Open access peer-reviewed chapter

The Contribution of Javanese Pharmacognosy to Suriname’s Traditional Medicinal Pharmacopeia: Part 2

Written By

Dennis R.A. Mans, Priscilla Friperson, Meryll Djotaroeno and Jennifer Pawirodihardjo

Reviewed: 15 April 2021 Published: 10 May 2021

DOI: 10.5772/intechopen.97751

From the Edited Volume

Natural Drugs from Plants

Edited by Hany A. El-Shemy

Chapter metrics overview

325 Chapter Downloads

View Full Metrics

Abstract

The Republic of Suriname (South America) is among the culturally, ethnically, and religiously most diverse countries in the world. Suriname’s population of about 600,000 consists of peoples from all continents including the Javanese who arrived in the country between 1890 and 1939 as indentured laborers to work on sugar cane plantations. After expiration of their five-year contract, some Javanese returned to Indonesia while others migrated to The Netherlands (the former colonial master of both Suriname and Indonesia), but many settled in Suriname. Today, the Javanese community of about 80,000 has been integrated well in Suriname but has preserved many of their traditions and rituals. This holds true for their language, religion, cultural expressions, and forms of entertainment. The Javanese have also maintained their traditional medical practices that are based on Jamu. Jamu has its origin in the Mataram Kingdom era in ancient Java, some 1300 years ago, and is mostly based on a variety of plant species. The many Jamu products are called jamus. The first part of this chapter presented a brief background of Suriname, addressed the history of the Surinamese Javanese as well as some of the religious and cultural expressions of this group, focused on Jamu, and comprehensively dealt with four medicinal plants that are commonly used by the Javanese. This second part of the chapter continues with an equally extensive narrative of six more such plants and concludes with a few remarks on the contribution of Javanese jamus to Suriname’s traditional medicinal pharmacopeia.

Keywords

  • Suriname
  • Javanese
  • ethnopharmacology
  • medicinal plants
  • ethnobotanical uses
  • phytochemistry
  • pharmacology

1. Introduction

The Republic of Suriname is a small independent country in South America that is renowned for its ethnic, cultural, and religious diversity [1]. The Javanese are currently the fourth most numerous ethnic group in Suriname, after the Hindustanis, the Creoles, and the Maroons [1]. The Javanese are the descendants of indentured laborers from particularly the Indonesian island of Java who were attracted by the Dutch colonizers from the former Dutch East Indies - modern-day Indonesia - at the end of the 19th century to work on the sugar cane plantations in Suriname following the abolition of slavery in the year 1863 [2, 3]. They had signed contracts for five years, and although some returned to their home country and others relocated to The Netherlands [2, 3], most remained in Suriname and settled in the district of Commewijne where the first groups of Javanese had been put to work [2, 3].

Today, only five generations later, the Javanese have integrated well in Suriname, actively participating in all sections of the society including politics, arts, entertainment, and sports. For instance, Iding and Willy Soemita and Paul Somohardjo were prominent Surinamese Javanese politicians. Iding Soemita was born in West Java and came as an indentured laborer to Suriname, and founded the political party Kerukunan Tulodo Pranatan Inggil (KTPI) in 1949, giving Surinamese Javanese for the first time a political voice. Iding Soemita’s son Willy succeeded his father as chairman of the KTPI in 1972 and served several times as a minister until 1996. As a more outspoken and assertive alternative to the KTPI, Paul Somohardjo founded the Javanese party Pendawa Lima in 1977 that was superseded in 1998 by the Pertjajah Luhur. Somohardjo became the first-ever Javanese Speaker of the National Assembly in 2005 and also served several terms as a minister.

The Surinamese-Javanese writer Karin Amatmoekrim studied Modern Literature at the University of Amsterdam, graduated with a thesis on ‘The ethnicity in literature in Suriname’, and won the 2009-Black Magic Woman Literature Prize for her novel ‘Titus’. The Surinamese-Javanese singers Ragmad Amatstam, Oesje Soekatma, and Eddy Assan are among the greatest and most beloved musicians Suriname has brought forth. Specializing in pop-Jawa songs, they reached a broad audience in both Suriname and The Netherlands. Notable Surinamese-Javanese sports heroes are Andy Atmodimedjo, Virgil Soeroredjo, and Mitchel Wongsodikromo. Andy Atmodimedjo was an impressive professional football player and became the successful manager of several clubs in Suriname’s highest soccer league as well as the head coach of the country’s senior and under-21 national soccer teams. And Virgil Soeroredjo and Mitchel Wongsodikromo were among the world’s top badminton players who excelled on various national, Caribbean, Central American, and South American competitions.

Nevertheless, the Javanese have preserved their own identity, speaking their own language and adhering to their own specific religious and cultural customs. This also holds true for their traditional medical customs which are based on Jamu, the centuries-old traditional form of medicine from Indonesia that mainly involves the use of plants with medicinal properties. The first part of this chapter gave some background on Suriname; then addressed some of the religious and cultural expressions of Surinamese Javanese; focused on Jamu, and concluded with an extensive account of the traditional, phytochemical, and pharmacological aspects of four medicinal plants that are mainly used by Surinamese Javanese. This second part of the chapter continues with a comprehensive narrative about six additional popular ‘Javanese’ medicinal plants and concludes with the contribution of the Javanese pharmacognostic knowledge to Suriname’s traditional medicinal pharmacopeia.

Advertisement

2. Plants used in Javanese pharmacognosy

Hereunder, six medicinal plants that are traditionally mainly used by Surinamese Javanese - in addition to four that have been addressed in the first part of this chapter - have in detail been assessed for their phytochemical contents and pharmacological activities in order to provide a scientific rationale for their ethnopharmacological applications. The plants have been selected on the basis of the number of times they have been dealt with in a number of comprehensive publications describing the use of medicinal plants in the country [4, 5, 6, 7, 8, 9, 10, 11, 12]. All the ten plants and their main traditional use by Surinamese Javanese have been given in Table 1.

FamilySpecies (vernacular names in English; Surinamese-Javanese)Part(s) usedTraditional indicationsReferences
AcanthaceaeStrobilanthes crispa Blume (black face general; ketji beling)LeafDisorders of the urinary system, diabetes mellitus[8]
AraceaeAcorus calamus L. (sweet flag; dlingo)RhizomeGastrointestinal disorders, intestinal parasites, common cold, convulsions and seizures in children, evil eye and evil spirits[7, 8, 12]
ArecaceaeCocos nucifera L. (coconut; klapa)Coconut oil, coconut meat, coconut water, husk fibersRespiratory problems, pimples, shingles due to herpes, diabetes mellitus, hypertension, gastrointestinal disorders, skin and hair care, skin lesions, burns, sprains, sore muscles, evil eye and luck[6, 7, 8, 9, 10]
AsclepiadaceaeCalotropis gigantea (L.) Aiton (crown flower; bidari, widuri)Latex, stemBleeding, skin lesions, burns, toothache, tonsillitis, colds, heart conditions[10, 12]
AsteraceaeAgeratum conyzoides L. 1753 not Hieron. 1895 nor Sieber ex Steud. 1840 (goatweed; wedusan)Whole plant, leaf, rootSymptioms of flu, gastrointestinal problems, gynecological disorders, gonorrhea, itching, skin lesions, burns, allergic conjunctivitis[6, 9, 11, 12]
CaesalpiniaceaeTamarindus indica L. (tamarind; asem)Leaf, fruit pulpHealth-promoting jamus, fever, gynecological conditions, gastrointestinal disorders, itching, skin lesions[4, 8, 12]
FabaceaeSesbania grandiflora (L.) Poiret (vegetable hummingbird; turi)Leaf, barkAbdominal disorders, throat and oral infections[12]
PortulacaceaePortulaca oleracea L. (green purslane; krokot)Whole plant, leafSkin lesions, sprains, swellings, stiff joints, pain, bronchitis, conjunctivitis, anemia[5, 9, 12]
ZingiberaceaeCurcuma longa L. (turmeric; kunjit)RhizomeHealth-promoting jamus, gynecological disorders, gastrointestinal diseases, fever, inflamed gums, conjunctivitis, skin lesions, pinworm infection[8, 12]
ZingiberaceaeZingiber officinale Roscoe (ginger; djahe)RhizomeHealth-promoting jamus, overweight, respiratory diseases, gastrointestinal disorders, gynecological problems, bruises, rheumatic joints, sore muscles[8, 12]

Table 1.

Plants commonly used in Javanese traditional medicine addressed in this chapter, parts preferentially used, and traditional indications in the Surinamese-Javanese community.

2.1 Asteraceae - Ageratum conyzoides L. 1753 not Hieron. 1895 nor Sieber ex Steud. 1840

The goat weed A. conyzoides, called wedusan in Surinamese-Javanese (Figure 1), is an annual herb that is native to northern Brazil. It is sometimes grown as an ornamental plant but has become a common invasive weed in many tropical regions in the Americas, the Caribbean, and Africa. It grows to a height of about 1 meter, has an erect stem covered with white hairs, carries leaves and flowers that emit a strong, unpleasant smell, and is commonly found in gardens and open spaces. The pungent smell from the leaf and flower is due to the presence of an essential oil with a strong nauseating odor [13]. Nevertheless, a decoction of fresh shoots and leaves is used as an ingredient of an ‘anti-ageing’ herbal shampoo [14]. A. conyzoides produces hepatotoxic pyrrolizidine alkaloids [15] that are regularly encountered as contaminants in, among others, grains, honey, milk, organ meats, and eggs [16]. For this reason, A. conyzoides is not consumed by humans except when taken for medicinal purposes.

Figure 1.

The goat weed Ageratum conyzoides L. 1753 not Hieron. 1895 nor Sieber ex Steud. 1840 (Asteraceae) (from: https://images.app.goo.gl/1tsLVnadhCW6kXSr6).

In Suriname, an infusion from the whole plant is drunk against a sore throat, colics, and atony of the digestive tract [11], as well as gynecological diseases and gonorrhea [9]. A tea prepared from the root as well as the juice from the leaf are also used against a sore throat and, in addition, for reducing fever and fighting colds [12]. Itching all over the body, diarrhea, and chest conditions are treated by drinking a tea from the leaves [6]. A concentrated leaf decoction is used against chiggers [17], the juvenile forms of a type of mite (Trombiculidae) that are also known as berry bugs and that produces itching and blisters on the skin [18]. An infusion from the leaves is externally used against allergic conjunctivitis [6]. And a paste from the crushed leaves is applied on abscesses or burn wounds [6].

Most of the traditional uses and supporting pharmacological activities of A. conyzoides have been attributed to its essential oil [19]. The oil contains monoterpenes and sesquiterpenes that are mainly responsible for its fragrance [19], as well as important bioactive constituents such as flavonoids, alkaloids, steroids, benzofurans, tannins, chromenes, and coumarins [19]. A number of alkaloids and flavonoids have been encountered in other parts of the plant [19]. Several of these compounds may be associated with some its traditional uses and pharmacological activities.

The use of preparations from A. conyzoides against a sore throat, fever, and colds [11, 12] may have its rationale in the analgesic, antiinflammatory, antimicrobial, and wound healing activities of the plant. Evidence for analgesic activity was provided by the inhibitory effects of aqueous and ethanolic whole-plant or leaf extracts on acetic acid-induced writhing and/or formalin-induced licking in laboratory rodents, and the increase in the threshold of pain the animals experienced on a hot plate [20, 21]. Notably, an aqueous extract of the whole plant had been reported to accomplish analgesic effects and improvements in articulation mobility in patients with arthrosis [22]. The antinociceptive effects of A. conyzoides might be due to saponins and/or flavonoids in the plant [20, 23].

Indications for antiiflammatory effects of A. conyzoides came from the significant reduction in carrageenin-induced hind paw edema in rats by methanolic and ethanolic root, whole-plant, aerial, and leaf extracts of the plant [21, 23]. A leaf extract also exerted antiinflammatory effects in cotton pellet-induced granuloma and formaldehyde-induced arthritis models of inflammation in rats [24]. The antiinflammatory activity of A. conyzoides might be attributed to certain flavonoids [25]. This supposition is based on the amplification of the inhibitory effect of a methanol extract of the aerial part of the plant on the carrageenan-triggered edema in rats by a flavonoid fraction [25].

Support for antimicrobial activities of A. conyzoides was provided by the broad activity of aqueous, methanolic, and ethanolic extracts from the leaf and the whole plant as well as the essential oil of the plant against both Gram-positive and Gram-negative pathogenic bacteria [26, 27]. The plant extracts were also active against bacteria from wound isolates [28], methicillin-resistant Staphylococcus aureus [29], and clinical isolates of Helicobacter pylori [30]. Furthermore, the essential oil exhibited meaningful fungitoxic effects [26, 31] including activity against Microsporum gypseum, the causative agent of ringworm [32], Candida spp. [26], and the aflatoxin B1-producing Aspergillus flavus [33]. That A. conyzoides may possess, in addition, would healing properties, has been suggested by the improved rates of epithelialization and wound contraction as well as the increased tensile strength of open excision wounds in Wistar rats accomplished by aqueous, methanolic, and ethanolic extracts of the leaf [34].

The analgesic, antiinflammatory, antimicrobial, and wound healing properties of A. conyzoides may also (partially) explain the traditional use of leaf preparations against burning eyes and skin lesions [6], itchy skin [6, 11], gonorrhea [9] as well as against gastrointestinal disorders caused by infectious microorganisms [6, 11]. The latter use is further supported by the in vitro schistosomicidal effect of the essential oil of the plant against the blood fluke Schistosoma mansoni [35]; the protective activity of aqueous and ethanolic leaf extracts against ethanol-induced gastric lesions in rats [36]; and the spasmolytic effect of an aqueous leaf extract on isolated rat intestine smooth muscles [37].

2.2 Caesalpiniaceae - Tamarindus indica L.

The tamarind T. indica, called asem in Surinamese-Javanese (Figure 2), is a long-lived, slow-growing, evergreen tree that can reach a height of 20 meters. The plant has a dense, spreading crown and an extensive root system that makes it very tolerant of windy conditions and drought. Its origin is uncertain but is thought to lie in tropical Africa. From there, it has long ago been introduced in more than fifty tropical and subtropical parts throughout the world. T. indica has fragrant flowers and is extensively cultivated for its edible, sweet–sour-tasting seedpods due to their relatively high concentrations of tartaric acid and reducing sugars. The pods, along with the young leaves, seedlings, and flowers, are extensively used for preparing a large variety of dishes, beverages, and confections, and the dried seeds can be roasted and ground as a coffee substitute.

Figure 2.

The tamarind Tamarindus indica L. Caesalpiniaceae (from: https://images.app.goo.gl/ViLrKniuZfAGExDb7).

T. indica also has a wide range of medicinal applications in various traditional systems throughout Asia, Africa, and the Americas. The plant has presumably been introduced in Suriname by enslaved Africans who used it to fight fever, diarrhea, and worm infections on board of the slave ships [11]. Since then, it is abundantly used medicinally in this country. Javanese use the leaves, together with the rhizomes from Curcuma spp., in various health-promoting jamus, and drink an infusion from the fruit pulp to ease menstrual pain and vaginal discharge [8]. The pulp is used for treating constipation as well as skin conditions, heartburn, and jaundice [4]. Preparations from the leaf are drunk to stimulate perspiration in patients with fever [4] and in herbal baths to ease itching and skin irritation caused by measles, chicken pox, or rubella [12]. And Maroon women who have recently given birth incorporate T. indica leaves in a hot steam bath to cleanse the uterus and the vagina [12], presumably because of the presence of astringent, antimicrobial, and wound healing properties of the tannins in the plant [38].

T. indica contains a variety of phytochemicals with nutritious and pharmacological properties which can account, at least partially, for the traditional uses of the plant and for the pharmacological studies supporting these uses [39]. Important bioactive constituents in the plant are terpenoids and phenolic compounds including tannins; citric, malic, and tartaric acid; pectin and various pentoses and hexoses; many essential dietary minerals; as well as amino acids and proteins [39].

The incorporation of T. indica parts in jamus for improving well-being [35] may be associated with its antioxidant, weight-reducing, and immunomodulatory properties. Evidence for the former property was provided by the potent antioxidant activity exhibited by seed, pericarp, and fruit pulp preparations in various in vitro assays [40, 41, 42], and the improved efficiency of the antioxidant defense system in hypercholesterolemic hamsters treated with an ethanolic extract of the seed coat [41]. The antioxidant activity could be attributed to the polyphenolic compounds such as proanthcyanidins in the preparations [40, 42].

Indications for weight-reducing properties of T. indica came from the hypolipidemic and antioxidant activities of the fruit pulp or a fruit extract in rats on a cholesterol-rich diet [41] and the hypolipidemic and slimming effects of an ethanolic fruit pulp extract in obese rats on a cafeteria diet or on sulpiride (an antipsychotic drug that causes weight gain) [43]. And suggestions for immunomodulatory activity of T. indica were given by the stimulatory effects of a polysaccharide isolated from the fruit pulp on the uptake of foreign bodies by phagocytes, the promotion of lymphocyte proliferation, and the inhibition of leukocyte proliferation in vitro [44], as well as the increase in total white blood cell count, CD4+ T-cell population, and bone marrow cellularity in BALB/c mice which had receiving the polysaccharide intraperitoneally [45].

The use of T. indica against skin lesions [12] as well as menstrual pain and vaginal discharge [8] may be accounted for by the analgesic, antiinflammatory, and antimicrobial properties of the plant. Indications for both former activities came from the increase in reaction time of laboratory rodents in a tail immersion, acetic acid-induced writhing, tail flicking, and hot-plate assay, and the reduction in carrageenan-induced hind paw edema in the animals accomplished by a petroleum ether stembark extract, an aqueous fruit extract, a hydroethanolic leaf extract, and a methanolic seed extract [46, 47]. Evidence for antimicrobial activity came from the broad in vitro antibacterial and antifungal activity of preparations from various parts of the plant [48, 49, 50]. The analgesic and antiinlammatory activities of T. indica have been attributed to sterols, triterpenes, and phenolic compounds in the plant [46, 47]. The antimicrobial activities have mainly been associated with phenolic compounds and the essential oil [48].

Further support for the traditional uses of T. indica mentioned in the preceding alinea lesions [12] are its antihistaminic potential as suggested by the inhibitory effects of a leaf methanolic extract on the histamine-induced contraction of goat tracheal chain and guinea pig ileum [51], and its wound healing activity, as indicated by the accelerated wound closure, epithelial migration, and re-epithelialization of various types of wounds in laboratory rodents caused by water and methanolin seed extracts seed [52], the fruit paste [53], and the cork and seed ash [54].

The claims of efficacy of T. indica against gastrointestinal problems [4] is supported by the efficacy of preparations from the fruit pulp against constipation [55] due to the presence of relatively high amounts of tartaric acid and malic acid in their salt form which improve the movement of the bowel and act as a mild laxative [38]. Furthermore, a methanolic seed coat extract elicited meaningful antiulcer effect on ibuprofen-, alcohol-, and pyloric ligation-induced gastric lesions in rats when compared to the antipeptic agent ranitidine [56]. Finally, the use of T. indica against fevers [4] is supported by the antipyretic activity of a water-soluble crude polysaccharide fraction from the fruit pulp against the raised body temperature in rats and mice which had subcutaneously been injected with yeast or intraperitoneally with a lipopolysaccharide (LPS) from Escherichia coli, respectively [57].

2.3 Fabaceae - Sesbania grandiflora (L.) Poiret

The vegetable hummingbird S. grandiflora, known as turi in Surinamese-Javanese (Figure 3), is a fast-growing, relatively short-lived, highly branched tree that can grow to about 4 meters tall, and has rounded leaves, white, red, or pink flowers, as well as flat, long, thin, and green bean-like fruits. The plant probably originates from Indonesia and/or Malaysia, but is abundantly found in other hot and humid locations throughout the world. It is cultivated in various countries for its many edible parts. S. grandiflora also has many traditional medicinal uses in various parts of the world [58]. In Suriname, the plant is mainly used by Javanese, who cook the flowers, leaves, and young pods and eat them as a vegetable [12]. They also prepare a tea from the astringent bark for treating abdominal disorders and use the leaf juice as a gargle against a sore throat and mouth sprue [12].

Figure 3.

The vegetable hummingbird Sesbania grandiflora (L.) Poiret (Fabaceae) (from: https://images.app.goo.gl/8oo6WbZ8MJsEr5e2A).

Phytochemical studies revealed the presence of several bioactive in S. grandiflora that may account for its traditional uses including alkaloids, flavonoids, tannins, terpenoids, glycosides, steroids, and saponins [59]. Some of these compounds have been associated with, among others, the antimicrobial, hepatoprotective, antioxidant, and hypolipidemic activities of the plant [60, 61, 62].

Pharmacological support for the use of a concoction from S. grandiflora stembark against gastrointestinal disorders [12] is provided by the preventive action of an ethanolic extract of this part of the plant against acute gastric injury in rats caused by stress or non-steroidal anti-inflammatory drugs [63]. Of note, the extract did not modify the volume, pH, and hydrochloric acid contents of the gastric secretion of the animals [63]. Importantly, ethanolic extracts of the leaf substantially inhibited the development of ulcers in adult albino rats caused by pylorus ligation and ethanol when compared to omeprazole [64]. Furthermore, the seed oil showed considerable anthelmintic activity [65], speaking in favor of its traditional use to expel parasitic worms and other internal parasites from the gastrointestinal tract. Moreover, an S. grandiflora ethanolic leaf extract exerted a substantial protective effect on liver injury in rats produced by the antibiotic erythromycin estolate [61].

The use of the leaf juice against conditions of the throat and the oral cavity [12] may be accounted for by the antibacterial and wound healing activity of this preparation. Support for the former suggestion came from the notable activity of extracts from the leaf - but also from the stembark, the flower, and other parts of the plant - against a variety of bacterial strains [66, 67, 68]. The antimicrobial activity was most prominent with extracts prepared with organic solvents [66, 67, 68] and was also directed against methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci, either in culture or in silkworms infected with these pathogens [68]. In some cases, the antimicrobial properties of the plant have been associated with the presence of bioactive constituents such as tannins, sterols, saponins, phenolic compounds including flavonoids, and alkaloids [66, 68, 69].

Indications for wound healing activity of S. grandiflora came from the rapid repair of excision and/or incision wounds in Wistar albino rats following the topical application of a methanolic extract of the stembark or an ethanolic extract from the flower when compared to the topical antibiotics framycetin sulphate and nitrofurazone, respectively [70, 71]. Furthermore, the use of a semi-purified extract of the stembark significantly stimulated the healing of incision wounds on the mucosa of the lower lip of rats when compared to the topical antiseptic and disinfectant dequalinium chloride [72]. This activity has been attributed to the astringent and antimicrobial properties of the stembark and has been interpreted as support for the traditional use of the plant against mouth ulcers [72]. As well, an ethanolic extract of S. grandiflora flower showed potent in vitro activity against Streptococcus mutans - that is commonly found in the human oral cavity and is a significant contributor to tooth decay - that was comparable to that of erythromycin [73].

2.4 Portulacaceae - Portulaca oleracea L.

The green purslane P. oleracea, called krokot in Surinamese-Javanese (Figure 4), is an erect annual succulent herb that can reach a height of about 40 centimeters. The origin of this plant is uncertain but may lie in western Asia, India, and/or even Africa. P. oleracea was probably one of the first plants to be domesticated as a food crop. Now, it often grows as a weed in fields, waste grounds, roadside verges, and cultivated grounds in many temperate tropical parts of the world. However, it is among the richest vegetable sources of omega-3 fatty acids and also contains high levels of vitamins C and E, β-carotene, and dietary minerals such as potassium, magnesium, calcium, phosphorus, and iron [74].

Figure 4.

The green purslane Portulaca oleracea L. (Portulacaceae) (from: https://images.app.goo.gl/wmwxjiB8FvgpzF7JA).

P. oleracea has also been used in various folk medicinal systems since ancient times and is still one of the most widely used medicinal plants in the world [75]. In Suriname, individuals from all ethnic groups including those from Javanese ancestry, mainly use preparations from P. oleracea as a dressing on abscesses and sores; as an ointment with coconut oil for sprains, swellings, stiff joints, as well as back and neck pain; to stop bronchitis, and against conjunctivitis [5, 9]. The plant is also widely used against anemia [12] because of its relatively high iron content [74]. Incidentally, administration of P. oleracea seeds to adolescent females with iron deficiency anemia led to appreciable increases in the girls’ hemoglobin, hematocrit, and mean corpuscular volume [76].

Some of the traditional uses may be associated with the presence in the plant of potent bioactive compounds such as flavonoids and alkaloids [77, 78]. Some of the flavonoids have been associated with antibacterial, antiviral, antiinflammatory, and/or antioxidant activities of the plant [77, 78, 79]. Potent alkaloids identified in the plant are dopa, dopamine, and noradrenalin as well as oleracimines which have been associated with the antiinflammatory properties of the plant [77, 78, 80].

The apparent usefulness of P. oleracea as a dressing on abscesses and sores and for treating sprains, swellings, stiff joints, and back and neck pain [12] may be explained by the wound healing, antiinflammatory, analgesic, antioxidant, and/or antimicrobial activity elicited by preparations of parts of the plant. Support for the wound healing-promoting properties of P. oleracea came from the stimulation of contraction, the decrease in surface area, and the increase in tensile strength of excision wounds in mice following topical treatment with an extract of the aerial parts of the plant [81]. An ethanolic extract of the aerial parts of the plant also accelerated the healing of excision wounds in streptozotocin-induced albino rats by stimulating wound contraction and promoting granulation and organization formation [82].

Indications for antiinflammatory and analgesic activities of P. oleraceae were provided by the inhibitory effects of extracts from aerial-parts, leaf, and seed extracts of the plant in the paw edema and ear edema tests, as well as the paw licking, writhing, tail flicking, and hot plate tests [83, 84]. Importantly, intraperitoneal administration of the alkaloid allantoin isolated from the seed gave comparable results [85]. The anti-inflammatory and analgesic efficacies of the plant-derived products were similar to those found with diclofenac and aspirin [83, 84, 85]. Furthermore, aqueous extracts of the dried aerial parts of the plant prevented the TNF-α-induced vascular inflammatory events in cultured human umbilical vein endothelial cells (HUVECs) [86], and substantially inhibited the protein expression of iNOS as well as the production of PGE2, IL-6, and TNF-α in LPS-activated cultured RAW 264.7 murine macrophages [87]. The antiinflammatory activities of the P. oleracea preparations have been attributed to oleracimines, since these compounds inhibited the secretion of various inflammatory mediators by LPS-stimulated macrophages [88].

Indications for antioxidant activity of P. oleracea have been provided by the protective effects of an aerial-parts aqueous extract against DNA strand breakage in human lymphocytes caused by hydrogen peroxide [89], and of aqueous and ethanolic seed extracts against hemolysis of erythrocytes caused by the free radical initiator 2,2′-azobis hydrochloride [90]. Furthermore, administration of leaf and stem preparations prevented oxidative damage in the liver of laboratory mice treated with steptozotocin [79] or fed with a high-fat diet, among others, by modulating blood and liver antioxidant enzyme activities [91]; increased the levels of reduced glutathione, catalase, superoxide dismutase, glutathione reductase, glutathione-S-transferase and glutathione peroxidase, and inhibited lipid peroxidation and nitric oxide in liver, kidney, and testis of male Wistar albino rats [92]; and improved serum levels of superoxide dismutase and catalase in sensitized rats [93]. The latter results were also achieved with the use of α-linolenic acid, one of the constituents of the leaf and stem of the plant [93].

P. oleracea probably also possesses antibacterial, antifungal, and antiviral activities. These suppositions are supported by the growth-inhibitory activity of extracts of the leaf and stem against a wide range of both Gram-negative and Gram-positive pathogenic bacteria [94, 95] as well as Candida albicans [94]. An ethyl acetate extract of the aerial parts of the plant also inhibited the growth of dermatophytes of the genera Trichophyton [96]. And a polysaccharide isolated from the aerial part of the plant was active against herpes simplex virus type 2 [97] and a methanolic extract of the aerial parts against hepatitis C virus genotype-3a infected in cultured Huh-7 human liver cells [98]. The apparent antiviral activities of P. oleracea, along with the above-mentioned antibacterial and antifungal properties of the plant may account for its traditional use in Suriname against conjunctivitis [5] and bronchitis [9].

2.5 Zingiberaceae - Curcuma longa L.

The turmeric C. longa, known as kunjit in Surinamese-Javanese (Figure 5), is an erect, herbaceous, perennial plant that grows to a height of about 1.5 meters. It is believed to have arisen in southern or south-eastern Asia by selection and vegetative propagation of a hybrid between the wild turmeric Curcuma aromatica Salisb. and other closely related species. As a result, C. longa is not found in the wild and is only known as a domesticated plant. The maternal plant carries yellow-white flowers that do not produce viable seed and multiplies by producing new sprouts from its underground rhizomes. The plant is abundantly cultivated for its pulpy orange-yellow rhizome in Indonesia and India as well as many other tropical and subtropical parts of the world. The pungent and bitter-tasting rhizome is dried and ground into a yellow powder, an essential ingredient of curry powders and pastes for coloring and flavoring many meat and fish dishes in Asian cuisine and as a bright yellow constituent that is used in the food industry as a natural food colorant.

Figure 5.

The turmeric Curcuma longa L. (Zingiberaceae) (from: https://images.app.goo.gl/1QbEoBg45CR8ub2m6).

C. longa preparations have also a centuries-long medicinal use in various traditional systems, particularly in Indonesian Jawa and Indian Ayurveda, Unani, and Siddha [99]. In Suriname, C. longa rhizome is an essential ingredient of many jamus to promote health and fitness and to enhance mental functioning and well-being, as well as in jamus for treating inflamed gums, abscesses, menstrual pains, and skin rash [8]. In addition, drinking a tea from the fresh rhizome would purify the blood and treat stomach pain, chewing on the fresh rhizome and swallowing the sap would help against diarrhea, drinking the sap mixed with chalk would help against menstrual pains, and having children drink the sap of the grated rhizome mixed with that of other plants would take care of pinworm infection [12]. Moreover, the sap from the fresh rhizome is rubbed on the abdomen to ease bellyache and fever, dripped on watering eyes and inflamed eyelids, and used to disinfect pierced ears and the navel of newborn babies [12].

The main constituents of C. longa rhizome that may be associated with these beneficial effects are polyphenolic compounds such as diarylheptanoids and diarylpentanoids, as well as terpenoids such as sesquiterpenes, monoterpenes, diterpenes, and triterpenoids [100, 101]. The most common diarylheptanoids are the yellow-colored curcuminoids curcumin along with its derivatives demethoxycurcumin and bisdemethoxycurcumin [100, 101]. These constituents impart the characteristic color and flavor to preparations from this part of the plant [100, 101]. The sesquiterpenes are the main constituents of the rhizome essential oil, while the monoterpenes dominate the essential oils from the leaves and the flowers [101]. The major volatile principles of the rhizome oil are the aromatic compounds α- and β-turmerone [101].

Several lines of evidence support the inclusion of C. longa rhizome in jamus to promote health and fitness and to enhance mental functioning and well-being [8]. Firstly, rhizome preparations reduced the deposition of plaques similar to those seen in Alzheimer’s disease in the brains of aged mice and the oxidative damage and amyloid pathology in transgenic mouse models of Alzheimer’s disease [102]. Secondly, curcumin therapy produced favorable responses in a transgenic mouse model of Alzheimer’s disease [103], and both curcumin and dimethoxycurcumin lessened lead-induced memory deficits in Wistar rats [104]. Furthermore, a water extract of the rhizome exerted anti-stress effects in laboratory rats which were comparable to those caused by the antidepressant fluoxetine [105]. Importantly, the results from clinical studies suggested that the daily intake of this extract had a positive influence on emotional fatigue in healthy individuals [106], that curcumin intake reduced fatigue, tension, anger, confusion, and total mood disturbance following 4 weeks of supplementation in non-depressed healthy elderly people [107, 108], and that curcumin reduced depressive symptoms in individuals suffering from depression [109].

The usefulness of C. longa against (inflammatory) gastrointestinal conditions [12] is sustained by the inhibitory effects of curcumin on the damage caused by indomethacin to the gastric mucosa of laboratory rats [110] and the production of inflammatory cytokines, intercellular adhesion molecule 1, and TNF-α in the animals [110]. Furthermore, curcumin substantially improved the profile of inflammatory markers, severity of diarrhea, and colonic architecture in laboratory mice with colitis induced by trinitrobenzenesulfonic acid [111]. Clinical trials indeed showed beneficial effects of curcumin or a standardized C. longa rhizome extract in patients with peptic ulcers [112] or inflammatory bowel disease [113]. In fact, a Cochrane analysis revealed that curcumin may be a safe and effective therapy for the maintenance of remission in quiescent ulcerative colitis [114].

These apparent antiinflammatory activities of C. longa also support its traditional use against primary dysmenorrhea [12]. The substantial reduction in the level of pain during menstruation accomplished by rhizome preparations in various clinical studies (see, for instance, ref. [115]) was presumably due to blockade of prostaglandin production by curcumin [116], producing analgesic and antiinflammatory effects [116, 117]. The same mechanisms may be involved in the apparent beneficial effects of C. longa preparations against inflamed gums, abscesses, pain, inflammatory skin conditions, and conjunctivitis [8], as well as the application of a C. longa-based Javanese ointment called bobok for alleviating the discomfort of, among others, toothache [8].

The aseptic properties of C. longa may contribute to these effects. Indeed, both rhizome preparations and curcumin inhibited the growth of various standard bacterial strains [118, 119] including common periopathogens [120], as well as pathogenic fungi such as Candida albicans [121] and Aspergillus flavus [122]. Curcumin and its derivatives were also active against a broad variety of pathogenic viruses [123] including the influenza virus [124], accounting for its traditional use for fighting fever [12]. Finally, the broad antiparasitic activity of curcumin [125] may explain the Surinamese-Javanese custom of including C. longa rhizome in preparations for treating pinworm infection in children [8].

2.6 Zingiberaceae - Zingiber officinale Roscoe

The ginger Z. officinale, called djahe in Surinamse-Javanese (Figure 6), is a slender, erect, herbaceous perennial plant that grows to a height of about 2 meters. It has a thick, branched rhizome that grows horizontally near the soil surface and gives rise to leafy shoots that grow close together. Z. officinale has a long history of cultivation and use, with records going back almost 2,000 years [126]. It has probably first been domesticated in tropical Asia, presumably China, and is believed to have spread via south-eastern Asia and Africa to the Neotropics. In all these parts of the world it is abundantly cultivated for its succulent, aromatic, and pungent rhizome. This part of the plant is widely used as a hot, spicy flavoring for a variety of oriental dishes, as well as cakes, candies, and hot and cold beverages. Z. officinale also has a very long use as a medicinal herb in many traditional systems, particularly those from Indonesia, India, and China, where the fresh or dried rhizome and the essential oil are ingredients of numerous medicaments [127].

Figure 6.

The ginger Zingiber officinale Roscoe Zingiberaceae (from: https://images.app.goo.gl/ETuwvyrgdVMDP5eY9).

Z. officinale has probably been introduced in Suriname by Javanese indentured laborers [8]. The rhizome is an ingredient of many health-promoting jamus including those for maintaining the functioning of heart, muscles, blood vessels, and intestines, as well as those to stimulate fertility, reduce the risk of diabetes mellitus, and decrease stress [8]. Furthermore, all ethnic groups in Suriname also use Z. officinale rhizome for treating coughing, influenza, cold, sore throat, hoarseness, laryngitis, and pneumonia; stomach cramps and other abdominal problems; overweight; menstrual pain and to cleanse the uterus after delivery; and externally to massage bruised limbs, rheumatic joints, and sore muscles [12].

The distinctive odor and flavor of Z. officinale rhizome is mainly the result of a combination of volatile oils and non-volatile phenolic compounds [128]. The volatile oils predominantly consist of zingiberol and other sequiterpene hydrocarbons [128], while the non-volatile phenolic phytochemicals comprise, among others, gingerols, shogaols and paradols [128]. Gingerols, including 6-gingerol - the best studied phytochemical in Z. officinale - are the major pungent compounds in the fresh rhizome [128]. The gingerols are thermally labile and easily undergo dehydration reactions during drying, heating, or prolonged storage of the rhizome to form the corresponding shogaols which are about twice as pungent as the gingerols [128].

Both gingerols and shogaols exhibit a host of biological activities [129, 130], supporting some of the traditional claims of Z. officinale [8, 12]. The incorporation of Z. officinale in many health-promoting jamus [8] may be related to the ameliorating effects of the essential oil on the antioxidant capacity, inflammatory response, and the building up of fat in the liver in cases of high-fat diet-induced non-alcoholic fatty liver disease [131]. The antioxidant compounds in Z. officinale rhizome are primarily gingerols, shogaols, and some related diarylheptanoids [132], and may help protect the cells from oxidative stress [133]. In addition, Z. officinale preparations accomplished hypocholesterolemic, hypolipidemic, and antiatherosclerotic effects in cholesterol-fed rabbits and rats [134] and inhibited LDL oxidation and attenuated the development of atherosclerosis in apolipoprotein E-deficient mice [133].

Furthermore, crude rhizome methanol extracts elicited potent positive-inotropic effects in an isolated guinea pig left atria preparation [135]. These extract also induced a dose-dependent fall in blood pressure in anesthetized rats, inhibited the spontaneous force and beating rate of atrial contractions in guinea pig atria similarly to the calcium antagonist verapamil, and caused endothelium-independent vasodilation in rabbit and rat aorta [136]. These apparent cardiotonic activities have been ascribed to the gingerols and the shogaols in the preparations [125, 136]. Additional support for the inclusion of Z. officinale rhizome in health-enhancing jamus is provided by the ameliorating activity of 6-gingerol on the genotoxicity (chromosomal aberrations and sister chromatid exchanges) caused in cultured human lymphocyte chromosomes by norethandrolone and oxandrolone [137], and the chemopreventive activities of gingerols and shogaols in animals treated with laboratory carcinogens [138].

The folkloristic use of Z. officinale for treating conditions of the respiratory system is supported by the broad in vitro antibacterial properties of rhizome preparations as well as gingerols against both Gram-positive and Gram-negative human pathogenic bacteria and fungi [139] including those associated with respiratory tract infections [140]; the potentiation of the antibacterial effect of some commonly used antibiotics by the rhizome extract [141]; and the activity of these substances and compounds against the influenza A/Aichi/2/68 virus [142], the human respiratory syncytial virus (HRSV) [143], and the influenza A (H1N1) virus [144].

Several pieces of evidence support the traditional use of Z. officinale against gastrointestinal disorders [12]. Firstly, preparations from the rhizome inhibited the growth of several strains of Helicobacter pylori [145] and improved dyspeptic symptoms in patients with H. pylori-positive functional dyspepsia [146]. Secondly, rhizome preparations had a stimulatory effect on antiinflammatory cytokines and an inhibitory effect on proinflammatory cytokines in inflammations associated with the alimentary channel such as colitis and inflammatory bowel disease [147]. The antiinflammatory activities have been associated with potent inhibitory effects on prostaglandin, thromboxane, and leukotriene biosynthesis [148] and COX-2 activity [149]. Furthermore, methanolic and aqueous extracts from the rhizome exerted meaningful activity against larvae of the rat long worm Angiostrongylus cantonensis and those of the parasitic fish nematode Anisakis simplex, the causative agents of angiostrongyliasis, the most common causes of eosinophilic meningitis or meningoencephalitis in south-eastern Asia and the tropical Pacific islands [150], and anisakiasis, a gastrointestinal infection characterized by severe abdominal cramps [151], respectively. This suggests that Z. officinale also has activity against parasites of the alimentary tract, further supporting its traditional use against conditions of this organ system [12].

Advertisement

3. Concluding remarks

The Javanese have been in Suriname for over a century. They have been brought to that country as poor and unaccustomed indentured laborers but have become successful individuals who have integrated well into the Surinamese community, actively participating in all its sections. Indeed, the Javanese are full-fledged citizens of the country and an integral part of the vibrant color palette represented by its unique cultural, religious, and ethnic diversity. This is reflected by the presence of the small, often family-owned Javanese restaurants called warungs in the smallest of towns, where widely appreciated savory dishes such as bami goreng (fried noodles), nasi goreng (fried rice), pityel (mixed blanched vegetables with a peanut sauce dressing), teloh (fried cassava), pejeh (prawn crackers), and saoto soup (chicken broth, meat, and vegetables) can be enjoyed, and can be washed down with refreshing beverages such as the lemongrass-, corn starch-, and coconut milk-based dawet.

Nevertheless, the Javanese have maintained most of their cultural traditions. This particularly holds true for their ancient form of medicine that is based on centuries-old Jamu. Jamu has added a unique element to the array of traditional forms of medicine throughout the Caribbean and Latin America, most of which are based on practices from the Indigenous American peoples as well as those from Africa, China, India, and various European countries. The influence of Jamu is already noticeable. The use of an infusion of the cat’s whiskers or kumis kutjing Orthosiphon aristatus (Blume) Miq. (Lamiaceae) for treating kidney stones and renal colics is no longer limited to Indonesia and Suriname but has expanded into many other parts of the world. It is foreseeable that many more Jamu recipes will one day contribute to Suriname’s traditional medicinal pharmacopeia as well as to the development of novel mainstream drugs for treating human diseases.

References

  1. 1. General Bureau of Statistics. Demographic data 2015-2018. Paramaribo: General Bureau of Statistics; 2019.
  2. 2. Hoefte R. In place of slavery: a social history of British Indian and Javanese laborers in Suriname. Gainesville: University Press of Florida; 1998.
  3. 3. Djasmadi L, Hoefte R, Mingoen H, editors. Migratie en cultureel erfgoed: verhalen van Javanen in Suriname, Indonesië en Nederland (Migration and cultural heritage: stories of Javanese in Suriname, Indonesia and The Netherlands). Leiden: KITLV Press; 2010.
  4. 4. Stephen HJM. Geneeskruiden van Suriname: hun toepassing in de volksgeneeskunde en in de magie (Herbal medicines from Suriname: their applications in folk medicine and wizardry). Amsterdam: De Driehoek; 1979.
  5. 5. Wooding CJ. Traditional healing and medicine in Winti: a sociological interpretation. Journal of Opinion. 1979;9:35-40.
  6. 6. May AF. Sranan oso dresi. Surinaams kruidenboek (Surinamese folk medicine. A collection of Surinamese medicinal herbs). Paramaribo: De Walburg Pers; 1982.
  7. 7. Titjari. Famiri-encyclopedia foe da natoera dresi-fasi. Gezinskruidenboek van de natuurgeneeswijzen. Natuurgeneeswijzen uit het zonnige Suriname (Encyclopedia of plant-based forms of treatment. Folk medicines from sunny Suriname). Amsterdam: Sangrafoe; 1985.
  8. 8. Tjong Ayong G. Het gebruik van medicinale planten door de Javaanse bevolkingsgroep in Suriname (The use of medicinal plants by the Javanese in Suriname). Paramaribo: Instituut voor de Opleiding van Leraren; 1989.
  9. 9. Sedoc NO. Afrosurinaamse natuurgeneeswijzen: bevattende meer dan tweehonderd meest gebruikelijke geneeskrachtige kruiden (Afro-Surinamese natural remedies: over two hundred commonly used medicinal herbs). Paramaribo: Vaco Press; 1992.
  10. 10. Raghoenandan UPD. Etnobotanisch onderzoek bij de Hindoestaanse bevolkingsgroep in Suriname (An ethnobotanical investigation among Hindustanis in Suriname). [MSc thesis]. Paramaribo: Anton de Kom Universiteit van Suriname; 1994.
  11. 11. DeFilipps R, Maina S, Crepin J, National Museum of Natural History (US), Department of Botany. Medicinal plants of the Guianas (Guyana, Surinam, French Guiana). Washington DC: Dept. of Botany, National Museum of Natural History, Smithsonian Institution; 2004.
  12. 12. Van Andel TR, Ruysschaert S. Medicinale en rituele planten van Suriname (Medicinal and ritual plants of Suriname). Amsterdam: KIT Publishers; 2011.
  13. 13. Wandji J, Bissangou MF, Ouambra JM, Silou T, Abena AA, Keita A. The essential oil from Ageratum conyzoides. Fitoterapia. 1996;67:427-431.
  14. 14. Singh SR, Phurailatpam AK, Senjam P. Identification of the plants use as natural herbal shampoo in Manipur. African Journal of Traditional, Complementary and Alternative Medicine. 2014;11:135-139.
  15. 15. Neuman MG, Cohen L, Opris M, Nanau RM, Hyunjin J. Hepatotoxicity of pyrrolizidine alkaloids. Journal of Pharmacy and Pharmaceutical Sciences. 2015;18:825-843.
  16. 16. Griffin CT, Sheehan A, Danaher M, Furey A. Pyrrolizidine alkaloids in food: Analytical, toxicological and health considerations. In: Wong Y-C, Lewis RJ, editors. Analysis of food toxins and toxicants. 2017. p. 267-318.
  17. 17. Diallo BO, Joly HI, McKey D, Hosaert-McKey M, Chevallier MH. Genetic diversity of Tamarindus indica populations: Any clues on the origin from its current distribution? African Journal of Biotechnology. 2007;6:853-860.
  18. 18. Elston DM. What’s eating you? Chiggers. Cutis. 2006;77:350-352.
  19. 19. Singh SB, Devi WR, Marina A, Devi WI, Swapana N, Singh CB. Review: Ethnobotany, phytochemistry and pharmacology of Ageratum conyzoides Linn (Asteraceae). Journal of Medicinal Plant Research. 2013;7:371-385.
  20. 20. Okemy-Andissa N, Ouamba JM, Koudou J, Diatewa M, Gleassor H, Abena AA. Comparative study of analgesic activities of Tetra® and an association of three plants: Ageratum conyzoides, Cymbopogon citratus and Lippia multiflora. International Journal of Pharmacology. 2006;2:42-44. DOI: 10.3923/ijp.2006.42.44.
  21. 21. Rahman MA, Akter N, Rashid MH, Nazim A, Uddin M, Islam M. Analgesic and anti-inflammatory effect of whole Ageratum conyzoides and Emilia sonchifolia alcoholic extracts in animal models. African Journal of Pharmacy and Pharmacology. 2012;6:1469-1476. DOI: 10.5897/AJPP12.083.
  22. 22. Marques-Neto JF, Lapa A, Kubota M. Efeitos do Ageratum conyzoides Linee no tratamento da artrose (Effects of Ageratum conyzoides Linn, on the treatment of arthrosis). Revista Brasileira de Reumatologia. 1988;28:34-37.
  23. 23. Sampson JH, Phillipson JD, Bowery NG, OíNeill MJ, Houston JG, Lewis JA. Ethnomedicinally selected plants as sources of potential analgesic compounds: Indication of in vitro biological activity in receptor binding assays. Phytotherapy Research. 2000;14:24-29.
  24. 24. Moura AC, Silva EL, Fraga MC, Wanderley AG, Afiatpour P, Maia MB. Anti-infl ammatory and chronic toxicity study of the leaves of Ageratum conyzoides L. in rats. Phytomedicine. 2005;12:138-142.
  25. 25. Galati EM, Miceli N, Taviano MF, Sanogo R, Raneri E. Anti-inflammatory and antioxidant activity of Ageratum conyzoides. Pharmaceutical Biology. 2001;39:336-339. DOI: 10.1076/phbi.39.5.336.5891.
  26. 26. Pattnaik S, Subramayam V, Perumal SR, Igancimuthu S, Patric RD. Preliminary screening of ethnomedicinal plants from India. Journal of Ethnopharmacology. 1999;66:235-290.
  27. 27. Neelabh C, Nahid A, Navneet K. Study on methanolic extract of Ageratum conyzoides for its ability to act as an antioxidant and to suppress the microbial growth. The Pharma Innovation Journal. 2017;6:170-173.
  28. 28. Odeleye OP, Oluyege JO, Aregbesola OA, Odeleye PO. Evaluation of preliminary phytochemical and antibacterial activity of Ageratum conyzoides (L) on some clinical bacterial isolates. International Journal of Engineering and Science. 2014;3:1-5.
  29. 29. Akinyemi KO, Oladapo O, Okwara CE, Ibe CC, Fasure K.A. Screening of crude extracts of six medicinal plants used in South- West Nigerian unorthodox medicine for antimethicillin resistant Staphylococcus aureus activity. BMC Complementary and Alternative Medicine. 2005;5:6-8.
  30. 30. Roland NN, Alertia EMT, Susan MM, Henry NL, Agnes M, Lucy MN, et al. In vitro anti-Helicobacter pylori activity of extracts of selected medicinal plants from North West Cameroon. Journal of Ethnopharmacology. 2007;114:452-457. DOI: 10.1016/j.jep.2007.08.037.
  31. 31. Fiori ACG, Schwan-Estrada KRF, Stangarlin JR, Vida JB, Scapim CA, Cruz MES, et al. Antifungal activity of leaf extracts and essential oils of some medicinal plants against Didymella bryoniae. Journal of Phytopathology. 2000;148:483-487. DOI: 10.1046/j.1439-0434.2000.00524.x.
  32. 32. Mishra DN, Dixit V, Mishra AK. Mycotoxic evaluation of some higher plants against ringworm causing fungi. Indian Drugs. 1991;28:300-303.
  33. 33. Nogueira JH, Gonçalez E, Galleti SR, Facanali R, Marques MO, Felício JD. Ageratum conyzoides essential oil as aflatoxin suppressor of Aspergillus flavus. International Journal of Food Microbiology. 2010;137:55-60. DOI: 10.1016/j.ijfoodmicro.2009.10.017.
  34. 34. Dash G, Pn M. Wound healing effects of Ageratum conyzoides Linn. International Journal of Pharma and Bio Sciences. 2011;2:369-383.
  35. 35. De Melo NI, Magalhaes LG, de Carvalho CE, Wakabayashi KA, de P Aguiar G, Ramos RC, et al. Schistosomicidal activity of the essential oil of Ageratum conyzoides L. (Asteraceae) against adult Schistosoma mansoni worms. Molecules. 2011;16:762-773. DOI: 10.3390/molecules16010762.
  36. 36. Shirwailkar A, Bhilegaonkar PM, Malini S, Kumar JS. The gastroprotective activity of the ethanol extract of Ageratum conyzoides. Journal of Ethnopharmacology. 2003;86:117-121. DOI: 10.1016/S0378-8741(03)00050-3.
  37. 37. Silva MJMe, Capaz FR, Vale MR. Effects of the water soluble fraction from leaves of Ageratum conyzoides on smooth muscle. Phytotherapy Research. 2000;14:130-132. DOI: 10.1002/(SICI)1099-1573(200003)14:2<130::AID-PTR594>3.0.CO;2-4.
  38. 38. Havinga RM, Hartl A, Putscher J, Prehsler S, Buchmann C, Vogl CR. Tamarindus indica L, (Fabaceae): Patterns of use in traditional African medicine. Journal of Ethnopharmacology. 2010;127:573-588. DOI: 10.1016/j.jep.2009.11.028.
  39. 39. Ahmad A, Ahmad W, Zeenat F, Sajid M. Therapeutic, phytochemistry and pharmacology of Tamarindus indica: A review. International Journal of Unani and Integrative Medicine. 2018;2:14-19.
  40. 40. Sudjaroen Y, Haubner R, Wurtele G, Hull WE, Erben G, Spiegelhalder B, et al. Isolation and structure elucidation of phenolic antioxidants from tamarind (Tamarindus indica L.) seeds and pericarp. Food and Chemical Toxicology. 2005;43:1673-1682.
  41. 41. Martinello F, Soares SM, Franco JJ, Santos AC, Sugohara A, Garcia SB, et al. Hypolipemic and antioxidant activities from Tamarindus indica L. pulp fruit extract in hypercholesterolemic hamsters. Food and Chemical Toxicology. 2006;44:810-818.
  42. 42. Siddhuraju P. Antioxidant activity of polyphenolic compounds extracted from defatted raw and dry heated Tamarindus indica seed coat. LWT Food Science and Technology. 2007;40:982-990.
  43. 43. Jindal V, Dhingra D, Sharma S, Parle M, Harna RK. Hypolipidemic and weight reducing activity of the ethanolic extract of Tamarindus indica fruit pulp extract in cafeteria diet and sulpiride-induced obese rats. Journal of Pharmacology and Pharmacother. 2011;2:80-84. DOI: 10.4103/0976-500X.81896.
  44. 44. Sreelekha TT, Vijayakumar T, Ankanthil R, Vijayan KK, Nair MK. Immunomodulatory effects of a polysaccharide from Tamarindus indica. Anticancer Drugs. 1993;4:209-212. DOI: 10.1097/00001813-199304000-00013.
  45. 45. Aravind SR, Joseph MM, Varghese S, Balaram P, Sreelekha TT. Antitumor and immunopotentiating activity of polysaccharide PST001 isolated from the seed kernel of Tamarindus indica: an in vivo study in mice. ScientificWorld Journal. 2012;361382. DOI: 10.1100/2012/361382.
  46. 46. Bhadoriya SS, Mishra V, Raut S, Jain S. Antiinflammatory and antinociceptive activities of hydroethanolic extract of Tamarindus indica leaves. Scientia Pharmaceutica. 2012;80:685-700.
  47. 47. Suralkar AA, Rodge KN, Kamble RD, Maske KS. Evaluation of anti-inflammatory and analgesic activities of Tamarindus indica seeds. International Journal of Pharmaceutical Sciences and Drug Research. 2012;4:213-217.
  48. 48. Escalona-Arranz JC, Péres-Roses R, Urdaneta-Laffita I, Camacho-Pozo MI, Rodríguez-Amado J, Licea-Jiménez I. Antimicrobial activity of extracts from Tamarindus indica L. leaves. Pharmacognosy Magazine. 2010;6:242-247. DOI: 10.4103/0973-1296.66944.
  49. 49. Nwodo UU, Obiiyeke GE, Chigor VN, Okoh AI. Assessment of Tamarindus indica extracts for antibacterial activity. International Journal of Molecular Sciences. 2011;12:6385-6396. DOI: 10.3390/ijms12106385.
  50. 50. Pratibha G, Gudipati T, Srivasthva A. Antifungal activity and phytochemical analysis of the alcoholic and aqueous extract of the aerial part of the plant Tamarindus indica L. Research Journal of Recent Sciences. 2017;6:8-13.
  51. 51. Tayade P, Borde N, Jagtap S, Patil V, Vaishna G, Girbane Y, et al. Effect of Tamarindus indica Linn. against isolated goat tracheal and guinea pig ilium preparation. International Journal Of Comprehensive Pharmacy. 2010;1:1-3.
  52. 52. Bin Mohamad MY, Akram HB, Bero DN. Tamarind seed extract enhances epidermal wound healing. International Journal of Biology. 2012;4:81-88. DOI: 10.5539/ijb.v4n1p81.
  53. 53. Attah MO, Ishaya HB, Chiroma MS, Amaza DS, Balogun SU, Jacks TW. Effect of Tamarindus indica (Linn) on the rate of wound healing in adult rabbits. IOSR Journal of Dental and Medical Sciences. 2015;14:80-84. DOI: 10.9790/0853-14818084.
  54. 54. Naik TI, Shrikanth P, Mundugaru R, Shridhara Bairy ST. Wound healing activity of Tamarindus indica Linn. seed and cork ash. Journal of Ayurveda Medical Sciences. 2017;2:129-135.
  55. 55. Bhadoriya SS, Ganeshpurkar A, Narwaria J, Rai G, Jain AP. Tamarindus indica: Extent of explored potential. Pharmacognosy Reviews. 2011;5:73-81. DOI: 10.4103/0973-7847.79102.
  56. 56. Kalra P, Sharma S, Suman Kumar S. Antiulcer effect of the methanolic extract of Tamarindus indica seeds in different experimental models. Journal of Pharmacy and Bioallied Sciences. 2011;3:236-241. DOI: 10.4103/0975-7406.80778.
  57. 57. Izquierdo T, García-Tamayo F, Soto C, Castrillón LE. A Tamarindus indica. Linn pulp polysaccharide inhibits fever in vivo and IL-1β release by murine peritoneal exudate cells. Pharmaceutical Biology. 2007;45:22-30.
  58. 58. Mohiuddin AK. Medicinal and therapeutic values of Sesbania grandiflora. Advanced Research Journal of Pharmacy and Pharmacology. 2019;4:87-93.
  59. 59. Wagh VD, Wagh KV, Tandale YN, Salve SA. Phytochemical, pharmacological and phytopharmaceutics aspects of Sesbania grandiflora (Hadga): A review. Journal of Pharmacy Research. 2009;2:889-892.
  60. 60. Goun E, Cunningham G, Chu D, Nguyen C, Miles D. Antibacterial and antifungal activity of Indonesian ethnomedical plants. Fitoterapia. 2003;76:592-596.
  61. 61. Pari L, Uma A. Protective effect of Sesbania grandiflora against erythromycin estolate-induced hepatotoxicity. Therapie. 2003;58:439-443.
  62. 62. Ramesh T, Begum VH. Protective effect of Sesbania grandiflora against cigarettes smoke-induced oxidative damage in rats. Journal of Medicinal Food. 2008;11:369-375.
  63. 63. Sertié JAA, Wiezel G, Woisky RG, Carvalho JCT. Antiulcer activity of the ethanol extract of Sesbania grandiflora. Brazilian Journal of Pharmaceutical Sciences. 2001;37:107-112.
  64. 64. Bhoumik D, Berhe AH, Mallik A. Evaluation of gastric anti-ulcer potency of ethanolic extract of Sesbania grandiflora Linn leaves in experimental animals. American Journal of Phytomedicine and Clinical Therapeutics. 2016;4:174-182.
  65. 65. Jalalpurea SS, Alagawadia KR, Mahajanshetty CS. In vitro anthelmintic property of various seed oils. Iranian Journal of Pharmaceutical Sciences. 2006;4:281-284.
  66. 66. China R, Mukherjee S, Sen S, Bose S, Datta S, Koley H, et al. Antimicrobial activity of Sesbania grandiflora flower polyphenol extracts on some pathogenic bacteria and growth stimulatory effect on the probiotic organism Lactobacillus acidophilus. Microbiological Research. 2012;167:500-506.
  67. 67. Vipin K, Gupta AK, Gupta R. Antimicrobial activity of Sesbania grandiflora (L.) Pers. International Research Journal of Pharmacy. 2011;2:85-87.
  68. 68. Anantaworasakul P, Hamamoto H, Sekimizu K, Okonogi S. In vitro antibacterial activity and in vivo therapeutic effect of Sesbania grandiflora in bacterial infected silkworms. Pharmaceutical Biology. 2017;55:1256-1262.
  69. 69. Roy A, Bhoumik D, Kumar Sahu R, Dwivedi J. Phytochemical screening and antioxidant activity of Sesbania grandiflora leaves extracts. Asian Journal of Research in Pharmaceutical Sciences. 2014;4:16-21.
  70. 70. Karthikeyan P, Suresh V, Suresh A. Wound healing activity of Sesbania grandiflora (L.) Poir. bark. International Journal of Pharmaceutical Research and Development. 2011;3:87-93.
  71. 71. Sheikh AA, Sayyed Z, Siddiqui AR, Pratapwar AS, Sheakh SS. Wound healing activity of Sesbania grandiflora Linn flower ethanolic extract using excision and incision wound model in Wistar rats. International Journal of PharmTech Research. 2011;3:895-898.
  72. 72. Reyes GL, Wong S, Uera J, Dimagiba MF, Timoteo MAC. Katuray (Sesbania grandiflora) bark extract as oral paint for ulcers. Poster 089. Meeting International Association for Dental Research-South East Asian Division. Bali, Indonesia: 2015.
  73. 73. Saifudin A, Forentin AM, Fadhilah A, Tirtodiharjo K, Melani WD, Widyasari D, et al. Bioprospecting for anti-Streptococcus mutans: the activity of 10% Sesbania grandiflora flower extract comparable to erythromycin. Asian Pacific Journal of Tropical Biomedicine. 2016;6:751-754.
  74. 74. Uddin MK, Juraimi, Hossain MS, Un Nahar MA, Ali ME, Rahman MM. Purslane weed (Portulaca oleracea): A prospective plant source of nutrition, omega-3 fatty acid, and antioxidant attributes. The Scientific World Journal. 2014;951019. DOI: 10.1155/2014/951019.
  75. 75. Dweck AC. Purslane - Portulaca oleracea: the global panacea. [Internet]. 2001. Available from: http://www.dweckdata.com/Published_papers/Portulaca_oleracea.pdf [Accessed: 25-01-2021].
  76. 76. Mokhtarifar A, Zeydabadi FA, Asili J, Kooshyar MM, Sahebkar A. The effect of Portulaca oleracea (purslane) seeds on hemoglobin levels in adolescent girls with iron deficiency anemia: A randomized comparative trial. Comparative Clinical Pathology. 2017;26:11-16.
  77. 77. Okafor IA, Ezejindu D. Phytochemical studies on Portulaca oleracea (purslane) plant. Global Journal of Biology, Agriculture and Health Sciences. 2014;3:132-136.
  78. 78. Zhou Y-X, Xin H-L, Rahman K, Wang S-J, Peng C, Zhang H. Portulaca oleracea L.: A review of phytochemistry and pharmacological effects. BioMed Research International. 2015; 925631. DOI: 10.1155/2015/925631.
  79. 79. Oliveira I, Valentão P, Lopes R, Andrade PB, Bento A, Pereira JA. Phytochemical characterization and radical scavenging activity of Portulaca oleraceae L. leaves and stems. Microchemical Journal. 2009;92:129-134.
  80. 80. Rafieian-Kopaei M, Alesaeidi S. Portulaca oleraceae: A review study with anti-inflammatory and muscle relaxant perspective. Indian Journal of Medical Research and Pharmaceutical Sciences. 2016;3:50-59.
  81. 81. Rashed AN, Afifi FU, Disi AM. Simple evaluation of the wound healing activity of a crude extract of Portulaca oleracea L. (growing in Jordan) in Mus musculus JVI-1. Journal of Ethnopharmacology. 2003;88:131-136. DOI: 10.1016/s0378-8741(03)00194-6.
  82. 82. Eldeighdye SM, Abdel-Khalek LG, Taha MS, Allam TM, Hassanian WF, Akaber TK, et al. The therapeutic effect of purslane (Portulaca oleracea) extract on wound healing in diabetic albino rats. World Journal of Pharmacy and Pharmaceutical Sciences. 2016;5:1454-1469.
  83. 83. Chan K, Islam MW, Kamil M, Radhakrisha R, Zakaria MN, Habibullah M, Attas A. The analgesic and anti-inflammatory effects of Portulaca oleracea L. subsp. Sativa (Haw.) Celak. Journal of Ethnopharmacology. 2000;73: 445-451. DOI: 10.1016/s0378-8741(00)00318-4.
  84. 84. Fatima N, Syed S, Ahmad M, Mehjabeen, Jahan N. A Comparison of analgesic and anti-inflammatory activities of Portulaca oleraceae leaf and seeds. RADS Journal of Pharmacy and Pharmaceutical Sciences. 2018;6:194-199.
  85. 85. Wang H-Z, Wang CJ. Isolation, characterization and analgesic activity of natural allantoin from Portulaca oleracea seed. Modern Chemistry and Applications. 2018;6:1. DOI: 10.4172/2329-6798.1000249.
  86. 86. Lee AS, Kim JS, Lee YJ, Kang DG, Lee HS. Anti-TNF-α activity of Portulaca oleraceae L in vascular endothelial cells. International Journal of Molecular Sciences. 2012;13:5628-5644.
  87. 87. Kim Y-O, Lee S-W, Na SW, Park HR, ES. Anti-inflammatory effects of Portulaca oleracea L. on the LPS-induced RAW 264.7 cells. Journal of Medicinal Plants Research. 2015;9:407-411. DOI: 10.5897/JMPR2012.1183.
  88. 88. Xu L, Ying Z, Wei W, Hao D, Wang H, Zhang W, et al. A novel alkaloid from Portulaca oleracea L. Natural Product Research. 2017;31:902-908.
  89. 89. Behravan J, Mosafa F, Soudmand N, Taghiabadi E, Razavi BM, Karimi G. Protective effects of aqueous and ethanolic extracts of Portulaca oleracea L. aerial parts on H2O2-induced DNA damage in lymphocytes by comet assay. Journal of Acupuncture and Meridian Studies. 2011;4:193-197.
  90. 90. Karimi G, Aghasizadeh M, Razavi M, Taghiabadi E. Protective effects of aqueous and ethanolic extracts of Nigella sativa L. and Portulaca oleracea L. on free radical induced hemolysis of RBCs. Daru: Journal of Faculty of Pharmacy, Tehran University of Medical Sciences. 2011;19:295-300.
  91. 91. Chen B, Zhou H, Zhao W, Zhou W, Yuan Q, Yang G. Effects of aqueous extract of Portulaca oleracea L. on oxidative stress and liver, spleen leptin, PARα and FAS mRNA expression in high-fat diet induced mice. Molecular Biology Reports. 2012;39:7981-7988.
  92. 92. Dkhil MA, Moniem AEA, Al-Quraishy S, Saleh RA. Antioxidant effect of purslane (Portulaca oleracea) and its mechanism of action. Journal of Medicinal Plants Research. 2011;5:1589-1593.
  93. 93. Kaveh M, Eidi A, Nemati A, Boskabady MH. The extract of Portulaca oleracea and its constituent, alpha linolenic acid affects serum oxidant levels and inflammatory cells in sensitized rats. Iranian Journal of Allergy, Asthma, and Immunology. 2017;16:256-270.
  94. 94. Elkhayat ES, Ibrahim SRM, Aziz MA. Portulene, a new diterpene from Portulaca oleracea L. Journal of Asian Natural Products Research. 2008;10:1039-1043.
  95. 95. Islam SM, Shawon JAM, Mahmud SAM. Antimicrobial activity of methanol, n-hexane and dichloromethane extract of Portulaca oleracea. Advances in Pharmacology and Clinical Trials. 2018;3:1-4.
  96. 96. Oh K-B, Chang I-M, Hwang K-J, Mar W. Detection of antifungal activity in Portulaca oleracea by a single-cell bioassay system. Phytotherapy Research. 2000;14:329-332.
  97. 97. Dong CX, Hayashi K, Lee JB, Hayashi T. Characterization of structures and antiviral effects of polysaccharides from Portulaca oleracea L. Chemical and Pharmaceutical Bulletin. 2010;58:507-510. DOI: 10.1248/cpb.58.507.
  98. 98. Noreen S, Hussain I, Tariq MI, Ijaz B, Iqbal S, Ul-Zaman Q, et al. Portulaca oleracea L. as a prospective candidate inhibitor of hepatitis C virus NS3 serine protease. Viral Immunology. 2015;28:282-289.
  99. 99. Kunnumakkara B, Bordoloi D, Padmavathi G, Monisha J, Roy NK. Curcumin, the golden nutraceutical: multitargeting for multiple chronic diseases. British Journal of Pharmacology. 2017;174:1325-1348.
  100. 100. Li S, Yuan W, Deng G, Wang P, Yang P. Chemical composition and product quality control of turmeric (Curcuma longa L.). Pharmaceutical Crops. 2011;2:28-54.
  101. 101. Sabale P, Modi A, Sabale V. Curcuma longa Linn. A phytochemical and phytopharmacological review. Research Journal of Pharmacognosy and Phytochemistry. 2013;5:59-68.
  102. 102. Rao R, Descamps O, John V, Bredesen DE. Ayurvedic medicinal plants for Alzheimer’s disease: a review. Alzheimer’s Research and Therapy. 2012;4:22. DOI: 10.1186/alzrt125.
  103. 103. Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. Journal of Neuroscience. 2001;21:8370-8377.
  104. 104. Dairam A, Limson JL, Walkins GM, Antunes E, Daya S. Curcuminoids, curcumin, and demethoxycurcumin reduce lead-induced memory deficits in male Wistar rats. Journal of Agricultural and Food Chemistry. 2007;55:1039-1044.
  105. 105. Zafir A, Banu N. Antioxidant potential of fluoxetine in comparison to Curcuma longa in restraint-stressed rats. European Journal of Pharmacology. 2007;572:23-31.
  106. 106. Kawasaki K, Muroyama K, Murosaki S. Effect of a water extract of Curcuma longa on emotional states in healthy participants. Bioscience of Microbiota, Food and Health. 2018;37:25-29. DOI: 10.12938/bmfh.17-020.
  107. 107. Cox KH, Pipingas A, Scholey AB. Investigation of the effects of solid lipid curcumin on cognition and mood in a healthy older population. Journal of Psychopharmacology. 2015;29:642-651.
  108. 108. Cox KHM, White DJ, Pipingas A, Poorun K, Scholey A. Further evidence of benefits to mood and working memory from lipidated curcumin in healthy older people: A 12-week, double-blind, placebo-controlled, partial replication study. Nutrients. 2020;4:1678. DOI: 10.3390/nu12061678.
  109. 109. Fusar-Poli L, Vozza L, Gabbiadini A, Vanella A, Concas I, Tinacci S, et al. Curcumin for depression: A meta-analysis. Critical Reviews in Food Science and Nutrition. 2020;60:2643-2653.
  110. 110. Thong-Ngam D, Choochuai S, Patumraj S, Chayanupatkul M, Klaikeaw N. Curcumin prevents indomethacin-induced gastropathy in rats. World Journal of Gastroenterology. 2012;18:1479-1484.
  111. 111. Ukil A, Maity S, Karmakar S, Datta N, Vedasiromoni JR, Das PK. Curcumin, the major component of food flavour turmeric, reduces mucosal injury in trinitrobenzene sulphonic acid-induced colitis. British Journal of Pharmacology. 2003;139:209-218.
  112. 112. 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 Journal of Tropical Medicine and Public Health. 2011;32:208-215.
  113. 113. Hanai H, Iida T, Takeuchi K, Watanabe F, Maruyama Y. Curcumin maintenance therapy for ulcerative colitis: randomized, multicenter, double-blind, placebo-controlled trial. Clinical Gastroenterology and Hepatology. 2006;4:1502-1506.
  114. 114. Kumar S, Ahuja V, Sankar MJ, Kumar A, Moss AC. Curcumin for maintenance of remission in ulcerative colitis. Cochrane Database of Systemic Reviews. 2012;17:CD008424.
  115. 115. Rahman SF, Hardi GW, Maras MAJ, Riva YR. Influence of curcumin and ginger in primary dysmenorrhea: A review. International Journal of Applied Engineering Research. 2020;15:634-638.
  116. 116. Kim SO, Kundu JK, Shin YK, Park J-H, Cho M-H, Kim T-Y, et al. [6]-Gingerol inhibits COX-2 expression by blocking the activation of p38 MAP kinase and NF-κB in phorbol ester-stimulated mouse skin. Oncogene. 2005;24:2558-2567. DOI: 10.1038/sj.onc.1208446.
  117. 117. Lakhan SE, Ford CT, Tepper D. Zingiberaceae extracts for pain: a systematic review and meta-analysis. Nutrition Journal. 2015;14;50. DOI: 10.1186/s12937-015-0038-8.
  118. 118. Gunes H, Gulen D, Mutlu R. Antibacterial effects of curcumin: an in vitro minimum inhibitory concentration study. Toxicology and Industrial Health. 2016;32:246-250.
  119. 119. Teow SY, Liew K, Ali SA, Khoo AS, Peh SC. Antibacterial action of curcumin against Staphylococcus aureus: A brief review. Journal of Tropical Medicine. 2016;2853045.
  120. 120. Bomdya RS, Shah MU, Doshi YS, Shah VA, Khirade SP. Antibacterial activity of curcumin (turmeric) against periopathogens - An in vitro evaluation. Journal of Advanced Clinical and Research Insights. 2017;4:175-180.
  121. 121. Petnual P, Sangvanich P, Karnchanatat A. A lectin from the rhizomes of turmeric (Curcuma longa L.) and its antifungal, antibacterial, and α-glucosidase inhibitory activities. Food Science and Biotechnology. 2010;19:907-916.
  122. 122. Hu Y, Luo J, Kong W, Zhang J, Logrieco FA, Wangd X, et al. Uncovering the antifungal components from turmeric (Curcuma longa L.) essential oil as Aspergillus flavus fumigants by partial least squares. Royal Society of Chemistry, Advances. 2015;5:41967-41976.
  123. 123. Singh RK, Rai D, Yadav D, Bhargava A, Balzarini J, De Clercq E. Synthesis, antibacterial and antiviral properties of curcumin bioconjugates bearing dipeptide, fatty acids and folic acid. European Journal of Medicinal Chemistry. 2010;45:1078-1086.
  124. 124. Chen D-Y, Shien J-H, Tiley Chiou S-S, Wang S-Y, Chang T-J, Lee Y-J, et al. Curcumin inhibits influenza virus infection and haemagglutination activity. Food Chemistry. 2010;119:1346-1351. DOI: 10.1016/j.foodchem.2009.09.011.
  125. 125. Cheraghipour K, Marzban A, Ezatpour B, Khanizadeh S, Koshki J. Antiparasitic properties of curcumin: a review. AIMS Agriculture and Food. 2019;4:1-18.
  126. 126. Langner E, Greifenberg S, Gruenwald J. Ginger: History and use. Advances in Therapy. 1998;15:25-44.
  127. 127. Imtiyaz S, Rahman K, Sultana A, Tariq M, Chaudhary SS. Zingiber officinale Rosc.: a traditional herb with medicinal properties. TANG Humanitas Medicine. 2013;3:e26. DOI: http://dx.doi.org/10.5667/tang.2013.0009.
  128. 128. Semwal RB, Semwal DK, Combrinck S, Viljoen AM. Gingerols and shogaols: Important nutraceutical principles from ginger. Phytochemistry. 2015;117:554-568. DOI: 10.1016/j.phytochem.2015.07.012.
  129. 129. Rahmani AH, Shabrmi FM, Aly SM. Active ingredients of ginger as potential candidates in the prevention and treatment of diseases via modulation of biological activities. International Journal of Physiology, Pathophysiology and Pharmacology. 2014;6:125-136.
  130. 130. Gupta R, Singh PK, Singh R, Singh RL. Pharmacological activities of Zingiber officinale (ginger) and its active ingredients: A review. International Journal of Innovation Science and Research. 2016;4:1-18.
  131. 131. Lai YS, Lee WC, Lin YE, Ho CT, Lu KH, Lin SH, Panyod S, Chu YL, Sheen LY. Ginger essential oil ameliorates hepatic injury and lipid accumulation in high fat diet-induced nonalcoholic fatty liver disease. Journal of Agricultural and Food Chemistry. 2016;64:2062-2071/
  132. 132. Masuda Y, Kikuzaki H, Hisamoto M, Nakatani N. Antioxidant properties of gingerol related compounds from ginger. Biofactors. 2004;21:293-296.
  133. 133. Fuhrman B, Rosenblat M, Hayek T, Coleman R, Aviram M. Ginger extract consumption reduces plasma cholesterol, inhibits LDL oxidation and attenuates development of atherosclerosis in atherosclerotic, apolipoprotein E-deficient mice. The Journal of Nutrition. 2000;130:1124-1131.
  134. 134. Thomson M, Al Qattan, KK Al Sawan SM, Alnaqeeb MA, Khan I, Ali M. The use of ginger (Zingiber officinale Rosc.) as a potential anti-inflammatory and antithrombotic agent. Prostaglandins, Leukotrienes and Essential Fatty Acids. 2002;67:475-478.
  135. 135. Shoji N, Iwasa A, Takemoto T, Ishida Y, Ohizumi Y. Cardiotonic principles of ginger (Zingiber officinale Roscoe). Journal of Pharmaceutical Sciences. 1982;71:1174-1175. https://doi.org/10.1002/jps.2600711025.
  136. 136. Ghayur MN, Gilani AH. Ginger lowers blood pressure through blockade of voltage-dependent calcium channels. Journal of Cardiovascular Pharmacology. 2005;45:74-80. DOI: 10.1097/00005344-200501000-00013.
  137. 137. Beg T, Siddiqe Y S, Ara G, Gupta J, Afzal M. Antigenotoxic effect of genistein and gingerol on genotoxicity induced by norethandrolone and oxandrolone in cultured human lymphocytes. International Journal of Pharmacology. 2008;4:177-183.
  138. 138. Vinothkumar R, Sudha M, Nalini N. Chemopreventive effect of zingerone against colon carcinogenesis induced by 1,2-dimethylhydrazine in rats. European Journal of Cancer Prevention. 2014;23:361-371.
  139. 139. Teles AM, dos Santos BA, Ferreira CG, Mouchreck AN, da Silva Calabrese K, Abreu-Silva AL, Almeida-Souza F. (December 6th 2019). Ginger (Zingiber officinale) antimicrobial potential: A review. In: Ginger cultivation and its antimicrobial and pharmacological potentials, Haiping Wang, IntechOpen. DOI: 10.5772/intechopen.89780.
  140. 140. Okiki AP, Oyetunji O, Benjamin O. Antibacterial activity of ginger (Zingiber officinale) against isolated bacteria from the respiratory tract infections. Journal of Biology, Agriculture and Healthcare. 2015;5:131-138.
  141. 141. Nagoshi C, Shiota S, Kuroda T, Hatano T, Yoshida T, Kariyama R, et al. Synergistic effect of [10]-gingerol and aminoglycosides against vancomycin-resistant enterococci (VRE). Biological and Pharmaceutical Bulletin. 2006;29:443-447. DOI: 10.1248/bpb.29.443.
  142. 142. Imanishi N, Andoh T, Mantani N, Sakai S, Terasawa K, Shimada Y, et al. Macrophage mediated inhibitory effect of Zingiber officinale Rosc, a traditional oriental herbal medicine, on the growth of influenza A/Aichi/2/68 virus. The American Journal of Chinese Medicine. 2006;34:157-169. DOI: 10.1142/S0192415X06003722.
  143. 143. Chang JS, Wang KC, Yeh CF, Shieh DE, Chiang LC. Fresh ginger (Zingiber officinale) has anti-viral activity against human respiratory syncytial virus in human respiratory tract cell lines. Journal of Ethnopharmacology. 2013;145:146-151.
  144. 144. Sahoo M, Jena L, Rath SN, Kumar S. Identification of suitable inhibitor against influenza A (H1N1) neuraminidase protein by molecular docking. Genomics and Informatics. 2016;14:96-103.
  145. 145. Mahady GB, Pendlan SL, Yun GS, Lu Z-Z, Stoia A. Ginger (Zingiber officinale Roscoe) and the gingerols inhibit the growth of Cag A+ strains of Helicobacter pylori. Anticancer Research. 2003;23:3699-3702.
  146. 146. Ebrahimzadeh Attari V, Somi MH, Asghari Jafarabadi M, Ostadrahimi A, Moaddab SY, Lotfi N. The gastro-protective effect of ginger (Zingiber officinale Roscoe) in Helicobacter pylori positive functional dyspepsia. Advanced Pharmaceutical Bulletin. 2019;9:321-324. DOI: 10.15171/apb.2019.038.
  147. 147. Zehsaz F, Farhangi N, Mirheidari L. The effect of Zingiber officinale R. rhizomes (ginger) on plasma pro-inflammatory cytokine levels in well-trained male endurance runners. Central European Journal of Immunology. 2014;39:174-180.
  148. 148. 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. Journal of Ethnopharmacology. 2018;214:113-123
  149. 149. 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. Bioorganic Chemistry. 2001;29:156-163.
  150. 150. Lin R-J, Chen C-Y, Chung L-Y, Yen C-M. Larvicidal activities of ginger (Zingiber officinale) against Angiostrongylus cantonensis. Acta Tropica. 2010;115:69-76. DOI: 10.1016/j.actatropica.2009.12.007.
  151. 151. Lin RJ, Chen C-Y, Lee J-D, Lu C-M, Chung L-Y. Larvicidal constituents of Zingiber officinale (ginger) against Anisakis simplex. Planta Medica. 2010;76:1852-1858.

Written By

Dennis R.A. Mans, Priscilla Friperson, Meryll Djotaroeno and Jennifer Pawirodihardjo

Reviewed: 15 April 2021 Published: 10 May 2021