Next Article in Journal
Electronic Structure Regulated Nickel-Cobalt Bimetal Phosphide Nanoneedles for Efficient Overall Water Splitting
Next Article in Special Issue
Antioxidant and Cytotoxic Properties of Berberis vulgaris (L.) Stem Bark Dry Extract
Previous Article in Journal
Research on the Interaction Mechanism and Structural Changes in Human Serum Albumin with Hispidin Using Spectroscopy and Molecular Docking
Previous Article in Special Issue
Antitumor Mechanism and Therapeutic Potential of Cordycepin Derivatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Extraction, Determination, and Bioactivity of Curcumenol: A Comprehensive Review

by
Jie Li
1,2,3,
Yitian Sun
1,2,
Guohua Li
1,2,
Chunsong Cheng
4,
Xinbing Sui
2 and
Qibiao Wu
1,5,6,*
1
State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
2
College of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
3
School of Environmental and Chemical Engineering, Zhaoqing University, Zhaoqing 526061, China
4
Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China
5
Zhuhai M.U.S.T. Science and Technology Research Institute, Zhuhai 519031, China
6
Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangzhou 510006, China
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(3), 656; https://doi.org/10.3390/molecules29030656
Submission received: 27 December 2023 / Revised: 18 January 2024 / Accepted: 26 January 2024 / Published: 30 January 2024
(This article belongs to the Special Issue Advances in Plant-Sourced Natural Compounds as Anticancer Agents)

Abstract

:
Curcuma wenyujin is a member of the Curcuma zedoaria (zedoary, Zingiberaceae) family, which has a long history in traditional Chinese medicine (TCM) due to its abundant biologically active constituents. Curcumenol, a component of Curcuma wenyujin, has several biological activities. At present, despite different pharmacological activities being reported, the clinical usage of curcumenol remains under investigation. To further determine the characteristics of curcumenol, the extraction, determination, and bioactivity of the compound are summarized in this review. Existing research has reported that curcumenol exerts different pharmacological effects in regard to a variety of diseases, including anti-inflammatory, anti-oxidant, anti-bactericidal, anti-diabetic, and anti-cancer activity, and also ameliorates osteoporosis. This review of curcumenol provides a theoretical basis for further research and clinical applications.

Graphical Abstract

1. Introduction

Curcuma wenyujin Y. H. Chen et C. Ling (Curcuma wenyujin) is a member of the Curcuma zedoaria (zedoary, Zingiberaceae) family, which is primarily distributed in Zhejiang Province, China [1]. It has a long history of extensive utilization in TCM. It is one of the members of the well-known TCM group named “Zhebawei”, which includes eight authentic Chinese herbal medicines from Zhejiang Province (Atractylodes, paeony, Zhejiang fritillaria, Chrysanthemum morifolium, Corydalis glaucescens, Scrophularia ningpoensis Hemsl., Ophiopogon japonicus (Thunb.) Ker-Gawl, and Wen Yujin) [2] (Figure 1). Curcuma wenyujin can be incorporated into three different Chinese medicines, “Acruginous Turmeric Rhizome”, “Wenyujin”, and “Wenyujin Rhizoma Concisum”, and exhibits anti-inflammatory [3], anti-tumor [4], anti-oxidant [5], anti-bacterial, anti-viral, and hepatoprotective properties. The pharmacological value of Curcuma wenyujin is derived from its constituents, which have been reported to comprise a total of 169 compounds, thus far. These include monoterpenoids, sesquiternoids, diterpenoids, curcuminoids, etc. [6]. Curcumin, an active ingredient of Curcuma wenyujin, has been widely investigated and applied [7]. Curcumenol is one of the other sesquiterpenoid active ingredients (following the Chinese Pharmacopoeia), which has been extracted from the edible rhizome of Curcuma zedoaria since the 1960s [8]; it has been reported as one of the primary constituents in Curcuma plant essential oils, as well as an important component of food and traditional remedies [9,10,11,12]. A growing number of studies have suggested that it exhibits useful biological effects, including anti-cancer [13,14,15,16], anti-inflammatory [17,18,19], anti-bactericidal [20,21], and liver protection activities [22], among others. Curcumina longa, another important member of the Curcuma zedoaria (zedoary, Zingiberaceae) family, has also attracted research interest. The differences in the components and physiological effects of the two herbs are shown in Table 1.
Despite the huge pharmacological potential of curcumenol, its further application has been hindered by unsatisfactory physical properties (e.g., low water solubility). In this research, relevant studies and reports were reviewed to summarize the methods related to the extraction, determination, and pharmacology of curcumenol, with the aim of promoting the investigation and utilization of curcumenol in human healthcare.
Table 1. Differences between the components and physiological effects of Curcuma wenyujin and Curcumina longa.
Table 1. Differences between the components and physiological effects of Curcuma wenyujin and Curcumina longa.
Curcuma wenyujinCurcumina longa
The Content of Unique Chemical Markers to Distinguish the Two Herbs [23]
Curcumenol> Curcumenol
Curcumenone> Curcumenone
Neocurdione> Neocurdione
Curdione> Curdione
Curcumin< Curcumin
Physiological effects
Anti-inflammatory [24]Anti-inflammatory [25]
Anti-oxidant [26]Anti-oxidant [27]
Antimicrobial [28] and antiviral activity [29]Antimicrobial [30] and antiviral activity [31]
Anti-diabetic [32]Anti-diabetic [33]
Hepatoprotective [34]Hepatoprotective [35]
Effects on the cardiovascular system [36]Effects on the cardiovascular system [37]
Analgesic effect [38]Analgesic effect [39]
Effect on the nervous system: Alzheimer’s disease [40]Effect on the nervous system: improve memory impairment [41]
Anti-cancer: glioma [42], breast cancer [43], liver cancer [44], gastric cancer [45], colon cancer [46], leukemia [47], cervical cancer [48], lung cancer [49]Anti-cancer: liver cancer [50], colon cancer [51], cervical cancer [52], human thyroid cancer [53], human nasopharyngeal carcinoma cells [54], lung cancer [55]
Effects on the respiratory system [56]
Effect on the digestive system [57]
Anticoagulant effect [58]

2. Traditional Medicinal Uses of Curcumenol-Related Herbs

Curcuma is an important herb in Chinese medicine, with a long history of use for about 1500 years. It has significance in healthcare due to its various medicinal effects. It has been reported that Curcuma can promote blood circulation, promote the flow of qi, unblock meridians, and relieve pain [1]. In-depth investigations have also been conducted to elucidate which exact compounds exert the abovementioned traditional curative effects, based on current medical theories. A series of compounds related to these treatment effects have been identified, including curcumenol. Curcumenol exhibits anti-inflammatory, anti-tumor, anti-virus, anti-oxidation, and hepatoprotective effects, as listed in Table 2. The above-described pharmacologica activity of curcumenol provides further evidence for the use of Curcuma in traditional medicines.
Table 2. Traditional uses of curcumenol-related herbal medicines.
Table 2. Traditional uses of curcumenol-related herbal medicines.
Scientific NameTraditional NamePlant Part UsedEffective Compounds and Ref.Traditional
Medicine Use
Monotherapy or
an Adjunct and Ref.
Curcuma zedoariaZedoary, ZingiberaceaeRhizomeCurcumenol [17]OsteoarthritisAn adjunct to allopathic medication [59]
Curcuma zedoariaWhite turmericRhizomeCurcumenol [18]Anti-inflammatoryAn adjunct to allopathic medication [60]
Curcuma zedoariaWhite turmericRhizomeCurcumenol and six other compounds [19]PsoriasisAn adjunct to allopathic medication [61]
CurcumaCurcuma phaeocaulis, Curcuma kwangsiensis, Curcuma wenyujinRhizomeCurcumenol, Curcumol, β-elemene, curdione [21]Anti-fungalAn adjunct to allopathic medication
[62]
Curcuma zedoariaZingiberaceaeRhizomeCurcumenol and some principal sesquiterpenes [22]HepatoprotectiveAn adjunct to allopathic medication [63]
Curcuma zedoariaZingiberaceaeRhizomeCurcumenol, dihydrocurdione [64,65]AnalgesicAn adjunct to allopathic medication [66]
CurcumaZingiberaceaeCurcuma aromatica Salisb. rhizomeCurzerene, isoprocurcumenol, and (+)-curcumenol [67]Coronary heart diseaseAn adjunct to allopathic medication [68]
Curcuma zedoariaZingiberaceaeRhizomeCurcumenol [69]OsteoporosisAn adjunct to allopathic medication [70]
Curcuma zedoary RhizomeCurcumenol [71]Liver cancerAn adjunct to allopathic medication [72]
Curcuma RhizomeCurcumenol [73]Cervical cancerAn adjunct to allopathic medication [74]
Curcuma wenyujinWenyujinRhizomeCurcumenol [13]Lung cancerAn adjunct to allopathic medication [75]
Curcuma zedoaria rhizomeZingiberaceaeC. zedoaria rhizomesCurcumenol, 4,8-dioxo-6β-methoxy-7α,11-epoxycarabrane, and zedoarofuran [15]Gastric cancerAn adjunct to allopathic medication [76]
Curcuma zedoariaTemu putihRhizomeCurcumenone,
Curcumenol [16]
Breast cancerAn adjunct to allopathic medication [77]

3. Sources of Curcumenol

Curcumenol was extracted from Curcuma zedoaria’s edible rhizome for the first time in the mid-20th century. In tropical forests, Zingerberaceae covers 52 genera, with 1500 species in total. Most of them are planted and reported in South East Asia [78]. Zingerberaceae, also known as “temu putih”, acts as a kind of additive; it is widely used in food preparation for nursing women after childbirth and as a spice in native dishes in Malaysia [79,80]. Curcumenol has also been identified in several other plants (e.g., the flower tea Chrysanthemum indicum [81], Neolitsea pallens [82], and Torilis japonica [83]). Zhu Shunying et al. [81] reported essential oils from samples of Chrysanthemum indicum, including monoterpene hydrocarbons (0.42–6.72%), sesquiterpenes (9.18–39.29%), and oxygenated monoterpenes (43.29–78.92%); curcumenol was derived from the oxygenated monoterpenes. G.C. Kharkwal et al. [83] investigated Torilis japonica (Houtt.) DC., which grows in the central Himalayan region. As revealed by their research, the oils are abundant in sesquiterpine hydrocarbons (approximately 60%). In total, 43 compounds with curcumenol accounted for 94.28% of the oils.
Rajendra C. Padalia et al. [82] investigated the composition of volatile constituents in the leaf, stem, and bark of N. pallens through nuclear magnetic resonance (NMR) spectroscopy, gas chromatography–mass spectrometry (GC–MS), and gas chromatography (GC) methods. Curcumenol (5.3%) was reported to occur in the oxygenated sesquiterpenoid, making up the major proportion of the bark oil. Curcumenol has also been extracted from bacterium and animal samples. Noor Akbar et al. [20] purified docosanedioic acid, Di-rhamnolipids, L-homotyrosine, N-acyl-homoserine lactone, and curcumenol from the gut bacteria of animals (e.g., cockroaches, water monitor lizards, and turtles).

4. Physicochemical Properties of Curcumenol

C15H22O2, 5β-Guaia-7 (11), 9-dien-8α-ol, 5, 8-epoxy-, 234.33, and 19431–84-6 are the molecular formula, chemical name, molecular weight, and chemical abstract service (CAS) number of curcumenol, respectively. Curcumenol has a white needle-like crystal structure at ambient temperatures, with a melting point of 113–115 °C, and it is insoluble in water, but soluble in methanol, ethanol, dimethyl sulfoxide, and some organic solvents. As indicated by the results achieved using spectroscopic and chemical methods, curcumenol exists in solution as a pair of hemiacetal–ketone tautomers [84]. More insights have been gained into the ultraviolet (UV) and electron-capture dissociation spectroscopic properties of curcumenol through theoretical time-dependent density functional theory calculations and modeling. Co-existing curcumenotone has a large tensional and unstable structure, contributing notably to the anti-oxidant process [84].

5. Extraction Methods

The extraction method for the components is of critical significance in the qualitative and quantitative process of herbal drug research [85]. Appropriate extraction methods should be developed to deal with different samples. Extraction methods for the different samples (e.g., biological samples, crude herb samples, and preparations) are detailed in Table 3.
A few investigations have reported on the extraction of curcumenol from biological samples. Salwa Mansur Ali et al. identified curcumenol and 11 other compounds from tissues and hemolymphs, which was extracted from the red-headed centipede, through the addition of protease inhibitors (serine/cysteine/metalloproteases) [86]. Subsequently, the samples were treated at 4 °C; then, ten cycles of freezing and thawing were performed on the gut and muscle tissues and, before analysis, the samples were subjected to sonication for further extraction [86].
Table 3. Sample preparation methods employed to extract curcumenol.
Table 3. Sample preparation methods employed to extract curcumenol.
MatrixPretreatment/Extraction
Approach
ProcedureRecovery (%)Ref.
Biological Samples
Crude hemolymph extractOrgan lysisThe exposed hemolymph was taken in an aseptic manner and suspended in a quantity of sterile distilled water. Ten cycles of freezing and thawing were applied to the gut and muscle tissues; then, they were homogenized, sonicated, centrifuged, and analyzed._[86]
Plant
Crude turmeric samplesPressurized liquid water extractionCurcuma Radix was dried at 60 °C and sieved with a 60-mesh sieve. The dried powder and diatomaceous earth were added to a stainless steel extraction cell. The extraction of the sample was obtained using the following parameters: 40% of the flush volume, methanol at 100 °C, static extraction for 5 min, 1000 psi._[23]
Powdered rhizomeUltrasonication in methanolThe powdered rhizome was extracted with methanol at 40 kHz, 200 W for 1 h using ultrasound, then filtered through 0.45 μm pores before analysis.103.62[87]
Powdered rhizomeSteam distillationPowdered rhizome was exposed to vapor at 90 °C, followed by extraction for 30 min._[88]
Powdered rhizomeExtraction with n-hexanen-Hexane was shaken with samples for 30 min at 40 °C, and centrifuged at 4000 rpm. _[89]
Powdered rhizomePressurized liquid extractionThe extraction of the sample was achieved with the following conditions: static cycle, 1, filtering through a 0.45 μm icon filter for 40% of the flush volume; pressure, 1000 psi; static extraction period, 5 min; particle size, 0.2–0.3 mm; temperature, 100 °C, with methanol.100.5[90]
Different parts of the plant, powderedExtraction with dichloromethaneDichloromethane was added and the sample was macerated for 7 days at 15–20 °C. _[64]
Powdered rhizomeCold immersion with methanolA 0.5 g sample of powder was placed in a 100 mL triangle bottle, then 50 mL methanol was added, before cold soaking for 12 h, shaking every several minutes, until the supernatant was poured out.99.6[91]
Four sources of Curcuma Radix decoction piecesHeating reflux with waterDecoction pieces were added in 10-fold amounts (m L/g) of water, soaked for 1 h, heated and reflowed twice, filtered, and concentrated.116.8[92]
Powdered rhizomeUltrasonication in ethanolUltrasound with 70% ethanol for 45 min.95.8–104.1[93]
Powdered rhizomePressurized liquid extractionMethanol; 120 °C; particle size, 0.2–0.3 mm; static extraction time, 5 min; pressure, 1500 psi; static cycle, 1; and 60% flush volume._[94]
Formulation
DecoctionImmersed in water and ethanolImmersed in 50% ethanol, 70% ethanol, 100% ethanol, and water for 1 h each, then 2 h reflux._[95]
In general, curcumenol is extracted from crude herb samples using various conditions and solvents (e.g., ultrasonication, pressurized liquid, and steam distillation) (Table 2). Zang Yuanfang et al. [96] developed an ultrasonic extraction method (power 600 W, frequency 40 kHz) with 70% methanol for 30 min, with a recovery rate of 99.8%. Yang F.Q. et al. indicated that a pressurized liquid extraction method could yield 90% recovery of curcumenol under methanol at 100 °C, with the particle size ranging from 0.2 to 0.3 mm, a static extraction time of 5 min, a pressure of 1000 psi, a static cycle, and a flush volume of 40% [97].

6. Methods for Qualitative and Quantitative Analysis

During the extraction of target compounds from different matrices, specific and effective analysis methods are also important. Curcumenol is a component of essential oils in many plant samples, and the accurate determination of volatile components in these samples is necessary. Thin-layer chromatography (TLC), colorimetry, HPLC, and GC with flame ionization detection or mass detection, have been used to perform quality control of essential oils in herbal medicines [98,99]. Nevertheless, the low separation efficiency of TLC and poor selectivity of colorimetry have hampered both their further application and development. A more powerful analysis tool was needed for the quality assessment of the abovementioned essential oils. Therefore, different qualitative and quantitative analysis methods have been established by researchers. Chromatography is a powerful and common tool used in research on curcumenol, involving different samples, covering GC and liquid chromatography (LC) (Table 4). For instance, Komatsu K. et al. [88] established a GC–MS method for the identification and detection of curcumenol in rhizomes of C. kwangsiensis from Guangdong Province, and C. wenyujin from Zhejiang Province, China, and C. kwangsiensis from the Guangxi Zhuangzu Autonomous Region (Guangxi A. R.). LC techniques have been widely applied for quantification in different fields, and are usually combined with other detectors (e.g., fluorescence, UV, and MS) [100,101,102]. F.Q. Yang et al. [90] developed a detection approach utilizing HPLC in combination with a diode array, with the aim of conducting rapid and simultaneous quantitative analysis of curcumenol and other compounds in the three species of Curcuma rhizomes. The measurement of the 11 above-described components in three different species of Curcuma rhizomes using the abovementioned method showed high repeatability, with inter-day and intra-day variances lower than 1.98% and 1.57%, respectively. It was effective in quantifying target components in samples of different Curcuma species. Gang Yin et al. [95] established UPLC-QQQ-MS to determine curcumenol and other bioactive components for the quality assessment of an AR–CR herb pair. The recovery (90.20–107.60%), repeatability (relative standard deviation (RSD) 5.98%), stability (RSD 4.29%), intra- and inter-day precision ((RSD) 3.64%, RSD 5.68%), limit of quantification (0.81–2.54 ng/mL), and limit of detection (0.33–10.78 ng/mL), were all used to optimize and validate the proposed method. Therefore, the primary bioactive ingredients in the AR–CR herb combination and the individual herbs were successfully compared using the established methodology.
Table 4. Spectroscopic and chromatographic methods used to assay the samples.
Table 4. Spectroscopic and chromatographic methods used to assay the samples.
Analytical MethodResearch Objectives/TitleMatrix and
Sample Preparation Method
ResultRef.
High-performance liquid chromatography (HPLC)Determination of curcumenol in Yujin.Powdered rhizomeRecovery value of curcumenol 103.62%.[87]
HPLCSimultaneous determination of 11 characteristic components in three species of Curcuma rhizomes using pressurized liquid extraction and HPLC.Powdered
rhizome
With intra- and inter-day fluctuations of less than 1.57% and 1.98%, respectively, this approach demonstrated good repeatability for the quantification of the abovementioned 11 components in three species of Curcuma rhizomes.[90]
HPLCQuantitation of Curcumenol extracted from water.Four sources of Curcuma Radix decoction piecesFor C. kwangsiensis, C. wenyujin, C. longa, and C. phaeocaulis, the contents of curcumenol in the water extract were 0.066, 0.271, 0.058, and 0.310 mg/g, respectively.[92]
Ultra-performance liquid chromatography (UPLC)Determination of the curcumenol content in Curcuma kwangsiensis, vinegar-boiled Curcuma kwangsiensis, and water extract residues using UPLC.Powdered rhizome and water residuesThe established analysis method is accurate, stable, and sensitive; it can be used for the quality assay of Curcuma kwangsiensis, and it provides a scientific basis for the resource utilization of water extract residues.[103]
High resolution gas chromatography (HRGC)Quantitation of curcumenol.Different parts of this plant, powderedStandard samples of curcumenol within the concentration range of 0.03–0.93 mg/mL.[64]
Liquid chromatography tandem–mass spectrometry (LC–MS)Fingerprint of Curcuma phaeocaulis using LC–MS.Powdered rhizomePeaks 4–6 and 9 were identified as curcumin, demethoxycurcumin, curcumenol, and curcumone, respectively, through the contrast of the retention time and the online UV spectra and the molecular weight of the chemical standards.[104]
Ultra-high-performance liquid chromatography coupled with the triple quadrupole tandem mass spectrometry method (UPLC-QQQ-MS)Identification of curcumenol.Decoction, dispensing granule decoctionIt was the first time that 17 chemicals in the Astragali Radix–Curcumae Rhizoma (AR–CR) herb pair were simultaneously analyzed using UPLC-QQQ-MS, offering a workable approach in terms of overall quality control.[95]
Ultra-high-performance liquid chromatography–quadrupole time-of-flight mass spectrometryIdentification of curcumenol.Crude turmeric samples, pressurized liquid extractionsThis new method led to the discovery of curcumenol and some other components as unique chemical markers for identification.[23]
Gas chromatography–mass spectrometry (GC/GC–MS)Identification of curcumenol.Powdered rhizomeIdentified curcumenol.[88]
GC–MSIdentification of curcumenol.Powdered rhizomeTwo isomeric forms of curcumenol comprised the GC chromatogram’s major components, taking up 28.68 ± 0.91% and 17.96 ± 0.69% of the control plants’ total volatiles, respectively.[89]
GC–MSIdentification and quantitation of curcumenol in Curcuma rhizomes.Powdered rhizomeCurcumenol and four other compounds were optimized as markers for the quality control of Ezhu.[94]
GC–MSDetermination of curcumenol in Curcuma rhizomes.Powdered rhizomeNine sesquiterpenoids were effectively quantified using the validated method in 18 samples of three Curcuma species, utilized as Ezhu.[97]
GC–MSDetermination of curcumenol in the fractionation of volatile constituents originating from the Curcuma rhizome using GC.Powdered rhizomeThe structures of the compound were identified as curcumenol using a mass spectrometer (MS) and NMR spectra, respectively.[105]

7. Other Factors Influencing the Content of Curcumenol

The contents of the components in plants are affected by their variety, place of origin, growth environment, processing methods, etc. [106,107,108,109]. Some factors relevant to the content of curcumenol (e.g., the constituents of minerals in the soil and the harvest season of herbs) have been illustrated. Rabia F. El-Hawaz et al. [89] examined the mineral concentration effects on the content of volatile constituents in rhizomes. The interaction of Ca2+ with KNO3 affected the contents of β-elemenone, isocurcumenol, germacrone, and curcumenol isomers I and II. The abovementioned findings demonstrated that minerals in the in vitro bioreactor medium during rhizome growth had an impact on the production of volatile components in turmeric when transferred to the greenhouse after six months. Christiane Regina Pamplona et al. [64] revealed that dihydrocurdione (2) and curcumenol (1) are two active terpenoids from different portions of Curcuma zedoaria cultivated in Brazil and that they displayed seasonal variations. As revealed by the results, the production of 1 and 2 was nearly three times greater in the mother rhizome in the autumn (Table 5 and Table 6). There are an insufficient number of studies focusing on the influencing factors of the content of curcumenol in Curcuma wenyujin. However, as a component of the volatile oil in Curcuma wenyujin, the content of volatile oil was affected by the areas of production [110], is shown in Table 7.

8. Bioactivity

Existing research has confirmed that curcumenol has numerous biological effects, including anti-tumor [13,14,15,16], anti-inflammatory [17,18], anti-virus [20,21], hepatoprotection [32], and neuroprotection activities [5]. Curcumin is another major bioactive substance contained in the Curcuma species; these two compounds have aroused increasing attention from chemists and biologists, and are expected to be investigated in-depth in the future. A comparison of the physiological effects of the two compounds is shown in Table 8.
Table 8. The physiological effects of curcumenol and curcumin.
Table 8. The physiological effects of curcumenol and curcumin.
CurcumenolCurcumin
Disease/
Curcumenol Activity
ModelTreatment DosesMechanismRef.Treatment DosesModelRef.
Anti-inflammatory activityBV-2 microglial cells5–20 μMInhibiting Akt-dependent nuclear factor kappa-B (NF-κB) activation and the downregulation of Akt and p38 mitogen-activated protein kinase (MAPK) signaling[18]5–20 μMBV-2 microglial cells[111]
Anti-inflammatory activityHaCaT cells200 μg/mLInhibits the overexpression of inflammatory factors[19]20 μMHaCaT cells[112]
Analgesic effectsMice10 mg/kg-[64]1000 mg/kg to 2000 mg/kgMice[113]
Analgesic effectsMice12, 22, 29 μmol/kgNo involvement with the opioid system[65]---
Anti-oxidant ability and neuroprotective activityNG108-15 cells4 μM-[5]25–100 μMNG108-15 cells[114]
Against coronary heart diseaseCoronary heart disease rats-Improving blood lipid level, blood stasis, and myocardial infarction, and controlling the signaling pathway of PI3K/AKT/mTOR[67]---
Anti-diabetic activityHuman hepatocellular carcinoma (HepG2) cells10 μM-[32]-Human[115]
Ameliorating osteoporosisMice100 μMImpairs the stability of TRAF6 enhanced by IPMK and suppresses excessive osteoclast activity[69]100 mg/kgMice[116]
SARS-CoV-2 infection--High-affinity interaction with proteins involved in coronavirus infection[117]10 µg/mLDG614 strain and Delta variant[118]
Anti-bacteriostatic effectsGram-negative and Gram-positive bacteria50 μg/mL-[20]---
Anti-bacteriostatic effectsP. capsici20 μg/mLDamages the cell membrane[21]---
Breast cancerMCF-7 cells9.3 ± 0.3 μg/mLAnti-proliferative activity and induces apoptotic cell death[16]20 μMMCF-7 cells[119]
Gastric cancerAGS cells263.34 ± 2.97 μMInhibition of proliferation[15]32 μMAGS cells[120]
Lung cancerH1299 and H460 cells100–400 μg/mLVia the lncRNA H19/miR-19b-3p/FTH1 axis for lung cancer cell ferroptosis[13]5–30 μMH460 cells[121]
Cervical cancerHeLa and C33A Cells-Through the tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein gamma polypeptide (YWHAG) pathway, facilitating cervical cancer cell apoptosis[73]20 μMHeLa and C33A Cells[122]
Inhibitory to cytochrome P450 (CYP) enzymesCytochrome P450 3A4 (CYP3A4)12.6 ± 1.3 μM-[123]11.93 ± 3.49 µMCYP3A4[124]
Binding to human serum albumin (HSA)HSA60 μM-[125]-HSA[126]

8.1. Anti-Inflammatory Activity

Inflammation is the defense response of the immune system to tissue damage arising from trauma, infection, chemical exposure, etc. [127,128,129]. It is helpful in the repair of damaged tissues and removes harmful substances, but it is also destructive and disease may be triggered under excessive inflammatory conditions [130,131]. The production of inflammatory cytokines, followed by a decline in the biomechanics and structural integrity of the knee joint, results in osteoarthritis (OA) [132]. Xiao Yang et al. suggested that curcumenol reduced inflammation in ATDC5 chondrocytes and primary mouse chondrocytes, and ameliorated OA in a mouse model of medial meniscus-induced destabilization [17]. They employed tumor necrosis factor alpha (TNF-α) and Interleukin-1β (IL-1β) to treat ATDC5 chondrocyte and primary chondrocyte cells to induce inflammation. Curcumenol inhibited the progression of inflammation through downregulation of the expression levels of matrix metalloproteinase-3 (MMP3), inactivation of the MAPK and NF-κB signaling pathways, and the amelioration of catalytic changes and degradation of the extracellular matrix. Curcumenol can, therefore, be adopted to treat OA and inflammatory-mediated neurodegenerative diseases. Jia Ye Lo et al. [18] investigated the regulatory effect of curcumenol on the lipopolysaccharide-induced inflammation of BV-2 microglia, and reported that it could downregulate the Akt and p38 MAPK signaling pathways by inhibiting Akt-dependent NF-κB activation. Thus, the expression of proinflammatory mediators and regulatory genes is downregulated. Wang Jiafeng [19] suggested that 10 μg/mL curcumenol and other active components in Curcuma could inhibit the excessive secretion of four inflammatory cytokines (IL-1β, interleukin-6, interleukin-8, and interleukin 33) induced by TNF-α to different degrees in the treatment of psoriasis. Moreover, IκB-α and p-p65 expression in the NF-κB pathway could be suppressed. As revealed by the above results, the possible mechanism in the treatment of psoriasis by curcumenol and other active components in Curcuma is the inhibition of the TNF-α pathway and the inducement of the NF-κB pathway; subsequently, the inflammation and excessive proliferation of epidermal cells in the affected area can be alleviated.

8.2. Analgesic Effects

Curcuma rhizomes have long served as analgesics throughout history [6]. Some researchers have conducted phytochemical and analgesic activity analyses and attempted to determine the antinociceptive active compounds of Curcuma zedoaria R. Br. over the past few years [64,65]. Navarro et al. [65] reported that curcumenol exhibited manifold more potent and dose-associated analgesic activities in mice when evaluated in several models as compared with reference drugs. However, the mechanism of curcumenol does not cover the participation of the opioid system, as revealed by insufficient effects in the hot plate test.

8.3. Anti-Oxidant Ability and Neuroprotective Activity

Oxidative stress has been reported to arise from mitochondrial dysfunction, and it is also considered one of the critical contributors to neurodegeneration [133,134]. Compounds extracted from Curcuma, ginger, Ginkgo biloba, cinnamon, and related plants are characterized by their preventive and therapeutic roles in neurodegenerative disorders [135,136,137,138,139]. Omer Abdalla Ahmed Hamdi et al. [5] investigated the effect of components from Curcuma zedoaria (Christm.) Rosc. on hydrogen peroxide-induced oxidative stress in NG108-15 cells. The results showed that when NG108-15 cells were exposed to H2O2 (400 μM) for 24 h, the cell viability decreased to 67.6%. If pretreated with curcumenol, the protective effect is 100% at 4 μM and 97.7% when the curcumenol concentration is increased to 30 μM. Therefore, curcumenol has neuroprotective effects. Curcumenol also showed anti-oxidant activity in the oxygen radical anti-oxidant capacity (ORAC) assay [5].

8.4. Coronary Heart Disease (CHD)

CHD is the most common cause of death globally [140]. The Curcuma aromatica Salisb. rhizome (CASR) has often been employed to treat CHD arising from blood stasis syndrome in clinics, exhibiting multi-component and multi-pathway effects [67]. Chenghao Fei et al. [67] suggested that CASR is capable of significantly improving blood stasis, myocardial infarction, and blood lipid levels, and regulating the PI3K/AKT/mTOR signaling pathway, in CHD rats. Lipidomics has further shown that CASR can control the metabolism of glycerophospholipids, aberrant sphingolipids, and glycerolipids. Its effects may be attributed to (+)-curcumenol, curzerene, and isoprocurcumenol, which are reportedly the major active CASR compounds and are capable of mitigating vascular endothelial damage, upregulating blood lipid levels, and reducing blood viscosity [67]. These components could become promising drugs in CHD treatment.

8.5. Anti-Diabetic Activity

Over the past 20 years, obesity has spread over the world like wildfire, endangering lives by impacting nearly every organ system. It is currently a major public health concern and one of the most prevalent non-communicable diseases (NCDs) [141,142,143]. Changxin Zhou et al. [32] acquired extract compounds from Curcuma wenyujin, and adopted a glucose consumption model of HepG2 cells to evaluate their anti-diabetic effect. As the results showed, curcumenol and eight other compounds exhibited promising activity, with an over 45% increase in glucose consumption at 10 μM.

8.6. Ameliorating Osteoporosis

Osteoporosis is characterized by reduced bone mass and damaged bone microstructure; it is a systemic bone disease, which can result in bone brittleness and easy fracturing [144]. Despite substantial research into the pharmaceutical management of osteoporosis, better and more efficient treatment approaches are still needed [145]. Shiyu Wang et al. [69] revealed for the first time that curcumenol inhibits osteoclast differentiation in vitro and ameliorates osteoporosis in ovariectomy mice. Curcumenol can inhibit the protective effect of inositol polyphosphate multikinase on osteoclast differentiation, while hindering key downstream pathway activation when the receptor activator of nuclear factor-κB ligand induces osteoclastogenesis. Given the novel involvement of inositol polyphosphate multikinase in osteoclastogenesis and the possible therapeutic effect of curcumenol in regulating osteoporosis, inhibitors targeting inositol polyphosphate multikinase are likely to represent a novel direction for developing anti-osteoporosis medications.

8.7. Anti-Viral and Anti-Bacteriostatic Effects

With the rapid spread of the novel coronavirus disease over the past few years, further effective treatments are still required [146]. Gaurav S. Dave et al. selected curcumenol and three other potent candidates from natural compounds to treat and prevent coronavirus infection. Comparing the open state of severe acute respiratory syndrome coronavirus 2 to the closed state, curcumenol achieved an impressive docking score. This finding provides evidence in favor of the preventative ability of curcumenol to reduce the binding capacity of severe acute respiratory syndrome coronavirus in healthy humans [117].
As revealed by Noor Akbar et al. [20], the gut bacteria of animals that live in polluted environments serve as a promising source of anti-bacterial compounds. Using purified chemicals (e.g., curcumenol) extracted from the gut bacteria of such animals, these researchers determined the anti-bacterial and cytotoxic effects of these chemicals; cell viability tests were performed using lactate dehydrogenase release and methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay, respectively. The findings showed that purified curcumenol and other compounds demonstrated significant anti-bacterial activities against several Gram-positive and Gram-negative bacteria, with effective minimal inhibitory concentrations of MIC50 and MIC90 at μg levels. Additionally, these concentration levels only exert minor effects on human cells, as indicated by the lactate dehydrogenase and MTT research [20].
As indicated by Wang Bi’s team, natural plant-derived zedoary turmeric oil (ZTO) (e.g., curcumenol) and several other components (curcumol, β-elemene, and curdione) serve as the main effective chemicals, exhibiting superb anti-fungal activity against P. capsici both in vitro and in vivo. It has been speculated that it exerts its effects by disrupting cell membrane integrity [21]. ZTO is a promising natural anti-fungal compound for the treatment of phytophthora blight arising from P. capsici.

8.8. Diversity of Anti-Cancer Effects

Cancer is a large category of diseases that can occur in nearly every organ or tissue in the body, and is characterized by uncontrollable abnormal cell growth, invading adjacent parts of the body outside of the usual boundaries, and/or spreading to other organs. Chinese herbal medicines have long served as anti-cancer treatments, since they contain ample anti-cancer chemicals, exert promising cytotoxicity effects, regulate the tumor microenvironment and cancer immunity, and result in improved chemotherapy effects [147]. Existing research has suggested that curcumenol extracted from Curcuma wenyujin exhibits appreciable anti-tumor efficacy (e.g., suppressing breast tumor cells [148,149], digestive tumor cells [150], liver cancer cells [71], and lung cancer cells [13]), as shown in Figure 2.

8.8.1. Effects on Breast Cancer

Breast cancer ranks highly among women with cancer-related mortality and is a major cause of death for women in their 40s [151,152]. Many researchers have identified effective compounds to treat breast cancer [153]. The chemical components in Curcuma zedoaria’s hexane and dichloromethane fractions were studied by Omer Abdalla Ahmed Hamdi et al. [16]. In total, 19 substances were examined using an MTT assay against Ca Ski, MCF-7, the human prostatic carcinoma cell line (PC-3), and HT-29 cancer cell lines, to determine whether they could inhibit cell proliferation. The two key ingredients of Curcuma zedoaria, curcumenone and curcumenol, both showed remarkable anti-proliferative action against MCF-7 cells, with the half-maximal inhibitory concentrations (IC50 values) reaching 8.3 ± 1.0 and 9.3 ± 0.3 μg/mL, respectively. As revealed by the results achieved based on the Hoechst 33342/PI double-staining experiment and phase contrast, they might cause apoptotic cell death. These findings underpin the ethnomedical use of Curcuma zedoaria as a breast cancer treatment.

8.8.2. Effects on Gastric Cancer

Gastric adenocarcinoma ranks third in cancer-related deaths, with approximately 800,000 fatalities worldwide, and is the fifth most frequent malignancy in terms of incidence [154]. Although many chemical therapies exist, a new and effective candidate compound is still needed. Curcumenol has demonstrated the dose-dependent suppression of gastric cancer cell proliferation [15]. Curcumenol, 11-epoxycarabrane, 4,8-dioxo-6-methoxy-7, and zedoarofuran, exert lethal effects against stomach cancer AGS cells, with IC50 values ranging from 212 to 392 μM, as found by Tae Kyoung Lee’s research team. As the primary cytotoxic components in C. zedoaria rhizomes, curcumenol and three other compounds offer additional experimental support for the traditional usage of C. zedoaria rhizomes in treating gastric cancer.

8.8.3. Effects on Lung Cancer

Lung cancer tumors have the highest mortality rate in the world; however, even though there have been many improvements in the therapy of lung cancer [155], developing new effective drugs is still urgently required. Zhang Ruonan et al. [13] reported that curcumenol could suppress cell proliferation and induce cell death in lung cancer cells. Their research showed that ferroptosis was the mechanism by which curcumenol induced lung cancer cell death, and that the lncRNA H19/miR-19b-3p/FTH1 axis was crucial for this process. In light of this, curcumenol could be used to treat lung cancer patients.

8.8.4. Synergistic Anti-Cancer Effects

Drug combinations have been demonstrated to alleviate toxic side effects and improve efficacy in the treatment of cancer [156,157,158,159]. In addition to directly acting on different cancer cells, it has been reported that curcumenol can increase anti-tumor effects in combination with other anti-cancer compounds, such as cisplatin and laminarin [71,73]. Zhijie Mao et al. [73] determined that curcumenol extracted from Curcuma could inhibit YWHAG expression in cervical cancer and reduce cervical cancer cell proliferation, invasion, and MMP2 and MMP9 expression when combined with cisplatin, compared with cisplatin alone. The abovementioned combination of drugs led to increased apoptosis, downregulated B cell lymphoma 2 (Bcl-2) expression, and the upregulated expression of Bcl-2 antagonist X, polyadenosine diphosphate-ribose polymerase, and caspase-3. Accordingly, curcumenol is capable of increasing the anti-tumor effect of cisplatin against cancer cell proliferation, migration, invasion, and apoptosis. Curcumenol also exhibits enhanced anti-cancer effects against human hepatoma HepG2 cells when combined with laminarin. Huanxiao Han et al. [71] suggested that the combination of curcumenol and laminarin could inhibit the proliferation, migration, and invasion of human hepatoma HepG2 cells; the levels of pSTAT3 and Bcl-2 decreased in cystathionine beta synthase knockdown HepG2 cells. Furthermore, it can notably contribute to the use of kelp and Curcuma zedoary in liver cancer treatment. The mechanism of synergistic anti-cancer effects is shown in Figure 3.

8.9. Pharmacokinetics Study

Curcumenol is the key component of extensively employed natural products, requiring the determination of the pharmacokinetic parameters in order to guarantee safety and effectiveness. However, few studies have been conducted on its pharmacokinetic characteristics, as depicted in the following paragraphs (e.g., the interactions with human liver cytochrome P450 enzymes and the binding effect on human serum albumin).

8.9.1. Inhibitory Effect on Human Liver Cytochrome P450 Enzymes

Curcumenol has been extensively used to inhibit cancer and inflammation, such that it is likely to be employed in combination with different drugs in many conditions to enhance treatment efficacy. It is imperative to evaluate the pharmacokinetics of drug–drug interactions induced by curcumenol. Dong-Xue Sun et al. [123] confirmed that CYP3A4 could be strongly inhibited (IC50 = 12.6 ± 1.3 μM) by curcumenol in the investigation of inhibitory effects on seven CYP isoforms. Curcumenol does not serve as a mechanism-based inhibitor, according to research into time- and nicotinamide adenine dinucleotide phosphate-dependent inhibition. However, given the limited pharmacokinetic information available, the promising clinical effects of curcumenol on patients are unlikely to have been adequately evaluated. Accordingly, more research should be conducted to determine the extent of the drug–drug interactions potentially arising due to curcumenol.

8.9.2. Binding to Human Serum Albumin (HSA) In Vitro

The interaction of a compound with serum albumin can influence its pharmacokinetic characteristics, such as distribution, metabolism, elimination, and bioavailability, in bodily processes [160,161]. Based on this theory, Omer Abdalla Ahmed Hamdi et al. [125] investigated the binding of curcumenol and curcumenone to HSA through fluorescence quench titration. Molecular docking was performed to gain thorough insights into the interactions with HSA. Analysis of the fluorescence data indicated that there was a moderate binding affinity between the ligands and HSA. This showed that the binding constants of curcumenone and curcumenol were 2.46 × 105 M−1 and 1.97 × 104 M−1, respectively. The binding properties of curcumenol to HSA offer useful parameters for the detection of pharmacokinetic profiles based on the fluorescence spectroscopy and molecular docking findings. Nevertheless, since there are scant in vivo pharmacokinetic data, the pharmacokinetic parameters of curcumol, an analogue of curcumenol, in beagle dogs [150] can serve as a reference if required. However, specific pharmacokinetic data, in vitro and in vivo, remain to be obtained more substantially in the future.

8.10. Other Effects

In addition to the abovementioned bioactivities, further pharmacology effects have been identified. Curcumenol and some principal sesquiterpenes isolated from Zedoariae Rhizoma can exert prominent protective effects against liver injury mice [22]. Curcumenol may also be an effective candidate agent for treating structural and functional problems involving the skeletal muscle [84]. Zhang et al. [84] investigated the effect of curcumenol on myogenic differentiation and mitochondrial function. After increasing the mitochondria mass and function in myotubes, it was reported to promote the initiation of myogenesis and the formation of functional myotubes. Guanghui Zhong found that curcumenol could improve renal function in 5/6 nephrectomy rats with chronic renal failure [162].
Overall, curcumenol has immense value in the health field, as shown in Figure 4, and further in-depth investigations are warranted.
Although there are scarce pharmacokinetics data concerning curcumenol, it has shown an inhibitory effect on CYP3A, and binding effects to HSA, which are two key factors in the pharmacokinetics process in vivo.

9. Conclusions and Prospects

Wenyujin, a member of the Curcuma zedoaria family, has been reported to comprise a total of 169 compounds, thus far. It has a long history and has been extensively employed in TCM due to its abundant components, including curcuminoids, monoterpenoids, sesquiterpenoids, diterpenoids, etc. To elucidate the exact compounds that exert the abovementioned traditional curative effects using current medical theories and to determine the targets of its biological effects, so as to change the dosage form, improve the effect of treatment, and reduce the waste of TCM resources, many researchers have conducted studies and found a series of compounds related to those treatment effects, including curcumenol. To more effectively extract curcumenol from different plant samples, researchers have developed several methods (e.g., pressurized liquid water extraction, steam distillation, ultrasonication in methanol or ethanol, extraction with n-hexane, extraction with dichloromethane, cold immersion with methanol, and heating reflux with water). Different detection methods have also been established (e.g., HPLC, UPLC, HRGC, LC-MS/MS, and GC–MS methods) to precisely measure and identify traces of curcumenol. Curcumenol also exhibits extensive bioactive effects (e.g., anti-cancer, anti-inflammatory, anti-bactericidal, and anti-diabetic activities, and the amelioration of osteoporosis).
In traditional applications of Curcuma, it is often used as an adjuvant of pre-existing therapies with other Chinese medicine to treat diseases. Moreover, curcumenol, as an important active ingredient, can be combined with the main chemical components of other Chinese medicine compounds in preparations of clinical applications to achieve better clinical effects. In anti-cancer treatments, it should be combined with chemotherapy drugs to reduce the toxicity of these medications to normal cells, while enhancing the sensitivity of tumor cells to chemotherapy drugs, reducing the drug resistance of tumor cells, and reducing side effects. However, it can dissolve in organic solvents but is insoluble in water, which has seriously limited its pharmacological effects in basic research and clinical treatment. To address this, new preparations should be developed to increase the water solubility and biological target effects of curcumenol. It is also possible to modify the structure of curcumenol to increase its solubility and biological effects.
More pharmacokinetic parameters of curcumenol should be determined to establish its safety and efficacy, such that further references can be provided for applications in clinical practice. Furthermore, it could facilitate the development of TCM and safeguard consumers’ health and safety.

Author Contributions

Conceptualization, J.L.; writing—original draft, J.L.; writing—review and editing, Y.S. and G.L.; supervision, C.C., X.S. and Q.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially funded by the Science and Technology Development Fund, Macau SAR (No: 0098/2021/A2 and 0048/2023/AFJ), Jiangxi Province Double Thousand Talents-Leader of Natural Science Talents Project, and the Jiangxi Province urgently needed overseas talent project (20223BCJ25027), Macau University of Science and Technology’s Faculty Research Grant (No: FRG-23-003-FC).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AGS cellshuman gastric adenocarcinoma cells
ARAstragali Radix
AR–CRAstragali Radix–Curcuma Rhizoma
ATDC5 chondrocytesmouse embryonic tumor cells
CASchemical abstracts service
CaSkihuman cervical cancer
CASRCurcuma aromatica salisb. rhizome
CHDcoronary heart disease
CRCurcuma Rhizoma
CYPcytochrome P450
CYP3A4cytochrome P450 3A4
GCgas chromatography
GC–MSgas chromatography–mass spectrometry
MSmass spectrometry
H460 cellshuman large cell lung cancer cells
HaCaT cellshuman immortalized keratinocytes
HeLahuman cervical carcinoma cell line
HPLChigh-performance liquid chromatography
HRGChigh-resolution gas chromatography
HSAhuman serum albumin
HT-29human colorectal adenocarcinoma cells
IL-1βInterleukin-1β
LC-MSliquid chromatography tandem mass spectrometry
MAPKmitogen-activated protein kinases
MCF-7 cellshuman breast cancer cell line
MICminimal inhibitory concentration
MMPmatrix metalloproteinase
MTTmethylthiazolyldiphenyl-tetrazolium bromide
NF-κBnuclear factor kappa-B
NMRnuclear magnetic resonance
OAosteoarthritis
pSTAT3signal transducer and activator of transcription 3 protein phosphorylation
PC-3human prostate cancer cell
RSDrelative standard deviation
TCMtraditional Chinese medicine
TLCthin-layer chromatography
TNF-αtumor necrosis factor alpha
UPLCultra-performance liquid chromatography;
UPLC-QQQ-MSultra-high-performance liquid chromatography coupled to triple quadrupole tandem mass spectrometry method
UVultraviolet
YWHAGtyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein gamma polypeptide
ZTOzedoary turmeric oil

References

  1. Fang, F.; Cheng, Z.H.; Guo, Y.L.; Cai, Y.B. Comparative Analysis of the Volatile Components in the Fresh Roots and Rhizomes of Curcuma wenyujin by Static Headspace Gas Chromatography Mass Spectrometry. Chin. J. Chem. 2006, 24, 1346–1351. [Google Scholar] [CrossRef]
  2. Yin, M.; Chu, S.; Zhao, Y.; Zheng, X.; HS, P. Materia medica illustrations related to the regional names in Zhejiang province in Ben Cao Tu Jing. Zhonghua Yi Shi Za Zhi 2022, 52, 131–139. [Google Scholar] [CrossRef] [PubMed]
  3. Cai, Y.; Li, W.; Tu, H.; Chen, N.; Zhong, Z.; Yan, P.; Dong, J. Curcumolide reduces diabetic retinal vascular leukostasis and leakage partly via inhibition of the p38MAPK/NF-κ B signaling. Bioorg. Med. Chem. Lett. 2017, 27, 1835–1839. [Google Scholar] [CrossRef]
  4. Ma, J.; Chen, J.; Zhao, B.; Jiang, Z.; Feng, L.; Jia, X. Advance in research on anticancer drug β-elemene and its derivatives. Chin. Tradit. Herb. Drug. 2018, 49, 1184–1191. [Google Scholar] [CrossRef]
  5. Hamdi, O.A.A.; Ye, L.J.; Kamarudin, M.N.A.; Hazni, H.; Paydar, M.; Looi, C.Y.; Shilpi, J.A.; Kadir, H.A.; Awang, K. Neuroprotective and Antioxidant Constituents from Curcuma zedoaria Rhizomes. Rec. Nat. Prod. 2015, 9, 349–355. [Google Scholar]
  6. Li, Y.; Wu, Y.; Li, Y.; Guo, F. Review of the traditional uses, phytochemistry, and pharmacology of Curcuma wenyujin Y. H. Chen et C. Ling. J. Ethnopharmacol. 2021, 269, 113689. [Google Scholar] [CrossRef] [PubMed]
  7. Kotha, R.R.; Luthria, D.L. Curcumin: Biological, Pharmaceutical, Nutraceutical, and Analytical Aspects. Molecules 2019, 24, 2930. [Google Scholar] [CrossRef] [PubMed]
  8. Hikino, H.; Sakurai, Y.; Numabe, S.; Takemoto, T. Structure of curcumenol. Chem. Pharm. Bull. 1968, 16, 39–42. [Google Scholar] [CrossRef] [PubMed]
  9. Xu, J.; Ji, F.; Kang, J.; Wang, H.; Li, S.; Jin, D.-Q.; Zhang, Q.; Sun, H.; Guo, Y. Absolute Configurations and NO Inhibitory Activities of Terpenoids from Curcuma longa. J. Agric. Food. Chem. 2015, 63, 5805–5812. [Google Scholar] [CrossRef] [PubMed]
  10. Asem, S.D.; Laitonjam, W.S. A new guaianolide sesquiterpene lactone from Curcuma leucorrhiza Roxb. Nat. Prod. Res. 2014, 28, 477–482. [Google Scholar] [CrossRef]
  11. Qiu, G.; Yan, P.; Shao, W.; Zhou, J.; Lin, W.; Fang, L.; Zhao, X.; Dong, J. Two new sesquiterpenoids including a sesquiterpenoid lactam from Curcuma wenyujin. Chem. Pharm. Bull. 2013, 61, 983–986. [Google Scholar] [CrossRef] [PubMed]
  12. Aspollah Sukari, M.; Wah, T.S.; Saad, S.M.; Rashid, N.Y.; Rahmani, M.; Lajis, N.H.; Hin, T.-Y.Y. Bioactive sesquiterpenes from Curcuma ochrorhiza and Curcuma heyneana. Nat. Prod. Res. 2010, 24, 838–845. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, R.; Pan, T.; Xiang, Y.; Zhang, M.; Xie, H.; Liang, Z.; Chen, B.; Xu, C.; Wang, J.; Huang, X.; et al. Curcumenol triggered ferroptosis in lung cancer cells via lncRNA H19/miR-19b-3p/FTH1 axis. Bioact. Mater. 2022, 13, 23–36. [Google Scholar] [CrossRef]
  14. Peng, B.; Zhou, X.; Shi, J.; Li, Z. Effects of volatile oil and three main components from Curcuma phaeocaulis Valeton on liver cancer and endometrial carcinoma cell lines. West China J. Pharm. Sci. 2007, 2007, 312–313. [Google Scholar] [CrossRef]
  15. Lee, T.K.; Lee, D.; Lee, S.R.; Ko, Y.-J.; Sung Kang, K.; Chung, S.J.; Kim, K.H. Sesquiterpenes from Curcuma zedoaria rhizomes and their cytotoxicity against human gastric cancer AGS cells. Bioorg. Chem. 2019, 87, 117–122. [Google Scholar] [CrossRef] [PubMed]
  16. Ahmed Hamdi, O.A.; Syed Abdul Rahman, S.N.; Awang, K.; Abdul Wahab, N.; Looi, C.Y.; Thomas, N.F.; Abd Malek, S.N. Cytotoxic Constituents from the Rhizomes of Curcuma zedoaria. Sci. World J. 2014, 2014, 321943. [Google Scholar] [CrossRef]
  17. Yang, X.; Zhou, Y.; Chen, Z.; Chen, C.; Han, C.; Li, X.; Tian, H.; Cheng, X.; Zhang, K.; Zhou, T.; et al. Curcumenol mitigates chondrocyte inflammation by inhibiting the NF-κB and MAPK pathways, and ameliorates DMM-induced OA in mice. Int. J. Mol. Med. 2021, 48, 192. [Google Scholar] [CrossRef]
  18. Lo, J.Y.; Kamarudin, M.N.A.; Hamdi, O.A.A.; Awang, K.; Kadir, H.A. Curcumenol isolated from Curcuma zedoaria suppresses Akt-mediated NF-κB activation and p38 MAPK signaling pathway in LPS-stimulated BV-2 microglial cells. Food. Funct. 2015, 6, 3550–3559. [Google Scholar] [CrossRef]
  19. Wang, J. Inhibition of HaCaT Cell Proliferation by Active Constituents of Rhizoma Curcumae and Its Mechanism of Action Based on NF-κB Signaling Pathway. Master’s Thesis, University of Electronic Science and Technology of China, Chengdu, China, 2019. [Google Scholar]
  20. Akbar, N.; Siddiqui, R.; Iqbal, M.; Khan, N.A. Antibacterial Activities of Selected Pure Compounds Isolated from Gut Bacteria of Animals Living in Polluted Environments. Antibiotics 2020, 9, 190. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, B.; Liu, F.; Li, Q.; Xu, S.; Zhao, X.; Xue, P.; Feng, X. Antifungal activity of zedoary turmeric oil against Phytophthora capsici through damaging cell membrane. Pestic. Biochem. Physiol. 2019, 159, 59–67. [Google Scholar] [CrossRef] [PubMed]
  22. Matsuda, H.; Ninomiya, K.; Morikawa, T.; Yoshikawa, M. Inhibitory effect and action mechanism of sesquiterpenes from Zedoariae Rhizoma on D-galactosamine/lipopolysaccharide-induced liver injury. Bioorg. Med. Chem. Lett. 1998, 8, 339–344. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, F.; Bai, X.; Yang, F.Q.; Zhang, X.J.; Hu, Y.; Li, P.; Wan, J.B. Discriminating from species of Curcumae Radix (Yujin) by a UHPLC/Q-TOFMS-based metabolomics approach. Chin. Med. 2016, 11, 21. [Google Scholar] [CrossRef] [PubMed]
  24. Hu, X.; Wang, L.; Zhang, L.; Zhang, T. β-Elemene inhibits 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate-induced skin tumorigenesis through suppression of NF-κB-associated signaling events in the mouse skin model. J. Biochem. Mol. Toxicol. 2020, 34, e22550. [Google Scholar] [CrossRef] [PubMed]
  25. Gouthamchandra, K.; Sudeep, H.V.; Chandrappa, S.; Raj, A.; Naveen, P.; Shyamaprasad, K. Efficacy of a Standardized Turmeric Extract Comprised of 70% Bisdemothoxy-Curcumin (REVERC3) against LPS-Induced Inflammation in RAW264.7 Cells and Carrageenan-Induced Paw Edema. J. Inflamm. Res. 2021, 14, 859–868. [Google Scholar] [CrossRef]
  26. Lee, H.Y.; Kim, S.W.; Lee, G.H.; Choi, M.K.; Chung, H.W.; Lee, Y.C.; Kim, H.R.; Kwon, H.J.; Chae, H.J. Curcumin and Curcuma longa L. extract ameliorate lipid accumulation through the regulation of the endoplasmic reticulum redox and ER stress. Sci. Rep. 2017, 7, 6513. [Google Scholar] [CrossRef] [PubMed]
  27. Mhillaj, E.; Tarozzi, A.; Pruccoli, L.; Cuomo, V.; Trabace, L.; Mancuso, C. Curcumin and Heme Oxygenase: Neuroprotection and Beyond. Int. J. Mol. Sci. 2019, 20, 2419. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, L.; Yang, Z.; Chen, D.; Huang, Z.; Li, Y.; Lan, X.; Su, P.; Pan, W.; Zhou, W.; Zheng, X.; et al. Variation on Composition and Bioactivity of Essential Oils of Four Common Curcuma Herbs. Chem. Biodivers. 2017, 14, e1700280. [Google Scholar] [CrossRef]
  29. Qin, Y.; Zhao, Q.; Zhao, Y.; Ren, X.; Bao, K.; Li, X.; Jiang, C. Exploring mechanism of Curcuma wenyujin against COVID-19. Chin. Tradit. Herb. Drugs 2020, 51, 1977–1983. [Google Scholar] [CrossRef]
  30. Salehi, B.; Rodrigues, C.F.; Peron, G.; Dall’Acqua, S.; Sharifi-Rad, J.; Azmi, L.; Shukla, I.; Singh Baghel, U.; Prakash Mishra, A.; Elissawy, A.M.; et al. Curcumin nanoformulations for antimicrobial and wound healing purposes. Phytother. Res. 2021, 35, 2487–2499. [Google Scholar] [CrossRef]
  31. Hu, Y.; Zhang, J.; Kong, W.; Zhao, G.; Yang, M. Mechanisms of antifungal and anti-aflatoxigenic properties of essential oil derived from turmeric (Curcuma longa L.) on Aspergillus flavus. Food Chem. 2017, 220, 1–8. [Google Scholar] [CrossRef]
  32. Zhou, C.; Zhang, L.; Chen, F.; Wu, H.; Mo, J.; Gan, L. Terpenoids from Curcuma wenyujin increased glucose consumption on HepG2 cells. Fitoterapia 2017, 121, 141–145. [Google Scholar] [CrossRef] [PubMed]
  33. Lekshmi, P.C.; Arimboor, R.; Indulekha, P.S.; Menon, A.N. Turmeric (Curcuma longa L.) volatile oil inhibits key enzymes linked to type 2 diabetes. Int. J. Food Sci. Nutr. 2012, 63, 832–834. [Google Scholar] [CrossRef] [PubMed]
  34. Xie, H.; Su, D.; Zhang, J.; Ji, D.; Mao, J.; Hao, M.; Wang, Q.; Yu, M.; Mao, C.; Lu, T. Raw and vinegar processed Curcuma wenyujin regulates hepatic fibrosis via bloking TGF-β/Smad signaling pathways and up-regulation of MMP-2/TIMP-1 ratio. J. Ethnopharmacol. 2020, 246, 111768. [Google Scholar] [CrossRef] [PubMed]
  35. Pivari, F.; Mingione, A.; Brasacchio, C.; Soldati, L. Curcumin and Type 2 Diabetes Mellitus: Prevention and Treatment. Nutrients 2019, 11, 1837. [Google Scholar] [CrossRef] [PubMed]
  36. Lin, W.; Tu, H.; Zhu, Y.; Guan, Y.; Liu, H.; Ling, W.; Yan, P.; Dong, J. Curcumolide, a unique sesquiterpenoid from Curcuma wenyujin displays anti-angiogenic activity and attenuates ischemia-induced retinal neovascularization. Phytomedicine 2019, 64, 152923. [Google Scholar] [CrossRef] [PubMed]
  37. Abolfazli, S.; Mortazavi, P.; Kheirandish, A.; Butler, A.E.; Jamialahmadi, T.; Sahebkar, A. Regulatory effects of curcumin on nitric oxide signaling in the cardiovascular system. Nitric Oxide 2023, 143, 16–28. [Google Scholar] [CrossRef] [PubMed]
  38. Ma, L.T.; Bai, Y.; Cao, P.; Ren, K.X.; Chen, J.; Zhang, T.; Fan, B.Y.; Qiao, Y.; Yan, H.Y.; Wang, J.J.; et al. The analgesic effects of β-elemene in rats with neuropathic pain by inhibition of spinal astrocytic ERK activation. Mol. Pain. 2022, 18, 17448069221121562. [Google Scholar] [CrossRef]
  39. Limcharoen, T.; Dasuni Wasana, P.W.; Hasriadi Muangnoi, C.; Vajragupta, O.; Rojsitthisak, P.; Towiwat, P. Curcumin Diglutaric Acid, a Prodrug of Curcumin Reduces Pain Hypersensitivity in Chronic Constriction Injury of Sciatic Nerve Induced-Neuropathy in Mice. Pharmaceuticals 2020, 13, 212. [Google Scholar] [CrossRef]
  40. Qi, Y.; Qin, W.; Kang, K.; Jiang, H.; Li, Z.; Wang, Y.; Jia, D. Effects of Wenyujin Essential Oil on tau Protein Phosphorylation in Mice with Aβ-induced Alzheimer Disease through PI3k/Akt Pathway. Chin. J. Inf. Tradit. Chin. Med. 2017, 24, 45–48. [Google Scholar] [CrossRef]
  41. Eun, C.S.; Lim, J.S.; Lee, J.; Lee, S.P.; Yang, S.A. The protective effect of fermented Curcuma longa L. on memory dysfunction in oxidative stress-induced C6 gliomal cells, proinflammatory-activated BV2 microglial cells, and scopolamine-induced amnesia model in mice. BMC Complement. Altern. Med. 2017, 17, 367. [Google Scholar] [CrossRef] [PubMed]
  42. Li, J.; Zeng, H.; You, Y.; Wang, R.; Tan, T.; Wang, W.; Yin, L.; Zeng, Z.; Zeng, Y.; Xie, T. Active targeting of orthotopic glioma using biomimetic liposomes co-loaded elemene and cabazitaxel modified by transferritin. J. Nanobiotechnol. 2021, 19, 289. [Google Scholar] [CrossRef] [PubMed]
  43. Zhong, Z.F.; Tan, W.; Tian, K.; Yu, H.; Qiang, W.A.; Wang, Y.T. Combined effects of furanodiene and doxorubicin on the migration and invasion of MDA-MB-231 breast cancer cells in vitro. Oncol. Rep. 2017, 37, 2016–2024. [Google Scholar] [CrossRef] [PubMed]
  44. Wu, J.; Tang, X.; Shi, Y.; Ma, C.; Zhang, H.; Zhang, J.; Lu, Y.; Wei, J.; Li, L.; Han, L. Crosstalk of LncRNA HOTAIR and SP1-mediated repression of PDK1 contributes to β-Elemene-inhibited proliferation of hepatocellular carcinoma cells. J. Ethnopharmacol. 2022, 283, 114456. [Google Scholar] [CrossRef] [PubMed]
  45. Deng, M.; Liu, B.; Song, H.; Yu, R.; Zou, D.; Chen, Y.; Ma, Y.; Lv, F.; Xu, L.; Zhang, Z.; et al. β-Elemene inhibits the metastasis of multidrug-resistant gastric cancer cells through miR-1323/Cbl-b/EGFR pathway. Phytomedicine 2020, 69, 153184. [Google Scholar] [CrossRef] [PubMed]
  46. Sun, R.; Gu, J.; Chang, X.; Liu, F.; Liang, Y.; Yang, X.; Liang, L.; Tang, D. Metabonomics study on orthotopic transplantion mice model of colon cancer treated with Astragalus membranaceus-Curcuma wenyujin in different proportions via UPLC-Q-TOF/MS. J. Pharm. Biomed. Anal. 2021, 193, 113708. [Google Scholar] [CrossRef] [PubMed]
  47. Ying, J.; Yang, W.; Xie, C.Y.; Ni, Q.C.; Pan, X.D.; Dong, J.H.; Liu, Z.M.; Wang, X.S. Induction of caspase-3-dependent apoptosis in human leukemia HL-60 cells by δ-elemene. Yakugaku Zasshi 2011, 131, 1383–1394. [Google Scholar] [CrossRef] [PubMed]
  48. Lim, C.B.; Ky, N.; Ng, H.M.; Hamza, M.S.; Zhao, Y. Curcuma wenyujin extract induces apoptosis and inhibits proliferation of human cervical cancer cells in vitro and in vivo. Integr. Cancer Ther. 2010, 9, 36–49. [Google Scholar] [CrossRef]
  49. Song, G.; Lu, H.; Chen, F.; Wang, Y.; Fan, W.; Shao, W.; Lu, H.; Lin, B. Tetrahydrocurcumin-induced autophagy via suppression of PI3K/Akt/mTOR in non-small cell lung carcinoma cells. Mol. Med. Rep. 2018, 17, 5964–5969. [Google Scholar] [CrossRef]
  50. Abdel-Lateef, E.; Mahmoud, F.; Hammam, O.; El-Ahwany, E.; El-Wakil, E.; Kandil, S.; Abu Taleb, H.; El-Sayed, M.; Hassenein, H. Bioactive chemical constituents of Curcuma longa L. rhizomes extract inhibit the growth of human hepatoma cell line (HepG2). Acta Pharm. 2016, 66, 387–398. [Google Scholar] [CrossRef]
  51. Lee, Y.H.; Song, N.Y.; Suh, J.; Kim, D.H.; Kim, W.; Ann, J.; Lee, J.; Baek, J.H.; Na, H.K.; Surh, Y.J. Curcumin suppresses oncogenicity of human colon cancer cells by covalently modifying the cysteine 67 residue of SIRT1. Cancer Lett. 2018, 431, 219–229. [Google Scholar] [CrossRef]
  52. Jiang, J.L.; Jin, X.L.; Zhang, H.; Su, X.; Qiao, B.; Yuan, Y.J. Identification of antitumor constituents in curcuminoids from Curcuma longa L. based on the composition-activity relationship. J. Pharm. Biomed. Anal. 2012, 70, 664–670. [Google Scholar] [CrossRef]
  53. Zhang, L.; Cheng, X.; Gao, Y.; Zhang, C.; Bao, J.; Guan, H.; Yu, H.; Lu, R.; Xu, Q.; Sun, Y. Curcumin inhibits metastasis in human papillary thyroid carcinoma BCPAP cells via down-regulation of the TGF-β/Smad2/3 signaling pathway. Exp. Cell Res. 2016, 341, 157–165. [Google Scholar] [CrossRef]
  54. Kuo, C.L.; Wu, S.Y.; Ip, S.W.; Wu, P.P.; Yu, C.S.; Yang, J.S.; Chen, P.Y.; Wu, S.H.; Chung, J.G. Apoptotic death in curcumin-treated NPC-TW 076 human nasopharyngeal carcinoma cells is mediated through the ROS, mitochondrial depolarization and caspase-3-dependent signaling responses. Int. J. Oncol. 2011, 39, 319–328. [Google Scholar] [CrossRef]
  55. Lin, C.Y.; Hung, C.C.; Wang, C.C.N.; Lin, H.Y.; Huang, S.H.; Sheu, M.J. Demethoxycurcumin sensitizes the response of non-small cell lung cancer to cisplatin through downregulation of TP and ERCC1-related pathways. Phytomedicine 2019, 53, 28–36. [Google Scholar] [CrossRef]
  56. Emami, B.; Shakeri, F.; Ghorani, V.; Boskabady, M.H. Relaxant effect of Curcuma longa on rat tracheal smooth muscle and its possible mechanisms. Pharm. Biol. 2017, 55, 2248–2258. [Google Scholar] [CrossRef]
  57. Bundy, R.; Walker, A.F.; Middleton, R.W.; Booth, J. Turmeric extract may improve irritable bowel syndrome symptomology in otherwise healthy adults: A pilot study. J. Altern. Complement. Med. 2004, 10, 1015–1018. [Google Scholar] [CrossRef] [PubMed]
  58. Kim, D.C.; Ku, S.K.; Bae, J.S. Anticoagulant activities of curcumin and its derivative. BMB Rep. 2012, 45, 221–226. [Google Scholar] [CrossRef]
  59. Feng, M.; Liao, T.; Yang, N.; Fang, Y.; Wang, P.; Liu, Z. LC-MS Combined with Network Pharmacology to Explore the Mechanism of “Wenyujin Rhizoma Concisum-Angelicae Sinensis Radix” Essential Oil in the Treatment of Knee Osteoarthritis. Chin. J. Mod. Appl. Pharm. 2023, 40, 2403–2413. [Google Scholar] [CrossRef]
  60. Wang, K.; Shi, X.; Ke, J.; Ma, Y.; Ye, M. Mechanism of action of Rhizoma Acori graminei-Curcuma aromatic in treatment of Alzheimer’s disease: A study based on network pharmacology. Hum. J. Tradit. Chin. Med. 2023, 39, 163–170. [Google Scholar] [CrossRef]
  61. Yu, J.; Li, Z.; He, Z.; Lu, C. Meta-analysis of Efficacy and Safety of Yinxieling Tablet and Its Optimized Formula in the Treatment of Psoriasis Vulgaris. Tradit. Chin. Drug Res. Clin. Pharmacol. 2021, 32, 1048–1054. [Google Scholar] [CrossRef]
  62. Liu, C.; Dong, Y. Clinical study on the vaginal flora of acrobic vaginitis and the effects of the treatments. Prog. Obstet. Gynecol. 2009, 18, 832–835. [Google Scholar] [CrossRef]
  63. Zhang, S.; Peng, Q.; Zhao, X.; Xie, M.; Guo, P. Combined Sanleng Ezhu Decoction with Western medicine in treating liver fibrosis in chronic hepatitis B of liver spleen deficiency and blood stasis syndrome. Hebei J. TCM. 2022, 44, 1834–1838. [Google Scholar] [CrossRef]
  64. Pamplona, C.R.; Souza, M.M.d.; Machado, M.d.S.; Filhoa, V.C.; Navarro, D.; Yunes, R.A.; Monache, F.D.; Niero, R. Seasonal Variation and Analgesic Properties of Different Parts from Curcuma zedoaria Roscoe (Zingiberaceae) Grown in Brazil. Z. Naturforsch. C. J. Biosci. 2006, 61, 6–10. [Google Scholar] [CrossRef]
  65. De Fátima Navarro, D.; De Souza, M.M.; Neto, R.A.; Golin, V.; Niero, R.; Yunes, R.A.; Monache, F.D.; Cechinel-Filho, V. Phytochemical analysis and analgesic properties of Curcuma zedoaria grown in Brazil. Phytomedicine 2002, 9, 427–432. [Google Scholar] [CrossRef] [PubMed]
  66. Qin, L.; Bai, M. Preparation and application of Yu Jin powder. J. Chin. Med. Mater. 1994, 7, 46. [Google Scholar] [CrossRef]
  67. Fei, C.; Ji, D.; Tong, H.; Li, Y.; Su, L.; Qin, Y.; Bian, Z.; Zhang, W.; Mao, C.; Li, L.; et al. Therapeutic mechanism of Curcuma aromatica Salisb. rhizome against coronary heart disease based on integrated network pharmacology, pharmacological evaluation and lipidomics. Front. Pharmacol. 2022, 13, 950749. [Google Scholar] [CrossRef]
  68. Li, J. Clinical observation on stable angina of coronary heart disease treated with Sanleng Ezhu decoction. Tianjin J. Tradit. Chin. Med. 2007, 24, 470–471. [Google Scholar]
  69. Wang, S.; Ma, Q.; Xie, Z.; Shen, Y.; Zheng, B.; Jiang, C.; Yuan, P.; An, Q.; Fan, S.; Jie, Z. An Antioxidant Sesquiterpene Inhibits Osteoclastogenesis Via Blocking IPMK/TRAF6 and Counteracts OVX-Induced Osteoporosis in Mice. J. Bone. Miner. Res. 2021, 36, 1850–1865. [Google Scholar] [CrossRef]
  70. Lu, J.; Li, Z.; Du, G.; Zhang, J.; Pan, F.; Zhang, Z.; Wang, Y.; Shi, J.; Lian, Y.; Zhan, H. Systematic review and Meta-analysis of Gusongbao preparation in treatment of primary osteoporosis. China J. Chin. Mater. Medica 2023, 48, 3086–3096. [Google Scholar] [CrossRef]
  71. Han, H.; Wang, L.; Liu, Y.; Shi, X.; Zhang, X.; Li, M.; Wang, T. Combination of Curcuma zedoary and kelp inhibits growth and metastasis of liver cancerin vivoandin vitro via reducing endogenous H2S levels. Food. Funct. 2019, 10, 224–234. [Google Scholar] [CrossRef]
  72. Wang, G. Curative Effect and Clinical Evaluation of the Anticancer of Compound Zedoary Turmeric Oil Hepatic Artery Embolism in Treatment of Liver Cancer. China Foreign Med. Treat. 2016, 35, 110–111. [Google Scholar] [CrossRef]
  73. Mao, Z.; Zhong, L.; Zhuang, X.; Liu, H.; Peng, Y.; Zhang, H. Curcumenol Targeting YWHAG Inhibits the Pentose Phosphate Pathway and Enhances Antitumor Effects of Cisplatin. Evid. Based. Complement. Alternat. Med. 2022, 2022, 3988916. [Google Scholar] [CrossRef] [PubMed]
  74. Ruan, Q.; Zhong, G. Effect analysis of Curcuma zedoary oil combined with conventional chemotherapy on ovarian cancer. J. Med. Theory Pract. 2018, 31, 554–555. [Google Scholar] [CrossRef]
  75. Li, P.; Ke, H. Effect Analysis of Zedoary Turmeric Oil Combined with Cisplatin and Docetaxel in Treatment of Advanced Non-Small Cell Lung Cancer. Chin. Community Dr. 2023, 39, 77–79. [Google Scholar]
  76. Gao, D.; Zhang, J.; Li, H. Effects of Chanpi Ezhu Decoction on Adjuvant Chemotherapy in the Treatment of Gastric Cancer. World J. Integr. Tradit. West. Med. 2021, 16, 2329–2333. [Google Scholar] [CrossRef]
  77. Zeng, X.; Cui, J.; Zhou, Q.; Li, N.; Liu, J.; Li, J.; Huang, D.; Cai, P.; Zhou, R.; Yan, J.; et al. Research status of Jinrong granule against breast hyperplasia and breast cancer and its clinical application. Chin. J. Clin. Pharmacol. 2023, 39, 295–299. [Google Scholar] [CrossRef]
  78. Lobo, R.; Prabhu, K.S.; Shirwaikar, A.; Shirwaikar, A. Curcuma zedoaria Rosc. (white turmeric): A review of its chemical, pharmacological and ethnomedicinal properties. J. Pharm. Pharmacol. 2009, 61, 13–21. [Google Scholar] [CrossRef]
  79. Etoh, H.; Kondoh, T.; Yoshioka, N.; Sugiyama, K.; Ishikawa, H.; Tanaka, H. 9-Oxo-neoprocurcumenol from Curcuma aromatica (Zingiberaceae) as an Attachment Inhibitor against the Blue Mussel, Mytilus edulis galloprovincialis. Biosci. Biotechnol. Biochem. 2003, 67, 911–913. [Google Scholar] [CrossRef]
  80. Yoshioka, T.; Fujii, E.; Endo, M.; Wada, K.; Tokunaga, Y.; Shiba, N.; Hohsho, H.; Shibuya, H.; Muraki, T. Antiinflammatory potency of dehydrocurdione, a zedoary-derived sesquiterpene. Inflamm. Res. 1998, 47, 476–481. [Google Scholar] [CrossRef] [PubMed]
  81. Shunying, Z.; Yang, Y.; Huaidong, Y.; Yue, Y.; Guolin, Z. Chemical composition and antimicrobial activity of the essential oils of Chrysanthemum indicum. J. Ethnopharmacol. 2005, 96, 151–158. [Google Scholar] [CrossRef] [PubMed]
  82. Padalia, R.C.; Chanotiya, C.S.; Thakuri, B.C.; Mathela, C.S. Germacranolide Rich Essential Oil from Neolitsea pallens. Nat. Prod. Commun. 2006, 2, 291–293. [Google Scholar] [CrossRef]
  83. Kharkwala, G.C.; Pandea, C.; Tewari, G.; Panwarb, A.; Pandeb, V. Terpenoid composition and antimicrobial activity of essential oil from Torilis japonica (Houtt.) DC. J. India. Chem. Soc. 2017, 94, 191–194. [Google Scholar]
  84. Zhang, L.; Shen, S.; Gao, Y.; Shi, S.; Zhou, C.; Mo, J.; Xu, Y.; Lin, L.; Gan, L. Tautomerism and bioactivities of curcumenol, a common sesquiterpenoid widely existing in edible plants. Food. Funct. 2019, 10, 1288–1294. [Google Scholar] [CrossRef] [PubMed]
  85. Indrayanto, G. The importance of method validation in herbal drug research. J. Pharm. Biomed. Anal. 2022, 214, 114735. [Google Scholar] [CrossRef]
  86. Ali, S.M.; Khan, N.A.; Sagathevan, K.; Anwar, A.; Siddiqui, R. Biologically active metabolite(s) from haemolymph of red-headed centipede Scolopendra subspinipes possess broad spectrum antibacterial activity. AMB. Express. 2019, 9, 95. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, L.; Wang, X.; Liu, X.; Wang, Y.; Ren, X.; Dong, Y.; Song, R.; Ma, J.; Fan, Q.; Wei, J.; et al. Fast discrimination and quantification analysis of Curcumae Radix from four botanical origins using NIR spectroscopy coupled with chemometrics tools. Spectrochim. Acta. A Mol. Biomol. Spectrosc. 2021, 254, 119626. [Google Scholar] [CrossRef] [PubMed]
  88. Komatsu, K.; Sasaki, Y.; Tanaka, K.; Kuba, Y.; Fushimi, H.; Cai, S.-Q. Morphological, genetic, and chemical polymorphism of Curcuma kwangsiensis. J. Nat. Med. 2008, 62, 413–422. [Google Scholar] [CrossRef]
  89. El-Hawaz, R.F.; Grace, M.H.; Janbey, A.; Lila, M.A.; Adelberg, J.W. In vitro mineral nutrition of Curcuma longa L. affects production of volatile compounds in rhizomes after transfer to the greenhouse. BMC. Plant. Biol. 2018, 18, 122. [Google Scholar] [CrossRef]
  90. Yang, F.Q.; Wang, Y.T.; Li, S.P. Simultaneous determination of 11 characteristic components in three species of Curcuma rhizomes using pressurized liquid extraction and high-performance liquid chromatography. J. Chromatogr. A 2006, 1134, 226–231. [Google Scholar] [CrossRef]
  91. Chen, S. Determination of Three Effective Constituents in Rhizoma Curcumae from Different Area by HPLC. Chin. Pharm. J. 2009, 44, 1742–1744. [Google Scholar]
  92. Shi, D.; Su, B.; Zhang, J.; Dai, Y. Study on Analgesic Effect of 4 Sources of Curcumae Radix Decoction Pieces and Comparison of Curcuminol Content in Its Water Extracts. Chin. Pharm. 2020, 31, 2209–2213. [Google Scholar] [CrossRef]
  93. Wang, M.; Yu, M.; Peng, M.; Yin, Z.; Mao, C.; Su, L.; Ji, D.; Lu, T. Quality evaluation of Curcumae Radix from different origins based on UPLC characteristic chromatogram, multicomponent content, and chemometrics. China J. Chin. Mater. Med. 2022, 47, 2964–2974. [Google Scholar] [CrossRef]
  94. Yang, F.Q.; Li, S.P.; Chen, Y.; Lao, S.C.; Wang, Y.T.; Dong, T.T.X.; Tsim, K.W.K. Identification and quantitation of eleven sesquiterpenes in three species of Curcuma rhizomes by pressurized liquid extraction and gas chromatography–mass spectrometry. J. Pharm. Biomed. Anal. 2005, 39, 552–558. [Google Scholar] [CrossRef]
  95. Yin, G.; Cheng, X.; Tao, W.; Dong, Y.; Bian, Y.; Zang, W.; Tang, D. Comparative analysis of multiple representative components in the herb pair Astragali Radix-Curcumae Rhizoma and its single herbs by UPLC-QQQ-MS. J. Pharm. Biomed. Anal. 2018, 148, 224–229. [Google Scholar] [CrossRef] [PubMed]
  96. Zang, Y.; Xu, L.; Liu, H.; Luan, Y.; Xu, X.; Yu, Y.; Lin, Y. Determination of 8 Main Active Compounds in Curcumae Rhizoma by HPLC Wavelength Switching Method. Chin. J. Mod. Appl. Pharm. 2021, 38, 2227–2233. [Google Scholar] [CrossRef]
  97. Yang, F.Q.; Li, S.P.; Zhao, J.; Lao, S.C.; Wang, Y.T. Optimization of GC–MS conditions based on resolution and stability of analytes for simultaneous determination of nine sesquiterpenoids in three species of Curcuma rhizomes. J. Pharm. Biomed. Anal. 2007, 43, 73–82. [Google Scholar] [CrossRef] [PubMed]
  98. Shiea, J.; Lin, H.; Bhat, S.M.; Lee, C.; Huang, M.; Ponnusamy, V.K.; Cheng, S. Thin layer chromatography/desorption flame-induced atmospheric pressure chemical ionization/mass spectrometry for the analysis of volatile and semi-volatile mixtures. Rapid. Commun. Mass. Spectrom. 2022, 36, e9409. [Google Scholar] [CrossRef] [PubMed]
  99. Ayoub, I.M.; Abdel-Aziz, M.M.; Elhady, S.S.; Bagalagel, A.A.; Malatani, R.T.; Elkady, W.M. Valorization of Pimenta racemosa Essential Oils and Extracts: GC-MS and LC-MS Phytochemical Profiling and Evaluation of Helicobacter pylori Inhibitory Activity. Molecules 2022, 27, 7965. [Google Scholar] [CrossRef] [PubMed]
  100. Depmeier, T.; Lange, T.; Hanekamp, W.; Strünker, T.; Lehr, M. HPLC fluorescence assay for measuring the activity of diacylglycerol lipases and the action of inhibitors thereof. Anal. Biochem. 2022, 657, 114889. [Google Scholar] [CrossRef] [PubMed]
  101. Sammut Bartolo, N.; Vella Szijj, J.; Ferrito, V.; Serracino-Inglott, A. HPLC-UV Method Development and Validation to Monitor Difluprednate Synthesis. J. Chromatogr. Sci. 2023, 61, 322–328. [Google Scholar] [CrossRef]
  102. Gajula, S.N.R.; Khairnar, A.S.; Jock, P.; Kumari, N.; Pratima, K.; Munjal, V.; Kalan, P.; Sonti, R. LC-MS/MS: A sensitive and selective analytical technique to detect COVID-19 protein biomarkers in the early disease stage. Expert. Rev. Proteom. 2023, 20, 5–18. [Google Scholar] [CrossRef]
  103. Liang, Z.; Liang, L.; Zeng, Z.; Wei, M.; Lei, Y.; Lin, W. Determination of curcumenol content in Curcuma kwangsiensis, Vingar-Boiled Curcuma kwangsiensis, and Water Residues by UPLC. Chem. Bio Eng. 2020, 37, 62–65. [Google Scholar] [CrossRef]
  104. Zhou, X.; Chen, G.; Zhao, C.; Gong, X.; Li, M. Fingerprint of Curcuma phaeocaulis by LC-MS. China J. Chin. Mater. Med. 2008, 33, 2218–2221. [Google Scholar]
  105. Yang, F.Q.; Wang, H.K.; Chen, H.; Chen, J.D.; Xia, Z.N. Fractionation of Volatile Constituents from Curcuma Rhizome by Preparative Gas Chromatography. J. Autom. Methods Manag. Chem. 2011, 2011, 942467. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, L.; Liu, Y.; Huang, X.; Zhu, Y.; Li, J.; Miao, Y.; Du, H.; Liu, D. Comparison of Chemical Constituents and Pharmacological Effects of Different Varieties of Chrysanthemum Flos in China. Chem. Biodivers. 2021, 18, e2100206. [Google Scholar] [CrossRef] [PubMed]
  107. Çakmakçı, R.; Mosber, G.; Milton, A.H.; Alatürk, F.; Ali, B. The Effect of Auxin and Auxin-Producing Bacteria on the Growth, Essential Oil Yield, and Composition in Medicinal and Aromatic Plants. Curr. Microbiol. 2020, 77, 564–577. [Google Scholar] [CrossRef] [PubMed]
  108. Saffariha, M.; Azarnivand, H.; Zare Chahouki, M.A.; Tavili, A.; Nejad Ebrahimi, S.; Jahani, R.; Potter, D. Changes in the essential oil content and composition of Salvia limbata C.A. Mey at different growth stages and altitudes. Biomed. Chromatogr. 2021, 35, e5127. [Google Scholar] [CrossRef] [PubMed]
  109. Gu, S.; Li, L.; Huang, H.; Wang, B.; Zhang, T. Antitumor, Antiviral, and Anti-Inflammatory Efficacy of Essential Oils from Atractylodes macrocephala Koidz. Produced with Different Processing Methods. Molecules 2019, 24, 2956. [Google Scholar] [CrossRef]
  110. Zhen, F.; Sun, X.; Jin, X. Phenological period and quality comparison of different producing areas of Wenyujin in southern Zhejiang. J. Zhejiang Agr. Sci. 2019, 60, 823–824. [Google Scholar] [CrossRef]
  111. Yu, Y.; Shen, Q.; Lai, Y.; Park, S.Y.; Ou, X.; Lin, D.; Jin, M.; Zhang, W. Anti-inflammatory Effects of Curcumin in Microglial Cells. Front. Pharmacol. 2018, 9, 386. [Google Scholar] [CrossRef]
  112. Cho, J.W.; Lee, K.S.; Kim, C.W. Curcumin attenuates the expression of IL-1beta, IL-6, and TNF-alpha as well as cyclin E in TNF-alpha-treated HaCaT cells; NF-kappaB and MAPKs as potential upstream targets. Int. J. Mol. Med. 2007, 19, 469–474. [Google Scholar]
  113. Sengupta, S.; Tripathi, A. Evaluation of analgesic and prophylactic activity of curcumin against chikungunya-infected acute/chronic arthralgic mice. J. Med. Virol. 2023, 95, e28661. [Google Scholar] [CrossRef]
  114. Mahakunakorn, P.; Tohda, M.; Murakami, Y.; Matsumoto, K.; Watanabe, H.; Vajaragupta, O. Cytoprotective and cytotoxic effects of curcumin: Dual action on H2O2-induced oxidative cell damage in NG108-15 cells. Biol. Pharm. Bull. 2003, 26, 725–728. [Google Scholar] [CrossRef]
  115. Marton, L.T.; Pescinini, E.S.L.M.; Camargo, M.E.C.; Barbalho, S.M.; Haber, J.; Sinatora, R.V.; Detregiachi, C.R.P.; Girio, R.J.S.; Buchaim, D.V.; Cincotto Dos Santos Bueno, P. The Effects of Curcumin on Diabetes Mellitus: A Systematic Review. Front. Endocrinol. 2021, 12, 669448. [Google Scholar] [CrossRef]
  116. Fan, D.; Lu, J.; Yu, N.; Xie, Y.; Zhen, L. Curcumin Prevents Diabetic Osteoporosis through Promoting Osteogenesis and Angiogenesis Coupling via NF-κB Signaling. Evid. Based Complement. Altern. Med. 2022, 2022, 4974343. [Google Scholar] [CrossRef]
  117. Dave, G.S.; Rakholiya, K.D.; Kaneria, M.J.; Galvadiya, B.P.; Vyas, S.R.; Kanbi, V.H.; Patel, M.P. High affinity interaction of Solanum tuberosum and Brassica juncea residue smoke water compounds with proteins involved in coronavirus infection. Phytother. Res. 2020, 34, 3400–3410. [Google Scholar] [CrossRef] [PubMed]
  118. Marín-Palma, D.; Tabares-Guevara, J.H.; Zapata-Cardona, M.I.; Flórez-Álvarez, L.; Yepes, L.M.; Rugeles, M.T.; Zapata-Builes, W.; Hernandez, J.C.; Taborda, N.A. Curcumin Inhibits In Vitro SARS-CoV-2 Infection In Vero E6 Cells through Multiple Antiviral Mechanisms. Molecules 2021, 26, 6900. [Google Scholar] [CrossRef] [PubMed]
  119. Liu, J.L.; Pan, Y.Y.; Chen, O.; Luan, Y.; Xue, X.; Zhao, J.J.; Liu, L.; Jia, H.Y. Curcumin inhibits MCF-7 cells by modulating the NF-κB signaling pathway. Oncol. Lett. 2017, 14, 5581–5584. [Google Scholar] [CrossRef] [PubMed]
  120. Zheng, R.; Deng, Q.; Liu, Y.; Zhao, P. Curcumin Inhibits Gastric Carcinoma Cell Growth and Induces Apoptosis by Suppressing the Wnt/β-Catenin Signaling Pathway. Med. Sci. Monit. 2017, 23, 163–171. [Google Scholar] [CrossRef]
  121. Ye, M.; Zhang, J.; Zhang, J.; Miao, Q.; Yao, L.; Zhang, J. Curcumin promotes apoptosis by activating the p53-miR-192-5p/215-XIAP pathway in non-small cell lung cancer. Cancer Lett. 2015, 357, 196–205. [Google Scholar] [CrossRef]
  122. Kim, B.; Kim, H.S.; Jung, E.J.; Lee, J.Y.; Tsang, B.; Lim, J.M.; Song, Y.S. Curcumin induces ER stress-mediated apoptosis through selective generation of reactive oxygen species in cervical cancer cells. Mol. Carcinog. 2016, 55, 918–928. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, D.; Fang, Z.; Zhang, Y.; Cao, Y.; Yang, L.; Yin, J. Inhibitory effects of curcumenol on human liver cytochrome P450 enzymes. Phytother. Res. 2010, 24, 1213–1216. [Google Scholar] [CrossRef]
  124. Shamsi, S.; Tran, H.; Tan, R.S.; Tan, Z.J.; Lim, L.Y. Curcumin, Piperine, and Capsaicin: A Comparative Study of Spice-Mediated Inhibition of Human Cytochrome P450 Isozyme Activities. Drug Metab. Dispos. 2017, 45, 49–55. [Google Scholar] [CrossRef]
  125. Hamdi, O.; Feroz, S.; Shilpi, J.; Anouar, E.; Mukarram, A.; Mohamad, S.; Tayyab, S.; Awang, K. Spectrofluorometric and Molecular Docking Studies on the Binding of Curcumenol and Curcumenone to Human Serum Albumin. Int. J. Mol. Sci. 2015, 16, 5180–5193. [Google Scholar] [CrossRef]
  126. Dezhampanah, H.; Shabanzade, Z. Investigation of binding interaction between human serum albumin with zirconium complex of curcumin and curcumin. J. Biomol. Struct. Dyn. 2022, 40, 722–732. [Google Scholar] [CrossRef] [PubMed]
  127. Roe, K. An inflammation classification system using cytokine parameters. Scand. J. Immunol. 2020, 93, e12970. [Google Scholar] [CrossRef]
  128. Krstanović, F.; Britt, W.J.; Jonjić, S.; Brizić, I. Cytomegalovirus Infection and Inflammation in Developing Brain. Viruses 2021, 13, 1078. [Google Scholar] [CrossRef] [PubMed]
  129. Pollard, K.M.; Cauvi, D.M.; Toomey, C.B.; Hultman, P.; Kono, D.H. Mercury-induced inflammation and autoimmunity. Biochim. Biophys. Acta. Gen. Subj. 2019, 1863, 129299. [Google Scholar] [CrossRef]
  130. Hong, Y.-K.; Chang, Y.-H.; Lin, Y.-C.; Chen, B.; Guevara, B.E.K.; Hsu, C.-K. Inflammation in Wound Healing and Pathological Scarring. Adv. Wound. Care 2023, 12, 288–300. [Google Scholar] [CrossRef]
  131. Nedunchezhiyan, U.; Varughese, I.; Sun, A.R.; Wu, X.; Crawford, R.; Prasadam, I. Obesity, Inflammation, and Immune System in Osteoarthritis. Front. Immunol. 2022, 13, 907750. [Google Scholar] [CrossRef]
  132. Motta, F.; Barone, E.; Sica, A.; Selmi, C. Inflammaging and Osteoarthritis. Clin. Rev. Allergy. Immunol. 2022, 64, 222–238. [Google Scholar] [CrossRef]
  133. Peoples, J.N.; Saraf, A.; Ghazal, N.; Pham, T.T.; Kwong, J.Q. Mitochondrial dysfunction and oxidative stress in heart disease. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [PubMed]
  134. Dionísio, P.A.; Amaral, J.D.; Rodrigues, C.M.P. Oxidative stress and regulated cell death in Parkinson’s disease. Ageing. Res. Rev. 2021, 67, 101263. [Google Scholar] [CrossRef] [PubMed]
  135. Nebrisi, E.E. Neuroprotective Activities of Curcumin in Parkinson’s Disease: A Review of the Literature. Int. J. Mol. Sci. 2021, 22, 11248. [Google Scholar] [CrossRef] [PubMed]
  136. El-Akabawy, G.; El-Kholy, W. Neuroprotective effect of ginger in the brain of streptozotocin-induced diabetic rats. Ann. Anat. 2014, 196, 119–128. [Google Scholar] [CrossRef] [PubMed]
  137. Singh, S.K.; Srivastav, S.; Castellani, R.J.; Plascencia-Villa, G.; Perry, G. Neuroprotective and Antioxidant Effect of Ginkgo biloba Extract Against AD and Other Neurological Disorders. Neurotherapeutics 2019, 16, 666–674. [Google Scholar] [CrossRef]
  138. Yulug, B.; Kilic, E.; Altunay, S.; Ersavas, C.; Orhan, C.; Dalay, A.; Tuzcu, M.; Sahin, N.; Juturu, V.; Sahin, K. Cinnamon Polyphenol Extract Exerts Neuroprotective Activity in Traumatic Brain Injury in Male Mice. CNS. Neurol. Disord. Drug Targets 2018, 17, 439–447. [Google Scholar] [CrossRef] [PubMed]
  139. Bian, Y.; Zhao, C.; Lee, S.M.-Y. Neuroprotective Potency of Saffron Against Neuropsychiatric Diseases, Neurodegenerative Diseases, and Other Brain Disorders: From Bench to Bedside. Front. Pharmacol. 2020, 11, 579052. [Google Scholar] [CrossRef]
  140. World Health Organization. The Top 10 Causes of Death. Available online: https://www.who.int/newsroom/fact-sheets/detail/the-top-10-causes-of-death (accessed on 1 December 2023).
  141. Tsai, A.G.; Bessesen, D.H. Obesity. Ann. Intern. Med. 2019, 170, ITC33–ITC48. [Google Scholar] [CrossRef]
  142. Shiriki Kumanyika, W.H.D. Solving Population-wide Obesity-Progress and Future Prospects. N. Engl. J. Med. 2020, 383, 2197–2200. [Google Scholar] [CrossRef]
  143. Blüher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef]
  144. Aspray, T.J.; Hill, T.R. Osteoporosis and the Ageing Skeleton. In Biochemistry and Cell Biology of Ageing: Part II Clinical Science; Subcellular Biochemistry; Springer: New York, NY, USA, 2019; pp. 453–476. [Google Scholar]
  145. Arceo-Mendoza, R.M.; Camacho, P.M. Postmenopausal Osteoporosis. Endocrinol. Metab. Clin. N. Am. 2021, 50, 167–178. [Google Scholar] [CrossRef] [PubMed]
  146. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef] [PubMed]
  147. Han, X.H.; Ye, Y.Y.; Guo, B.F.; Liu, S. Effects of platycodin D in combination with different active ingredients of Chinese herbs on proliferation and invasion of 4T1 and MDA-MB-231 breast cancer cell lines. Chin. J. Integr. Med. 2012, 10, 67–75. [Google Scholar] [CrossRef] [PubMed]
  148. Al-Amin, M.; Eltayeb, N.M.; Khairuddean, M.; Salhimi, S.M. Bioactive chemical constituents from Curcuma caesia Roxb. rhizomes and inhibitory effect of curcuzederone on the migration of triple-negative breast cancer cell line MDA-MB-231. Nat. Prod. Res. 2019, 35, 3166–3170. [Google Scholar] [CrossRef]
  149. Jung, E.B.; Trinh, T.A.; Lee, T.K.; Yamabe, N.; Kang, K.S.; Song, J.H.; Choi, S.; Lee, S.; Jang, T.S.; Kim, K.H.; et al. Curcuzedoalide contributes to the cytotoxicity of Curcuma zedoaria rhizomes against human gastric cancer AGS cells through induction of apoptosis. J. Ethnopharmacol. 2018, 213, 48–55. [Google Scholar] [CrossRef]
  150. Zhang, R.; Wang, B.; Zhao, H.; Li, X.; Wei, C.; Wei, R.G. Determ ination of curcumol in plasma by HPLC-MS/MS method and its pharmacokinetics in Beagle dogs. Acta Pharm. Sin. B. 2007, 42, 973–977. [Google Scholar] [CrossRef]
  151. Center for Disease Control. Breast Cancer Statistics. Available online: https://www.cdc.gov/cancer/breast/statistics/index.htm (accessed on 1 December 2023).
  152. American Cancer Society. Key Statistics for Breast Cancer. Available online: https://www.cancer.org/cancer/breast-cancer/about/how-common-is-breast-cancer.html (accessed on 1 December 2023).
  153. Siddiqui, J.A.; Singh, A.; Chagtoo, M.; Singh, N.; Godbole, M.M.; Chakravarti, B. Phytochemicals for breast cancer therapy: Current status and future implications. Curr. Cancer. Drug Targets 2015, 15, 116–135. [Google Scholar] [CrossRef]
  154. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef]
  155. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  156. Prager, G.W.; Taieb, J.; Fakih, M.; Ciardiello, F.; Van Cutsem, E.; Elez, E.; Cruz, F.M.; Wyrwicz, L.; Stroyakovskiy, D.; Pápai, Z.; et al. Trifluridine-Tipiracil and Bevacizumab in Refractory Metastatic Colorectal Cancer. N. Engl. J. Med. 2023, 388, 1657–1667. [Google Scholar] [CrossRef] [PubMed]
  157. Duarte, D.; Rêma, A.; Amorim, I.; Vale, N. Drug Combinations: A New Strategy to Extend Drug Repurposing and Epithelial-Mesenchymal Transition in Breast and Colon Cancer Cells. Biomolecules 2022, 12, 190. [Google Scholar] [CrossRef] [PubMed]
  158. Ducrey, E.; Castrogiovanni, C.; Meraldi, P.; Nowak-Sliwinska, P. Forcing dividing cancer cells to die; low-dose drug combinations to prevent spindle pole clustering. Apoptosis 2021, 26, 248–252. [Google Scholar] [CrossRef] [PubMed]
  159. Nair, N.U.; Greninger, P.; Zhang, X.; Friedman, A.A.; Amzallag, A.; Cortez, E.; Sahu, A.D.; Lee, J.S.; Dastur, A.; Egan, R.K.; et al. A landscape of response to drug combinations in non-small cell lung cancer. Nat. Commun. 2023, 14, 3830. [Google Scholar] [CrossRef]
  160. Yang, F.; Zhang, Y.; Liang, H. Interactive Association of Drugs Binding to Human Serum Albumin. Int. J. Mol. Sci. 2014, 15, 3580–3595. [Google Scholar] [CrossRef]
  161. Ashraf, G.M.; Gupta, D.D.; Alam, M.Z.; Baeesa, S.S.; Alghamdi, B.S.; Anwar, F.; Alqurashi, T.M.A.; Al Abdulmonem, W.; Alyousef, M.A.; Alhumaydhi, F.A.; et al. Unravelling Binding of Human Serum Albumin with Galantamine: Spectroscopic, Calorimetric, and Computational Approaches. ACS. Omega 2022, 7, 34370–34377. [Google Scholar] [CrossRef]
  162. Zhong, G.; Cai, X.; Wei, R.; Wei, S.; Cao, X. Curcumenol improves renal function in 5/6 nephrectomy-induced chronic renal failure rats via the SIRT1/NF-κB pathway. Anat. Rec. 2023, 306, 3189–3198. [Google Scholar] [CrossRef]
Figure 1. Picture of Curcuma wenyujin and the structure of curcumenol.
Figure 1. Picture of Curcuma wenyujin and the structure of curcumenol.
Molecules 29 00656 g001
Figure 2. Diversity of anti-cancer effects. In this figure, we can conclude that curcumenol exerts five anti-cancer effects: suppressing breast tumor cells, digestive tumor cells, liver cancer cells, cervical cancer cells, and lung cancer cells. In cervical cancer, curcumenol can inhibit YWHAG expression in cervical cancer and reduce cervical cancer cell proliferation, invasion, and MMP2 and MMP9 expression when combined with cisplatin; the abovementioned combination of drugs leads to increased apoptosis. In liver cancer, the combination of curcumenol and laminarin can inhibit the proliferation, migration, and invasion of human hepatoma HepG2 cells. In lung cancer, curcumenol can suppress cell proliferation and induce cell death in lung cancer cells. Ferroptosis is the mechanism by which curcumenol induces lung cancer cell death; the lncRNA H19/miR-19b-3p/FTH1 axis was found to be crucial for this process. In gastric cancer, curcumenol has demonstrated the dose-dependent suppression of gastric cancer cell proliferation. In breast cancer, curcumenol showed remarkable anti-proliferative action against MCF-7 cells, which might cause apoptotic cell death. The increasing and decreasing expression of proteins were signed as ↑ and ↓, respectively.
Figure 2. Diversity of anti-cancer effects. In this figure, we can conclude that curcumenol exerts five anti-cancer effects: suppressing breast tumor cells, digestive tumor cells, liver cancer cells, cervical cancer cells, and lung cancer cells. In cervical cancer, curcumenol can inhibit YWHAG expression in cervical cancer and reduce cervical cancer cell proliferation, invasion, and MMP2 and MMP9 expression when combined with cisplatin; the abovementioned combination of drugs leads to increased apoptosis. In liver cancer, the combination of curcumenol and laminarin can inhibit the proliferation, migration, and invasion of human hepatoma HepG2 cells. In lung cancer, curcumenol can suppress cell proliferation and induce cell death in lung cancer cells. Ferroptosis is the mechanism by which curcumenol induces lung cancer cell death; the lncRNA H19/miR-19b-3p/FTH1 axis was found to be crucial for this process. In gastric cancer, curcumenol has demonstrated the dose-dependent suppression of gastric cancer cell proliferation. In breast cancer, curcumenol showed remarkable anti-proliferative action against MCF-7 cells, which might cause apoptotic cell death. The increasing and decreasing expression of proteins were signed as ↑ and ↓, respectively.
Molecules 29 00656 g002
Figure 3. The mechanism of synergistic anti-cancer effects [71,73]. The combination of curcumenol and laminarin could inhibit the proliferation, migration, and invasion of HepG2 cells, and it may inhibit the angiogenesis of hepatoma via H2S and the VEGF pathway, consequently inhibiting the metastasis of HepG2 cells. In cervical cancer, curcumenol could inhibit YWHAG expression and reduce cervical cancer cell proliferation, invasion, and MMP2 and MMP9 expression when combined with cisplatin. The abovementioned combination of drugs led to increased apoptosis, downregulated Bcl-2 expression, and the upregulated expression of Bax, PARP, and caspase-3. The increasing and decreasing trend were signed as ↑ and ↓, respectively.
Figure 3. The mechanism of synergistic anti-cancer effects [71,73]. The combination of curcumenol and laminarin could inhibit the proliferation, migration, and invasion of HepG2 cells, and it may inhibit the angiogenesis of hepatoma via H2S and the VEGF pathway, consequently inhibiting the metastasis of HepG2 cells. In cervical cancer, curcumenol could inhibit YWHAG expression and reduce cervical cancer cell proliferation, invasion, and MMP2 and MMP9 expression when combined with cisplatin. The abovementioned combination of drugs led to increased apoptosis, downregulated Bcl-2 expression, and the upregulated expression of Bax, PARP, and caspase-3. The increasing and decreasing trend were signed as ↑ and ↓, respectively.
Molecules 29 00656 g003
Figure 4. Biological activities of curcumenol. In this figure, we can conclude that the biological activities of curcumenol are very abundant, ranging from pharmacology to pharmacokinetics; in addition to anti-cancer effects, curcumenol exerts anti-inflammatory activity, analgesic effects, anti-oxidant ability, neuroprotective activity against coronary heart disease, anti-diabetic activity, osteoporosis-ameliorating, anti-viral activity, and anti-bacteriostatic effects. The anti-cancer mechanism is detailed in Figure 2. Neuroprotective effects, involving anti-inflammatory and anti-oxidant effects, and NO levels, were decreased. The amelioration of osteoporosis is associated with decreased TRAF expression and decreased osteoclast activity. Curcumenol has also exhibited anti-viral effects in various studies. The anti-bacteriostatic effects mechanism of curcumenol involves damaging the cell membrane. The anti-inflammation effect of curcumenol involves decreases in Akt, Nrf2, p38, and MAPK expression. It has also shown anti-diabetes effects and analgesic effects, but its mechanism does not involve the participation of the opioid system. The mechanism of action against coronary heart disease involves the PI3K/AKT/mTOR pathway. The increasing and decreasing trend were signed as ↑ and ↓, respectively.
Figure 4. Biological activities of curcumenol. In this figure, we can conclude that the biological activities of curcumenol are very abundant, ranging from pharmacology to pharmacokinetics; in addition to anti-cancer effects, curcumenol exerts anti-inflammatory activity, analgesic effects, anti-oxidant ability, neuroprotective activity against coronary heart disease, anti-diabetic activity, osteoporosis-ameliorating, anti-viral activity, and anti-bacteriostatic effects. The anti-cancer mechanism is detailed in Figure 2. Neuroprotective effects, involving anti-inflammatory and anti-oxidant effects, and NO levels, were decreased. The amelioration of osteoporosis is associated with decreased TRAF expression and decreased osteoclast activity. Curcumenol has also exhibited anti-viral effects in various studies. The anti-bacteriostatic effects mechanism of curcumenol involves damaging the cell membrane. The anti-inflammation effect of curcumenol involves decreases in Akt, Nrf2, p38, and MAPK expression. It has also shown anti-diabetes effects and analgesic effects, but its mechanism does not involve the participation of the opioid system. The mechanism of action against coronary heart disease involves the PI3K/AKT/mTOR pathway. The increasing and decreasing trend were signed as ↑ and ↓, respectively.
Molecules 29 00656 g004
Table 5. Content of curcumenol in various seasons and in a wide range of parts of C. zedoaria (mg/100 g dried plant) [64]. Reproduced with permission from Christiane Regina Pamplona, et al., Zeitschrift fur Naturforschung. C, Journal of biosciences; published by Verl. d. Zeitschrift für Naturforschung, 2006.
Table 5. Content of curcumenol in various seasons and in a wide range of parts of C. zedoaria (mg/100 g dried plant) [64]. Reproduced with permission from Christiane Regina Pamplona, et al., Zeitschrift fur Naturforschung. C, Journal of biosciences; published by Verl. d. Zeitschrift für Naturforschung, 2006.
PartAutumnWinterSpringSummer
Roots15.70 ± 0.148.90 ± 0.158.70 ± 0.121.5 ± 0.10
Mother rhizome33.10 ± 0.129.10 ± 0.085.90 ± 0.056.0 ± 0.01
Rugous rhizome10.40 ± 0.033.10 ± 0.032.00 ± 0.042.9 ± 0.03
Table 6. Content of dihydrocurdione in various seasons and in a wide range of parts of C. zedoaria (mg/100 g dried plant) [64]. Reproduced with permission from Christiane Regina Pamplona, et al., Zeitschrift fur Naturforschung. C, Journal of biosciences; published by Verl. d. Zeitschrift für Naturforschung, 2006.
Table 6. Content of dihydrocurdione in various seasons and in a wide range of parts of C. zedoaria (mg/100 g dried plant) [64]. Reproduced with permission from Christiane Regina Pamplona, et al., Zeitschrift fur Naturforschung. C, Journal of biosciences; published by Verl. d. Zeitschrift für Naturforschung, 2006.
PartAutumnWinterSpringSummer
Roots8.50 ± 0.273.10 ± 0.154.40 ± 0.061.60 ± 0.10
Mother rhizome25.00 ± 0.259.40 ± 0.176.10 ± 0.157.40 ± 0.14
Rugous rhizome6.70 ± 0.032.90 ± 0.081.50 ± 0.061.50 ± 0.03
Table 7. Comparison of the content of volatile oil in wenyujin from different producing areas [110].
Table 7. Comparison of the content of volatile oil in wenyujin from different producing areas [110].
Producing AreasContent of Volatile Oils
Taoshan Town, Ruian City, Zhejiang Province, China2.0%
Mayu Town, Ruian City, Zhejiang Province, China1.8%
Siqian Town, Wenzhou City, Zhejiang Province, China1.5%
Yongjia County, Wenzhou City, Zhejiang Province, China3.1%
Leqing City, Zhejiang Province, China2.0%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, J.; Sun, Y.; Li, G.; Cheng, C.; Sui, X.; Wu, Q. The Extraction, Determination, and Bioactivity of Curcumenol: A Comprehensive Review. Molecules 2024, 29, 656. https://doi.org/10.3390/molecules29030656

AMA Style

Li J, Sun Y, Li G, Cheng C, Sui X, Wu Q. The Extraction, Determination, and Bioactivity of Curcumenol: A Comprehensive Review. Molecules. 2024; 29(3):656. https://doi.org/10.3390/molecules29030656

Chicago/Turabian Style

Li, Jie, Yitian Sun, Guohua Li, Chunsong Cheng, Xinbing Sui, and Qibiao Wu. 2024. "The Extraction, Determination, and Bioactivity of Curcumenol: A Comprehensive Review" Molecules 29, no. 3: 656. https://doi.org/10.3390/molecules29030656

Article Metrics

Back to TopTop