Next Article in Journal
Daily Fructose Traces Intake and Liver Injury in Children with Hereditary Fructose Intolerance
Previous Article in Journal
A Study of the Food Environment at Australian Family Day Care
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Kaempferia Phytochemistry and Biological Activity: A Comprehensive Review

by
Abdelsamed I. Elshamy
1,2,
Tarik A. Mohamed
3,
Ahmed F. Essa
2,
Ahmed M. Abd-El Gawad
4,5,
Ali S. Alqahtani
6,*,
Abdelaaty A. Shahat
3,6,
Tatsuro Yoneyama
1,
Abdel Razik H. Farrag
7,
Masaaki Noji
1,
Hesham R. El-Seedi
8,9,10,
Akemi Umeyama
1,
Paul W. Paré
11 and
Mohamed-Elamir F. Hegazy
3,12,*
1
Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
2
Chemistry of Natural Compounds Department, National Research Centre, 33 El Bohouth St., Dokki, Giza 12622, Egypt
3
Chemistry of Medicinal Plants Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
4
Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
5
Plant Production Department, College of Food & Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
6
Pharmacognosy Department, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
7
Pathology Department; National Research Centre, Dokki, Giza 12622, Egypt
8
Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Box 574, SE-75 123 Uppsala, Sweden
9
Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom 32512, Egypt
10
College of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
11
Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
12
Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, University of Mainz, Staudinger Weg 5, 55128 Mainz, Germany
*
Authors to whom correspondence should be addressed.
Nutrients 2019, 11(10), 2396; https://doi.org/10.3390/nu11102396
Submission received: 12 September 2019 / Revised: 26 September 2019 / Accepted: 1 October 2019 / Published: 7 October 2019

Abstract

:
Background: Plants belonging to the genus Kaempferia (family: Zingiberaceae) are distributed in Asia, especially in the southeast region, and Thailand. They have been widely used in traditional medicines to cure metabolic disorders, inflammation, urinary tract infections, fevers, coughs, hypertension, erectile dysfunction, abdominal and gastrointestinal ailments, asthma, wounds, rheumatism, epilepsy, and skin diseases. Objective: Herein, we reported a comprehensive review, including the traditional applications, biological and pharmacological advances, and phytochemical constituents of Kaempheria species from 1972 up to early 2019. Materials and methods: All the information and reported studies concerning Kaempheria plants were summarized from library and digital databases (e.g., Google Scholar, Sci-finder, PubMed, Springer, Elsevier, MDPI, Web of Science, etc.). The correlation between the Kaempheria species was evaluated via principal component analysis (PCA) and agglomerative hierarchical clustering (AHC), based on the main chemical classes of compounds. Results: Approximately 141 chemical constituents have been isolated and reported from Kaempferia species, such as isopimarane, abietane, labdane and clerodane diterpenoids, flavonoids, phenolic acids, phenyl-heptanoids, curcuminoids, tetrahydropyrano-phenolic, and steroids. A probable biosynthesis pathway for the isopimaradiene skeleton is illustrated. In addition, 15 main documented components of volatile oils of Kaempheria were summarized. Biological activities including anticancer, anti-inflammatory, antimicrobial, anticholinesterase, antioxidant, anti-obesity-induced dermatopathy, wound healing, neuroprotective, anti-allergenic, and anti-nociceptive were demonstrated. Conclusions: Up to date, significant advances in phytochemical and pharmacological studies of different Kaempheria species have been witnessed. So, the traditional uses of these plants have been clarified via modern in vitro and in vivo biological studies. In addition, these traditional uses and reported biological results could be correlated via the chemical characterization of these plants. All these data will support the biologists in the elucidation of the biological mechanisms of these plants.

Graphical Abstract

1. Introduction

From the first known civilization, medicinal plants have met primary care and health needs around the world [1,2,3]. Natural products, derived from plants, have enriched the pharmaceutical industry since time immemorial. So far, people of the developing countries depend upon the traditional medicines to cure daily aliments [4]. The medicinal plants are characterized by a diversity of chemical and pharmacological constituents, owing to their complicity and the abundance of secondary metabolites. There are several factors that caused the variations of the secondary metabolites such as ecological zones, weather, climates, and other natural factors via the effects on the biosynthetic pathways [1,2,3].
Zingiberaceae (the ginger family) is distributed worldwide comprising 52 genera and more than 1300 plant species [5,6]. Kaempferia is a diverse family with members distributed widely throughout Southeast Asia and Thailand, including some 60 species [5]. Several Kaempferia species are used widely in folk medicine, including K. parviflora, K. pulchra, and K. galanga, (Figure 1). In Laos and Thai, traditional medicines derived from K. parviflora rhizomes are reported for the treatment of inflammation, hypertension, erectile dysfunction, abdominal ailments [6,7], and improvement of the vitality and blood flow [8]. Japanese use the extract of K. parviflora as a food supplement and for the treatment of metabolic disorders [9]. K. pulchra is used extensively as a carminative, diuretic, deodorant, and euglycemic, as well as for the treatment of urinary tract infections, fevers, and coughs [4]. The rhizomes of K. galanga are used as an anti-tussive, expectorant, anti-pyretic, diuretic, anabolic, and carminative, as well as for the curing of gastrointestinal ailments, asthma, wounds, rheumatism, epilepsy, and skin diseases [10].
Extracts and purified compounds from select Kaempferia species are used for the treatment of knee osteoarthritis and the inhibition of a breast cancer resistance protein (BCRP), anti-inflammatory, anti-acne, anticholinesterase, anti-obesity-induced dermatopathy, wound healing, anti-drug resistant strains of Mycobacterium tuberculosis, neuroprotective, anti-nociceptive, human immunodeficiency virus type-1 (HIV-1) inhibitory activity, in vitro anti-allergenic, and larvicidal activity against Aedes aegypti [4,6,7,8,9,10,11]. The scientific literature such as, Google Scholar, Scifinder, PubMed, Springer, Elsevier, Wiley, Web of Science, were screened in the period between 1972–2019 in order to collect the up-to-date information of the traditional uses/applications, biological studies, and chemical characterization of Kaempheria species. All these collected data were addressed and summarized in our review article to highlight the potential ethnopharmacological importance of these plants.

2. Materials and Methods

The scientific literature such as Google Scholar, Scifinder, PubMed, Springer, Elsevier, Wiley, Web of Science, etc., including all the traditional uses/applications, biological studies, and chemical characterization of Kaempheria species were collected between 1972–2019. All these collected data were adjusted and summarized in our review article due to the potential ethnopharmacological importance of these plants.
The correlation between the Kaempheria species was evaluated based on the main chemical classes of compounds. The data matrix of seven Kaempferia species (K. angustifolia, K. elegans, K. galanga, K. marginata, K. parviflora, K. pulchra, and K. roscoeana) and six chemical classes (abietanes, labdanes and clerodanes, flavonoids, phenolic compounds, and chalcones) were subjected to principal component analysis (PCA) to identify correlation between different Kaempferia species. In addition, the similarity based on the Pearson correlation coefficient was determined via subjecting the dataset to an agglomerative hierarchical cluster (AHC). The PCA and AHC were performed using an XLSTAT statistical computer software package (version 2018, Addinsoft, NY, USA, www.xlstat.com).

3. Distribution

Zingiberaceae (the ginger family) comprises 52 genera and more than 1300 plant species. Kaempferia is distributed worldwide with diverse members occurring throughout southeast tropical Asian countries such as Indonesia, India, Malaysia, Myanmar, Cambodia, and China, as well as Thailand, including some 60 species [5]. K. pulchra is a perennial herbal plant and widely cultivated in numerous tropical countries, involving Indonesia, Malaysia, Myanmar, and Thailand [12].

4. Traditional Uses

Several Kaempferia species are used widely in folk medicine, including K. parviflora, K. pulchra, and K. galanga (Figure 1). In Laos and Thai, traditional medicines derived from K. parviflora rhizomes are reported for the treatment of inflammation, hypertension, erectile dysfunction, abdominal ailments [6,7], and improvement of the vitality and blood flow [8]. Japanese folk medicine documented a positive effect of K. parviflora extract when used as a food supplement and for the treatment of metabolic disorders [9]. K. pulchra is used extensively as a carminative, diuretic, deodorant, and euglycemic, as well as for the treatment of urinary tract infections, fevers, and coughs [4]. K. galanga is sold as an industrial crop in the market, and its rhizome has been used as a flavor spice of various cooking [13]. The rhizomes of K. galanga is used as an anti-tussive, expectorant, anti-pyretic, diuretic, anabolic, carminative, as well as for curing of gastrointestinal ailments, asthma, wounds, rheumatism, epilepsy, and skin diseases [10]. In Malaysian folk medicines, several gingers belonging to the Zingiberaceae family especially, Kaempheria genus, are used in the treatment of several diseases such as stomach ailments, vomiting, cough, bruises, epilepsy, nausea, rheumatism, sore throat, wounds, eyewash, sore eyes, childbirth, liver complaints, muscular pains, ringworm, asthma, fever, malignancies, swelling, and several other disorders [14].

5. Biological Activity

Extracts and purified compounds of Kaempferia species are used for the treatment of knee osteoarthritis and the inhibition of a breast cancer resistance protein (BCRP), anti-inflammatory, anti-acne, anticholinesterase, anti-obesity-induced dermatopathy, wound healing, anti-drug resistant strains of Mycobacterium tuberculosis, neuroprotective, anti-nociceptive, human immunodeficiency virus type-1 (HIV-1) inhibitory activity, in vitro anti-allergenic, and larvicidal activity against Aedes aegypti [11]. Kaempheria plant extracts and isolated compounds demonstrate numerous and promising biological and pharmaceutical activities, which are summarized in Figure 2.

5.1. Anticancer Activity

Rhizome ethanolic extracts of K. galanga and the purified component ethyl trans p-methoxycinnamate (105) demonstrate moderate cytotoxic activity against human cholangiocarcinoma (CL-6) cells with IC50 of 64.2 and 49.4 μg mL−1, respectively. Significant cholangiocarcinoma (CCA) efficacy as indicated by suppressing tumor growth and lung metastasis in CL6-xenografed mice [15] is also observed. Swapana et al. [16] documented that K. galanga isopimarene diterpenoids, sandaracopimaradiene-9α-ol (2), kaempulchraol I (14), and kaempulchraol L (17) exhibit promising activity against human lung cancer with IC50 of 75 µM, 74 µM, and 76 µM, respectively, and mouth squamous cell carcinoma (HSC-2) inhibition with IC50 of 70 µM, 53 µM, and 58 µM, respectively [16]. The latter compound, isolated from K. pulchra, is reported to have weak anti-proliferative activity against human pancreatic and cervix cancers [17]. Chawengrum et al. [18] stated that K. pulchra labdene diterpenoids, (−)-kolavelool (81), and (−)-2β-hydroxykolavelool (82) exhibit cytotoxic activity against human leukemia cells (HL-60) with IC50 values of 9.0 ± 0.66 and 9.6 ± 0.88 μg mL−1, respectively [18]. Acetone, petroleum ether, chloroform, and MeOH extracts of K. galanga rhizomes show moderate cytotoxicity in a brine shrimp lethality bioassay compared with vincristine sulfate as the reference compound [19]. Moreover, a methanolic extract of K. galanga rhizomes induces Ehrlich ascites carcinoma (EAC) cell death in a dose-dependent manner [20]. 5,7-Dimethoxyflavone (86) isolated from K. galanga was found to reduce cancer resistance to tyrosine kinase inhibitors (TKI) by inhibiting breast cancer resistance protein (BCRP), one of the efflux transporters that increased efflux of TKI out of cancer cells. This was observed both in vitro with a dose-dependent increase in the intracellular concentration of sorafenib in MDCK/BCRP1 breast cancer resistance cells, with an EC50 of 8.78 μM as well as in vivo by increasing sorafenib AUC in mice tissues when co-administered with compound 88, as reported by kinetic results [21]. The isolated methyl-β-D-galactopyranoside specific lectin from the rhizome of K. rotunda exhibited in vitro antitumor activity against Ehrlich ascites carcinoma cells at a pH between 6–9 and a temperature range between 30–80 °C. Tumor inhibition was also observed in vivo in EAC-bearing mice [22].
The cytotoxicity of MeOH, petroleum ether, and EtOAc extracts against C33A cancer cells via MTT and scratch assays compared with essential oils of K. galanga rhizomes showed activity for the EtOAc and MeOH fractions at 1000 μg mL−1 with 11% and 14% cell viability and weak efficacy with petroleum ether extracted essential oils in a MTT assay. Cell growth inhibition was observed with all extracts in the scratch assay [23]. Compound (140) isolated from K. angustifolia was described to have strong activity with an IC50 of 1.4 µg mL−1, which was comparable to 5-fluorouracil as a reference drug. Compound (138) also showed moderate inhibition against human lung cancer. 2′-Hydroxy-4,4′,6′-trimethoxychalcone (flavokawain A; 119) exhibited potent activity against HL-60 and MCF-7 cell lines. The results of Tang et al. [24] revealed that flavokawain A (119) exhibited cytotoxic activity against MCF-7 and HT-29 cell lines with GI50 values of 17.5 µM (5.5 µg mL−1) and 45.3 μM (14.2 µg mL−1), respectively. Kaempfolienol (65) and zeylenol (133) were also found to have moderate activity against HL-60 and MCF-7 cells with IC50 values <30 µg mL−1 and against HL-60 only with an IC50 value of 11.6 µg mL−1 respectively [24].

5.2. Anti-Obesity Activity

An ethanolic extract, a polymethoxyflavonoid-rich fraction (PMF) and a polymethoxyflavonoid-poor fraction from K. parviflora were screened against an obesity-induced dermatopathy system using Tsumura Suzuki obese diabetes (TSOD) mice as an obesity model (Hidaka, Horikawa, Akase, Makihara, Ogami, Tomozawa, Tsubata, Ibuki, and Matsumoto) [11]. The ethanolic extract reduced mouse body weight and the thickness of the subcutaneous fat layer more than the PMF fraction that is used as a dietary supplement in controlling skin disorders caused by obesity [11].

5.3. Anti-HIV Activity

Viral protein R (Vpr) is one of the HIV accessory proteins that can be targeted for controlling viral replication and pathogenesis. A CHCl3 fraction of K. pulchra exhibits Vpr-inhibitory activity at 25l g mL−1. In addition, isopimarene type diterpenoids isolated from the rhizomes of the plants, kaempulchraol B (43), kaempulchraol D (45), kaempulchraol G (46), kaempulchraol Q (20), kaempulchraol T (36), kaempulchraol U (50), and W (22) inhibit the expression of Vpr at concentrations from 1.56 to 6.25 µM [25].

5.4. Antimicrobial Activity

Arabietatriene (62) isolated from K. roscoeana exhibits antibacterial activity against Gram-positive bacteria Staphylococcus epidermidis and Bacillus cereus [26]. Anticopalic acid (72), anticopalol (77), and 8(17)-labden-15-ol (68) isolated from K. elegans also exhibited antibacterial activity against B. cereus [18]. Acetone, petroleum ether, chloroform, and MeOH extracts of K. galanga rhizomes exhibit moderate antibacterial activity against Gram-positive and Gram-negative bacteria in comparison with ciprofloxacin [19]. Ethyl p-methoxycinnamate (105) also isolated from K. galanga rhizomes have been shown based on a resazurin micro-titer assay to inhibit Mycobacterium tuberculosis H37Ra, H37Rv, multidrug-resistant, and drug-susceptible isolates with MIC 0.242–0.485 mM [27]. Its essential oil also displays strong antibacterial activity against Staphylococcus aureus and Salmonella typhimurium, and weak activity against Escherichia coli [28]. Moreover, essential oils extracted from three varieties of K. galanga exhibited potent larvicidal activity [29]. An ethyl acetate extract of K. rotunda inhibits S. aureus and E. coli [30]. A rhizomes extract of K. galanga inhibits Epstein–Barr virus with no cytotoxic effect in Raji cells [14]. In contrast, isolated diterpenoids from K. roscoeana exhibited no activity against Plasmodium falciparum (Chloroquine-resistant) [26]. Fauziyah et al. [31] described that an ethanolic extract of K. galanga alone exhibits 100% growth inhibition of the multi-drug resistant (MDR) Mycobacterium tuberculosis (isolates at 500 µg mL−1). However, a combination of this extract with streptomycin, ethambutol, and isoniazid showed inhibition values of 55%, 76%, and 50%, respectively. Ethanol, methanol, petroleum ether, chloroform, and aqueous extracts of K. galanga rhizome showed antimicrobial activity against human pathogenic bacteria and fungi, while the ethanolic extract exhibited the strongest inhibition of S. aureus using an inhibition zone assay [32]. However, flavokawain A (119) and other compounds reported from K. angustifolia had no antimicrobial activity against tested microbes [24].

5.5. Antioxidant Activity

The CHCl3 and MeOH extracts of the rhizomes of K. angustifolia showed strong antioxidant activity against DPPH expressed with 615.92 mg trolox equivalent (TE)/g of extract. In an azinobis (3-ethyl-benzothiazoline-6-sulfonic acid) (ABTS) assay, MeOH extracts showed good antioxidant properties with a value of 38.87 mg TE/g. However, n-hexane extract exhibited significant antioxidant activity with 901.76 mg TE/g in a cupric-reducing antioxidant capacity assay, while EtOAc extract exhibited significant reduction ability against ferric reducing antioxidant power (FRAP) with a value of 342.23 mg TE/g. Also, kaempfolienol (65) showed potent free radical scavenging activity in a DPPH assay, as well as, 2′-hydroxy-4,4′,6′-trimethoxychalcone (119) in ABTS, CUPRAC, and FRAP assays [33,34]. A methanol extract of rhizomes of K. galanga exhibited a concentration-dependent antioxidant activity in DPPH, ABTS, and nitric oxide (NO) radical scavenging assays [20]. Moreover, the essential oil extracts of conventionally propagated and in vitro propagated K. galanga had significant DPPH radical scavenging activity [35]. As well, the ethanol extract of K. rotunda exhibited antioxidant activity in a DPPH assay with IC50 (67.95 μg mL−1) [30].

5.6. Anti-Inflammatory Activity

The cyclohexane, chloroform, and ethyl acetate extracts with diarylheptanoids isolated from K. galanga showed a pronounced inhibition of Lipopolysaccharides (LPS)-induced nitric oxide in macrophage RAW 264.7 cells compared with indomethacin [13]. The EtOH extract and compounds (1, 52, 53, 119, 120) isolated from K. marginata had promising anti-inflammatory activity based on the suppression of NO production and inducible nitric oxide synthase (iNOS) mRNA and cyclooxygenase-2 (COX-2) genes expression [36,37]. Diterpenoids (910) isolated from K. pulchra had topical anti-inflammatory activity in 12-O-tetradecanoylphorbol-13-acetate-induced ear edema in rats with ID50 330 and 50 µg/ear, respectively. Biological activity may be due to the activation of Maxi-K channels in neurons and smooth muscles [38]. The ethanol extract of K. parviflora exhibited potent inhibition of PGE2. The plant extract and 3′,4′,5,7-tetramethoxyflavone (86) were also reported to exhibit a dose-dependent inhibition of iNOS-mRNA expression. Additionally, H2O, EtOH, EtOAC, CHCl3, and n-hexane soluble sub-fractions exhibited good in vivo anti-inflammatory activity by decreasing rat paw edema [39]. An 80% EtOH extract reduced UV-induced COX-2 expression in mice skin that was attributed to the anti-oxidative activity of polyphenolics against the oxidizing properties of UV radiation [40]. A 60% EtOH and EtOAc-soluble fraction of 100% methanol extracts of K. parviflora decreased knee osteoarthritis, which was likely due to methoxylated flavones [41]. Ethyl p-methoxycinnamate (105) isolated from K. galana inhibited cytokines as IL-1 and TNFα and endothelial function in rats [42].
Tewtrakul, et al. [43] found that the isolated methoxylated flavonoids from K. parviflora, 5-hydroxy-3,7,3′,4′-tetramethoxyflavone (96), 5-hydroxy-7,4′-dimethoxyflavone (93), and 5-hydroxy-3,7,4′-trimethoxyflavone (95) exhibited anti-inflammatory activity against the PGE2 production, with IC50 values of 16.1 μM, 24.5 μM, and 30.6 μM, respectively [43]. Tewtrakul and Subhadhirasakul [44] described methoxyflavones 96, 93, and 95 from a hexane extract of K. parviflora rhizomes that exhibited activity against NO release in RAW264.7 cells with IC50 values of 16.1 μM, 24.5 μM, and 30.6 μM, respectively. In addition, 5-hydroxy-3,7,3′,4′-tetramethoxyflavone (96) inhibited PGE2 release with an IC50 value of 16.3 μM, with negative activity on Tumor Necrosis Factor alpha (TNF-α) with IC50 >100 μM [44]. Petroleum ether extract from K. galanga was active against acute inflammation at 300 mg/kg in rats and inhibited the inflammation and MPO levels at 100 mg kg−1 in the chronic model [45].

5.7. Anticholinesterase Activity

According to Sawasdee et al. [46], a MeOH extract as well as compounds (8687) isolated from K. parviflora rhizomes inhibited acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) with greater cholinesterase inhibitory toward AChE and BChE for (86), which was an observation of significance in the treatment of Alzheimer’s disease [46].

5.8. Anti-Mutagenicity Activity

CH2Cl2 and EtOAc soluble fractions of K. parviflora showed anti-mutagenicity and α-glucosidase inhibitory activity. Isolated methoxylated compounds (86, 97, 84, and 92) from these extracts exhibited potent activity with IC50 values of 0.40, 0.40, 0.42, and 0.47 nmol/plate, respectively. Compounds (88, 87, and 91), also showed significant activity with IC50 values of 20.4 μM, 54.3 μM, and 64.3 μM, respectively [47].

5.9. Effect on Cytochromes CYP 450

The results listed by Ochiai et al. [48] stated that the continued ingestion of (88) isolated from K. parviflora decreases liver CYP3A expression, which in turn increased levels of compounds metabolized by CYP3As such as midazolam [48].

5.10. Vascular Activity

The oral administration of CH2Cl2 extract of K. parviflora in middle-aged rats was found to decrease vascular responses to phenylephrine, increase acetylcholine-induced vasorelaxation and the production of nitric oxide (NO) from blood vessels, and decrease visceral, subcutaneous fat, fasting serum glucose, triglyceride, and liver lipid accumulation [49]. The effect of intravenous administration of a CH2Cl2 extract of K. galanga to rats reduced the mean arterial blood pressure [50]. This anti-hypertensive effect was attributed to ethyl cinnamate, which is a major compound in the extract [50]. The ethanol extract of rhizomes of K. parviflora caused dose-dependent relaxation on aortic rings as well as ileum pre-contracted with phenylephrine and acethylcholine [51].

5.11. Adaptogenic Activity

Hexane, chloroform, methanol, and ethanol extracts of K. parviflora exhibited adaptogenic activity compared with a crude ginseng root powder used as a reference [52]. A single oral dose of K. parviflora rhizome (60% EtOH extract) increased the whole-body potential expenditure in humans [53]. K. parviflora was also found to improvement physical fitness and health by decreasing oxidative stress [54].

5.12. Xanthine Oxidase Inhibitory Activity

Among the isolated methoxylated flavonoids from K. parviflora, (87 and 86) inhibit xanthine oxidase activity with IC50 values of 0.9 and >4 mM, respectively [9].

5.13. Allergenic Activity

Isolated polymethoxyflavones from K. parviflora (86, 97), in addition to CH2Cl2, EtOAc, and H2O extracts, alleviated type I allergy symptoms through suppressing Rat Basophilic Leukemia cells (RBL-2H3) cell degranulation, with (92) and (94) showing the highest anti-allergenic activity [55].

5.14. Neurological Activity

A methanolic extract (95% MeOH) of K. parviflora exhibited neuroprotective activity by increasing rat hippocampus serotonin, norepinephrine, and dopamine levels in comparison with a vehicle-treated group [56]. An acetone extract of K. galanga rhizomes and leaves also exhibited central nervous system depressant activity [57].

5.15. Nociceptive Activity.

A K. galanga rhizome extract exhibited anti-nociceptive activity in rats that was stronger than aspirin but weaker than morphine. The efficacy was abolished by naloxone, suggesting that the analgesic effect may be centrally and peripherally mediated [58].

5.16. Wound-Healing Activity

The co-administration of a K. galanga rhizomes extract (95% EtOH) with dexamethazone was found to have wound-healing activity in mice comparable to dexamethazone only [59].

5.17. Effects on Sexual Performance

Several 7-methoxyflavones (86, 87, 89, 91, 93–95) isolated from K. parviflora rhizomes improved sexual activity in males through the inhibition of PDE5, with 86 being the most potent [60]. The activity was attributed to methoxyls present at positions C5 and C7 [60]. K. parviflora rhizome extracts, standardized to 5% DMF, also improve erectile function in healthy men [61]. A K. parviflora extract as well as 5,7-dimethoxyflavones augment testosterone production, which decreases age-related diseases and hypogonadism [62]. Improved testosterone levels, sperm count, and sexual performance was observed in streptozotocin (STZ)-induced diabetic rats when treated with a K. parviflora extract (aqueous with 1% Tween-80) [63].

5.18. Miscellaneous

The rhizome extract (95% ethanolic) of K. parviflora reduced obesity via the inhibition of adipogenesis, lipogenesis, and muscle atrophy in mice [64]. In contrast, the K. parviflora derivatives of 5-hydroxy-7-methoxyflavone induce skeletal muscle hypertrophy [65]. A K. parviflora extract (95% EtOH) served as a potential anti-acne agent with anti-inflammatory, sebostatic, and anti-propioni bacteria activity [66].
Recently, K. parviflora alcoholic extract at 3–30 µg mL−1 was evaluated regarding the molecular mechanisms associated with rheumatoid arthritis for up to 72 h compared with the dexamethasone as positive control [67]. They documented that the EtOH extract significantly decreased the gene expression levels of pro-inflammatory cytokines, inflammatory mediators, and matrix-degraded enzymes, but neither induced apoptosis nor altered the cell cycle. They also reported that the alcoholic extract inhibits cell migration, reduces the mRNA expression of cadherin-11, and selectively reduces the phosphorylation of mitogen-activated protein kinases (P38, MAPKs), signal transducers, and activators of transcription 1 (STAT1) and 3 (STAT3) signaling molecules, without interfering with the NF-κB pathway [67].
A K. galanga extract (acetone, petroleum ether, chloroform, or methanolic) exhibited dose-dependent anthelmintic activity with strong paralytic activity within one hour and death within 80 min at a 25 mg mL−1 concentration [68].

6. Chemical Metabolites of Kaempferia Species

Chemical profiles of Kaempferia exhibited the presence of different types of secondary metabolites such as terpenoids, especially isopimarane phenolic compounds, diarylheptanoids [13], flavonoids [69,70,71], and essential oils [72,73]. This review summarized the reported variety of compound types, including isopimarane, abietane, labdan, and clerodane diterpenoids, flavonoids, phenolic acids, phenyl-heptanoids, curcuminoids, tetrahydropyrano-phenolic, and steroids. Diterpenoids, especially isopimarane types, were the most reported compounds from the plants of this genus, in addition to phenolics, flavonoids, and essential oils. Each class will be described and listed in the following items, and the structures will be summarized in Table 1, Table 2 and Table 3.

6.1. Diterpenoids

Kaempferia plants were characterized with a predominance of diterpenoids, especially the isopimaranes in addition to abietane, labdane, and clerodane types (Table 1).

6.1.1. Isopimarane-Type Diterpenoids

The isopimaranes reported from the Kaempheria species (Table 1) are characterized with the presence of two double bonds; one is mostly ∆15(16), while the other is between ∆8(9) or ∆8(14) [4,25,74]. From the rhizomes of K. galanga, 12 usual isopimarenes (1–8, 10, 11, 14, and 17) were observed that contained a ∆8(14),15 motif in addition to the rarely reported oxygenated seco-isopimarane (56) [16]. From the rhizomes of K. marginata, five isopimarenes with a ∆8(14),15 motif were observed (1, 2, 5254) [36]. Only one thumbing isopimarenes, roscorane A (57), was reported from K. roscoeana, which was characterized by only one double bond ∆8(9) and (7-8)-epoxy, as well as the absence of the exomethylene ∆15(16) [26].

Biosynthesis of Isopimarane-Type Diterpenoids

Isopimarane diterpenoids are the most characteristic compounds for Kaempheria plants. (E,E,E)-Geranylgeranyl diphosphate (GGPP) is a well-known biosynthesized intermediate of diterpenoids as described by [80]. GGPP is firstly cyclized via copalyl diphosphate (CPP) synthases (CPS), and then by the unknown enzyme (PS), affording a charged intermediate (INM). Then, this intermediate is completely cyclized by the enzymatic reactions via the bifunctional (iso) pimaradiene synthases (AoCPS-PS, NfCPS-PS, and AfCPS-PS) (Scheme 1), as described by Xu et al. [81].

6.1.2. Abietane-Type Diterpenoids

Seven abietanes (58–64) (Table 1) have been isolated and characterized from the rhizomes of K. roscoeana, and one (65) was isolated and characterized from K. angustifolia [26,33,34]. These highly oxygenated metabolites contain one or more double bonds and an absence of exomethylenes, except for roscotane D (61), which contains no double bonds.

6.1.3. Labdane and Clerodane Diterpenoids

After isopimarenes, labdane and clerodane represent major diterpenoid classes from the Kaempheria species. Nineteen highly oxygenated labdanes and clerodanes (6686) have been reported from Kaempheria rhizomes (Table 1) [18,26]. From these isolated labdanes, only (12Z,14R)-labda-8(17),12-dien-14,15,16-triol (66) has been isolated from K. roscoeana rhizomes. In contrast, several labdane and clerodane types of diterpenoids have been isolated from K. elegans and K. pulchra rhizomes collected in Thailand.

6.1.4. Flavonoids

Kaempheria species are characterized by rich biological activity due in part to the presence of a diversity of flavonoids (86105) and phenolic compounds (106–137) (Table 2). K. parviflora rhizomes with flavonoid nuclei contain methoxy groups in specific positions (8697) [9,55]. Pyrano-flavone, 2”,2”-dimethylpyrano-[5”,6”:8,7]-flavone (105), has been isolated from K. pulchra rhizomes collected from Thailand [18], and flavanones (97–99) have been isolated and identified from K. parviflora rhizomes [70,71]. K. galanga contains kaempferol and kaempferide (98, 99) [78].

6.1.5. Phenolic Compounds

From K. galanga rhizomes, diarylheptanoid compounds (116, 117, 122–125) are reported by Yao, Huang, Wang, and He [13]. From K. marginata rhizomes, curcuminoid (121) was characterized by Kaewkroek, Wattanapiromsakul, Kongsaeree, and Tewtrakul [36]. From K. galanga, rhizomes phenolic acids (106113) were the major compounds isolated, including methoxylated cinnamic acid derivatives. Two (4-methoxyphenyl)-propanoates (114–115) were also isolated from the K. galanga rhizomes [13,50]. S- and R-isomers at C-4 of phenolic glycosides (135 and 136) as well as a rare phenolic glycoside (137) were observed in K. previflora rhizomes [79]. All the phenolic compounds (106–137) are summarized in Table 2.

6.1.6. Steroids and Triterpenes

Steroids represent a minor class of compounds reported from Kaempheria species. Only three steroids, β-sitosterol (138), β-sitosterol-β-D-glucoside (139), and stigmasterol (140) (Table 3) have been reported from K. marginata rhizomes [36]. Moreover, only one lanostane type triterpene, (24S)-24-methyl-lanosta-9(11), 25-dien-3β-ol (141), was isolated from K. angustifolia [24].

6.1.7. Volatile Oils

Kaempheria species were documented as very rich plants with volatile oils such as K. galanga [29,73,82,83], K. angustiflora [29], and K. marginata [29]. The volatile oil of K. galanga has been reported as a potential market product in India and over all the world with market values around 600–700 US$/kg on the international market [83]. Phenylpropanoids and/or cinamates were represented as major constituents of volatile oils derived from Kaempheria species followed by monoterpenes [29,73,82]. The phenylpropanoid compound, trans-ethyl cinnamate, was documented as a principal component of volatile oils of all the studied Kaempheria species up to date with concentrations varied from 16–35% of the total identified [29,73,82,83]. The volatile oils of Kaempheria species were reported to have numerous biological activities such as anti-microbial [83], antioxidant [35], nutraceutical [83], nematicidal toxicity [82], and larvicide activities [29]. Table 4 summarized the main components (142–157) of the reported volatile oils of Kaempheria species.

7. Principal Components Analysis (PCA) and Agglomerative Hierarchical Clustering (AHC) for Kaempferia Species

To assess the correlation between the various Kaempferia species, chemical classes of different compounds were subjected to PCA and AHC (Figure 3). According to the similarity, the analysis showed that we can group the Kaempferia species under three groups: the first group comprised K. galanga, K. marginata, K. pulchra, and K. roscoeana, and these species are correlated to isopimaranes compounds. The Pearson correlation coefficient (r) between K. marginata and K. pulchra was the highest with r = 0.938, while between K. marginata and K. roscoeana, it was 0.771, between K. roscoeana and K. pulchra, it was 0.766, and between K. marginata and K. galanga, it was 0.615 (Table 5).
The second group contained K. angustifolia and K. parviflora (r = 0.833), and this group showed a close correlation to flavonoids and phenolics. However, the K. elegans was separated alone, and showed a close relation to labdane and clerodane compounds. The similarities within each group might be ascribed to the genetic relations, as well as the environmental and microclimatic conditions [1,2,3].
In a study of a genetic variation of Kaempferia species based on chloroplast DNA [5], K. marginata and K. galanga were grouped together, which is agreeable with our results (r = 0.615) according to the PCA data of the present study based on the chemical composition. However, in contrast to the data from the PCA, K. angustifolia and K. parviflora were separated in different groups, but K. elegans and K. parviflora were grouped together. In another recent study, based on the DNA and morphological characteristics [84], K. angustifolia and K. parviflora were grouped together in agreement with the chemical variation of the present study.

8. Conclusions

Kaempheria species are widely used plants in traditional medicine worldwide. All the biological activity data for these plants and their isolated constituents have resulted in numerous leads for medicinal drugs. Mainly, seven rhizomes of Kaempheria plants afforded a vast array of diterpenoids, especially the isopimarane type, along with significant bioactive methoxylated flavonoids. From all these documented chemical and biological results, these plants have been and continue to be a promising source for medicinal natural products and food industrial products.

Author Contributions

A.I.E., T.A.M. and M.-E.F.H. suggested and designed the study. A.I.E., T.A.M., A.F.E., and A.M.A.-E.G. did the data preparation and structure drawing. A.I.E., T.A.M., A.M.A.-E.G., and M.-E.F.H. collected the data. A.S.A., A.A.S., T.Y., A.R.H.F., and M.N. wrote the original manuscript. A.I.E., T.A.M., M.-E.F.H., P.W.P., A.U., A.S.A., A.A.S., and H.R.E.-S. edited the final manuscript. All the authors reviewed and approved the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors are thankful to the Deanship of the Scientific Research and Research Center, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia. Dr. Elshamy gratefully acknowledges the Takeda Science Foundation, Japan for financial support. Prof. Mohamed Hegazy gratefully acknowledges the financial support from Alexander von Humboldt Foundation “Georg Foster Research Fellowship for Experienced Researcher”.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

K.Kaempheria
Sp.Species
PCAPrincipal component analysis
AHCAgglomerative hierarchical clustering
CCASignificant Cholangiocarcinoma
HSC-2Mouth squamous cell carcinoma
EACEhrlich ascites carcinoma cancer cells
HL-60Human leukemia cancer cells
CL-6Human cholangiocarcinoma cells
TKITyrosine kinase inhibitors
BCRPBreast cancer resistance protein
MTT(3-[4,5,[4,5-Dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide) assay
CH2Cl2Dichloromethane
BChEButyrylcholinesterase
NONitric oxide
CUPRACModified cupric reducing antioxidant capacity
PDE5Phosphodiesterase type 5 inhibitor
APAerial parts
AChEAcetylcholinesterase
RBL-2H3Rat Basophilic Leukemia cells
P38Type of mitogen-activated protein kinases
STAT1 and 3Signal transducers and activators of transcription 1 and 3
MeOHMethanol
EtOAcEthyl acetate
MCF-7Breast cancer cells
HT-29Colorectal adenocarcinoma cell
PMFPolymethoxyflavonoid-rich fraction
TSODTsumura Suzuki obese diabetes
GGPP(E,E,E)-Geranylgeranyl diphosphate
CPPCopalyl diphosphate
CPSCopalyl diphosphate synthases
EtOHEthanol
AChEAcetylcholinesterase
ABTS2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulphonic acid) assay
VprViral protein R
DPPH1,1-Diphenyl-2-picrylhydrazyl assay
FRAPFerric reducing antioxidant power
CYP3ACytochrome P450, family 3, subfamily A
NF-κBNuclear factor pathway
RhRhizomes
BChEButyrylcholinesterase
STZStreptozotocin
MPOmyeloperoxidase
DMFDimethylformamide
MAPKsType of mitogen-activated protein kinases
PGE2Prostaglandin E2

References

  1. Vinceti, B.; Loo, J.; Gaisberger, H.; van Zonneveld, M.J.; Schueler, S.; Konrad, H.; Kadu, C.A.; Geburek, T. Conservation priorities for Prunus africana defined with the aid of spatial analysis of genetic data and climatic variables. PLoS ONE 2013, 8, e59987. [Google Scholar] [CrossRef] [PubMed]
  2. Pérez-Sánchez, R.; Gálvez, C.; Ubera, J.L. Bioclimatic influence on essential oil composition in South Iberian Peninsular populations of Thymus zygis. J. Essent. Oil Res. 2012, 24, 71–81. [Google Scholar] [CrossRef]
  3. Elshamy, A.; Mohamed, T.A.; Al-Rowaily, S.L.; Abd El-Gawad, A.M.; Dar, B.A.; Shahat, A.A.; Hegazy, M.F. Euphosantianane E–G: Three new premyrsinane type 2 diterpenoids from Euphorbia sanctae-catharinae with 3 contribution to chemotaxonomy. Molecules 2019, 24, 2412. [Google Scholar] [CrossRef] [PubMed]
  4. Ekor, M. The growing use of herbal medicines: Issues relating to adverse reactions and challenges in monitoring safety. Front Pharmacol. 2013, 4, 177. [Google Scholar] [CrossRef] [PubMed]
  5. Techaprasan, J.; Klinbunga, S.; Ngamriabsakul, C.; Jenjittikul, T. Genetic variation of Kaempferia (Zingiberaceae) in Thailand based on chloroplast DNA (psbA-trnH and petA-psbJ) sequences. Genet. Mol. Res. 2010, 9, 1957–1973. [Google Scholar] [CrossRef]
  6. Wutythamawech, W. Encyclopedia of Thai Herbs; OS Printing: Bangkok, Thailand, 1997; p. 365. [Google Scholar]
  7. Yenjai, C.; Prasanphen, K.; Daodee, S.; Wongpanich, V.; Kittakoop, P. Bioactive flavonoids from Kaempferia parviflora. Fitoterapia 2004, 75, 89–92. [Google Scholar] [CrossRef]
  8. Akase, T.; Shimada, T.; Terabayashi, S.; Ikeya, Y.; Sanada, H.; Aburada, M. Antiobesity effects of Kaempferia parviflora in spontaneously obese type II diabetic mice. J. Nat. Med. 2011, 65, 73–80. [Google Scholar] [CrossRef]
  9. Nakao, K.; Murata, K.; Deguchi, T.; Itoh, K.; Fujita, T.; Higashino, M.; Yoshioka, Y.; Matsumura, S.-I.; Tanaka, R.; Shinada, T. Xanthine oxidase inhibitory activities and crystal structures of methoxyflavones from Kaempferia parviflora rhizome. Biol. Pharm. Bull. 2011, 34, 1143–1146. [Google Scholar] [CrossRef]
  10. Sivarajan, V.; Balachandran, I. Ayurvedic Drugs and their Plant Sources Oxford and IBH Publishing Co; Oxford & IBH Pub. Co: New Delhi, India, 1994. [Google Scholar]
  11. Hidaka, M.; Horikawa, K.; Akase, T.; Makihara, H.; Ogami, T.; Tomozawa, H.; Tsubata, M.; Ibuki, A.; Matsumoto, Y. Efficacy of Kaempferia parviflora in a mouse model of obesity-induced dermatopathy. J. Nat. Med. 2017, 71, 59–67. [Google Scholar] [CrossRef]
  12. Win, N.N.; Ito, T.; Matsui, T.; Aimaiti, S.; Kodama, T.; Ngwe, H.; Okamoto, Y.; Tanaka, M.; Asakawa, Y.; Abe, I. Isopimarane diterpenoids from Kaempferia pulchra rhizomes collected in Myanmar and their Vpr inhibitory activity. Bioorganic Med. Chem. Lett. 2016, 26, 1789–1793. [Google Scholar] [CrossRef]
  13. Yao, F.; Huang, Y.; Wang, Y.; He, X. Anti-inflammatory diarylheptanoids and phenolics from the rhizomes of kencur (Kaempferia galanga L.). Ind. Crop. Prod. 2018, 125, 454–461. [Google Scholar] [CrossRef]
  14. Vimala, S.; Norhanom, A.; Yadav, M. Anti-tumour promoter activity in Malaysian ginger rhizobia used in traditional medicine. Br. J. Cancer 1999, 80, 110. [Google Scholar] [CrossRef] [PubMed]
  15. Amuamuta, A.; Plengsuriyakarn, T.; Na-Bangchang, K.J. Anticholangiocarcinoma activity and toxicity of the Kaempferia galanga Linn. rhizome ethanolic extract. BMC Complementary Altern. Med. 2017, 17, 213. [Google Scholar] [CrossRef] [PubMed]
  16. Swapana, N.; Tominaga, T.; Elshamy, A.I.; Ibrahim, M.A.; Hegazy, M.-E.F.; Singh, C.B.; Suenaga, M.; Imagawa, H.; Noji, M.; Umeyama, A. Kaemgalangol A: Unusual seco-isopimarane diterpenoid from aromatic ginger Kaempferia galanga. Fitoterapia 2018, 129, 47–53. [Google Scholar] [CrossRef] [PubMed]
  17. Win, N.N.; Ito, T.; Aimaiti, S.; Kodama, T.; Imagawa, H.; Ngwe, H.; Asakawa, Y.; Abe, I.; Morita, H. Kaempulchraols I–O: New isopimarane diterpenoids from Kaempferia pulchra rhizomes collected in Myanmar and their antiproliferative activity. Tetrahedron 2015, 71, 4707–4713. [Google Scholar] [CrossRef]
  18. Chawengrum, P.; Boonsombat, J.; Kittakoop, P.; Mahidol, C.; Ruchirawat, S.; Thongnest, S. Cytotoxic and antimicrobial labdane and clerodane diterpenoids from Kaempferia elegans and Kaempferia pulchra. Phytochem. Lett 2018, 24, 140–144. [Google Scholar] [CrossRef]
  19. Dash, P.R.; Nasrin, M.; Ali, M.S. In vivo cytotoxic and In vitro antibacterial activities of Kaempferia galanga. J. Pharmacogn. Phytochem. 2014, 3, 172–177. [Google Scholar]
  20. Ali, H.; Yesmin, R.; Satter, M.A.; Habib, R.; Yeasmin, T. Antioxidant and antineoplastic activities of methanolic extract of Kaempferia galanga Linn. Rhizome against Ehrlich ascites carcinoma cells. J. King Saud Univ. Sci. 2018, 30, 386–392. [Google Scholar] [CrossRef]
  21. Bae, S.; D’cunha, R.; Shao, J.; An, G. Effect of 5, 7-dimethoxyflavone on Bcrp1-mediated transport of sorafenib in vitro and in vivo in mice. Eur. J. Pharm. Sci. 2018, 117, 27–34. [Google Scholar] [CrossRef]
  22. Ahmed, F.R.S.; Amin, R.; Hasan, I.; Asaduzzaman, A.; Kabir, S.R. Antitumor properties of a methyl-β-d-galactopyranoside specific lectin from Kaempferia rotunda against Ehrlich ascites carcinoma cells. Int. J. Biol. Macromol. 2017, 102, 952–959. [Google Scholar] [CrossRef]
  23. Omar, M.N.; Rahman, S.A.; Ichwan, S.; Hasali, N.; Rasid, F.A.; Halim, F.A. Cytotoxicity effects of extracts and essential oil of Kaempferia galanga on cervical cancer C33A cell line. Orient. J. Chem. 2017, 33, 1659–1664. [Google Scholar] [CrossRef]
  24. Tang, S.W.; Sukari, M.A.; Neoh, B.K.; Yeap, Y.S.Y.; Abdul, A.B.; Kifli, N.; Cheng Lian Ee, G. Phytochemicals from Kaempferia angustifolia Rosc. and their cytotoxic and antimicrobial activities. Biomed Res. Int. 2014, 2014. [Google Scholar] [CrossRef] [PubMed]
  25. Win, N.N.; Ngwe, H.; Abe, I.; Morita, H. Naturally occurring Vpr inhibitors from medicinal plants of Myanmar. J. Nat. Med. 2017, 71, 579–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Boonsombat, J.; Mahidol, C.; Chawengrum, P.; Reuk-Ngam, N.; Chimnoi, N.; Techasakul, S.; Ruchirawat, S.; Thongnest, S. Roscotanes and roscoranes: Oxygenated abietane and pimarane diterpenoids from Kaempferia roscoeana. Phytochemistry 2017, 143, 36–44. [Google Scholar] [CrossRef]
  27. Lakshmanan, D.; Werngren, J.; Jose, L.; Suja, K.; Nair, M.S.; Varma, R.L.; Mundayoor, S.; Hoffner, S.; Kumar, R.A. Ethyl p-methoxycinnamate isolated from a traditional anti-tuberculosis medicinal herb inhibits drug resistant strains of Mycobacterium tuberculosis in vitro. Fitoterapia 2011, 82, 757–761. [Google Scholar] [CrossRef]
  28. Yang, Y.; Tian, S.; Wang, F.; Li, Z.; Liu, L.; Yang, X.; Bao, Y.; Wu, Y.; Huang, Y.; Sun, L. Chemical composition and antibacterial activity of Kaempferia galanga essential oil. Int. J. Agric. Biol. 2018, 20, 457–462. [Google Scholar] [CrossRef]
  29. Panyakaew, J.; Sookkhee, S.; Rotarayanont, S.; Kittiwachana, S.; Wangkarn, S.; Mungkornasawakul, P. Chemical variation and potential of Kaempferia oils as larvicide against Aedes aegypti. J. Essent. Oil Bear. Plants 2017, 20, 1044–1056. [Google Scholar] [CrossRef]
  30. Malahayati, N.; Widowati, T.W.; Febrianti, A. Total phenolic, antioxidant and antibacterial activities of curcumin extract of kunci pepet (Kaempferia rotunda L). Res. J. Pharm. Biol. Chem. Sci. 2018, 9, 129–135. [Google Scholar]
  31. Fauziyah, P.N.; Sukandar, E.Y.; Ayuningtyas, D.K. Combination effect of antituberculosis drugs and ethanolic extract of selected medicinal plants against multi-drug resistant Mycobacterium tuberculosis isolates. Sci. Pharm. 2017, 85, 14. [Google Scholar] [CrossRef]
  32. Kochuthressia, K.; Britto, S.J.; Jaseentha, M.; Raphael, R. In vitro antimicrobial evaluation of Kaempferia galanga L. rhizome extract. Am. J. Biotechnol. Mol. Sci. 2012, 2, 1–5. [Google Scholar] [CrossRef]
  33. Yeap, Y.S.Y.; Kassim, N.K.; Ng, R.C.; Ee, G.C.L.; Saiful Yazan, L.; Musa, K.H. Antioxidant properties of ginger (Kaempferia angustifolia Rosc.) and its chemical markers. Int. J. Food Prop. 2017, 20, 1158–1172. [Google Scholar] [CrossRef]
  34. Tang, S.W.; Sukari, M.A.; Rahmani, M.; Lajis, N.H.; Ali, A.M. A new abietene diterpene and other constituents from Kaempferia angustifolia Rosc. Molecules 2011, 16, 3018–3028. [Google Scholar] [CrossRef] [PubMed]
  35. Sahoo, S.; Parida, R.; Singh, S.; Padhy, R.N.; Nayak, S. Evaluation of yield, quality and antioxidant activity of essential oil of in vitro propagated Kaempferia galanga Linn. J. Acute Dis. 2014, 3, 124–130. [Google Scholar] [CrossRef]
  36. Kaewkroek, K.; Wattanapiromsakul, C.; Kongsaeree, P.; Tewtrakul, S. Nitric oxide and tumor necrosis factor-alpha inhibitory substances from the rhizomes of Kaempferia marginata. Nat. Prod. Commun. 2013, 8, 1205–1208. [Google Scholar] [CrossRef] [PubMed]
  37. Kaewkroek, K.; Wattanapiromsakul, C.; Matsuda, H.; Nakamura, S.; Tewtrakul, S. Anti-inflammatory activity of compounds from Kaempferia marginata rhizomes. Songklanakarin J. Sci. Technol. 2017, 39, 91–99. [Google Scholar]
  38. Sematong, T.; Reutrakul, V.; Tuchinda, P.; Claeson, P.; Pongprayoon, U.; Nahar, N. Topical antiinflammatory activity of two pimarane diterpenes from Kaempferia pulchra. Phytother. Res. 1996, 10, 534–535. [Google Scholar] [CrossRef]
  39. Sae-wong, C.; Tansakul, P.; Tewtrakul, S. Anti-inflammatory mechanism of Kaempferia parviflora in murine macrophage cells (RAW 264.7) and in experimental animals. J. Ethnopharmacol. 2009, 124, 576–580. [Google Scholar] [CrossRef]
  40. Lee, M.-h.; Han, A.-R.; Jang, M.; Choi, H.-K.; Lee, S.-Y.; Kim, K.-T.; Lim, T.-G. Antiskin inflammatory activity of black ginger (Kaempferia parviflora) through antioxidative activity. Oxidative Med. Cell. Longev. 2018, 2018, 5967150. [Google Scholar] [CrossRef]
  41. Kobayashi, H.; Suzuki, R.; Sato, K.; Ogami, T.; Tomozawa, H.; Tsubata, M.; Ichinose, K.; Aburada, M.; Ochiai, W.; Sugiyama, K. Effect of Kaempferia parviflora extract on knee osteoarthritis. J. Nat. Med. 2018, 72, 136–144. [Google Scholar] [CrossRef]
  42. Umar, M.I.; Asmawi, M.Z.; Sadikun, A.; Majid, A.M.S.A.; Al-Suede, F.S.R.; Hassan, L.E.A.; Altaf, R.; Ahamed, M.B.K. Ethyl-p-methoxycinnamate isolated from Kaempferia galanga inhibits inflammation by suppressing interleukin-1, tumor necrosis factor-α, and angiogenesis by blocking endothelial functions. Clinics 2014, 69, 134–144. [Google Scholar] [CrossRef]
  43. Tewtrakul, S.; Subhadhirasakul, S.; Karalai, C.; Ponglimanont, C.; Cheenpracha, S. Anti-inflammatory effects of compounds from Kaempferia parviflora and Boesenbergia pandurata. Food Chem. 2009, 115, 534–538. [Google Scholar] [CrossRef]
  44. Tewtrakul, S.; Subhadhirasakul, S. Effects of compounds from Kaempferia parviflora on nitric oxide, prostaglandin E2 and tumor necrosis factor-alpha productions in RAW264. 7 macrophage cells. J. Ethnopharmacol. 2008, 120, 81–84. [Google Scholar] [CrossRef] [PubMed]
  45. Jagadish, P.C.; Latha, K.P.; Mudgal, J.; Nampurath, G.K. Extraction, characterization and evaluation of Kaempferia galanga L.(Zingiberaceae) rhizome extracts against acute and chronic inflammation in rats. J. Ethnopharmacol. 2016, 194, 434–439. [Google Scholar] [CrossRef] [PubMed]
  46. Sawasdee, P.; Sabphon, C.; Sitthiwongwanit, D.; Kokpol, U. Anticholinesterase activity of 7-methoxyflavones isolated from Kaempferia parviflora. Phytother. Res. 2009, 23, 1792–1794. [Google Scholar] [CrossRef]
  47. Azuma, T.; Kayano, S.-i.; Matsumura, Y.; Konishi, Y.; Tanaka, Y.; Kikuzaki, H. Antimutagenic and α-glucosidase inhibitory effects of constituents from Kaempferia parviflora. Food Chem. 2011, 125, 471–475. [Google Scholar] [CrossRef]
  48. Ochiai, W.; Kobayashi, H.; Kitaoka, S.; Kashiwada, M.; Koyama, Y.; Nakaishi, S.; Nagai, T.; Aburada, M.; Sugiyama, K. Effect of the active ingredient of Kaempferia parviflora, 5, 7-dimethoxyflavone, on the pharmacokinetics of midazolam. J. Nat. Med. 2018, 72, 607–614. [Google Scholar] [CrossRef]
  49. Yorsin, S.; Kanokwiroon, K.; Radenahmad, N.; Jansakul, C. Effects of Kaempferia parviflora rhizomes dichloromethane extract on vascular functions in middle-aged male rat. J. Ethnopharmacol. 2014, 156, 162–174. [Google Scholar] [CrossRef]
  50. Othman, R.; Ibrahim, H.; Mohd, M.A.; Mustafa, M.R.; Awang, K. Bioassay-guided isolation of a vasorelaxant active compound from Kaempferia galanga L. Phytomedicine 2006, 13, 61–66. [Google Scholar] [CrossRef]
  51. Wattanapitayakul, S.K.; Chularojmontri, L.; Herunsalee, A.; Charuchongkolwongse, S.; Chansuvanich, N. Vasorelaxation and antispasmodic effects of Kaempferia parviflora ethanolic extract in isolated rat organ studies. Fitoterapia 2008, 79, 214–216. [Google Scholar] [CrossRef]
  52. Pripdeevech, P.; Pitija, K.; Rujjanawate, C.; Pojanagaroon, S.; Kittakoop, P.; Wongpornchai, S. Adaptogenic-active components from Kaempferia parviflora rhizomes. Food Chem. 2012, 132, 1150–1155. [Google Scholar] [CrossRef]
  53. Matsushita, M.; Yoneshiro, T.; Aita, S.; Kamiya, T.; Kusaba, N.; Yamaguchi, K.; Takagaki, K.; Kameya, T.; Sugie, H.; Saito, M. Kaempferia parviflora extract increases whole-body energy expenditure in humans: Roles of brown adipose tissue. J. Nutr. Sci. Vitaminol. 2015, 61, 79–83. [Google Scholar] [CrossRef] [PubMed]
  54. Wattanathorn, J.; Muchimapura, S.; Tong-Un, T.; Saenghong, N.; Thukhum-Mee, W.; Sripanidkulchai, B. Positive modulation effect of 8-week consumption of Kaempferia parviflora on health-related physical fitness and oxidative status in healthy elderly volunteers. Evid. Based Complementary Altern. Med. 2012, 2012, 732816. [Google Scholar] [CrossRef] [PubMed]
  55. Kobayashi, S.; Kato, T.; Azuma, T.; Kikuzaki, H.; Abe, K. Anti-allergenic activity of polymethoxyflavones from Kaempferia parviflora. J. Funct. Foods 2015, 13, 100–107. [Google Scholar] [CrossRef]
  56. Plaingam, W.; Sangsuthum, S.; Angkhasirisap, W.; Tencomnao, T. Kaempferia parviflora rhizome extract and Myristica fragrans volatile oil increase the levels of monoamine neurotransmitters and impact the proteomic profiles in the rat hippocampus: Mechanistic insights into their neuroprotective effects. J. Tradit. Complementary Med. 2017, 7, 538–552. [Google Scholar] [CrossRef] [PubMed]
  57. Ali, M.S.; Dash, P.R.; Nasrin, M. Study of sedative activity of different extracts of Kaempferia galanga in Swiss albino mice. BMC Complementary Altern. Med. 2015, 15, 158. [Google Scholar] [CrossRef] [PubMed]
  58. Ridtitid, W.; Sae-Wong, C.; Reanmongkol, W.; Wongnawa, M. Antinociceptive activity of the methanolic extract of Kaempferia galanga Linn. in experimental animals. J. Ethnopharmacol. 2008, 118, 225–230. [Google Scholar] [CrossRef] [PubMed]
  59. Shanbhag, T.V.; Sharma, C.; Adiga, S.; Bairy, K.; Shenoy, S.; Shenoy, G. Wound healing activity of alcoholic extract of Kaempferia galanga in Wistar rats. Indian J. Physiol. Pharmacol. 2006, 50, 384–390. [Google Scholar]
  60. Temkitthawon, P.; Hinds, T.R.; Beavo, J.A.; Viyoch, J.; Suwanborirux, K.; Pongamornkul, W.; Sawasdee, P.; Ingkaninan, K. Kaempferia parviflora, a plant used in traditional medicine to enhance sexual performance contains large amounts of low affinity PDE5 inhibitors. J. Ethnopharmacol. 2011, 137, 1437–1441. [Google Scholar] [CrossRef]
  61. Stein, R.A.; Schmid, K.; Bolivar, J.; Swick, A.G.; Joyal, S.V.; Hirsh, S.P. Kaempferia parviflora ethanol extract improves self-assessed sexual health in men: A pilot study. J. Integr. Med. 2018, 16, 249–254. [Google Scholar] [CrossRef]
  62. Horigome, S.; Maeda, M.; Ho, H.-J.; Shirakawa, H.; Komai, M. Effect of Kaempferia parviflora extract and its polymethoxyflavonoid components on testosterone production in mouse testis-derived tumour cells. J. Funct. Foods 2016, 26, 529–538. [Google Scholar] [CrossRef]
  63. Lert-Amornpat, T.; Maketon, C.; Fungfuang, W. Effect of Kaempferia parviflora on sexual performance in streptozotocin-induced diabetic male rats. Andrologia 2017, 49, e12770. [Google Scholar] [CrossRef]
  64. Lee, S.; Kim, C.; Kwon, D.; Kim, M.-B.; Hwang, J.-K. Standardized Kaempferia parviflora Wall. ex Baker (Zingiberaceae) extract inhibits fat accumulation and muscle atrophy in ob/ob mice. Evid. Based Complementary Altern. Med. 2018, 2018, 8161042. [Google Scholar] [CrossRef]
  65. Ono, S.; Yoshida, N.; Maekawa, D.; Kitakaze, T.; Kobayashi, Y.; Kitano, T.; Fujita, T.; Okuwa-Hayashi, H.; Harada, N.; Nakano, Y. 5-Hydroxy-7-methoxyflavone derivatives from Kaempferia parviflora induce skeletal muscle hypertrophy. Food Sci. Nutr. 2019, 7, 312–321. [Google Scholar] [CrossRef]
  66. Jin, S.; Lee, M.-Y. Kaempferia parviflora extract as a potential anti-acne agent with anti-inflammatory, sebostatic and anti-propionibacterium acnes activity. Int. J. Mol. Sci. 2018, 19, 3457. [Google Scholar] [CrossRef]
  67. Kongdang, P.; Jaitham, R.; Thonghoi, S.; Kuensaen, C.; Pradit, W.; Ongchai, S. Ethanolic extract of Kaempferia parviflora interrupts the mechanisms-associated rheumatoid arthritis in SW982 culture model via p38/STAT1 and STAT3 pathways. Phytomedicine 2019, 59, 152755. [Google Scholar] [CrossRef]
  68. Dash, P.R.; Mou, K.M.; Erina, I.N.; Ripa, F.A.; Al Masud, K.N.; Ali, M.S. Study of anthelmintic and insecticidal activities of different extracts of Kaempferia galanga. Int. J. Pharm. Sci. Res. 2017, 8, 729–733. [Google Scholar]
  69. Jaipetch, T.; Reutrakul, V.; Tuntiwachwuttikul, P.; Santisuk, T. Flavonoids in the black rhizomes of Boesenbergia panduta. Phytochemistry 1983, 22, 625–626. [Google Scholar] [CrossRef]
  70. Herunsalee, A.; Pancharoen, O.; Tuntiwachwuttikul, P. Further studies of flavonoids of the black rhizome Boesenbergia pandurata. J. Sci. Soc. Thail. 1987, 13, 119–122. [Google Scholar]
  71. Trakoontivakorn, G.; Nakahara, K.; Shinmoto, H.; Takenaka, M.; Onishi-Kameyama, M.; Ono, H.; Yoshida, M.; Nagata, T.; Tsushida, T. Structural analysis of a novel antimutagenic compound, 4-hydroxypanduratin A, and the antimutagenic activity of flavonoids in a Thai spice, fingerroot (Boesenbergia pandurata Schult.) against mutagenic heterocyclic amines. J. Agric. Food Chem. 2001, 49, 3046–3050. [Google Scholar] [CrossRef]
  72. Raina, A.P.; Abraham, Z. Chemical profiling of essential oil of Kaempferia galanga L. germplasm from India. J. Essent. Oil Res. 2016, 28, 29–34. [Google Scholar] [CrossRef]
  73. Wong, K.; Ong, K.; Lim, C. Compositon of the essential oil of rhizomes of kaempferia galanga L. Flavour Fragr. J. 1992, 7, 263–266. [Google Scholar] [CrossRef]
  74. Win, N.N.; Ito, T.; Aimaiti, S.; Imagawa, H.; Ngwe, H.; Abe, I.; Morita, H. Kaempulchraols A–H, diterpenoids from the rhizomes of Kaempferia pulchra collected in Myanmar. J. Nat. Prod. 2015, 78, 1113–1118. [Google Scholar] [CrossRef]
  75. Thongnest, S.; Mahidol, C.; Sutthivaiyakit, S.; Ruchirawat, S. Oxygenated pimarane diterpenes from Kaempferia marginata. J. Nat. Prod. 2005, 68, 1632–1636. [Google Scholar] [CrossRef]
  76. Prawat, U.; Tuntiwachwuttikul, P.; Taylor, W.C.; Engelhardt, L.M.; Skelton, B.; White, A. Diterpenes from a Kaempferia species. Phytochemistry 1993, 32, 991–997. [Google Scholar] [CrossRef]
  77. Sutthanut, K.; Sripanidkulchai, B.; Yenjai, C.; Jay, M. Simultaneous identification and quantitation of 11 flavonoid constituents in Kaempferia parviflora by gas chromatography. J. Chromatogr. A 2007, 1143, 227–233. [Google Scholar] [CrossRef]
  78. Umar, M.I.; Asmawi, M.Z.B.; Sadikun, A.; Altaf, R.; Iqbal, M.A. Phytochemistry and medicinal properties of Kaempferia galanga L. (Zingiberaceae) extracts. Afr. J. Pharm. Pharmacol. 2011, 5, 1638–1647. [Google Scholar] [CrossRef]
  79. Azuma, T.; Tanaka, Y.; Kikuzaki, H. Phenolic glycosides from Kaempferia parviflora. Phytochemistry 2008, 69, 2743–2748. [Google Scholar] [CrossRef]
  80. Davis, E.M.; Croteau, R. Cyclization enzymes in the biosynthesis of monoterpenes, sesquiterpenes, and diterpenes. Top. Curr. Chem. 2000, 209, 53–95. [Google Scholar]
  81. Xu, M.; Hillwig, M.L.; Tiernan, M.S.; Peters, R.J. Probing labdane-related diterpenoid biosynthesis in the fungal genus Aspergillus. J. Nat. Prod. 2017, 80, 328–333. [Google Scholar] [CrossRef]
  82. Li, Y.C.; Ji, H.; Li, X.H.; Zhang, H.X.; Li, H.T. Isolation of nematicidal constituents from essential oil of Kaempferia galanga L rhizome and their activity against Heterodera avenae Wollenweber. Trop. J. Pharm. Res. 2017, 16, 59–65. [Google Scholar] [CrossRef]
  83. Munda, S.; Saikia, P.; Lal, M. Chemical composition and biological activity of essential oil of Kaempferia galanga: A review. J. Essent. Oil Res. 2018, 30, 303–308. [Google Scholar] [CrossRef]
  84. Labrooy, C.D.; Abdullah, T.L.; Stanslas, J. Identification of ethnomedicinally important Kaempferia L. (Zingiberaceae) species based on morphological traits and suitable DNA region. Curr. Plant Biol. 2018, 14, 50–55. [Google Scholar] [CrossRef]
Figure 1. Traditional medicinal used Kaempheria species.
Figure 1. Traditional medicinal used Kaempheria species.
Nutrients 11 02396 g001
Figure 2. Reported biological activities for Kaempheria species.
Figure 2. Reported biological activities for Kaempheria species.
Nutrients 11 02396 g002
Scheme 1. Plausible isopimaradiene biosynthesis [81] starting with (E,E,E)-geranylgeranyl diphosphate (GGPP): (E,E,E)-geranylgeranyl diphosphate; CPS: copalyl diphosphate (CPP) synthases; PS: Unknown enzyme; AoCPS-PS, NfCPS-PS, and AfCPS-PS: bifunctional (iso) pimaradiene synthases.
Scheme 1. Plausible isopimaradiene biosynthesis [81] starting with (E,E,E)-geranylgeranyl diphosphate (GGPP): (E,E,E)-geranylgeranyl diphosphate; CPS: copalyl diphosphate (CPP) synthases; PS: Unknown enzyme; AoCPS-PS, NfCPS-PS, and AfCPS-PS: bifunctional (iso) pimaradiene synthases.
Nutrients 11 02396 sch001
Figure 3. (A) Agglomerative hierarchical clustering (AHC) and (B) Principal component analysis (PCA) based on the chemical composition of different chemical classes of seven Kaempferia species (K. angustifolia, K. elegans, K. galanga, K. marginata, K. parviflora, K. pulchra, and K. roscoeana).
Figure 3. (A) Agglomerative hierarchical clustering (AHC) and (B) Principal component analysis (PCA) based on the chemical composition of different chemical classes of seven Kaempferia species (K. angustifolia, K. elegans, K. galanga, K. marginata, K. parviflora, K. pulchra, and K. roscoeana).
Nutrients 11 02396 g003
Table 1. Diterpenoids.
Table 1. Diterpenoids.
Nutrients 11 02396 i001
NoNameRR1R2R3R4R5R6R7PlantRef
1SandaracopimaradieneHHHHHHHHK. galanga
K. roscoeana
K. marginata
[4,16,17,25,26,36,74,75,76]
2Sandaracopimaradiene-9α-olα-OHHHHHHHH
38(14),15-Sandaracopimaradiene-1α,9α-diolα-OHHα-OHHHHHHK. galanga
K. pulchra
K. sp.
41,11-Dihydroxypimara-8(14),15-dieneHHα-OHHHα-OHHH
56β-Hydroxypimara-8(14),15-diene-1-oneHβ-OH=OHHHHHK. galanga
K. marginata
6Sandaracopimaradien-6β,9α-diol-l-oneα-OHβ-OH=OHHHHHK. galanga
7Boesenberol Iα-OHH=OHHHα-OHH
8Boesenberol Jα-OHβ-OH=OHHHHHK. galanga
9Sandaracopimaradien-1α,2α-diolHHα-OHα-OHHHHHK. roscoeana
K. pulchra
K. marginata
[26,38,75]
102α-Acetoxy-sandaracopimaradien-1α-olHHα-OHα-OAcHHHHK. pulchra
K. marginata
11Kaempulchraol Eα-Hβ-OHα-OHHHHHHK. galanga
K. pulchra
12Kaempulchraol FHHα-OHHα-OHHHHK. pulchra[4,16,17,25,26,74]
13Kaempulchraol HHβ-OHα-OHHα-OHHHH
14Kaempulchraol IHHα-OHHHHHHK. galanga
K. pulchra
K. roscoeana
15Kaempulchraol JHHα-OHHHH=O K. pulchra
16Kaempulchraol Kα-OHβ-OAcHHHHHH
17Kaempulchraol Lα-OMeβ-OHHHHHHHK. galanga
K. pulchra
18Kaempulchraol Mα-OHHα-OHα-OHHHHHK. pulchra
19Kaempulchraol PHβ-OHHHHHHH
20Kaempulchraol Qα-OAcβ-OHHHHHHH
21Kaempulchraol Rα-OHHHHHHα-OAcH
22Kaempulchraol THβ-OHHHHHα-OAcH
23Kaempulchraol Vα-OHβ-OHHHHHβ-OAcH
24Kaempulchraol Wα-OHβ-OHHHHHβ-OHH
259 α-Hydroxyisopimara-8(14),15-dien-7-oneα-OHHHHHH=OH
267β,9 α-Dihydroxypimara-8(14),15-dieneα-OHHHHHHβ-OHH
27Isopimara-8(14),15-dien-7-oneHHHHHH=OHK. roscoeana[26]
28(1S,5S,9S,10S,11R,13R)-1,11-Dihydroxypimara-8(14),15-dieneHHα-OHHHα-OHHHK. roscoeana
K. marginata
K. pulchra
[4,17,25,26,74,75]
29(1R,2S,5S,9S,10S,11R,13R)-1,2,11-Trihydroxypimara-8(14),15-dieneHHα-OHα-OHHα-OHHH
307α-Hydroxyisopimara-8(14),15-dieneHHHHHHα-OHHK. roscoeana
K. pulchra
31Sandaracopimaradien- 9α-ol-l-oneα-OHH=OHHHHHK. sp[76]
326β-Acetoxysandaracopimaradien-9α-ol-l-oneα-OHβ-OAc=OHHHHH
33Sandaracopimaradien-6β,9α-diol-l-oneα-OHβ-OH=OHHHHH
346β-Acetoxysandaracopimaradien-lα,9α-diolα-OHβ-OAcα-OHHHHHH
35Sandaracopimaradien- lα,6β,9α-triolα-OHβ-OHα-OHHHHHH
36Roscorane BHHHHHα-OHHOHK. roscoeana[26]
37Roscorane CHβ-OHHOHHHOHH
38Roscorane DHHHOHHHOHOH
39(1R,2S,5S,7S,9R,10S,13R)-1,2,7-Trihydroxypimara-8(14),15-dieneHHHα-OHHHβ-OHHK. marginata[75]
40(1S,5S,7R,9R,10S,11R,13R)-1,7,11-Trihydroxypimara-8(14),15-dieneHβ-OHHHHHα-OHH
41(1R,2S,5S,7S,9R,10S,13R)-1,2-Dihydroxypimara-8(14),15diene-7-oneHHHα-OHHHHH
Nutrients 11 02396 i002
52–54
Nutrients 11 02396 i003
42–51
NoNameR1R2R3R4PlantRef
42Kaempulchraol AHβ-OHHα-OMeK. pulchra[4,17,25,74]
43Kaempulchraol BHβ-OHHβ-OMe
44Kaempulchraol CHβ-OHHα-OH
45Kaempulchraol DHβ-OHHβ-OH
46Kaempulchraol GHβ-OHH=O
47Kaempulchraol Nα-OHβ-OHHα-OH
48Kaempulchraol Oα-OHβ-OHHβ-OMe
49Kaempulchraol SHH=Oα-OH
50Kaempulchraol UHHHα-OH
51Isopimara-8(9),15-dien-7-oneHH=OHK. roscoeana[26]
528(14),15-Isopimaradiene-6α-olHα-OHH---K. marginata[36]
531α-Acetoxy-sandaracopimaradieneα-OAcHH-
541α-Acetoxy-sandaraco pimaradien-2-oneα-OAc=OH-
NoNameStructurePlantRef
55(2R)-ent-2-Hydroxyisopimara-8(14),15-diene Nutrients 11 02396 i004K. pulchra[4,17,25,74]
56Kaemgalangol A Nutrients 11 02396 i005K. galanga[16]
57Roscorane A Nutrients 11 02396 i006K. roscoeana[26]
58R=OMe; Roscotane A Nutrients 11 02396 i007K. roscoeana[26]
59R=H; Roscotane B
60Roscotane C Nutrients 11 02396 i008
61Roscotane D Nutrients 11 02396 i009
62R=H; Ar-abietatriene Nutrients 11 02396 i010
63R=[=O]; 7-Dehydroabietanone
64R=α-OH; Abieta-8,11,13-trien-7α-ol
65Kaempfolienol Nutrients 11 02396 i011K. angustifolia[33,34]
66(12Z,14R)-Labda-8(17),12-dien-14,15,16-triol Nutrients 11 02396 i012K. roscoeana[26]
67Propadane A Nutrients 11 02396 i013K. elegans[18]
68R=H --- 8(17)-Labden-15-ol Nutrients 11 02396 i014
69R=OH; Propadane B
70Propadane C Nutrients 11 02396 i015K. pulchra
71Cleroda-2,4(18),14-trien-13-ol Nutrients 11 02396 i016K. pulchra
72R=H; Anticopalic acid Nutrients 11 02396 i017K. elegans
73R=Me; Methyl anticopalate
74(+)-15,16-Eoxy-8(17),13(16),14-labdatriene Nutrients 11 02396 i018[18]
75(+)-Pumiloxide Nutrients 11 02396 i019
7613-Oxo-14,15-bis-nor-labd-8(17)-ene Nutrients 11 02396 i020
77Anticopalol Nutrients 11 02396 i021K. elegans
78Labda-8(17),13(14)-diene-15,16-olide Nutrients 11 02396 i022
79(+)-Labda-8(17),13(Z)-diene-15,16-diol Nutrients 11 02396 i023
80Calcaratarin A Nutrients 11 02396 i024K. pulchra
81R=H; (-)-Kolavelool Nutrients 11 02396 i025[18]
82R= β-OH; (-)-2β-Hydroxykolavelool
83R=β-OMe; Dysoxydensin E
8413-Epi-roseostachenone Nutrients 11 02396 i026
85(+)-13-Epi-2α-hydroxykolavelool (13-epi-roseostachenol) Nutrients 11 02396 i027K. pulchra
Table 2. Flavonoids and phenolics (Flavonoids).
Table 2. Flavonoids and phenolics (Flavonoids).
Nutrients 11 02396 i028
86–97
Nutrients 11 02396 i029
98–101
Nutrients 11 02396 i030
102–103
NoNameR1R2R3R4PlantRef
865,7-DimethoxyflavoneHMeHHK. parviflora [9,55,71,77]
874‘,5,7-TrimethoxyflavoneHMeHOMe
883‘,4‘,5,7-TetramethoxyflavoneHMeOMeOMe
893,5,7-TrimethoxyflavoneOMeMeHH
903,5,7,4‘-TetramethoxyflavoneOMeMeHOMe
913,5,7,3‘,4‘-PentamethoxyflavoneOMeMeOMeOMe
925-Hydroxy-7-methoxyflavoneHHHH
935-hydroxy-7,4‘-dimethoxyflavoneHHHOMe
945-Hydroxy-3,7-dimethoxyflavoneOMeHHH
955-Hydroxy-3,7,4‘-trimethoxyflavoneOMeHHOMe
965-Hydroxy-3,7,3‘,4‘-tetramethoxy flavoneOMeHOMeOMe
975,3‘-Dihydroxy-3,7,4‘-trimethoxyflavoneOMeHOHOMe
98KaempferolHOH--K. galanga[42]
99KaempferideHOMe-
100TectochrysinMeH--K. parviflora
101GenkwaninMeOH--[46]
102PinocembinH---K. parviflora
K. angustifolia
[71]
103PinostrobinMe---
104Sakuranetin Nutrients 11 02396 i031
1052″,2″-Dimethylpyrano-[5″,6″:8,7]-flavone Nutrients 11 02396 i032K. pulchra[18]
Nutrients 11 02396 i033
106–109
Nutrients 11 02396 i034
110–113
Nutrients 11 02396 i035
114–115
Nutrients 11 02396 i036
116–117
Nutrients 11 02396 i037
118–119
NoNameR1R2R3PlantRef
106Ethyl trans-p-methoxycinnamateHOMeCH2MeK. galanga[13]
107Ferulic acidOMeOHH
108trans-p-Hydroxy-cinnamic acidHOHH
109trans-p-Methoxy cinnamic acidHHCH2Me
110p-Hydroxybenzoic acidHOHCOOH[13,50]
111p-Methoxybenzoic acidHOMeCOOH
112Vanillic acidOMeOHCOOH
113Methyl 3,4-dihydroxybenzoateOHOHCOOMe
114Methyl (2R,3S)-2,3-dihydroxy-3-(4-methoxyphenyl) propanoateMe--
115Ethyl-(2R,3S)-2,3-dihydroxy-3-(4-methoxyphenyl) propanoateCH2Me--
116(1R,3R,5R)-1,5-Epoxy-3-hydroxy-1-(3,4-dihydroxyphenyl)-7-(3,4-dihydroxy phenyl) heptaneH--[13]
117(1R,3R,5R)-1,5-Epoxy-3-hydroxy-1-(3,4-dihydroxyphenyl)-7-(4-hydroxyphenyl) heptane 3-O-β-D-glucopyranosideD-glc--
1182‘-hydroxy-4‘,6‘-dimethoxychalconeH--K. parviflora
K. angustifolia
[24,71]
1192‘-hydroxy-4,4‘,6‘-trimethoxychalconeMe--
NoNameStructurePlantRef
120Desmethoxyyangonin Nutrients 11 02396 i038K. marginata[36]
121Bisdemethoxycurcumin Nutrients 11 02396 i039
1221-(4-Hydroxy-3-methoxyphenyl)-7-(4-hydroxyphenyl)heptane-1,2,3,5,6-pentaol Nutrients 11 02396 i040K. galanga[13]
123(3R,5S)-3,5-Dihydroxy-1-(3,4-dihydroxyphenyl)-7-(4-hydroxy phenyl) heptane Nutrients 11 02396 i041K. galanga
124Phaeoheptanoxide Nutrients 11 02396 i042
125Hedycoropyran B Nutrients 11 02396 i043
1261-O-4-Carbonxylphenyl-(6-O-4-hydroxybenzoyl)-β-D-glucopyranoside Nutrients 11 02396 i044
127Dihydro-5,6-dehydrokawain Nutrients 11 02396 i045K. parviflora[13,50]
128R=OH; (-)-Hydroxypanduratin A Nutrients 11 02396 i046
129,R=OMe; (-)-Panduratin A
130Cinnamaldehyde Nutrients 11 02396 i047K. galanga[78]
131R=Me; Crotepoxide Nutrients 11 02396 i048K. angustifolia[24,33]
132R=Benzen; Boesenboxide
133 134 R=H; Zeylenol
R=Me; 6-methylzeylenol
Nutrients 11 02396 i049
135rel-(5aS,10bS)-5a,10b-Dihydro-1,3,5a,9-tetrahydroxy-8-methoxy-6H-benz[b]indeno[1–d]furan-6-one 5a-O-[α-l-rhamnopyranosyl-(1→6)-β-d-glucopyranoside] Nutrients 11 02396 i050
R=α-L-rha-(1→6)-β-D-glc
K. parviflora[79]
136rel-(5aS,10bR)-5a,10b-Dihydro-1,3,5a,9-tetrahydroxy-8-methoxy-6H-benz[b]indeno[1–d]furan-6-one 5a-O-[α-l-rhamnopyranosyl-(1→6)-β-d-glucopyranoside] Nutrients 11 02396 i051
R=α-L-rha-(1→6)-β-D-glc
137(2R,3S,4S)-3-O-[α-l-Rhamnopyranosyl-(1 → 6)-β-d-glucopyranosyl]-3′-O-methyl-ent-epicatechin-(2αO→3,4α→ 4)-(5aS,10bS)-5a,10b-dihydro-1,3,5a,9-tetrahydroxy-8-methoxy-6H-benz[b]indeno[1,2-d]furan-6-one 5a-O-[α-l-rhamnopyranosyl-(1→ 6)-β-d-glucopyranoside] Nutrients 11 02396 i052
R=α-L-rha-(1→6)-β-D-glc
Table 3. Steroid and triterpenes.
Table 3. Steroid and triterpenes.
Nutrients 11 02396 i053
138–139
NoNameStructurePlantRef
138β-SitosterolR=HK.marginata
K. angustifolia
[24,33,36]
139β-Sitosterol-β-D-glucosideR=β-D-glc
140Stigmasterol Nutrients 11 02396 i054
141(24S)-24-methyl-lanosta-9(11), 25-dien-3β-ol Nutrients 11 02396 i055K. angustifolia[24]
Table 4. Main components of volatile oils of Kaempferia species.
Table 4. Main components of volatile oils of Kaempferia species.
NoNamePlantRef
142δ-3-CareneK. galanga[29,35,82,83]
143E-Ethyl cinnamate
144Ethyl-p-methoxycinnamate
145γ-Cadinene
1461,8-Cineole
147Trans-cinnamaldehyde
148Borneol
149Pentadecane
150γ-car-3-ene
151Linoleoyl chloride
152Caryophyllene oxide
153Cubenol
154Caryophyllene
155Limonene
156Camphene
157α-Pinene
Table 5. Proximity matrix (Pearson correlation coefficient) of the seven Kaempferia species based on the chemical classes reported.
Table 5. Proximity matrix (Pearson correlation coefficient) of the seven Kaempferia species based on the chemical classes reported.
K. angustifoliaK. elegansK. galangaK. marginataK. parvifloraK. pulchra
K. elegans−0.539
K. galanga−0.042−0.339
K. marginata−0.500−0.2410.615
K. parviflora0.833−0.3120.075−0.280
K. pulchra−0.6750.0530.3780.938−0.372
K. roscoeana−0.643−0.2250.2060.771−0.5130.766

Share and Cite

MDPI and ACS Style

Elshamy, A.I.; Mohamed, T.A.; Essa, A.F.; Abd-El Gawad, A.M.; Alqahtani, A.S.; Shahat, A.A.; Yoneyama, T.; Farrag, A.R.H.; Noji, M.; El-Seedi, H.R.; et al. Recent Advances in Kaempferia Phytochemistry and Biological Activity: A Comprehensive Review. Nutrients 2019, 11, 2396. https://doi.org/10.3390/nu11102396

AMA Style

Elshamy AI, Mohamed TA, Essa AF, Abd-El Gawad AM, Alqahtani AS, Shahat AA, Yoneyama T, Farrag ARH, Noji M, El-Seedi HR, et al. Recent Advances in Kaempferia Phytochemistry and Biological Activity: A Comprehensive Review. Nutrients. 2019; 11(10):2396. https://doi.org/10.3390/nu11102396

Chicago/Turabian Style

Elshamy, Abdelsamed I., Tarik A. Mohamed, Ahmed F. Essa, Ahmed M. Abd-El Gawad, Ali S. Alqahtani, Abdelaaty A. Shahat, Tatsuro Yoneyama, Abdel Razik H. Farrag, Masaaki Noji, Hesham R. El-Seedi, and et al. 2019. "Recent Advances in Kaempferia Phytochemistry and Biological Activity: A Comprehensive Review" Nutrients 11, no. 10: 2396. https://doi.org/10.3390/nu11102396

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop