Next Article in Journal
In Vitro–In Vivo Correlation of Tianeptine Sodium Sustained-Release Dual-Layer Tablets
Next Article in Special Issue
Nutraceutical Difference between Two Popular Thai Namwa Cultivars Used for Sun Dried Banana Products
Previous Article in Journal
On the Development of Phenol-Formaldehyde Resins Using a New Type of Lignin Extracted from Pine Wood with a Levulinic-Acid Based Solvent
Previous Article in Special Issue
Optimization Extraction and Antioxidant Activity of Crude Polysaccharide from Chestnut Mushroom (Agrocybe aegerita) by Accelerated Solvent Extraction Combined with Response Surface Methodology (ASE-RSM)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plants of the Genus Zingiber: A Review of Their Ethnomedicine, Phytochemistry and Pharmacology

1
College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
2
Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(9), 2826; https://doi.org/10.3390/molecules27092826
Submission received: 18 March 2022 / Revised: 18 April 2022 / Accepted: 20 April 2022 / Published: 29 April 2022
(This article belongs to the Special Issue Chemical Compositions and Bioactivities of Foods)

Abstract

:
Plant of the genus Zingiber (Zingiberaceae) have primarily distributed in subtropical and tropical Asia, South America and Africa. The species of this genus have been widely used as food and in folk with a long history for treating various diseases. Reports related to the phytochemistry and phytochemistry of Zingiber species are numerous, but articles on the summary of the genus Zingiber remain scarce. This review aims at presenting comprehensive information about the genus Zingiber and providing a reference for the future application by systematically reviewing the literature from 1981 to 2020. Currently, a total of 447 phytochemical constituents have been isolated and identified from this genus, in which volatile oils, diarylheptanoids, gingerols, flavonoids and terpenoids are the major components. Gingerols, which are the main functional components, are the spicy and aromatic ingredients in the Zingiber species. Extracts and single compounds from Zingiber plants have been discovered to possess numerous biological functions, such as anti-inflammatory, anticancer, antimicrobial, larvicidal, antioxidant and hypoglycemic activities. This review provides new insights into the ethnomedicine, phytochemistry and pharmacology of the genus Zingiber and brings to the forefront key findings on the functional components of this genus in food and pharmaceutical industries.

1. Introduction

The genus Zingiber is the third largest of the family Zingiberaceae, whose members are mostly edible and medical plants [1]. It comprises 141 species, of which 12 species are native to China, southwest China in particular [2]. The plants of this genus are mostly perennial herbs with a fibrous rhizome, erectedg stem, and aromatic odor. The roots of Zingiber plants are mainly used for food and medicine, and the stems, leaves and roots are also used for extracting aromatic oils. Many types of chemical compounds of Zingiber have been discovered in current studies, such as volatile oils, organic acid, sterides, flavonoids, diarylheptanoids, gingerols and terpenoids. Although numerous chemical constituents of the genus Zingiber have been reported, including some well-known compounds named 6-gingerol (325), zerumbone (1) and curcumin (294), a systematic summary of the chemical constituents of this genus was rarely reported.
As traditional medical and edible herbs, numerous studies have focused on five plants of the genus Zingiber (Zingiber zerumbet (L.) Smith (Figure 1a), Zingiber officinale Rosc (ginger) (Figure 1b), Zingiber corallinum Hance (Figure 1c), Zingiber mioga (Thunb.) Rosc (Figure 1d) and Zingiber striolatum Diels (Figure 1e). Modern pharmacological studies demonstrated that they exhibit extensive biological activities, such as antimicrobial, larvicidal, antioxidant, anti-obesity, anti-inflammatory, hypoglycemic, neuroprotective, cardiovascular protective and anti-tumor effects. According to their usage in history, Zingiber plants have been used to treat various symptoms and diseases, including nausea, vomiting, cough, common cold, and headache, relieving joint pain and menstrual cramp, and preventing stomach ulcers, neurodegenerative diseases, eye inflammation, cardiovascular diseases, diuretic and respiratory disorders [3,4,5].
Despite scholars having conducted extensive research on this genus in recent years, reports on summaries of the genus Zingiber remain scarce. Most of the research is especially concentrated on the Z. officinale, which is abundant in China, and is distributed mainly in the southwest to southeast of China, especially in the Guangdong, Guangxi, Yunnan, Sichuan and Guizhou provinces [6]. However, the details on the other species of this genus are scanty. Therefore, in this review, the ethnomedicine, chemical compositions and pharmacological activities from the available research reports on the genus Zingiber were systematically summarized and presented.

2. Methods of Data Collection

2.1. Methods

All of the available information presented in this review, concerning the genus Zingiber, was gathered via the scientific database, including PubMed, Google Scholar, Web of Science and Chinese National Knowledge Infrastructure (CNKI) between 1981 and 2020. In addition, part of the information was obtained from some local books, PhD and master’s dissertations. Keywords, such as Zingiber, phytochemical composition, bioactivities of Zingiber, and Zingiberaceae, were used for the online search. The structures of the Zingiber chemical contents, which were mentioned in the articles, were obtained from theses, books, databases and other reliable sources.

2.2. Inclusion Criteria

Using the above-mentioned methods, we selected literature (1981–2020) concerning the ethnomedicine, phytochemistry and pharmacology of the genus Zingiber. Non-English articles were included if they included eligible study design and relevant outcomes. Considering the comprehensiveness of this review, all types of articles (books, research, reviews, clinical trials and meta-analysis) are included and ultimately 130 eligible articles as the result of the selection process. The details of screening and selecting eligible articles are available in Figure 2.

2.3. Exclusion Criteria

We excluded the articles whose study design is unreasonable or whose outcome is ambiguous.

3. Traditional Uses and Modern Applications

3.1. Traditional Medical Uses and Modern Applications of Zingiber Plants in China

Plants of the genus Zingiber have been used in China for centuries, which are recorded for treating cough, emesis, rhinobyon and typhia in the monographs of traditional Chinese medicine, such as Sheng Nong’s herbal classic and the Compendium of Materia Medica [7]. Z. officinale is the representative herbal medicine of the genus Zingiber, which is used as a common medication to relieve cough, cold, vomiting, diarrhea and abdominal pain in the traditional Chinese medicine system. Another species (Z. zerumbet) in this genus was recorded to have treatment effects of joint pain, stomachache, cold and dysmenorrhea. Z. striolatum has been documented to relieve diabetes and constipation in the Compendium of Materia Medica.
In addition to their traditional uses, plants of Zingiber can be used as prescription oral treatment for coronary heart disease, myocardial infarction, cardiovascular disease and enteritis [8,9]. Z. officinale has been found as a safe and well tolerated alternative to anti-emetic medications, which can decrease the severity and incidence of PONV (postoperative nausea and vomiting) [10]. Moreover, Z. officinale, as the best-known plant of the genus Zingiber, has different uses through different processing methods. Dried Z. officinale was used to cure enteritis, diarrhea and emesis, whereas baked Z. officinale was cured hemafecia, metrorrhagia and metrostaxis in the Chinese medicine system [11]. Ethyl-acetate extract of Z. zerumbet has a protective effect against ethanol-induced brain damage, which is mediated through its antioxidant properties [12]. With the unique fragrance and strong antimicrobial and insecticidal functions, Z. corallinum Hance has been widely used in the washing, fragrance and cosmetic industry.

3.2. Traditional Medical Uses and Modern Applications of Zingiber Plants in Other Countries

Plants of the genus Zingiber have played significant roles in the medicine system of many other countries. Z. mioga (Figure 1d) was used to ameliorate inflammation, rheumatic disorders and gastrointestinal discomforts in traditional Oriental medicines [13]. Z. officinale (Figure 1b) is employed as an important medicine for treating catarrh, rheumatism, nervous diseases, gingivitis toothache, asthma, stroke, constipation, and diabetes in the Ayurvedic and Tibb-Unani herbal medicines [14]. Moreover, Z. officinale has the effects of treating asthma, bronchitis, piles, eructation, constipation, ascites and relieving flatulence in the Indian medicine system [15,16]. The rhizome powder of Z. zerumbet (Figure 1a) mixed with ripe Morinda citrifolia is used for the treatment of severe pain in India. The fresh rhizome of Z. zerumbet is served as an anti-flatulent agent in Thailand. In addition, it also has a long history in the treatment of headache, toothache, ringworm, arthralgia, sprains and stomach-ache by Hawaiians [5].
Besides their medicinal uses, plants of Zingiber are the vital ingredient in the daily diets in other countries. The flower buds of Z. mioga, which have a pungent aroma, are used as spices, pickles and health supplements in Eastern Asia. As well, it is an excellent food ingredient for increasing the functionality and consumer acceptability of kimchi [17,18]. The leaves of Z. mioga can be used to wrap and preserve manjyu, which is a traditional Japanese confection [19].

4. Phytochemical Contents

A total of 447 compounds have been isolated and identified from the genus Zingiber, which can be classified into seven categories, including volatile oils, terpenoids analogues, flavonoids, gingerol analogues, diarylheptanoids, organic acids and sterides. Those compounds and their origins have been summarized in Table S1 from Supplementary Materials, and their chemical structures have been described in Figure 3, Figure 4, Figure 5, Figure 6, Figure 7, Figure 8, Figure 9 and Figure 10.

4.1. Volatile Oils

Compounds 1106 are the volatile oils that were isolated and identified from Zingiber plants (Figure 3). The volatile oils were extracted from the rhizome of Zingiber plants, which account for roughly 0.25~3.0%. The α-zingiberene (219) is the main component of the Z. officinale volatile oil [20,21]. Zerumbone (1) is the predominant component of Z. zerumbet volatile oils, whose analogs account for approximately 60.3% [22]. Palmitic acid (33) was identified as the highest content of the Z. striolatum Diels volatile oils, which account for about 30.5% [23].

4.2. Terpenoid Analogues

Compounds 107261 are the terpenoid analogues. The odor of Zingiber plants can be attributed to the terpenoids’ compounds, which mainly consist of monoterpene (Figure 4) and sesquiterpene (Figure 5), such as ar-curcumene (224) and β-sesquiphellandrene (225) [24]. Three diterpene dialdehydes’ compounds, named galanal A (260), galanal B (261) and miogadial (198), were isolated from Z. mioga, in which the compound 198 was the main contribution to the pungent taste of the flower buds [25].

4.3. Flavonoids

A total of 32 flavonoids (compounds 262293) have been identified from the Zingiber plants (Figure 6). The flavonoids’ content of the Z. mioga bud was 0.48%, which was much higher than that of common vegetables (0.001–0.1%). Two anthocyanin compounds, delphinidin (292) and cyanidin (293), were considered as the potential hypoglycemic components in Z. mioga [26].

4.4. Diarylheptanoids

Twenty-seven diarylheptanoid compounds (294320) were detected in the genus Zingiber, and curcumin (294) is the first diarylheptanoid compound that was isolated from this species (Figure 7). Diarylheptanoids are a class of compounds with 1,7-disubstituted phenyl groups and heptane framework, which can be subdivided into acyclic (294314) and cyclic diarylheptanoids. The aromatic rings of diarylheptanoids are easily hydroxylated and methoxylated, with the alkyl chains containing ketone, alkene, alcohol, and acetyl functionalities, and 1,5- or 3,6-oxy bridges (315320).

4.5. Gingerol Analogues

Forty-five gingerol analogues compounds (321367) were isolated and identified from the Zingiber species, which were the main metabolites of Zingiber plants (Figure 8). Gingerol analogues can be classified into six categories, including gingerol, shogaol, zingerone, paradol, gingerdione and gingerdiol, based on the difference of hydrocarbon chains [27]. Different gingerol analogues demonstrated similar biological activity, which may be influenced by the lengths of alkyl side chains. 6-gingerol (325) is the highest-content chemical component of gingerol analogues and the main pungency constituent of the fresh Z. officinale rhizome [28]. Due to their thermal instability, gingerols easily undergo dehydration reactions to form the corresponding shogaols (332338), which are the non-volatile pungent ingredients in the Zingiber species. 6-paradol (355) is produced from 6-shogaol (333) by a biotransformation process and possesses similar biological activities to 6-shogaol (333) [29]. 6-gingesulfonic acid (365) accounts for 0.0013% of Z. officinale, with weak pungency and strong anti-ulcer activity [30].

4.6. Organic Acids

Compounds 368395 were isolated and identified as organic acids in Zingiber plants (Figure 9). These ingredients can be divided into fatty acids (compound 390) and aromatic acids (compound 368). Oxalic and tartaric acids (394395) are two major acids of Z. officinale rhizomes [31].

4.7. Sterides

Only two sterides compounds have been found in the genus Zingiber, including daucosterol (396) and 24-propylcholesterol (397) (Figure 10).

4.8. Others

Compounds 398447 have been isolated and identified from the genus of Zingiber, including anthraquinones (441), furans (400401), pyrimidine (408409) and saccharides (407 and 447) (Figure 10).

5. Pharmacological Properties

Zingiber plants possess anti-inflammatory, anticancer, antimicrobial, larvicidal, antioxidant, hypoglycemic, analgesic, neuroprotective and cardiovascular protective effects. These pharmacological properties have been summarized in Table 1 in the following subsections.

5.1. Anti-Inflammatory Activity

The extracts of Zingiber plants have been used to treat inflammation. The extract of Z. zerumbet inhibited the inflammatory-mediated signaling pathways (NF-κB, MAPK and PI3K-Akt) in human macrophages via suppression of the release of pro-inflammatory mediators and the mRNA expression of pro-inflammatory factors [47]. The total essential oil of Z. officinale (28 mg/kg/d) prevents chronic joint inflammation, which may be attributed to the combined effects of both the aromatic essential oil and gingerols [48].
The anti-inflammatory effects of the single metabolites originated from the Zingiber plants have been well investigated. Zerumbone (1) has the effect of reducing the inflammatory response of acute lung injury (ALI) by inhibiting the Akt-NFκB activation [49]. 6-shogaol (333) and 6-dehydrogingerdione (357) display the inhibiting effect for binding between sICAM-1 (cell adhesion molecules) and VLA-4 (very late antigen) of the THP-1 (human monocytic cell line) cells, which are the main features of inflammation initiation. In addition, 10-gingerol (327), 6-shogaol (333), 8-shogaol (334) and 6-dehydrogingerdione (357) can inhibit direct binding between sICAM-1 and LFA-1 (lymphocyte function-associated antigen) of the THP-1 cells with IC50 value of 57.6, 27.1, 65.4 and 62.0 μM, respectively [32]. Moreover, the selective inhibition of pro-inflammatory cytokines for 6-gingerol (325) was also detected [33]. A few papers suggest that the strong anti-neuroinflammatory effects of Z. officinale are owing to 10-gingerol (327), not 6-gingerol (325). Those effects can be attributed to the inhibition of proinflammatory gene expression by blocking NF-κB activation, which can conduce to a reduced level of NO, IL-1β, IL-6 and TNF-α [50].

5.2. Anticancer Activity

The extracts and single metabolites originating from the Zingiber plants, especially in the essential oils, diarylheptanoids and gingerols, display significant anticancer activity. The essential oils from the fresh rhizome of Z. zerumbet displayed obvious cytotoxicity against K562 (human leukemia cell lines), PC-3 (human prostate cancer cell lines), A549 (human lung cancer cell lines) and MRC-5 (human fetal lung fibroblasts cell lines) cells with IC50 values range from 35.73 to 216.99 μM, which is stronger than that of the dry rhizome [22]. Zerumbone (1) is the main essential oil of Z. zerumbet, which exhibits a significant inhibitory effect on Hela (human cervical cancer cell lines), H460 (human lung cancer cell lines) and A549 cell lines with the IC50 value of 6.4 μg/mL, 15 and 25 μM, respectively [38]. β-myrcene (171) exhibited in vitro cytotoxicity on HepG2 (human liver cancer cell line), HCT116 (colon cancer cell line) and MCF7 (breast cancer cell line) cells with an IC50 value ranging from 2.51 to 3.28 μg/mL [35]. α-pinene (160) and β-pinene (182) also demonstrate strong cytotoxicity to SK-OV-3, HO-8910 and Bel-7402 cells (human tumor cell lines) [51]. Galanals (260261) can induce the death of Jurkat human T-cell leukemia cells, which are characterized by DNA fragmentation and caspase-3 activation.
3,5-dioxo-1,7-bis(3-methoxy-4-hydroxy)-phenyl-heptane (313), which belongs to the diarylheptanoid type compound, displayed cytotoxic effect on BEL7404 (human lung cancer cell lines), CNE (human nasopharyngeal carcinoma cell lines), Hela and KB (human oral epithelial cancer cell lines) cells with IC50 values of 49.4 ± 3.4, 76.7 ± 5.4, 86.8 ± 10.5 and 27.7 ± 2.7 μM, respectively. Curcumin (294) and Gingerenone A (311) also exhibited a stronger inhibitory effect in those human cancer cell lines [36].
Gingerols, as the major active components of Zingiber plants, possess remarkable anti-cancer activities as well. The aliphatic chain and hydroxyl moieties that existed in 6-gingerol (325) and 6-shogaol (333) were proven to be responsible for the anti-cancer activities, and 6-gingerol (325) had the potential to bind with DNA and induced cell death by autophagy and caspase-3 mediated apoptosis [28]. The HepG2 cell lines can be induced by 6-gingerol (325) via autophagy and caspase-3 mediated apoptosis, and 6-gingerol (325) also demonstrates cytotoxic effect on K562 as well [34,39]. 10-gingerol (327), 6-shogaol (333) and 6-dehydrogingerdione (357) exhibited a cytotoxic effect on multiple cancer cells (BEL7404, Hela and KB cell lines), while 6-shogaol (333) has a stronger inhibitory effect than the other two compounds.
The quantitative structure–activity relationship (QSAR) models found that the cytotoxicity was related to compound lipophilicity because it may increase the permeability of the cancer cell membrane [36]. This means that gingerols with longer unbranched alkyl side chains may have greater anticancer potential because of their increased lipophilicity (Figure 11a).

5.3. Antimicrobial Activity

The essential oils and gingerols originating from the Zingiber plants display significant antimicrobial activities and the details are available in Table S2 from Supplementary Materials. Essential oils of the Z. officinale rhizome have the significant effect of inhibiting the growth of both Gram-negative and Gram-positive bacteria [45]. Zerumbone (1) is the main antimicrobial ingredient of the Z. zerumbet essential oils, with a mid-to-high IC50 value against staphylococcus aureus, bacillus subtilis, escherichia coli and proteus vulgaris [52]. It also demonstrates an anti-virulence effect by inhibiting the biofilm formation and hyphal growth of C. albicans in a concentration-dependent manner, and it exhibited antimicrobial activity at the MIC (minimum inhibitory concentration) of 250 μg/mL against the H. pylori strain [53,54]. Three compounds named miogadial (198), galanal A (260) and galanal B (261) that isolated from Z. mioga, had antimicrobial activities against different strains of bacteria, yeasts and molds. However, miogadial (198) demonstrated stronger antimicrobial activity against Gram-positive bacteria and yeasts than compounds galanal A (260) and galanal B (261) [25]. The essential oil of Z. corallinum Hance also can inhibit the growth of numerous plant pathogenic fungi with a low concentration.
The majority of gingerol analogues displayed strong antimicrobial activity. For instance, 10-gingerol (327) has the stronger inhibitory effect on three periodontal bacteria than 12-gingerol (328), with a MIC range from 6 to 14 μg/mL and a MBC (minimum bactericidal concentration) range from 4 to 14 μg/mL. Bacteria of Mycobacterium avium and Mycobacterium tuberculosis, which may cause tuberculosis, can also be suppressed by 10-gingerol (327) as well. Furthermore, 10-gingerol (327) and 12-gingerol (328) from the Z. officinale rhizome have antibacterial activity against periodontal bacteria, with the MBC ranging from 4 to 20 μg/mL. The compound 6-gingerol (325) demonstrated a strong antimicrobial effect on Helicobacter pylori, with a MIC of 20 μg/mL. The structure–activity relationships (SARs) demonstrated that the shorter alkyl side chain of gingerols play a key role for their microbial inhibition effect (Figure 11a) [55].

5.4. Larvacidal Activity

The essential oils of Zingiber plants are proven to be an effective tool for mosquito larval control and the details are available in Table S3. The Z. corallinum Hance essential oil exhibited a stronger poison effect against the larval of Aedes albopictus and Culex quinquefasciatus than pupa [56]. Both methanol and dichloromethane extracts of Z. zerumbet rhizomes exhibited significant larvacidal effect. The dichloromethane extracts displayed more toxicity to the larvae of Aedes aegypti and Anopheles nuneztovari than the methanol extracts, which may be attributed to the essential oil. In addition, the gingerols were the vital insecticidal composition of the genus Zingiber. 4-gingerol (324) demonstrated strong larvicidal activity against Aedes aegypti and Culex quinquefasciatus [57]. β-sitosterol (397) was highly effective against Aedes aegypti, Anopheles stephensi and C. quinquefasciatus, with an LC50 value of 11.49, 3.58 and 26.67 ppm, respectively [58].

5.5. Antioxidant Activity

The polyphenols, flavonoids, gingerols and essential oils originating from the Zingiber plants demonstrated significant antioxidant activity. The polyphenols and flavonoids of Z. mioga account for approximately 0.5% and 4.6%, respectively, which were far higher than that in common vegetables (0.001~0.1%), manifesting its potential antioxidant properties. The ethanol extract of Z. mioga, compared to the water extract, exhibited stronger peroxyl radical scavenging linked antioxidant activity (0.53/TE 1 μM) with 2697.31 ± 118.25 mg/100 g of the total antioxidant capacity (TAC) [59]. The ethyl-acetate extract of Z. zerumbet at 400 mg/kg has a protective effect against ethanol-induced brain damage because of its antioxidant properties [11]. The antioxidant capacity of the methanolic extract of Z. officinale has been assessed with the DPPH assay (86.26 ± 0.97%), ABTS assay (91.04 ± 0.96%) and nitric oxide assay (86.72 ± 1.51%) [60].
The essential oils, such as zerumbone (1), display significant antioxidant power with a FRAP (Ferric-reducing antioxidant power) value of 58.3 ± 2.08, which is higher than that of ascorbic acid, by enhancing the cellular antioxidant pathway [53]. In addition, some gingerol analogues exhibited conspicuous antioxidant activity. 6-gingerol (325), 8-gingerol (326), 10-gingerol (327) and 6-shogaol (333) represented antioxidant effects with a IC50 value range from 8.05 to 26.3 μM against the DPPH radical. 10-gingerol (327) demonstrates a stronger quenching ability of DPPH radicals than curcumin (294), but a weaker quenching ability than quercetin (286). The highest antioxidant activity of 6-shogaol (333) can be attributed to the presence of unsaturated ketones moieties. The SAR demonstrated that the substituent groups and the length of the alkyl chain play a crucial role for their antioxidant effects and the presence of α; the β-unsaturated ketone moiety is predominant to that of the alkyl side chains’ length in exhibiting the antioxidant and anti-inflammatory properties (Figure 11b) [61,62].

5.6. Hypoglycemic Activity

The extracts and single metabolites originating from the Zingiber plants display significant hypoglycemic activity. The sucrase, maltase and α-amylase were significantly suppressed by the ethanol extract of Z. mioga. Moreover, the ethanol extract of Z. mioga exhibited it’s possibility of acting as an intestinal α-glucosidase inhibitor by using SD rat and db/db mice models [52]. The hypoglycemic activity of Z. mioga was considered to be the presence of the anthocyanin compound (292293) [26]. The ethanol extract of Z. striolatum has a dose-dependent hypoglycemic effect on insulin-resistant HepG2 cells with low cytotoxicity [63]. A daily feeding of 200 mg/kg ethanolic extract of Z. officinale for 20 days can significantly decrease blood glucose [64]. In addition, it also inhibits LDL (low-density lipoprotein) oxidation [65] and HMG-COA (3-hydroxy-3-methylglutaryl coenzyme A) reductase and increases insulin release [66].
Gingerols exert their anti-diabetic effects primarily by activating AMPK (AMP-activated protein kinase), which regulates the glucose and lipid metabolism energy sensor. The steaming process would enhance the anti-diabetic potential of Z. officinale via increasing the content of 6-dehydrogingerdione (357), which could stimulate the insulin secretion by the closure of KATP (ATP-sensitive potassium channels) in pancreatic β-cells [67]. Furthermore, Gingerenone A (311) is equipped to sensitize the insulin receptor and increase glucose uptake by inhibiting the activity of p70 S6 kinase [68].

5.7. Prevention of Nausea and Vomiting Activity

Z. officinale has been used as an antiemetic in various traditional medicine systems for over 2000 years, and it remains considered as an alternative therapy for nausea and vomiting in modern medicine [69]. Doctors in Thailand used Z. officinale as a drug to prevent nausea and vomiting after laparoscopic surgery for gynecological outpatients [7]. Besides its medical application on postoperative nausea, Z. officinale was used for motion sickness and pregnancy-induced nausea and vomiting as well; those effects of reducing nausea and vomiting might be associated with a weak inhibitory effect of gingerols and shogaols on M3 and 5-HT3 receptors or exert their anti-emetic effect by acting on the 5-HT3 receptor ion-channel complex [70].

5.8. Others

Several other pharmacological activities of the components or extracts of the Zingiber species have been found in previous research, such as analgesic, anti-ulcer, neuroprotective and cardiovascular protection. The genus Zingiber, especially Z. officinale, possess remarkable analgesic activity, which could be due to their phytoconstituents binding to TRPA1 (ankyrin receptors) and TRPV1 (vanilloid receptors) ion channels [71]. The significant central and peripheral antinociceptive effects of the Z. zerumbet essential oil has been detected [72]. Zerumbone (1) displayed anti-hyperalgesic properties via suppressing the pain transmission from primary afferent neurons to the ascending tract and modulating pain impulses reaching the supraspinal regions [73].
An intraperitoneal injection of 25 mg/kg–50 mg/kg of 6-gingerol (325) into a rat can generate the inhibitory effect of the acetic acid-induced writhing response and formalin-induced licking time [74]. Z. zerumbet enhanced the protection of the ethanol-induced gastric ulcer by the effects of maintaining mucus integrity, antioxidant activity and HSP-70 induction [42]. Furthermore, Z. officinale powder can obviously improve the gastric mucosa injury caused by aspirin as well [75]. Moreover, 6-gingerol (325) was tested and has the ability of preventing the acrylonitrile-induced cerebral cortex lesion as well, with the increase in the brain immunohistochemical expression of caspases-9 and caspases-3. It would be a better fungible drug for the prevention of neurodegenerative diseases when compared to some synthetic drugs in [76]. The presence of a double bond and the linear chain of 6-shogaol (333) may enhance the neuroprotective effects of this compound (Figure 11b).

6. Conclusions

In this work, the phytochemical constituents and pharmacological effects of the Zingiber species was first reviewed systematically, based on the literature from 1981 to 2020. A total of 447 metabolites are included in this review, of which 34.78% are volatile oils, 23.70% are terpenoids, 6.96% are flavonoids, 5.87% are diarylheptanoids, 9.78% are gingerols analogues, 6.30% are organic acids and 12.61% are classified as other compounds. These compounds, including zerumbone (1), zingerone (322), curcumin (294) and gingerols (321367), are considered the characteristic constituents of this genus. Gingerols are the main pungency components of the genus Zingiber, in which the 6-gingerol (325) accounts for more than 75% [7].
The anti-inflammatory, anticancer and antimicrobial effects are the main biological activities of the extract or single compound of these genus plants. As we all know, most plants of the genus Zingiber are medicine-food homology herbs. Therefore, the extract or single compound has a huge potential for the development of new food additives for their obvious biological activity. Some bioactive constituents of the Zingiber plants, such as 6-gingerol, 10-gingerol and 12-gingerol, displayed stronger antimicrobial activity, and are regarded as attractive targets in food contaminations management. In addition, the essential oil of Z. corallinum Hance holds great potential as an environmentally friendly pesticide, with a remarkable inhibitory effect of numerous plant pathogenic fungi. There is some evidence that Zingiber plants (Z. officinale, Z. mioga and Z. striolatum) may provide potential benefits on metabolic syndromes (obesity and type-2 diabetes). It is noteworthy that the ethanol extract demonstrates stronger hypoglycemic activity compared to the water extract in most animal studies. This may be due to ethanol extracting more flavonoid compounds, such as delphinidin (292) and cyanidin (293). As extensively used hypoglycemic drugs, such as acarbose, can cause side effects such as nausea, vomiting, gastrointestinal swelling and kidney function disorders, plants of the genus Zingiber could be used as a complementary or alternative medicine to diabetes therapy. Z. officinale has been used as an antiemetic for over 2000 years, which would be associated with gingerols and shogaols, and it remains considered as an alternative therapy for nausea and vomiting in motion sickness by the Committee on Herbal Medicinal Products (HMPC).
Despite possessing the significant pharmacological activity of some Zingiber constituents, the clinical applications are still rare, considering the uncertain safety of their consumption in humans. Furthermore, as products and studies related to the mechanisms underlying the pharmacological activity derived from those herbs remain scarce, more laboratory investigations and product developments are needed.
Structural modification by heating and dehydration, as well as enzyme reactions, may be helpful in enhancing the biological activities of Zingiber plants. Shogaols (332336) are produced from gingerols (323327) by heating, which are more effective in inhibiting the inflammatory mediators and ROS production and possess better thermostability as compared to gingerols (323327). 6-paradol (355) is the non-pungent metabolite of 6-shogaol (333), which may avoid side effects such as gastric irritation. However, the structure–activity information related to Zingiber plants remains scarce, due to the variability of experimental outcomes. These metabolite contents were very low in Zingiber plants or difficult to isolate due to their structural similarity. Therefore, more extensive studies in this direction are needed for future clinical applications or for serving as adjuvants.
In conclusion, Zingiber plants are the herbs in homologous medicine and food that have been widely used in different countries for centuries. Our present paper provides comprehensive information on the traditional uses, phytochemistry and pharmacology of the genus Zingiber. We highlight the enormous potential of the Zingiber plants to serve as potent clinical drug candidates, in order to provide a scientific foundation for future research and application on this genus.

Supplementary Materials

The following supporting information can be downloaded: https://www.mdpi.com/article/10.3390/molecules27092826/s1, Table S1: A comprehensive list of chemical constituents of Zingiber plants.; Table S2: Antimicrobial effect of Zingiber plants.; Table S3: Larvicidal effect of Zingiber plants. References from [77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128] are cited in the Supplementary Materials.

Author Contributions

M.D. wrote the original review and drew the chemical structures; X.Y. and S.R. helped to collect the data; F.L. discussed the data and gave some revised suggestions; Z.Q. designed the outline of the review and gave some revised suggestions. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financially supported by the Hong Kong Scholars Program (2021-164), the Hunan Provincial Natural Science Foundation of China (2021JJ40245), the Hunan Provincial Key Research and Development Project (2020NK2031), the National Key Research and Development Program of China (2021YFD1600301), and the Scientific Research Project of Hunan Education Department (19C0895).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Wu, Q.G.; Liao, J.P. Anatomy and Histochemistry of the Seeds of Zingiber boehm. Acta Bot. Boreali-Occident. Sin. 1995, 15, 32–39. [Google Scholar] [CrossRef]
  2. Tan, J.W.; Israf, D.A.; Tham, C.L. Major Bioactive Compounds in Essential Oils Extracted from the Rhizomes of Zingiber zerumbet (L.) Smith: A Mini-Review on the Anti-allergic and Immunomodulatory Properties. Front. Pharm. 2018, 20, 652. [Google Scholar] [CrossRef] [Green Version]
  3. Mao, Q.Q.; Xu, X.Y.; Cao, S.Y.; Gan, R.Y.; Corke, H.; Beta, T.; Li, H.B. Bioactive Compounds and Bioactivities of Ginger (Zingiber officinale Roscoe). Foods 2019, 8, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Li, Z.H.; Bai, X.; Du, J.X.; Li, J.Y.; He, J.; Ai, H.L. Zingiberols A–C, three rare diterpenols and their cytotoxicities from Zingiber mioga. Phytochem. Lett. 2019, 33, 106–109. [Google Scholar] [CrossRef]
  5. Yob, N.J.; Jofrry, S.M.; Affandi, M.M.; Teh, L.K.; Salleh, M.Z.; Zakaria, Z.A. Corrigendum to “Zingiber zerumbet (L.) Smith: A Review of Its Ethnomedicinal, Chemical, and Pharmacological Uses”. Evid.-Based Complement. Altern. Med. 2016, 2016, 8621824. [Google Scholar] [CrossRef] [Green Version]
  6. Wang, Z.; Jin, Y.R.; Li, X.W. Study on the Chemical Constituents of the Zingiberis Rhizoma. China Pharm. 2017, 28, 3403–3405. [Google Scholar] [CrossRef]
  7. Xiang, J.M.; Xu, L.J.; Xiao, W.; Xiao, P.G. Research Progress in Zingiber officinale. Chin. Pharm. J. 2017, 52, 353–357. [Google Scholar] [CrossRef]
  8. Tang, Q.M.; Yan, J.; Wu, Z.J.; Li, W.Q.; Qing, Z.X.; Luo, F.L. Nutritional & Healthy Function and Processing Status of Zingiber mioga Rosc. Farm Prod. Process. 2019, 18, 89–92. [Google Scholar] [CrossRef]
  9. Zhang, S.J.; Zhang, Y.G.; Xin, E.D.; Xin, E.D.; Zhang, A.X.; Niu, J.T.; Bian, T.T.; Si, X.L.; Li, Y.F. Pharmacological action of Zingiberis Rhizoma Recens. J. Gansu Univ. Chin. Med. 2020, 37, 79–81. [Google Scholar] [CrossRef]
  10. TóTh, B.; Lantos, T.; Hegyi, P.; Viola, R.; Vasas, A.; Benkő, R.; Gyöngyi, Z.; Vincze, Á.; Csécsei, P.; Mikó, A.; et al. Ginger (Zingiber officinale): An alternative for the prevention of postoperative nausea and vomiting. A meta-analysis. Phytomedicine 2018, 50, 8–18. [Google Scholar] [CrossRef]
  11. Sun, F.J.; Li, Z.L.; Qian, S.H.; Pu, S.B. Research Advances on the Zingiberis Rhizoma. Chin. Wild Plant Resour. 2015, 34, 34–37. [Google Scholar] [CrossRef]
  12. Hamid, A.; Ibrahim, F.W.; Ming, T.H.; Nasrom, M.N.; Eusoff, N.; Husain, K.; Abdul, L.M. Zingiber zerumbet L. (Smith) extract alleviates the ethanol-induced brain damage via its antioxidant activity. BMC Complement. Altern. Med. 2018, 18, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Shin, N.R.; Shin, I.S.; Jeon, C.M.; Hong, J.M.; Kwon, O.K.; Kim, H.S.; Oh, S.R.; Hahn, K.W.; Ahn, K.S. Zingiber mioga (Thunb.) Roscoe attenuates allergic asthma induced by ovalbumin challenge. Mol. Med. Rep. 2015, 12, 4538–4545. [Google Scholar] [CrossRef] [PubMed]
  14. Mehdi, S.R.; Maria, V.E.; Bahare, S.; Javad, S.R.; Karl, R.M.; Seyed, A.A.; Farzad, K.; Salam, A.I.; Dima, M.; Zainul, A.Z.; et al. Plants of the Genus Zingiber as a Source of Bioactive Phytochemicals: From Tradition to Pharmacy. Molecules 2017, 22, 2145. [Google Scholar] [CrossRef] [Green Version]
  15. Ali, B.H.; Blunden, G.; Tanira, M.O.; Nemmar, A. Some phytochemical, pharmacological and toxicological properties of ginger (Zingiber officinale Roscoe): A review of recent research. Food Chem. Toxicol. 2008, 46, 409–420. [Google Scholar] [CrossRef] [PubMed]
  16. Banihani, S.A. Ginger and Testosterone. Biomolecules 2018, 8, 119. [Google Scholar] [CrossRef] [PubMed]
  17. Kim, H.G.; Lim, S.; Hong, J.; Kim, A.J.; Oh, M.S. Effects of Myoga on Memory and Synaptic Plasticity by Regulating Nerve Growth Factor-Mediated Signaling. Phytother. Res. 2016, 30, 208–213. [Google Scholar] [CrossRef]
  18. Kim, H.S.; Kang, S.A. Study of Quality Characteristics of Kimchi Added with Yangha (Zingiber mioga Rosc). J. Korea Acad.-Ind. Coop. Soc. 2017, 18, 400–407. [Google Scholar]
  19. Lee, D.H.; Ahn, J.; Jang, Y.J.; Jung, C.H. Zingiber mioga reduces weight gain, insulin resistance and hepatic gluconeogenesis in diet-induced obese mice. Exp. Med. 2016, 12, 369–376. [Google Scholar] [CrossRef] [Green Version]
  20. Zhao, H.B.; Wang, Z.H.; He, F.; Meng, H.; Peng, J.H.; Shi, J.L. Analysis of Volatile Oils from Different Processed Products of Zingiber officinale Rhizome by GC-MS. J. Chin. Med. Mater. 2015, 38, 723–726, PMID: 26672336. [Google Scholar]
  21. Qi, X.; Zhang, Y.Y. Progress in Chemical and Pharmacological Research of Ginger. Shandong Chem. Ind. 2018, 47, 41–42. [Google Scholar] [CrossRef]
  22. Tian, M.Y.; Wu, X.H.; Hong, Y.; Wang, H.J.; Deng, G.D.; Zhou, Y. Comparison of Chemical Composition and Bioactivities of Essential Oils from Fresh and Dry Rhizomes of Zingiber zerumbet (L.) Smith. Biomed. Res. Int. 2020, 2020, 9641284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Cai, Y.; Xu, J.L.; Guo, B.Z.; Li, X.; Duan, J.H.; Ruan, J.L. Optimum Extraction Process and GC-MS Analysis of Volatile Oil from Zingiber striolatum Diels. Food Res. Dev. 2018, 39, 49–53. [Google Scholar] [CrossRef]
  24. Tan, J.N. Study on the Chemical Components of Volatile Oil in Zingiber officinale Rosc from Different Habitats by GC-MS. Asia-Pac. Tradit. Med. 2011, 7, 23–25. [Google Scholar]
  25. Abe, M.; Ozawa, Y.; Uda, Y.; Yamada, F.; Morimitsu, Y.; Nakamura, Y.; Osawa, T. Antimicrobial Activities of Diterpene Dialdehydes, Constituents from Myoga (Zingiber mioga Roscoe), and Their Quantitative Analysis. Jpn. Soc. Biosci. Biotechnol. Agrochem. 2004, 68, 1601–1604. [Google Scholar] [CrossRef] [Green Version]
  26. Kim, M.H.; Han, Y.S. Anti-Oxidative and Anti-Diabetic Effects of Butanol Faction from Yangha (Zingiber mioga ROSC). Korean J. Food Cook. Sci. 2018, 34, 105–112. [Google Scholar] [CrossRef]
  27. Zhu, M.M.; Liu, D.; Lu, K.X.; Cao, X.F.; Huang, X.Z.; Li, J.; Zhao, H.Y. Research progress on pharmacological effects of gingerols. J. Hubei Univ. Med. 2018, 37, 390–394. [Google Scholar] [CrossRef]
  28. Semwal, R.B.; Semwal, D.K.; Combrinck, S.; Viljoen, A.M. Gingerols and shogaols: Important nutraceutical principles from ginger. Phytochemistry 2015, 117, 554–568. [Google Scholar] [CrossRef] [PubMed]
  29. Jin, G.C.; Sun, Y.K.; Minsun, J.; Myung, S.O. Pharmacotherapeutic potential of ginger and its compounds in age-related neurological disorders. Curr. Med. Chem. 2018, 182, 56–69. [Google Scholar] [CrossRef]
  30. Yoshikawa, M.; Yamaguchi, S.; Kunimi, K.; Matsuda, H.; Okuno, Y.; Yamahara, J.; Murakmi, N. Stomachic Principles in Ginger. III. An Anti-ulcer Principle, 6-Gingesulfonic Acid, and Three Monoacyldigalactosylglycerols, Gingerglycolipids A, B, and C, from Zingiberis Rhizoma Originating in Taiwan. Chem. Pharm. Bull. 1994, 42, 1226–1230. [Google Scholar] [CrossRef] [Green Version]
  31. Yeh, H.Y.; Chuang, C.H.; Chen, H.C.; Wan, C.J.; Chen, T.L.; Lin, L.Y. Bioactive components analysis of two various gingers (Zingiber officinale Roscoe) and antioxidant effect of ginger extracts. LWT—Food Sci. Technol. 2014, 55, 329–334. [Google Scholar] [CrossRef]
  32. Lee, S.W.; Lim, J.H.; Kim, M.S.; Jeong, J.H.; Song, G.Y.; Lee, W.S.; Rho, M.C. Phenolic compounds isolated from Zingiber officinale roots inhibit cell adhesion. Food Chem. 2011, 128, 778–782. [Google Scholar] [CrossRef]
  33. Tripathi, S.; Maier, K.G.; Bruch, D.; Kittur, D. Effect of 6-Gingerol on Pro-Inflammatory Cytokine Production and Costimulatory Molecule Expression in Murine Peritoneal Macrophages. J. Surg. Res. 2006, 138, 209–213. [Google Scholar] [CrossRef] [PubMed]
  34. Hwang, S.; Jo, M.; Hong, J.E.; Park, C.O.; Lee, C.G.; Yun, M.Y.; Rhee, K.J. Zerumbone Suppresses Enterotoxigenic Bacteroides fragilis Infection-Induced Colonic Inflammation through Inhibition of NF-κΒ. Int. J. Mol. Sci. 2019, 20, 4560. [Google Scholar] [CrossRef] [Green Version]
  35. Tian, M.Y.; Liu, T.T.; Wu, X.G.; Hong, Y.; Liu, X.L.; Lin, B.; Zhou, Y. Chemical composition, antioxidant, antimicrobial and anticancer activities of the essential oil from the rhizomes of Zingiber striolatum Diels. Nat. Prod. Res. 2020, 34, 2621–2625. [Google Scholar] [CrossRef] [PubMed]
  36. Peng, F.; Tao, Q.F.; Wu, X.M.; Dou, H.; Spencer, S.; Mang, C.Y.; Xu, L.; Sun, L.L.; Zhao, Y.; Li, H.B.; et al. Cytotoxic, cytoprotective and antioxidant effects of isolated phenolic compounds from fresh ginger. Fitoterapia 2012, 83, 568–585. [Google Scholar] [CrossRef] [PubMed]
  37. Zeng, H.L.; Han, X.A.; Gu, C.; Huang, X.S.; Gu, J.Q.; Zhong, Q.; Ming, W.J.; Cai, X.N. Comparative protein analysis of K562 cell apoptosis induced by 6-gingerol. J. Chin. Med. Mater. 2010, 33, 753–758. [Google Scholar] [CrossRef]
  38. Hu, Z.H.; Zeng, Q.L.; Zhang, B.; Liu, H.C.; Wang, W. Promotion of p53 expression and reactive oxidative stress production is involved in zerumbone-induced cisplatin sensitization of non-small cell lung cancer cells. Biochimie 2014, 107 Pt B, 257–262. [Google Scholar] [CrossRef]
  39. Jegannathan, S.D.; Arul, S.; Dayalan, H. Zerumbone, a Sesquiterpene, Controls Proliferation and Induces Cell Cycle Arrest in Human Laryngeal Carcinoma Cell Line Hep-2. Nutr. Cancer 2016, 68, 865–872. [Google Scholar] [CrossRef] [PubMed]
  40. Zainal, N.S.; Gan, C.P.; Lan, B.F.; Yee, P.S.; Tiong, K.H.; Rahman, Z.A.A.; Patel, V.; Cheong, S.C. Zerumbone targets the CXCR4-RhoA and PI3K-mTOR signaling axis to reduce motility and proliferation of oral cancer cells. Phytomedicine 2018, 39, 33–41. [Google Scholar] [CrossRef] [PubMed]
  41. Jorvig, J.E.; Chakraborty, A. Zerumbone inhibits growth of hormone refractory prostate cancer cells by inhibiting JAK2/STAT3 pathway and increases paclitaxel sensitivity. Anticancer Drugs 2015, 26, 160–166. [Google Scholar] [CrossRef] [PubMed]
  42. Deorukhkar, A.; Ahuja, N.; Mercado, A.L.; Diagaradjane, P.; Raju, U.; Patel, N.; Mohindra, P.; Diep, N.; Guha, S.; Krishnan, S. Zerumbone increases oxidative stress in a thiol-dependent ROS-independent manner to increase DNA damage and sensitize colorectal cancer cells to radiation. Cancer Med. 2015, 4, 278–292. [Google Scholar] [CrossRef] [PubMed]
  43. Girisa, S.; Shabnam, B.; Monisha, J.; Fan, L.; Halim, C.E.; Arfuso, F.; Ahn, K.S.; Sethi, G.; Kunnumakkara, A.B. Potential of Zerumbone as an Anti-Cancer Agent. Molecules 2019, 24, 734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chakraborty, D.; Bishayee, K.; Ghosh, S.; Mandal, S.K.; Khuda-Bukhsh, A.R. [6]-Gingerol induces caspase 3 dependent apoptosis and autophagy in cancer cells: Drug-DNA interaction and expression of certain signal genes in HeLa cells. Eur. J. Pharm. 2012, 694, 20–29. [Google Scholar] [CrossRef]
  45. Akoachere, J.F.T.K.; Ndip, R.N.; Chenwi, E.B.; Ndip, L.M.; Njock, T.E.; Anong, D.N. Antibacterial effect of Zingiber officinale and Garcinia kola on respiratory tract pathogens. East Afr. Med. J. 2002, 79, 588–892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Murakami, A.; Takahashi, M.; Jiwajinda, S.; Koshimizu, K.; Ohigashi, H. Identification of Zerumbone in Zingiber zerumbet Smith as a Potent Inhibitor of 12-O-Tetradecanoylphorbol-13-acetate-induced Epstein-Barr Virus Activation. Jpn. Soc. Biosci. Biotechnol. Agrochem. 1999, 63, 1811–1812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Areeful, H.M.; Ibrahim, J.; Hemavathy, H.; Shazliana, G. Standardized extract of Zingiber zerumbet suppresses LPS-induced pro-inflammatory responses through NF-κB, MAPK and PI3K-Akt signaling pathways in U937 macrophages. Phytomedicine Int. J. Phytother. Phytopharm. 2019, 54, 195–205. [Google Scholar] [CrossRef]
  48. Funk, J.L.; Frye, J.B.; Oyarzo, J.N.; Chen, J.L.; Zhang, H.P.; Timmermann, B.N. Anti-inflammatory effects of the essential oils of ginger (Zingiber officinale Roscoe) in experimental rheumatoid arthritis. PharmaNutrition 2016, 4, 123–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Ho, Y.C.; Lee, S.S.; Yang, M.L.; Liu, R.H.; Lee, C.Y.; Li, Y.C.; Kuan, Y.H. Zerumbone reduced the inflammatory response of acute lung injury in endotoxin-treated mice via Akt-NFkappaB pathway. Chem. Biol. Interact. 2017, 271, 9–14. [Google Scholar] [CrossRef] [PubMed]
  50. Ho, S.C.; Chang, K.S.; Lin, C.C. Anti-neuroinflammatory capacity of fresh ginger is attributed mainly to 10-gingerol. Food Chem. 2013, 141, 3183–3191. [Google Scholar] [CrossRef] [PubMed]
  51. Wang, W.; Li, N.; Luo, M.; Shui, X.; Hua, Y. Determination of Volatile Oil from Zingiber striolatum Diels by GC-MS. Nat. Prod. Res. Dev. 2012, 24, 916–919. [Google Scholar] [CrossRef]
  52. Moreira, D.S.T.; Pinheiro, C.D.; Puccinelli, O.P.; Pinheiro, C.C.; Soares, P.G. Zerumbone from Zingiber zerumbet (L.) smith: A potential prophylactic and therapeutic agent against the cariogenic bacterium Streptococcus mutans. BMC Complement. Altern. Med. 2018, 18, 301. [Google Scholar] [CrossRef] [Green Version]
  53. Shin, D.S.; Eom, Y.B. Zerumbone inhibits Candida albicans biofilm formation and hyphal growth. Can. J. Microbiol. 2019, 65, 713–721. [Google Scholar] [CrossRef] [PubMed]
  54. Sidahmed, H.M.; Hashim, N.M.; Abdulla, M.A.; Ali, H.M.; Mohan, S.; Abdelwaha, S.I.; Taha, M.M.E.; Fai, L.M.; Vadivelu, J. Antisecretory, gastroprotective, antioxidant and anti-Helicobcter pylori activity of zerumbone from Zingiber zerumbet (L.) Smith. PLoS ONE 2015, 10, e0121060. [Google Scholar] [CrossRef] [Green Version]
  55. Park, M.; Bae, J.; Lee, D.S. Antibacterial activity of [10]-gingerol and [12]-gingerol isolated from ginger rhizome against periodontal bacteria. Phytother. Res. 2008, 22, 1446–1449. [Google Scholar] [CrossRef] [PubMed]
  56. Zhou, C.R.; Liu, L.; Wen, X.J. Toxic activity of essential oil from Zingiber Corallinum Hance of Aedes albopictus and Culex pipiens quinquefasciatus. Guizhou Med. J. 2016, 40, 38–39. [Google Scholar] [CrossRef]
  57. Bucker, A.; Falcao-Bucker, N.C.; Nunez, C.V.; Pinheiro, C.C.S.; Tadei, W.P. Evaluation of larvicidal activity and brine shrimp toxicity of rhizome extracts of Zingiber zerumbet (L.) Smith. Rev. Soc. Bras. Med. Trop. 2013, 46, 377–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Rahuman, A.A.; Gopalakrishnan, G.; Venkatesan, P.; Geetha, K. Isolation and identification of mosquito larvicidal compound from Abutilon indicum (Linn.) Sweet. Parasitol. Res. 2008, 102, 981–988. [Google Scholar] [CrossRef] [PubMed]
  59. Jo, S.H.; Cho, C.Y.; Lee, J.Y.; Ha, K.S.; Kwon, Y.I.; Apostolidis, E. In vitro and in vivo reduction of post-prandial blood glucose levels by ethyl alcohol and water Zingiber mioga extracts through the inhibition of carbohydrate hydrolyzing enzymes. BMC Complement. Altern. Med. 2016, 16, 111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Murugesan, S.; Venkateswaran, M.R.; Jayabal, S.; Periyasamy, S. Evaluation of the antioxidant and anti-arthritic potential of Zingiber officinale Rosc. by in vitro and in silico analysis. South Afr. J. Bot. 2020, 130, 45–53. [Google Scholar] [CrossRef]
  61. Swarnalatha, D.; Rao, P.M.; Devi, N.V.; Madhu, K.B.; Satyanarayana, T.; Jayaveera, N.K. Comparative antioxidant and anti-inflammatory effects of [6]-gingerol, [8]-gingerol, [10]-gingerol and [6]-shogaol. J. Ethnopharmacol. 2010, 127, 515–520. [Google Scholar] [CrossRef]
  62. Masuda, Y.; Kikuzaki, H.; Hisamoto, M.; Nakatani, N. Antioxidant properties of gingerol related compounds from ginger. BioFactors 2004, 21, 293–296. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, T.H.; Cai, J.Y.; Ni, J.; Yang, F. An UPLC-MS/MS application to investigate chemical compositions in the ethanol extract with hypoglycemic activity from Zingiber striolatum Diels. J. Chin. Pharm. Sci. 2016, 25, 116–121. [Google Scholar]
  64. Bhandari, U.; Kanojia, R.; Pillai, K.K. Effect of ethanolic extract of Zingiber officinale on dyslipidaemia in diabetic rats. J. Ethnopharmacol. 2005, 97, 227–230. [Google Scholar] [CrossRef] [PubMed]
  65. 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. J. Nutr. 2000, 130, 1124–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Bahman, M.P.; Alireza, K.; Fatame, A.; Nasrin, L.D.; Seyede-Arefe, P.; Ghorban, M. Effects of Zingiber officinale hydro-alcoholic extract on HMG-COA reductase level in the testis of streptozotocin-induced diabetic rats. J. Herbmed Pharmacol. 2018, 7, 94–99. [Google Scholar] [CrossRef] [Green Version]
  67. Nam, Y.H.; Hong, B.N.; Rodriguez, I.; Park, M.S.; Jeong, S.Y.; Lee, Y.G.; Shim, J.H.; Yasmin, T.; Na Kim, W.; Koo, Y.T.; et al. Steamed Ginger May Enhance Insulin Secretion through KATP Channel Closure in Pancreatic β-Cells Potentially by Increasing 1-Dehydro-6-Gingerdione Content. Nutrients 2020, 12, 324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Chen, J.; Sun, J.; Prinz, R.A.; Li, Y.; Xu, X. Gingerenone A Sensitizes the Insulin Receptor and Increases Glucose Uptake by Inhibiting the Activity of p70 S6 Kinase. Mol. Nutr. Food Res. 2018, 62, e1800709. [Google Scholar] [CrossRef]
  69. Palatty, P.L.; Haniadka, R.; Valder, B.; Arora, R.; Baliga, M.S. Ginger in the prevention of nausea and vomiting: A review. Crit. Rev. Food Sci. Nutr. 2013, 53, 659–669. [Google Scholar] [CrossRef]
  70. Giacosa, A.; Morazzoni, P.; Bombardelli, E.; Riva, A.; Bianchi, P.G.; Rondanelli, M. Can nausea and vomiting be treated with ginger extract? Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 1291–1296. [Google Scholar] [PubMed]
  71. Iryna, K.; Lidiya, E.; Mariia, N.; Alona, K. Anti-inflammatory and analgesic activity of ointment based on dense ginger extract (Zingiber officinale). J. Herbmed Pharmacol. 2019, 8, 126–132. [Google Scholar] [CrossRef]
  72. Sulaiman, M.; Mohamad, T.T.; Mossadeq, W.S.; Moin, S.; Yusof, M.; Mokhtar, A.F.; Zakaria, Z.A.; Israf, D.A.; Lajis, N. Antinociceptive Activity of the Essential Oil of Zingiber zerumbet. Planta Med. 2010, 76, 107–112. [Google Scholar] [CrossRef] [PubMed]
  73. Gopalsamy, B.; Farouk, A.A.O.; Tengku, M.T.A.S.; Sulaiman, M.R.; Perimal, E.K. Antiallodynic and antihyperalgesic activities of zerumbone via the suppression of IL-1beta, IL-6, and TNF-alpha in a mouse model of neuropathic pain. J. Pain Res. 2017, 10, 2605–2619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Young, H.Y.; Luo, Y.L.; Cheng, H.Y.; Hsieh, W.C.; Liao, J.C.; Peng, W.H. Analgesic and anti-inflammatory activities of [6]-gingerol. J. Ethnopharmacol. 2005, 96, 207–210. [Google Scholar] [CrossRef]
  75. Ding, S.; Wang, X. The protective effect of ginger on the gastric mucosa of asprin induced gastric ulcer rats. Chin. J. Ethnomed. Ethnopharm. 2012, 21, 22+24. [Google Scholar] [CrossRef]
  76. Farombi, E.O.; Abolaji, A.O.; Adetuyi, B.O.; Awosanya, O.; Fabusoro, M. Neuroprotective role of 6-Gingerol-rich fraction of Zingiber officinale (Ginger) against acrylonitrile-induced neurotoxicity in male Wistar rats. J. Basic Clin. Physiol. Pharmacol. 2018, 30. [Google Scholar] [CrossRef]
  77. Le, T.H.; Hoang, V.C.; Nguyen, T.G.A.; Nguyen, T.V.; Nguyen, H.H.; Nguyen, T.H.T.; Abdulatif, O.G.A.; Isiaka, A.O. Zingiber zerumbet Rhizome Essential Oil: Chemical Composition, Antimicrobial and Mosquito Larvicidal Activities. Eur. J. Med. Plants 2020, 30, 1–12. [Google Scholar] [CrossRef] [Green Version]
  78. Yu, D.S.; Ye, F.F.; Yang, X.Q. Antioxidant Activity of Essential Oil from Zingiber corallinum Hance. Food Sci. 2011, 32, 164–167. [Google Scholar]
  79. Sun, Q. The experimental study of Chemical Constituents and the Pharmacological Actions of Zingiberis Rhizoma. Ph.D. Thesis, China Medical University, Shenyang, China, 2018. [Google Scholar]
  80. Zhou, L.; Xie, W.S.; Jiang, M. Volatile Components of Two Main Kinds of Edible Gingers in Yunnan Province. J. Anhui Agric. Sci. 2016, 44, 95–97, 146. [Google Scholar] [CrossRef]
  81. Lee, J.W.; Chon, S.U.; Han, S.K.; Ryu, J.; Choi, D.G. Effects of Antioxidant and Flavor Components of Zingiber mioga Rosc. Korean J. Med. Crop Sci. 2007, 15, 203–209. [Google Scholar]
  82. Yuan, G.; Xian, J.M.; Yuan, J.M.; Wu, H.M. The Chemical constituents of zingiber corallinum by supercritical carbondioxide extraction. Plant Divers. 1997, 19, 102–104. [Google Scholar]
  83. Wang, Z. Chemical Constituents from Zingiber officinale and Siegesbeckia orientalis. Ph.D. Thesis, Anhui University, Hefei, China, 2012. [Google Scholar] [CrossRef]
  84. Rana, V.S.; Ahluwalia, V.; Shakil, N.A.; Prasad, L. Essential oil composition, antifungal, and seedling growth inhibitory effects of zerumbone from Zingiber zerumbet Smith. J. Essent. Oil Res. 2016, 29, 320–329. [Google Scholar] [CrossRef]
  85. Shi, Q.R.; Wang, L.; Yan, H.; Yan, H.; Yang, F.; Chen, Y.; Sun, X.D.; Sun, S.G. Gas Chromatography-Mass Spectrometry Analysis of the CO2 Supercritical Fluid Extract of Zingiber officinale Rosc.And Tracing of gingerol. Pharm. J. Chin. People’s Lib. Army 2011, 27, 296–298, 302. [Google Scholar] [CrossRef]
  86. Dai, D.N.; Thang, T.D.; Chau, L.T.M.; Isiaka, A.O. Chemical Constituents of the Root Essential Oils of Zingiber rubens Roxb., and Zingiber zerumbet (L.) Smith. Am. J. Plant Sci. 2013, 4, 7–10. [Google Scholar] [CrossRef] [Green Version]
  87. Yuan, G.F.; Wei, J.; Zheng, E.L.; Cai, X.K.; Xu, C.H. Study on Water-Soluble Flavor Material in Fresh Ginger and Dry Ginger by GC-MS. J. Anhui Agric. Sci. 2014, 42, 12304–12306. [Google Scholar] [CrossRef]
  88. Zhang, J.; Chang, Y.S.; Zeng, C.; Li, Y.; Xu, L.L.; Gu, Z.B. Chemical Constituents of Ginger Extract. J. Anhui Agric. Sci. 2015, 43, 287–288+290. [Google Scholar] [CrossRef]
  89. Tan, J.N.; Wang, R.; Huang, J.; Yan, K.J.; Liang, C.Y. The Changes of Chemical Compositions in Volatile Oil from Zingiber officinale Rosc during Processing. Lishizhen Med. Mater. Med. Res. 2012, 23, 569–573. [Google Scholar] [CrossRef]
  90. Virendra, S.; Ranaa, M.V.; Maria, A. Blazquez. Chemical Composition of the Essential Oil of Zingiber zerumbet var. darcyi. Nat. Prod. Commun. 2012, 7, 1369–1370. [Google Scholar] [PubMed]
  91. Wu, C.W.; Tu, Y.H.; Li, M.E.; Ye, Z.Y.; Xue, Y.C.; Cao, Y. Effect of Zingiber corallinum Oil on Proliferation and Apoptosis of Cervical Carcinoma Cell Line HeLa. Her. Med. 2017, 36, 32–36. [Google Scholar] [CrossRef]
  92. Gao, Y.Q.; Yang, N.J.; Liu, J.H.; Zhao, D.G.; Song, B.A. Antifungal Activity of the Oil from Zingiber corallinum Hance by Supercritical CO2 Extraction. Acta Sci. Nat. Univ. Sunyatseni 2008, 47 (Suppl. S1), 53–57. [Google Scholar] [CrossRef]
  93. Zhang, J.W.; Mao, Q. Preliminary study on the constituents of essential oil from Zingiber Corallinum Hance. Chin. Bull. Bot. 1988, 5, 108–109. [Google Scholar]
  94. Luo, S.Q.; Peng, Q.C.; Yang, X.Y.; Wu, D. Volatiles and inhibitory phytopathogens fungi activities of essential oil extracted from Zingiber corallinum Hance. Guangdong Agric. Sci. 2013, 40, 84–86. [Google Scholar] [CrossRef]
  95. Gao, Y.Q. Study on the Constituents and Some Bioactivities of Volatile Oils from 19 Species of Plants. Ph.D. Thesis, Guizhou University, Guiyang, China, 2018. [Google Scholar] [CrossRef]
  96. Wang, Y. Chemical Compositions Analysis of Essential Oil for Fresh Gingers from Different Districts by GC/MS. Guangdong Chem. Ind. 2016, 43, 176+191. [Google Scholar] [CrossRef]
  97. Mohd, Y.Y.A. Gingerol and Its Role in Chronic Diseases. Adv. Exp. Med. Biol. 2016, 929, 177–207. [Google Scholar] [CrossRef]
  98. Cui, W.H.; Wang, Y.Z.; Li, Z.Z. Chemical Constituents from Rhizoma Zingiberis. Chin. Pharm. J. 2018, 53, 1160–1164. [Google Scholar] [CrossRef]
  99. Cui, W.H.; Wang, Y.Z.; Li, Z.Z. Separation and Identification of Chemical Constituents from Zingiberis Rhizoma. J. Chin. Med. Mater. 2018, 41, 334–337. [Google Scholar] [CrossRef]
  100. Cui, W.H.; Wang, Y.Z.; Li, Z.Z.; Guo, Y. Two new diarylheptane derivatives from Rhizoma Zingiberis. Acta Pharm. Sin. 2018, 53, 100–103. [Google Scholar] [CrossRef]
  101. Long, Q.J.; Xu, X.Q. Pharmacological action of chemical constituents of dried ginger and literature analysis on processing and processing. Res. Pract. Chin. Med. 2015, 29, 82–83. [Google Scholar] [CrossRef]
  102. Guan, J.; Zhang, Y.; Zhao, Y.Q. Phytochemical Study on SFE Extract of Zinger officinale. Mod. Chin. Med. 2015, 17, 114–117. [Google Scholar] [CrossRef]
  103. Li, B.Z.; Li, F.; Piao, Y.G.; Wang, R.T.; Li, H.L.; Tong, Y.N.; Fu, Q.; Liu, Y.J.; Chen, M. Extraction of Chemical Constituents from Rhizoma Zingiberis and Its Application in Cigarette. Guangzhou Chem. Ind. 2019, 47, 95–97. [Google Scholar] [CrossRef]
  104. Fan, Q.H.; Zhang, W.Q.; He, Q.; Wang, W.; Tang, X.H. Progress in clinical application of zingiber zerumbet abroad. Asia Pac. Tradit. Med. 2014, 10, 39–40. [Google Scholar]
  105. Yang, X.; Wang, Q.H.; Wang, Y.; Kuang, H.X. Analysis on chemical constituents of volatile oils from zingiberis rhizoma. Chem. Eng. 2015, 29, 16–18. [Google Scholar] [CrossRef]
  106. Wu, J.F.; Zhang, C.; Zhang, X.M.; Xu, S.Y. Extraction and Analyse is of flavor compounds in ginger. Food Mach. 2006, 22, 94–96+99. [Google Scholar] [CrossRef]
  107. Liu, H.X.; Chen, S.D.; Chen, F.B.; You, M.L. Studies on the chemical component and extracting volatile oil from Zingiber officinale Rosc by soxhelt methods. China Condiment 2010, 35, 105–106, 112. [Google Scholar] [CrossRef]
  108. Luo, T.; Wang, J.Q.; Zhang, C.L.; Fan, S.M.; Yu, L.Y. Comparative Analysis of Quality of Simmered Ginger and Fresh Ginger. China Condiment 2020, 45, 160–164, 171. [Google Scholar] [CrossRef]
  109. Huo, W.L.; Li, Z.T. Study on fresh and dried ginger by GC-MS of supercritical fluid extraction. Appl. Chem. Ind. 2015, 44, 184–186, 189. [Google Scholar] [CrossRef]
  110. Han, J.S.; Lee, S.; Kim, H.Y.; Lee, C.H. MS-Based Metabolite Profiling of Aboveground and Root Components of Zingiber mioga and Officinale. Molecules 2015, 20, 16170–16185. [Google Scholar] [CrossRef] [Green Version]
  111. Miyoshi, N.; Nakamura, Y.; Ueda, Y.; Abe, M.; Ozawa, Y.; Uchida, K.; Osawa, T. Dietary ginger constituents, galanals A and B, are potent apoptosis inducers in Human T lymphoma Jurkat cells. Cancer Lett. 2003, 199, 113–119. [Google Scholar] [CrossRef]
  112. Jang, D.S.; Han, A.R.; Park, G.; Jhon, G.J.; Seo, E.K. Flavonoids and aromatic compounds from the rhizomes of Zingiber zerumbet. Arch. Pharmacal Res. 2004, 27, 386–389. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, C.; Geng, J.L.; Kuang, H.X.; Wang, Q.H. Study on Variation of Non-Volatile Compounds of Raw Ginger after Processing by UHPLC-Orbitrap/HRMS. J. Nanjing Univ. Tradit. Chin. Med. 2020, 36, 387–395. [Google Scholar] [CrossRef]
  114. Nakatani, N.; Jitoe, A.; Masuda, T.; Yonemori, S. Flavonoid Constituents of Zingiber zerumbet Smith. Agric. Biol. Chem. 1991, 55, 455–460. [Google Scholar] [CrossRef] [Green Version]
  115. Sun, F.J.; Li, Z.L.; Qian, S.H.; Pu, S.B. Chemical Constituents of Rhizomes of Dried Zingiber officinale. Chin. Wild Plant Resour. 2016, 35, 20–24, 60. [Google Scholar] [CrossRef]
  116. Mei, X.D.; Wang, Y.Q.; Liu, Z.H.; Zhang, Y.Z.; Song, S.; Lin, F.; Zhang, J.Y. Identification and analysis of gingerols in ginger before and after fermentation by UHPLC-HRMS. China J. Tradit. Chin. Med. Pharm. 2020, 35, 1990–1995. [Google Scholar]
  117. Zhao, W.Z.; Zhang, R.X.; Yu, Z.P.; Wang, X.K.; Li, J.R.; Liu, J.B. Research process in ginger chemical composition and biological activity. Sci. Technol. Food Ind. 2016, 37, 383–389. [Google Scholar] [CrossRef]
  118. Li, Y. Study on the Chemical Constituents of Rhizome Zingiberis and Maca. Ph.D. Thesis, Henan University of Traditional Chinese Medicine, Zhengzhou, China, 2016. [Google Scholar]
  119. Chen, T.H.; Cai, J.Y.; Yang, F.; Ni, J.; Zhang, T. Study on Hypoglycemic Activity and Chemical Constituents of Zingiber striolatum Diels. Food and Drug. 2015, 17, 413–416. [Google Scholar] [CrossRef]
  120. Qu, S.L.; Xia, L.; Song, L.D.; Feng, W.Y. Advances in the study of Zingiber striolatum Diels. Yunnan J. Tradit. Chin. Med. Mater. Med. 2015, 36, 111–113. [Google Scholar] [CrossRef]
  121. Guo, Y.; Wang, Y.Z.; Xu, Z.P.; Li, M.Q.; Zhang, X.J.; Liu, Y.F.; Hu, X.Y.; Feng, W.S. Two new monoterpenoids of Zingiber officinale peel. Acta Pharm. Sin. 2020, 55, 484–488. [Google Scholar] [CrossRef]
  122. Hao, N.M.; Zhao, B.F.; Hu, G.H.; Zhang, T. Study on Chemical Constituents from Zingiber Strioatum. J. Wenshan Univ. 2004, 17, 379–381. [Google Scholar] [CrossRef]
  123. Bao, L.; Deng, A.J.; Li, Z.H.; Qin, H.L. Study on the chemical constituents of ginger. China, J. Chin. Mater. Med. 2010, 35, 598–601. [Google Scholar] [CrossRef]
  124. Gu, G.Y. Cytotoxic components of Zingiber zerumbet (L.) Smith, Curcuma aeruginosa Roxb and Curcuma longa L. Foreign Pharm. (Plant Med. Vol.) 1981, 2, 20–21. [Google Scholar]
  125. Zhang, Y.L.; Zheng, Y.M.; Hu, S.N.; Liu, H. Anti-Helicobacter pylori effect of 6-gingerol in vitro. Mod. Food Sci. Technol. 2013, 29, 1259–1261, 1305. [Google Scholar] [CrossRef]
  126. Jyothilakshmi, M.; Jyothis, M.; Narayanan, G.N.H.; Mukalel, S.L. Antidermatophytic and Protease-inhibiting Activities of Zerumbone: A Natural Sesquiterpene from the Rhizome of Zingiber zerumbet (L.) Roscoe ex J.E; Smith. Pharm. Mag. 2017, 13, 2–6. [Google Scholar] [CrossRef]
  127. Yang, C.; Zhou, L.L.; Wang, H.Y.; Huang, S.N.; Liu, Q.; Hu, S.L.; Li, T.R.; Chen, Y.B.; Jiang, J.X. The inhibitory effect of Zingiber corallinum Hance essential oil on drug-resistant bacteria and evaluation of its acute toxicity. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2011, 17, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Cao, Y.; Zhu, S.Z. Experimental study on antifungal and antibacterial effects of total essential oil from Zingiber corallinum Hance. J. Guizhou Med. Univ. 1988, 13, 336–339. [Google Scholar] [CrossRef]
Figure 1. The characteristics of some Zingiber plants. ((a) Zingiber zerumbet (L.) Smith; (b) Zingiber officinale Rosc; (c) Zingiber corallinum Hance; (d) Zingiber mioga (Thunb.) Rosc; (e) Zingiber striolatum Diels).
Figure 1. The characteristics of some Zingiber plants. ((a) Zingiber zerumbet (L.) Smith; (b) Zingiber officinale Rosc; (c) Zingiber corallinum Hance; (d) Zingiber mioga (Thunb.) Rosc; (e) Zingiber striolatum Diels).
Molecules 27 02826 g001
Figure 2. Flowchart of screening and choosing eligible articles.
Figure 2. Flowchart of screening and choosing eligible articles.
Molecules 27 02826 g002
Figure 3. Chemical structures of compounds 1106.
Figure 3. Chemical structures of compounds 1106.
Molecules 27 02826 g003aMolecules 27 02826 g003b
Figure 4. Chemical structures of monoterpenes (107182).
Figure 4. Chemical structures of monoterpenes (107182).
Molecules 27 02826 g004
Figure 5. Chemical structures of sesquiterpenes, diterpenes and triterpenes (183261).
Figure 5. Chemical structures of sesquiterpenes, diterpenes and triterpenes (183261).
Molecules 27 02826 g005aMolecules 27 02826 g005b
Figure 6. Chemical structures of the flavonoid compounds (262293).
Figure 6. Chemical structures of the flavonoid compounds (262293).
Molecules 27 02826 g006
Figure 7. Chemical structures of the diphenylheptanes (294320).
Figure 7. Chemical structures of the diphenylheptanes (294320).
Molecules 27 02826 g007
Figure 8. Chemical structures of the gingerol analogues (321367).
Figure 8. Chemical structures of the gingerol analogues (321367).
Molecules 27 02826 g008
Figure 9. Chemical structures of the organic acids (368395).
Figure 9. Chemical structures of the organic acids (368395).
Molecules 27 02826 g009
Figure 10. Chemical structures of the sterides and other compounds (396447).
Figure 10. Chemical structures of the sterides and other compounds (396447).
Molecules 27 02826 g010
Figure 11. The structure–activity relationship of anticancer for gingerol (a) and shogaol (b).
Figure 11. The structure–activity relationship of anticancer for gingerol (a) and shogaol (b).
Molecules 27 02826 g011
Table 1. Pharmacological effects of Zingiber plants.
Table 1. Pharmacological effects of Zingiber plants.
Pharmacological EffectsDetailsExtracts/CompoundsInhibitory Concentrations/DoseReferences
Anti-inflammatory activityInhibition of the direct binding between intercellular adhesion molecules and lymphocyte function-associated antigen-1 of the THP-1 cells10-gingerolIC50: 57.6 μM[32]
8-shogaolIC50: 65.4 μM
Inhibitory effect on direct binding between sVCAM-1 and VLA-4 of THP-1 cells6-shogaolIC50: 27.1 μM
Dehydro-6-gingerdioneIC50: 62.0 μM
Inhibition of the produc-tion of pro-inflammatory cytokines from LPS stim-ulated macrophages6-gingerol50 mg/kg[33]
Decreased ETBF-induced colitis via inhibition of NF-κB signalingZerumboneMIC: 32–48 μg/mL[34]
Anti-cancer activityCytotoxic effect on MRC-5
(human fetal lung fibroblasts cell linse)
Z. zerumbet fresh rhizome essential oilIC50: 216.99 ± 8.27 μM for 24 h[22]
Z. zerumbet dry rhizome essential oilIC50: 159.47 ± 9.34 μM for 24 h
ZerumboneIC50: 117.96 ± 5.67 μM for 24 h
Cytotoxic effect on PC-3
(human prostate cancer cell lines)
Z. zerumbet fresh rhizome essential oilIC50: 53.32 ± 1.34 μM for 24 h
Z. zerumbet dry rhizome essential oilIC50: 77.45 ± 0.46 μM for 24 h
ZerumboneIC50: 30.78 ± 1.31 μM for 24 h
Z. striolatum essential oilIC50: 86.05 μM[35]
6-shogaolIC50: 100.0 ± 13.1 μM[36]
6-dehydrogingerdioneIC50: 106.4 ± 12.5 μM
10-gingerolIC50: 59.7 ± 8.2 μM
3,5-dioxo-1,7-bis(3-methoxy-4-hydroxy)-phenyl-heptaneIC50: 153.5 ± 13.8 μM
Gingerenone AIC50: 114.3 ± 14.2 μM
3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 86.6 ± 7.5 μM
CurcuminIC50: 16.5 ± 2.7 μM
Cytotoxic effect on K562
(human leukemia cell lines)
Z. zerumbet fresh rhizome essential oilIC50: 35.73 ± 1.72 μM for 24 h[22]
Z. zerumbet dry rhizome essential oilIC50: 41.79 ± 1.18 μM for 24 h
ZerumboneIC50: 10.08 ± 0.61 μM
Z. striolatum essential oilIC50: 29.67 μM[35]
6-gingerolIC50: 22.86 μM[37]
Gingerenone AIC50: 33.3 ± 5.1 μM
3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 39.6 ± 4.8 μM
1,5-epoxy-3-hydroxy-1-(3,4-hydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 160.3 ± 14.1 μM
CitrylidenmalonsaeureIC50: 119.8 ± 9.9 μM
CurcuminIC50: 30.5 ± 5.3 μM
Cytotoxic effect on A-549
(human lung cancer cell lines)
Z. zerumbet fresh rhizome essential oilIC50: 44.88 ± 1.21 μM for 24 h[27]
Z. zerumbet dry rhizome essential oilIC50: 68.06 ± 1.09 μM for 24 h
ZerumboneIC50: 25 μM[38]
Z. striolatum essential oilIC50: 48.87 μM[35]
6-shogaolIC50: 22.9 ± 2.1 μM[36]
6-dehydrogingerdioneIC50: 81.2 ± 9.6 μM
10-gingerolIC50: 85.4 ± 10.2 μM
3,5-dioxo-1,7-bis(3-methoxy-4-hydroxy)-phenyl-heptaneIC50: 248 ± 17.9 μM
Gingerenone AIC50: 44.5 ± 5.8 μM
3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 96.3 ± 7.8 μM
7-(3,4-dihydroxy-5-methoxyphenyl)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl) heptan-3-oneIC50: 83.6 ± 8.4 μM
1,5-epoxy-3-hydroxy-1-(3,4-hydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 230.8 ± 17.9 μM
5-[4-hydroxy-6-(4-hydroxyphenethyl) tetrahydro-2H-pyran-2-yl]-3-methoxybenzene-1,2-diolIC50: 212.0 ± 15.2 μM
CurcuminIC50: 58.8 ± 9.4 μM
Cytotoxic effect on H-460
(human lung cancer cell lines)
ZerumboneIC50: 15 μM[38]
Cytotoxic effect on BEL7404
(human lung cancer cell lines)
6-shogaolIC50: 11.8 ± 2.6 μM[36]
6-dehydrogingerdioneIC50: 115.2 ± 13.7 μM
10-gingerolIC50: 95.2 ± 12.2 μM
3,5-dioxo-1,7-bis(3-methoxy-4-hydroxy)-phenyl-heptaneIC50: 49.4 ± 3.4 μM
Gingerenone AIC50: 9.0 ± 2.3 μM
3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 101.9 ± 13.1 μM
7-(3,4-dihydroxy-5-methoxyphenyl)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl) heptan-3-oneIC50: 180.6 ± 16.5 μM
1,5-epoxy-3-hydroxy-1-(3,4-hydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 180.6 ± 12.5 μM
CurcuminIC50: 38.2 ± 3.9 μM
Cytotoxic effect on CNE
(human nasopharyngeal cancer cell lines)
6-shogaolIC50: 43.8 ± 5.0 μM
6-dehydrogingerdioneIC50: 119.7 ± 7.9 μM
10-gingerolIC50: 88.1 ± 7.3 μM
3,5-dioxo-1,7-bis(3-methoxy-4-hydroxy)-phenyl-heptaneIC50: 76.7 ± 5.4 μM
Gingerenone AIC50: 27.7 ± 3.9 μM
3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 62.0 ± 10.7 μM
7-(3,4-dihydroxy-5-methoxyphenyl)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl) heptan-3-oneIC50: 75.4 ± 6.6 μM
1,5-epoxy-3-hydroxy-1-(3,4-hydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 174.2 ± 15.1 μM
5-[4-hydroxy-6-(4-hydroxyphenethyl) tetrahydro-2H-pyran-2-yl]-3-methoxybenzene-1,2-diolIC50: 247.9 ± 20.1 μM
CurcuminIC50: 33.5 ± 10.1 μM
Cytotoxic effect on Hep-2
(human laryngeal carcinoma cell lines)
ZerumboneIC50: 15 μM[39]
Anti-proliferative effect on HepG2
(human liver cancer cell lines)
ZerumboneIC50: 3.45 ± 0.026 μg/ml[40]
Cytotoxic effect on 16 human oral squamous cell carcinoma linesZerumboneIC50: average 2 µM; range: 0.8–4.9 µM
Cytotoxic effect on DU145
(human prostate cancer cell lines)
Zerumbone IC50: 24 μM[41]
Cytotoxic effect on HCT116
(human colorectal cancer cell lines)
Zerumbone IC50: 30 ± 1.5 μM[42]
Cytotoxic effect on SW620
(human colorectal cancer cell lines)
ZerumboneIC50: > 46 μM
Cytotoxic effect on MCF-7
(human breast cancer cell lines)
ZerumboneIC50: 23.0 μg/ml[43]
Cytotoxic effect on MDA-MB 231
(human breast cancer cell lines)
ZerumboneIC50: 24.3 μg/ml
Cytotoxic effect on Hela
(human cervical cancer cell lines)
ZerumboneIC50: 6.4 μg/mL
6-gingerolIC50: 126.89 μM[44]
6-dehydrogingerdioneIC50: 62.5 ± 4.7 μM[45]
ZingeroneIC50: 114.6 ± 9.3 μM
10-gingerolIC50: 52.4 ± 7.1 μM
3,5-dioxo-1,7-bis(3-methoxy-4-hydroxy)-phenyl-heptaneIC50: 86.8 ± 10.5 μM
Gingerenone AIC50: 15.4 ± 3.2 μM
3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 110.0 ± 9.8 μM
1,7-bis(4-hydroxy-3-methoxyphenyl) heptane-3,5-diolIC50: 191.0 ± 16.5 μM
7-(3,4-dihydroxy-5-methoxyphenyl)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl) heptan-3-oneIC50: 133.2 ± 16.1 μM
5-[4-hydroxy-6-(4-hydroxyphenethyl) tetrahydro-2H-pyran-2-yl]-3-methoxybenzene-1,2-diolIC50: 231.8 ± 13.7 μM
CurcuminIC50: 18.9 ± 2.8 μM
Inhibitory effect on epstein-barr virus
(human herpesvirus 4)
ZerumboneIC50: 0.14 μM[46]
Cytotoxic effect on KB
(human oral epithelial cancer cell lines)
6-shogaolIC50: 7.4 ± 2.2 μM[37]
6-dehydrogingerdioneIC50: 229.5 ± 17.5 μM
10-gingerolIC50: 89.5 ± 8.7 μM
3,5-dioxo-1,7-bis(3-methoxy-4-hydroxy)-phenyl-heptaneIC50: 27.7 ± 2.7 μM
Gingerenone AIC50: 8.8 ± 2.6 μM
CurcuminIC50: 34.7 ± 6.7 μM
3,5-diacetoxy-1-(3-methoxy-4,5-dihydroxy-phenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC50: 75.1 ± 10.5 μM
7-(3,4-dihydroxy-5-methoxyphenyl)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl) heptan-3-oneIC50: 90.3 ± 10.1 μM
1,5-epoxy-3-hydroxy-1-(3,4-hydroxy-5-methoxyphenyl)-7-(4-hydroxy-3-methoxyphenyl) heptaneIC5: 78.5 ± 11.3 μM
CurcuminIC50: 34.7 ± 6.7 μM
Cytotoxic effect on HT29
(human colorectal cancer cell lines)
ZerumboneIC50: 38.8 ± 1.2 μM[45]
IC50: the half maximal inhibitory concentration.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Deng, M.; Yun, X.; Ren, S.; Qing, Z.; Luo, F. Plants of the Genus Zingiber: A Review of Their Ethnomedicine, Phytochemistry and Pharmacology. Molecules 2022, 27, 2826. https://doi.org/10.3390/molecules27092826

AMA Style

Deng M, Yun X, Ren S, Qing Z, Luo F. Plants of the Genus Zingiber: A Review of Their Ethnomedicine, Phytochemistry and Pharmacology. Molecules. 2022; 27(9):2826. https://doi.org/10.3390/molecules27092826

Chicago/Turabian Style

Deng, Miao, Xuan Yun, Shurui Ren, Zhixing Qing, and Fenglian Luo. 2022. "Plants of the Genus Zingiber: A Review of Their Ethnomedicine, Phytochemistry and Pharmacology" Molecules 27, no. 9: 2826. https://doi.org/10.3390/molecules27092826

Article Metrics

Back to TopTop