rong HQ, Naitoh Y, Kitani T, Fujimura H. Inhibition of cytotoxic
drug-induced vomiting in suncus by a ginger constituent. J Ethnopharmacol.
1989;27:353-5.
183. Sharma SS, Kochupillai V, Gupta SK, Seth SD, Gupta YK. Antiemetic efficacy
of ginger (Zingiber officinale) against cisplatin-induced emesis in dogs. J
Ethnopharmacol. 1997;57:93-6.
184. Sharma SS, Gupta YK. Reversal of cisplatin-induced delay in gastric emptying
in rats by ginger. J Ethnopharmacol. 1998;62:49-55.
185. Adbel-Aziz H, Windeck T, Ploch M, Verspohl EJ. Mode of action of gingerols
and shogaols on 5-HT3 receptors: binding studies, cation uptake by the
receptor channel and contraction of isolated guinea-pig ileum. Eur J
Pharmacol. 2006;530:136-43.
186. Sontakke S, Thawani V, Naik MS. Ginger as an antiemetic in nausea and
vomiting induced by chemotherapy: a randomized, cross-over, double-blind
study. Indian J Pharmacol. 2003;35:32-6.
187. Fahimia F, Khodadad K, Amini S, Naghibi F, Salamzadeh J. Evaluating the
effect of Zingiber officinalis on nausea and vomiting in patients receiving
cisplatin-based regimens. Iran J Pharm Res. 2011;10:379-84.
188. Pertz HH, Lehmann J, Roth-Ehrang R, Elz S. Effects of ginger constituents on
the gastrointestinal tract: role of cholinergic M3 and serotonergic 5-HT3 and
5-HT4 receptors. Planta Med. 2011;77:973-8.
189. Walstab J, Krüger D, Stark T, Hofmann T, Demir IE. Ginger and its pungent
constituents non-competitively inhibit activation of human recombinant
and native 5-HT3 receptors of enteric neurons. Neurogastroenterol Motil.
2013;25:439-47.
190. Lete I, Allué J. The effectiveness of ginger in the prevention of nausea
and vomiting during pregnancy and chemotherapy. Integr Med Insights.
2016;11:11-7.
191. Malu SP, Obochi GO, Tawo EN, Nyong BE. Antibacterial activity and medicinal
properties of ginger (Zingiber officinale). Global J Pure Appl Sci. 2009;15:365-
8.
192. Atai Z, Atapour M, Mohseni M. Inhibitory effect of ginger extract on Candida
albicans. Am J Appl Sci. 2009;6:1067-9.
193. Gao D, Zhang Y. Comparative antibacterial activities of crude polysaccharides
and flavonoids from Zingiber officinale and processed ginger. Pharmacogn J.
2010;2:41-4.
194. 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;22:1035-9.
195. Iqbal Z, Nadeem QK, Khan MN, Akhtar MS, Waraich FN. In vitro anthelmintic
activity of Allium sativum, Zingiber officinale, Curcurbita mexicana and Ficus
religiosa. Int J Agric Biol. 2001;3:454-7.
196. Dubey RD, Verma S, Rane D, Wani VK, Pandey AK. Comparative studies of
anthelmintic activity of Zingiber officinale and Cassia tora. Int J Chem Pharm
Sci. 2010;1:1-4.
197. Lin RJ, Chen CY, Lee JD, Lu CM, Chung LY. Larvicidal constituents of Zingiber
officinale (ginger) against Anisakis simplex. Planta Med. 2010;76:1852-58.
198. Raji Y, Udoh US, Oluwadara OO, Akinsomisoye OS, Awobajo O. Anti-
inflammatory and analgesic properties of the rhizome extract of Zingiber
officinale. Afr J Biomed Res. 2002;5:121-4.
199. Yong-liang J, Jun-ming Z, Lin-hui Z, Bao-shan S, Meng-jing B. Analgesic and
anti-inflammatory effects of ginger oil. Chin Herb Med 2011;3:150-5.
200. Ezzat SM, Ezzat MI, Okba MM, Menze ET, Abdel-Naim AB. The hidden
mechanism beyond ginger (Zingiber officinale Rosc.) potent in vivo and in
vitro anti-inflammatory activity. J Ethnopharmacol. 2018;214: 113-23.
201. Tjendraputra E, Tran VH, Liu-Brennan D, Roufogalis BD, Duke CC. Effect of
ginger constituents and synthetic analogues on cyclooxygenase-2 enzyme in
intact cells. Bioorg Chem. 2001;29:156-63.
202. Jolad SD, Lantz RC, Solyom AM, Chen GJ, Bates RB. Fresh organically grown
ginger (Zingiber officinale): composition and effects on LPS-induced PGE2
production, Phytochemistry. 2004;65:1937-54.
203. Ficker CE, Smith ML, Susiarti S, Leaman DJ, Irawati C. Inhibition of human
pathogenic fungi by members of Zingiberaceae used by the Kenyah
(Indonesian Borneo). J Ethnopharmacol. 2003;85:289-93.
204. Willenbacher E, Khan SZ, Mujica SCA, Trapani D, Hussain S. Curcumin: new
insights into an ancient ingredient against cancer. Int J Mol Sci. 2019;20(8):pii:
E1808.
15 Pharmacognosy Journal, Vol 11, Issue 6(Suppl), Nov-Dec, 2019
Mans, et al.: Phytochemical and Pharmacological Support for the Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature
ABOUT AUTHORS
GRAPHICAL ABSTRACT
Dennis RA Mans: Dennis Mans is Full Professor of Pharmacology at the Faculty of Medical Sciences of the
Anton de Kom University of Suriname, Paramaribo, Suriname. He holds the academic chair in pharmacognosy
research. He has more than thirty years of teaching and research experience, particularly in the area of medicinal
plants. He has published more than one hundred full papers in international and national journals.
Jennifer Pawirodihardjo: Jennifer Pawirodihardjo has a teaching degree in Chemistry and is working for five
years as a chemical analyst at the Faculty of Medical Sciences, Department of Pharmacology, Anton de Kom
University of Suriname, Paramaribo, Suriname.
Priscilla Friperson: Priscilla Friperson has studied Higher Laboratory Sciences at the Polytechnic College and is
working as a chemical analyst at the Faculty of Medical Sciences, Department of Pharmacology, Anton de Kom
University of Suriname, Paramaribo, Suriname.
Meryll Djotaroeno: Meryll Djotaroeno has a BTech degree in Higher Laboratory Sciences and is working as a
chemical analyst at the Faculty of Medical Sciences, Department of Pharmacology, Anton de Kom University of
Suriname, Paramaribo, Suriname.
Cite this article: Mans DRA, Djotaroeno M, Friperson P, Pawirodihardjo J. Phytochemical and Pharmacological Support for the
Traditional Uses of Zingiberacea Species in Suriname - A Review of the Literature. Pharmacog J. 2019;11(6)Suppl:
... 2,3 Several species of the Zingiberaceae family (eg, Alpinia galanga, Curcuma longa, Zingiber officinale, Hedychium coronarium, Kaempferia galanga, etc) were well-known for their effects in treating various diseases, such as digestive disorders, hypertension, diabetes mellitus (DM), infection, inflammation, antioxidant, anti-microbial, anticancer, antiparasitic, analgesic, hepatoprotective activities, etc, as 1 reported in previous studies. 3,4 In terms of chemical composition profiles, some species (eg, A. galanga, C. longa, Curcuma thorelii, Curcuma rhabdota, Curcuma petiolata, Distichochlamys citrea, Z. officinale, H. coronarium, K. galanga, etc) were previously reported to be rich in essential oils. Moreover, various chemical components, such as flavonoids, tannins, resins, phenols, volatile fatty acids, etc, were isolated from their different parts. ...
... Moreover, various chemical components, such as flavonoids, tannins, resins, phenols, volatile fatty acids, etc, were isolated from their different parts. [4][5][6][7] Essential oils (EOs) are volatile substances with low molecular weight and various biological activities that have been known. 8 As a source of natural chemicals for treating illnesses including diabetes, anti-cancer, hepatoprotective, antioxidant, and antimicrobial activities, 4 EOs are currently gaining popularity as a natural substitute in the health industry. ...
... [4][5][6][7] Essential oils (EOs) are volatile substances with low molecular weight and various biological activities that have been known. 8 As a source of natural chemicals for treating illnesses including diabetes, anti-cancer, hepatoprotective, antioxidant, and antimicrobial activities, 4 EOs are currently gaining popularity as a natural substitute in the health industry. EOs are therefore extremely important in the pharmaceutical, food, agricultural, cosmetic, and health industries. ...
Article
Full-text available
The ginger family (Zingiberaceae) is one of the most diverse and abundant families in the plant kingdom in terms of number of species. In terms of chemistry, this is a family of plants containing much essential oil with mainly monoterpene and sesquiterpene derivatives, which has been widely used in medicine, food, and flavor industries. In our series of studies on ginger family plants, the two new species including Wurfbainia schmidtii (K. Schum.) Škorničk. & A.D. Poulsen and Zingiber atroporphyreus Škorničk. & Q.B. Nguyen has been investigated for the first time. This study aimed to identify the chemical compositions of essential oils extracted from the rhizomes and leaves of these two species using gas chromatography-mass spectrometry (GC/MS) analysis and evaluate their hypoglycemic effects through the α-glucosidase and α-amylase inhibitory assays. In terms of chemical composition, in general, the main compounds appearing in the essential oils of both leaves and rhizomes of each species are similar but they are differences in the percentage of the main components, for example, 1,8-cineole is a main component in leaves whereas fenchyl acetate is the main one in rhizomes of W. schmidtii. Several distinctive compounds, in comparison with other members in the genus Zingiber, namely β-pinene, α-pinene, β-elemene, and sabinene, were found in the essential oil of Z. atroporphyreus. Regarding hypoglycemic effects, the essential oils from the two species tested possessed weak α-glucosidase and α-amylase inhibitory effects. For the first time, this study was designed to investigate the chemical composition as well as the hypoglycemic effect of essential oils distilled from the rhizomes and leaves of two species W. schmidtii and Z. atroporphyreus have been reported, serving as a premise for further systematic research on their phytochemistry and pharmacological effects being conducted.
... Aromatic ginger (Kaempferia galanga L.) is a Kaempheria species widely used in traditional medicine worldwide [11]. The rhizome of K. galanga contains several pharmacologically active compounds, including phenylpropanoids, flavanols, and terpenoids [12]. With antimicrobial activity [12,13], antioxidants [14], and anti-inflammatory activities [14], K. galanga rhizome may be exploited as a phytogenic feed additive for broilers. ...
... The rhizome of K. galanga contains several pharmacologically active compounds, including phenylpropanoids, flavanols, and terpenoids [12]. With antimicrobial activity [12,13], antioxidants [14], and anti-inflammatory activities [14], K. galanga rhizome may be exploited as a phytogenic feed additive for broilers. According to a recent study, fermentation enhances the bioavailability of bioactive components (such as polyphenols) and the synthesis of health-promoting products [15]. ...
... Considering the potential pathogenicity of lactose-negative Enterobacteriaceae, the decrease in such a bacterial population may be beneficial for broilers' health condition. The antibacterial properties of probiotics, prebiotics, and herbal products [1,5,12] in FERMIX were most likely to decrease the lactose-negative Enterobacteriaceae proliferation in the ileum of broiler in this investigation. ...
Article
Full-text available
Background and Aim: Natural feed additives are important for broiler sustainability during the post-antibiotic era. This study aimed to evaluate the possible effects of the dietary supplementation of Lactobacillus casei-fermented mixture of red rice and aromatic ginger (FERMIX) on growth, blood profiles, intestinal bacterial content, and morphological measurements of broilers. Materials and Methods: Two hundred broiler chicks were allotted to four groups with five replications, including CONTROL (chicks provided with basal feed), FERMIX025 (basal feed supplemented with 0.25% FERMIX), FERMIX050 (0.50% FERMIX), and FERMIX100 (1.00% FERMIX). FERMIX is an anaerobic fermentation product from a mixture of red rice flour and aromatic ginger using L. casei. Blood, organs, digesta, and carcass were collected on day 35. Results: Final body weight, weight gain, feed intake, and feed conversion ratio did not differ (p>0.05) across treatments. FERMIX at 0.5% increased (p
... Species of the Curcuma have been used in India for over 4,000 years, being initially exploited for its nutritional value, but its use has spread due to the identification of pharmacological activities, including antioxidant, anti-inflammatory, hypoglycaemic, antifungal, antimicrobial and healing, as well as action on the respiratory, nervous and digestive systems [1][2][3][4][5][6]. ...
Article
Curcuma longa L. leaves are a residual material of the rhizome harvest and arouse interest in the investigation of their properties. This study aimed to extract and characterise the essential oil (EO) from C. longa leaves and compare the feasibility of delivering of the EO and its major compound in nanoemulsions for the further evaluation of in vitro anti-inflammatory activity and preliminary safety profile. The EO was obtained by hydrodistillation and analysed by gas chromatography (GC-MS). The nanoemulsions were prepared in a high-pressure homogeniser and characterised. The in vitro anti-inflammatory activity was measured using an LPS-stimulated macrophage model (RAW 264.7). The preliminary safety profile was evaluated by cell culture (HaCaT and MRC-5) and by the hen’s egg-chorioallantoic membrane test (HET-CAM). In the CG-MS analysis, 10 compounds were identified: α-phellandrene (49.93%), terpinolene (27.10%), and 1,8-cineole (15.41%) being the main compounds. All nanoemulsions showed droplet size < 300 nm, polydispersity index < 0.2, and zeta potential <-30 mV, indicative of monodisperse formulations without the tendency for droplet aggregation. The nanoemulsions showed an α-phellandrene content greater than 91.00%. The assessment of the irritant potential by HET-CAM assay shows no irritating effects on the membrane. In the cytotoxicity evaluation, no significant alterations were observed in HaCaT and MRC-5 cells. Regarding the in vitro anti-inflammatory activity, the inhibition of NO production by the EO nanoemulsion was evidenced with a low cytotoxicity rate for α-phellandrene concentrations below 125 µg/mL. These results show the potential of the EO extracted from leaves, which are a waste product from rhizome harvesting.
... Monoterpenes such as phellandrene, camphene, cineole, linalool, limonene, citral, geraniol, citronellol, borneol, and sesquiterpenes such as α-zingiberene, ar-curcumene, β-bisabolene, β-sesquiphellandrene, zingiberol, and zingiberenol are the main constituents of ginger oil [17]. It also contains anticancer compounds such as terpenoids, phenylpropanoids, flavonoids, and sesquiterpenes [18]. Bioactive chemicals included in this oil, such as 6-gingerol and zerumbone, have been shown to cause apoptosis in cancer cells [19]. ...
Article
Full-text available
Cancer is a disease that is characterized by uncontrolled cell proliferation. Breast cancer is the most prevalent cancer among women. Ginger oil is a natural cancer fighter and anti-oxidant. However, the minimal absorption of ginger oil from the gastrointestinal tract accounts for its limited medicinal efficacy. The present study was designed to evaluate the efficacy of a nanoemulsion preparation of ginger oil on its oral bioavailability and in vivo anti-cancer efficacy. Ginger oil nanoemulsion was prepared by a high-pressure homogenization technique using different surfactants (Tween 20, 40, and 80). The prepared formulations were evaluated for droplet size, polydispersity index (PDI), zeta potential (ZP), pH, viscosity, and stability by calculating the creaming index percentage. The best formulation was evaluated for shape by TEM. The antitumor activity of the best nano-formulation was determined in comparison with the free oil using the in vivo Ehrlich solid tumor (EST) model. The prepared ginger oil nanoemulsion formulations exhibited acceptable droplet size in the range from 56.67 ± 3.10 nm to 357.17 ± 3.62 nm. A PDI of less than 0.5 indicates the homogeneity of size distribution. The oil globules possessed a negative charge ranging from −12.33 ± 1.01 to −39.33 ± 0.96 mV. The pH and viscosity were in the acceptable range. The TEM image of the best formulation appeared to be spherical with a small size. The ginger oil nanoemulsion reduced in vivo tumor volume and weight, extended animals’ life span, and ameliorated liver and kidney function in EST-bearing mice. These effects were superior to using free ginger oil. Collectively, the present study demonstrated that the ginger oil nanoemulsion improved oral absorption with a subsequent enhancement of its anti-proliferative efficacy in vivo, suggesting a nano-formulation of ginger oil for better therapeutic outcomes in breast cancer patients.
... Zingiberaceae is a large, fragrant pantropical family consisting of 1,600 species divided among about 50 genera (Christenhusz and Byng, 2016). The plants are distributed throughout tropical Africa, Asia, and the Americas (Ra Mans et al., 2019). Zingiberaceae plants contain many bioactive terpenoids, flavonoids, and polyphenols that are economically important as traditional medicines, spices, and cosmetics. ...
Article
Full-text available
Introduction Alpinia oxyphylla Miquel (A. oxyphylla), one of the “Four Famous South Medicines” in China, is an essential understory cash crop that is planted widely in the Hainan, Guangdong, Guangxi, and Fujian provinces. Particularly, A. oxyphylla from Hainan province is highly valued as the best national product for geo-herbalism and is an important indicator of traditional Chinese medicine efficacy. However, the molecular mechanism underlying the formation of its quality remains unspecified. Methods To this end, we employed a multi-omics approach to investigate the authentic quality formation of A. oxyphylla. Results In this study, we present a high-quality chromosome-level genome assembly of A. oxyphylla, with contig N50 of 76.96 Mb and a size of approximately 2.08Gb. A total of 38,178 genes were annotated, and the long terminal repeats were found to have a high frequency of 61.70%. Phylogenetic analysis demonstrated a recent whole-genome duplication event (WGD), which occurred before A. oxyphylla’s divergence from W. villosa (~14 Mya) and is shared by other species from the Zingiberaceae family (Ks, ~0.3; 4DTv, ~0.125). Further, 17 regions from four provinces were comprehensively assessed for their metabolite content, and the quality of these four regions varied significantly. Finally, genomic, metabolic, and transcriptomic analyses undertaken on these regions revealed that the content of nootkatone in Hainan was significantly different from that in other provinces. Discussion Overall, our findings provide novel insights into germplasm conservation, geo-herbalism evaluation, and functional genomic research for the medicinal plant A. oxyphylla.
... However, the greatest problem is that compost fermentation does not proceed smoothly. This is because the branches and leaves of ginger contain volatile oils, phenols, pyrones, flavonoids, diterpenes, and other compounds with strong antibacterial effects [7,8]. Therefore, there is a need to construct a bacterial agent that can quickly detoxify the branches and leaves of Yanshan ginger for efficient fermentation and production of bio-organic fertilizer [9,10]. ...
Article
Full-text available
Background: Yanshan ginger (Alpinia zerumbet) is a perennial herb used as a medicine and spice, and is beneficial for soil and water conservation in karst areas. Given the widespread utilization of Yanshan ginger in China and continuing expansion of the planting area, disposal of waste materials is problematic. The branches and leaves of Yanshan ginger contain a variety of potent antibacterial compounds, such as volatile oils, phenols, and diterpenoids, which hinder their rapid degradation by microorganisms. In this study, we screened and constructed a composite microbial system to provide a technical reference for production of organic fertilizer from the branches and leaves of Yanshan ginger. Methods: A composite microbial system, "YanShan Jun" (YSJ), was developed by screening for efficient detoxification and degradation of the branches and leaves of Yanshan ginger. High-throughput sequencing technology was used to investigate the stability and diversity of YSJ subcultures. The culture conditions for YSJ were optimized by sequential single-factor experiments and response surface analysis. Yanshan ginger leaves and branches were inoculated with YSJ to study its effects on composting efficiency. Results: The microbial composition of YSJ was stable and rich in diversity through continuous subculture. Through response surface analysis, the optimized culture conditions for YSJ were determined as follows: peptone 8.0 g/L, sodium chloride 9.0 g/L, calcium carbonate 5.2 g/L, yeast powder 1.6 g/L, cultivation temperature 56.1°C, and culture duration 6 d. Under these conditions, the degradation rate of Yanshan ginger was 58.32%, which was 14.22% higher than that before optimization. The ability of YSJ to degrade the antibacterial compounds of ginger after optimization was significantly enhanced. Inoculation of Yanshan ginger compost with YSJ increased the fermentation temperature, prolonged the high-temperature period, and reduced the water content and pH of the compost in the early stage. Conclusions: Inoculation of plant compost with YSJ bacteria improves the nutritional environment of the compost, promotes the composting reaction, promotes the rapid formation of a strong indigenous microflora, forms a beneficial microecological environment, and increases the composting efficiency. This study provides a theoretical basis for practical application of YSJ for organic fertilizer production from Yanshan ginger.
Article
Neurodegenerative diseases encompass a collection of neurological disorders originating from the progressive degeneration of neurons, resulting in the dysfunction of neurons. Unfortunately, effective therapeutic interventions for these diseases are presently lacking. Copper (Cu), a crucial trace element within the human body, assumes a pivotal role in various biological metabolic processes, including energy metabolism, antioxidant defense, and neurotransmission. These processes are vital for the sustenance, growth, and development of organisms. Mounting evidence suggests that disrupted copper homeostasis contributes to numerous age-related neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), Menkes disease (MD), prion diseases, and multiple sclerosis (MS). This comprehensive review investigates the connection between the imbalance of copper homeostasis and neurodegenerative diseases, summarizing pertinent drugs and therapies that ameliorate neuropathological changes, motor deficits, and cognitive impairments in these conditions through the modulation of copper metabolism. These interventions include Metal-Protein Attenuating Compounds (MPACs), copper chelators, copper supplements, and zinc salts. Moreover, this review highlights the potential of active compounds derived from natural plant medicines to enhance neurodegenerative disease outcomes by regulating copper homeostasis. Among these compounds, polyphenols are particularly abundant. Consequently, this review holds significant implications for the future development of innovative drugs targeting the treatment of neurodegenerative diseases.
Article
Background Traumatic brain injury (TBI) is a worldwide problem. Almost about sixty-nine million people sustain TBI each year all over the world. Repetitive TBI linked with increased risk of neurodegenerative disorder such as Parkinson, Alzheimer, traumatic encephalopathy. TBI is characterized by primary and secondary injury and exerts a severe impact on cognitive, behavioral, psychological and other health problem. There were various proposed mechanism to understand complex pathophysiology of TBI but still there is a need to explore more about TBI pathophysiology. There are drugs present for the treatment of TBI in the market but there is still need of more drugs to develop for better and effective treatment of TBI,because no single drug is available which reduces the further progression of this injury. Objective The main aim and objective of structuring this manuscript is to design, develop and gather detailed data regarding about the pathophysiology of TBI and role of medicinal plants in its treatment. Method This study is a systematic review conducted between January 1995 to June 2021 in which a consultation of scientific articles from indexed periodicals was carried out in Science Direct, United States National Library of Medicine (Pubmed), Google Scholar, Elsvier, Springer and Bentham. Results A total of 54 studies were analyzed, on the basis of literature survey in the research area of TBI. Conclusion Recent studies have shown the potential of medicinal plants and their chemical constituents against TBI therefore, this review targets the detailed information about the pathophysiology of TBI and role of medicinal plants in its treatment.
Article
Full-text available
Amomum tsao-ko is an economically important spice plant in the ginger family (Zingiberaceous). The dried ripe fruit has been widely used as spice and medicine in Southeast Asia due to its distinct flavor metabolites. However, there is little genomic information available to understand the biosynthesis of its characteristic flavor compounds. Here, we present a high-quality chromosome-level genome of A. tsao-ko with a total length of 2.08 Gb assembled into 24 chromosomes. Potential relationships between genetic variation and chemical constituents were analyzed by genome-wide association study (GWAS) of 119 representative A. tsao-ko specimens in China. Metabolome and transcriptome correlation analysis of different plant organs and fruit developmental stages revealed the proposed biosynthesis of the characteristic bicyclononane aldehydes and aromatic metabolites in A. tsao-ko fruit. Transcription factors of 20 families may be involved in the regulatory network of terpenoids. This study provides genomic and chemical insights into the biosynthesis of characteristic aroma and flavor constituents, which can be used to improve the quality of A. tsao-ko as food and medicine.
Article
Full-text available
Crude extracts of flowers of Hedychium coronarium were examined for their antifungal activity against three fungi viz. Alternaria, Fusarium and Aspergillus flavus. Antifungal activity of flowers in terms of Minimal Fungicidal Concentration [MFC](µg/mL) ranged between 200-1000 µg/mL. The extracts performed as good as or even better than the standard drugs like Nystatin and Greseofulvin. Highest antifungal activity of flower extract is found to be the most effective against Alternaria. This works seems to be a promising work in the recent existing problem of Glucoma as H. coronarium extracts are having flavanoids which are key factors in curing the disease.
Article
Full-text available
Introduction: Lactobacillus acidophilus is one of the bacteria causes dental caries. The previous study has shown that Kaempferia galanga extract has a potential to inhibit the growth of Lactobacillus acidophilus.Objective: To determine the antibacterial effect of Kaempferia galanga extract to Lactobacillus acidophilus.Methods:Kaempferia galanga is extracted in 3 different solvents:dichlormethane, ethanol, and aquades. For each solvent, 0.2 μl Kaempferia galanga extractdroped into 6 mm steril paper dics. 0.1 ml Lactobacillus acidophilus inoculated on MRS agar. Each disc contains extract were impragnated into the agar media, then incubated at 370C for 24 hours, and inhibition zone measured.Results: Mean scores of Kaempferia galanga extract in 3 different solvents are: Kaempferia galanga (dichlormethane) is 1.6400; Kaempferia galanga (ethanol) is 1.7440; Kaempferia galanga extract is 1.6600; boiled Kaempferia galanga is 1.7000. Using Mann-Whitney Test, the results are: negative controls have no inhibition effect on Lactobacillus acidophilus compaired to Kaempferia galanga extract, comparation of those 4 Kaempferia galangal treatments shows no significant difference, those 4 Kaempferia galanga treatments compaired to erythromycin antibacterial effect shows significant difference, otherwise 4 Kaempferia galanga treatments compaired to penicillin shows no significant difference except Kaempferia galanga (ethanol).Conclusions: Kaempferia galanga extract can kill Lactobacillus acidophilus. Inhibition effect of Kaempferia galanga extract has no significant difference to penicillin but lower inhibition effect than erythromycin. The Kaempferia galanga extracts showed better antibacterial activity than penicillin.
Article
Full-text available
Objective: The purpose of this study was to compare the effectiveness of the rhizome extract against inflammatory markers such as tumor necrosis factor (TNF)-alpha and prostaglandin E2 (PGE2) with meloxicam in patients with osteoarthritis at knee. Materials and Methods: This study designed two phases as follows: The preparation of rhizome extract in a capsule dosage form which was then followed by double-blind clinical trials, randomized controlled, pre- and post-test design (18 cases) using the WOMAC scores for pain, to markers of inflammation using an enzyme-linked immunosorbent assay method. Results: The results did not reveal any significant differences in pain scores, stiffnesses, and physical function impairment between the intervention (the rhizome extract) and control groups (meloxicam), it either did not reveal any significant differences in the parameters of inflammatory marker TNF alpha and PGE2 among the groups. Conclusion: The rhizome extract had the same effect on pain, stiffness, physical interference as meloxicam, and on PGE2 as well as on the inflammatory marker TNF-alpha. Therefore, this extract can be used as an alternative herbal medicine for osteoarthritis.
Article
Full-text available
Cancer patients frequently use complementary medicine. Curcumin (CUR) and its derivates (from the extract of Curcuma longa L.) represent some of the most frequently used ones, having a long history in traditional Asian medicine. CUR was demonstrated, both in vitro and in vivo, to have significant anti-inflammatory effects, thus potentially counteracting cancer-promoting inflammation, which is a hallmark of cancer. CUR modulate a plethora of signaling pathways in cancer cells, comprising the NF-κB (nuclear factor k-light-chain-enhancer of activated B cells), the JAK/STAT (Janus-Kinase/Signal Transducers and Activators of Transcription), and the TGF-β (transforming growth factor-β) pathways. Furthermore, CUR confers properties of electron receptors, which destabilize radical oxygen species (ROS), explaining its antioxidant and anti-apopototic effects. Although CUR has a low bioavailability, its role in advanced cancer treatment and supportive care was addressed in numerous clinical trials. After promising results in phase I–II trials, multiple phase III trials in different indications are currently under way to test for direct anti-cancer effects. In addition, CUR exerts beneficial effects on cancer treatment-related neurotoxcity, cardiotoxicity, nephrotoxicity, hemato-toxicity, and others. More efficient galenic formulations are tested to optimze CUR’s usability in cancer treatment. This review should provide a comprehensive overview of basic science, and pre-clinical and clinical data on CUR in the field of oncology.
Article
Full-text available
Background Hedychium coronarium Koen. (Zingiberaceae) is traditionally used as medicine in countries such as India, China, and Vietnam for treatment of various ailments including cancer. However, in spite of its implied significance in cancer treatment regimes, there are no reports so far involving the anticancerous attributes of H, coronarium ethanol extract (HCEE) on cancer cells and a more comprehensive study on its mechanism is still lacking. Materials and methods The cytotoxicity of HCEE was evaluated by MTT and clonogenic survival assay. Annexin V/propidium iodide (PI), Hoechst 33342 staining, and TUNEL assay were performed to detect apoptosis. Cell cycle analysis was performed using PI staining. JC-1 and 2′,7′-dichlorodihydrofluorescein diacetate assay were used to check the levels of MMP and ROS, respectively. Western blot analysis was carried out to measure the expression levels of proteins. Migration and invasion activity were assessed by wound healing and Transwell membrane assay, respectively. Results Antiproliferative effect of HCEE was investigated in various cancerous and normal cell lines. Among these, HCEE significantly inhibited the survival of HeLa cells without affecting the viability of normal human umbilical vein endothelial cells. Annexin V/PI, Hoechst staining, and TUNEL assay showed HCEE induced apoptosis in HeLa cells in a dose-dependent manner. HCEE promoted cell cycle arrest at G1 phase in HeLa cells by upregulating the levels of p53 and p21 and downregulating the levels of cyclin D1, CDK-4, and CDK-6. Moreover, HCEE treatment upregulated the expression of Bax and downregulated the expression of Bcl-2. Additionally, HCEE activated the caspase cascade by increasing the activities of caspase-9, caspase-8, and caspase-3. The expression levels of Fas ligand and Fas were also upregulated. Further, HCEE inhibited the migratory potential of HeLa cells by downregulating MMP-2 and MMP-9 expression levels. Conclusion Our results indicate H. coronarium exerts antiproliferative and apoptotic effects against HeLa cells, and therefore may be used for treatment against cervical cancer.
Article
Full-text available
Medicinal plants are known as one of the most effective approaches to overcome parasitic infections, which has been used several years ago. Curcuma longa (C. longa) that is commonly used as a food additive in cooking. Curcumin as the major compound extracted from C. longa serves for the various therapeutic and preventive purposes. The present review paper is aimed to investigate the antiparasitic effects of curcumin reported in recent years. The data was collected from several databanks including ISI, Google Scholar, Pubmed, Scopus, and SID (Scientific Information Database, Iran). After a primary study of the retrieved data, the most relevant literature was subjectively classified based on the type of parasite. Then, the effect of curcumin treatment on various parasites was assessed regarding the kind of parasite. According to the results, curcumin manifested a high potential to serve as an effective drug against various parasites. Therefore, further studies in detail on curcumin might offer a new perspective that helps to design efficient formulations for hampering the infections caused by parasites in both human and animals.
Article
Full-text available
Kaempferia galanga L. belonging to the family Zingiberaceae is an endangered medicinal plant with potent medicinal activities. The leaves, rhizome and root tubers of the plant possess a number of medicinal applications. The plant is economically important and is over exploited to the extent that there is always scarcity of propagating material (rhizomes) which is the consumable part too. The present review provides broad information of Kaempferia galanga throwing light on its current status, ethnobotany, phytochemistry and pharmacology. Extracts of Kaempferia galanga have anti-inflammatory, analgesic, anti-diarrheal, anti-bacterial, sedative, cytotoxic, insecticidal and anthelmintic properties which are reported here. Keywords: Kaempferia galanga, zingiberaceae, phytochemistry, pharmacological activity
Article
Besides other benefits, curcumin is getting more recognized for its antioxidant and anti‐inflammatory properties, highlighting the importance of curcumin application for chronic disease prevention. This systematic review and meta‐analysis of randomized controlled trials (RCTs) was conducted to assess the influence of curcumin‐containing supplements on biomarkers of inflammation and oxidative stress. MEDLINE, EMBASE, Web of Science, and Cochrane Central Register of Controlled Trials were searched till January 2018 for eligible studies. The selected studies were evaluated for their quality using the Cochrane risk of bias tool and relevant data were extracted from included studies. Data were pooled using the inverse variance method and expressed as standardized mean difference (SMD) with 95% confidence intervals (95% CI). Fifteen RCTs were included in the final analysis. The meta‐analysis indicated that curcumin supplementation significantly decreased interleukin 6 (IL‐6) (SMD −2.08; 95% CI [−3.90, −0.25]; p = 0.02), high‐sensitivity C‐reactive protein (hs‐CRP) (SMD −0.65; 95% CI [−1.20, −0.10], p = 0.02), and malondialdehyde (MDA) concentrations (SMD −3.14; 95% CI [‐4.76, −1.53], p < 0.001). Though, curcumin supplementation had no significant effect on tumor necrosis factor‐alpha (SMD −1.62; 95% CI [−3.60, 0.36]; p = 0.10) and superoxide dismutase levels (SMD 0.34; 95% CI [−1.06, 1.74], p = 0.63). Overall, this meta‐analysis suggests that taking curcumin‐containing supplements may exert anti‐inflammatory and antioxidant properties through a significant reduction in IL‐6, hs‐CRP, and MDA levels.