Next Article in Journal
Evaluation of the Antibacterial, Anti-Cervical Cancer Capacity, and Biocompatibility of Different Graphene Oxides
Previous Article in Journal
Recent Advances in Chemistry and Antioxidant/Anticancer Biology of Monoterpene and Meroterpenoid Natural Product
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Preliminary Study on Total Component Analysis and In Vitro Antitumor Activity of Eucalyptus Leaf Residues

1
Chenguang Biotech Group Co., Ltd., Handan 057250, China
2
Chenguang Biotech Group HanDan Co., Ltd., Handan 056000, China
3
Hebei Key Laboratory of Comprehensive Utilization of Plant Resources, Handan 057250, China
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(2), 280; https://doi.org/10.3390/molecules29020280
Submission received: 28 November 2023 / Revised: 22 December 2023 / Accepted: 28 December 2023 / Published: 5 January 2024

Abstract

:
Eucalyptus globulus is widely introduced and cultivated in Yunnan province. Its foliage is mainly used to extract eucalyptus oil, but the by-product eucalyptus residue has not been fully utilized. Based on the above reasons, in this study, we sought to explore the comprehensive utilization potential of eucalyptus resources. The total composition of eucalyptus residue was analyzed by ultra performance liquid chromatography-time-of-flight mass spectrometry (UPLC-Q/TOF MS), and the active components and nutrient components of eucalyptus leaf residue were determined by chemical methods and liquid phase techniques. Meanwhile, the antitumor activity of triterpenoids in eucalyptus leaves was evaluated by tetramethylazazole blue colorimetric assay (MTT). The results of qualitative analysis indicated that 55 compounds were identified from eucalyptus residue, including 28 phloroglucinols, 17 terpenoids, 3 flavonoids, 5 fatty acids, 1 amino acid and 2 polyphenols. Among them, the pentacyclic triterpenoids, in eucalyptus residue, were mainly oleanane type and urthane type. The results of quantitative determination indicated that the content of triterpenoid compounds was 2.84% in eucalyptus residue, which could be enhanced to 82% by silicone separation. The antitumor activity results showed that triterpenoid compounds have moderate inhibitory effects on human breast cancer cell MDA-MB-231, gastric adenocarcinoma cell SGC-7901 and cervical cancer cell Hela. The half maximal inhibitory concentration (IC50) was 50.67, 43.12 and 42.65 μg/mL, respectively. In this study, the triterpenoids from eucalyptus leaf residues were analyzed to reveal that the triterpenoids from eucalyptus leaf have antitumor effects and have potential to be developed as antitumor drugs.

1. Introduction

Eucalypt is the general name of the species of genus Angophora, Corymbia, and Eucalyptus of the Myrtaceae family, containing a total of 945 species and varieties that are important economic species naturally distributed in countries such as Australia [1]. Known for their fast growth, high yield, strong adaptability, and wide-ranging applications, eucalyptus trees have been introduced to China for over a century. Currently, more than 600 counties in over 20 provinces across China cultivate eucalyptus plantations [2]. Commonly planted eucalyptus species include Eucalyptus globulus, Eucalyptus citriodora, Eucalyptus grandis, and Eucalyptus urophylla, with major production regions in Guangdong, Guangxi, Yunnan, and Hainan [3].
One significant application of eucalyptus trees is for the production of eucalyptus essential oil. According to statistics, eucalyptus essential oil is a crucial export commodity for China, consistently ranking among the top four in export share from 2016 to 2020. Eucalyptus essential oil is well recognized for its anti-inflammatory, analgesic, anticancer, and antiviral properties, making it a valuable resource in the pharmaceutical industry [4]. Notably, Eucalyptus globulus cultivated in Yunnan has a high content of 1,8-cineole in its eucalyptus essential oil, exceeding 70%, making it a primary source for domestic eucalyptus essential oil production [5]. However, the residue after the extraction of eucalyptus essential oil, namely eucalyptus leaf residue, is not fully utilized. At present, eucalyptus residue is usually burned, which causes serious waste of resources and environmental pollution [6].
In this study, we focus on exploring the potential comprehensive utilization of eucalyptus leaf residue. Firstly, compounds, extracted from eucalyptus leaf residue with organic solvents of different polarity, were analyzed by ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF MS) and chemical methods. Subsequently, the extract was separated and purified by silica gel column chromatography, and high purity triterpenoids were obtained. Then, the efficacy of triterpenoids was evaluated through in vitro antitumor activity, aiming to provide insights into the pharmacological research of active components in eucalyptus leaf residue and offer a basis for the comprehensive utilization of eucalyptus leaf resources.

2. Results and Discussion

2.1. Qualitative Analysis of Components in Eucalyptus Leaf Residue

As described in Section 3.3, the chemical components in n-hexane and 70% ethanol extracts of eucalyptus leaf residue were separated and identified using UPLC-Q/TOF MS. Figure 1 and Figure 2 display the total ion chromatograms of n-hexane and 70% ethanol extracts of eucalyptus leaf residue in negative ion mode. From the figures, it can be observed that both extracts are primarily composed of phenolic compounds and triterpenoids, with similar compositions but varying in component concentrations. A total of 55 compounds were identified in this study, including 28 phloroglucinols, 17 terpenoids, 3 flavonoids, 5 fatty acids, 2 polyphenols, and 1 amino acid. The specific components are listed in Table 1 and Table 2.

2.1.1. Triterpenoids

Triterpenoids are a class of molecules with the molecular formula (C5H8)n that are derivatives of isoprene. Depending on the number of isoprene units in the molecular structure, they can be classified as monoterpenes (n = 2), sesquiterpenes (n = 3), diterpenes (n = 4), triterpenes (n = 6), tetraterpenes (n = 8), and polyterpenes (n > 8), and are widely distributed in nature. Most terpenoids in nature are oxygen-containing derivatives, typically alcohols, aldehydes, ketones, carboxylic acids, esters, and glycosides.
The compounds were identified through the analysis of secondary mass spectrometry fragment ion information, literature review, and the understanding of mass spectrometry fragmentation patterns. According to Table 1 and Table 2, a total of 17 terpenoids were identified in eucalyptus leaf residue, mainly comprising monoterpenes, sesquiterpenes, and triterpenes. Terpenoids in negative ion mode were primarily present in the form of [M − H]. Among them, nine compounds belonging to monoterpenes and sesquiterpenes were identified, including eucalyptol, eucaglobulin, eucalmaidin D, and some formyl-substituted terpene components. Eight triterpenoids were identified, including five triterpenes mainly of the dammarane and ursane types, and three containing coumaryl substitutions [7].
Analysis of pentacyclic triterpenoids: Taking compound peak 10 (Figure 1) as an example, its retention time was 13.46 min, and the quasi-molecular ion peak in negative ion mode was [M − H] at m/z 455. In the secondary mass spectrum, the sub-ion [M−H−HCHO−H2O] at m/z 407 was observed, indicating the loss of formaldehyde and water. Further removal of C2H6 formed the sub-ion peak [M−H−HCHO−H2O−C2H6] at m/z 377, and subsequent removal of CH2 resulted in the sub-ion peak [M−H−HCHO−H2O-C2H6−CH2] at m/z 363, with an additional ion peak at m/z 248 due to the Retro-Diels–Alder reaction. Based on mass spectrometric information and literature [8], it was inferred that compound 10 is dammarane/ursane/betulinic acid, and its fragmentation pattern is shown in Figure 3.
Analysis of coumaryl-substituted pentacyclic triterpenoids: Taking compound peak 37 (Figure 1) as an example, its retention time was 23.62 min, and the quasi-molecular ion peak in negative ion mode was [M − H] at m/z 633. In the secondary mass spectrum, a triterpene product ion peak [M−H−C9H6O3] at m/z 471 was observed, corresponding to the loss of O-p-coumaryl (162 Da), and an additional ion peak at m/z 248 due to the Retro-Diels–Alder reaction. Based on the above information and literature [9], it was inferred that peak 37 is 3-O-trans-p-Coumaroyltormentic acid, and its fragmentation pattern is shown in Figure 4.

2.1.2. Phloroglucinols

Phloroglucinol and its derivatives are a unique class of compounds specific to Eucalyptus plants, characterized by distinctive structures. They often combine with monoterpenes, sesquiterpenes, and diterpenes to form novel compounds. Due to the diverse structures of the associated monoterpenes, sesquiterpenes, and diterpenes, a wide variety of phloroglucinol derivatives are formed. According to literature reports, phloroglucinol derivatives are mainly classified into three types of compounds: phloroglucinols, phloroglucinol dimers, and hybrids formed by phloroglucinols with monoterpenes, sesquiterpenes, or diterpenes. In eucalyptus leaf residue, a total of 28 phloroglucinol components were identified. Some are phloroglucinol dimers, primarily including Macrocarpal A and Sideroxylonal A/B/C [10,11,12]. Others are hybrids formed by phloroglucinols with monoterpenes, sesquiterpenes, or diterpenes, mainly including eucalyptone, eucalyptal, Macrocarpal A/B/D/E, Macrocarpal C, and Macrocarpal I/J [7,12,13].
Analysis of phloroglucinol dimers: Taking compound peak 13 (Figure 1) as an example, its retention time was 15.14 min, and the quasi-molecular ion peak in negative ion mode was [M − H] at m/z 499. In the secondary mass spectrum, the quasi-molecular ion peak [M−H−CO] at m/z 471, formed by the removal of CO, was observed. This ion further dehydrated to form the [M−H−CO−H2O] at m/z 453 sub-ion. Additionally, the base peak at m/z 249 of isopentyl dimethylphloroglucinol [C13H13O5] was observed. Based on the above information and literature [14], it was inferred that compound 13 is Sideroxylonal A/B/C, and its fragmentation pattern is shown in Figure 5a.
Analysis of hybrids formed by phloroglucinols with monoterpenes, sesquiterpenes, or diterpenes: Taking compound peak 11 (Figure 1) as an example, its retention time was 14.07 min, and the quasi-molecular ion peak in negative ion mode was [M − H] at m/z 471. In the secondary mass spectrum, base peaks [M−H−H2O] at m/z 453 and [M−H−CHO] at m/z 443, formed by dehydration and removal of CHO, respectively, were observed. Additionally, the base peak at m/z 249 of isopentyl dimethylphloroglucinol [C13H13O5] was detected. The fragmentation pathways observed were consistent with the mass spectrometric fragmentation reported for Macrocarpal A/B/D/E, suggesting that compound 11 is composed of Macrocarpal A/B/D/E, and its fragmentation pattern is shown in Figure 5b.

2.1.3. Flavonoids

Flavonoids are widely present in plants in nature and belong to the secondary metabolites of plants. Their basic skeleton is C6-C3-C6, and depending on the different substituents, they can be classified as flavones, anthocyanins, chalcones, flavonols, and so on. Different types of flavonoid glycosides and aglycones exhibit characteristic fragmentation patterns. Flavonoid aglycones are prone to undergoing neutral losses (such as the loss of sugar, -CH3, -CO, -H2O, -CH2O, and other free radicals) or undergoing Retro-Diels–Alder (RDA) reactions at the glycoside position, producing characteristic fragment ions.
In eucalyptus leaf residue, three flavonoid compounds were identified, including Sideroxylin, Eucalmaidin D/cypellogin A/B, and leptospermone. The fragmentation patterns of these flavonoids are consistent with the literature-reported data for O-flavonoid glycosides.
Analysis of flavonoids: Taking compound 4 (Figure 2) as an example, its retention time was 1.43 min, and the quasi-molecular ion peak in negative ion mode was [M − H] at m/z 629. In the secondary mass spectrum, the base peak [M−H−C10H14O2] at m/z 463, formed by the loss of olivanic acid, was observed. This fragment ion further lost deoxyhexose to form the base peak [M−H−C10H14O2−C6H10O4] at m/z 301 (quercetin). Combining the fragmentation pattern of the compound with literature data [15,16], it was inferred that the compound was Eucalmaidin D/cypellogin A/B, and its fragmentation pattern is shown in Figure 6.

2.2. Quantitative Analysis of Components in Eucalyptus Leaf Residue

Based on the quantitative method in Section 3.4.4, the active components (total sugars, total terpenes, phloroglucinol derivatives) and nutritional components (crude fiber, crude protein, crude fat, crude ash, moisture) of eucalyptus leaf residue were determined, with a total content of 76.99%. The content of each component in the eucalyptus leaf residue is shown in Table 3.
The content of terpenoids in eucalyptus leaf residue accounted for 2.84% of the raw material. Studies displayed that pentacyclic triterpenes, found in Eucalyptus, have inhibitory effects on cancer cells in the esophagus, lung [17], liver [18], breast [19], pancreas [20], colon, and stomach. The anti-tumor mechanism was mainly achieved by blocking the cell cycle of tumor cells, regulating the protein expression of related genes, inhibiting cell proliferation, inducing cell differentiation, and regulating the body’s immune response. In the early stages of cancer treatment, patients might choose treatment drugs with minimal adverse effects or high efficiency at lower doses [21]. Therefore, the pentacyclic triterpenes, from eucalyptus leaves, are expected to be developed as candidate anti-tumor drugs.
The content of phloroglucinol derivatives in eucalyptus leaf residue accounted for 1.93% of the raw material. Research has shown that phloroglucinol derivatives have good biological activities such as antibacterial, antiviral, and anti-tumor effects, with broad prospects for medical research, and are expected to become new anti-tumor and antibacterial drugs [22].
Eucalyptus leaf residue contains a small amount of flavonoids. Studies indicated that flavonoids in eucalyptus leaves have antibacterial, anti-cardiovascular, anti-inflammatory, analgesic and significant antioxidant properties, which could be developed for new, natural antioxidant-containing functional foods and applied widely in the pharmaceutical field [23].
The content of crude fiber in eucalyptus leaf residue was 27.6%, and the content of crude protein was 5.64%. Due to the high content of crude fiber, it could be used as fiber feed (the content of crude fiber ≥ 18%) in the feed industry.

2.3. In Vitro Antitumor Activity

In order to evaluate the antitumor activity of triterpenoids (content of 82.55%) from eucalyptus leaf residue, fluorouracil (5-FU) was chosen as the positive control drug. MDA-MB-231, SGC-7901, and Hela tumor cells were selected as experimental subjects, and the triterpenoids were tested for activity using the MTT (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) colorimetric method.
The results, as shown in Table 4, demonstrated a certain antitumor activity of triterpenoids from eucalyptus leaf residue. Compared to positive drug (5-FU), they demonstrated moderate inhibitory activity against MDA-MB-231, SGC-7901, and Hela cells, with half-maximal inhibitory concentration (IC50) values of 50.67, 43.12, and 42.65 μg/mL, respectively. Additionally, Wu et al. [24] studied the growth inhibition rate of mango saibao total triterpenes (TTC) on H22 liver cancer cells. Results showed tumor inhibition rates of 42.65% and 43.63% for the high-dose groups (200 mg/kg and 400 mg/kg), respectively. Liang et al. [25] investigated the effects of total triterpenes from Celastrus orbiculatus on the proliferation, apoptosis, and invasion of human esophageal cancer Eca-109 cells. Studies revealed an inhibitory effect, with a maximum inhibition rate of 44.69% at a drug concentration of 160 μg/mL, indicating a dose–response relationship. Ma et al. [26] explored the antitumor activity of different doses of triterpenoid compounds from Laurencia, and results showed an inhibition rate of 39.1% in the high-dose group (63.29%, 100 mg/kg).
Compared to positive drug (5-FU) and triterpenoid compounds from other plant sources, the antitumor activity of the triterpenoids from eucalyptus leaf residue was stronger than that of other plant triterpenes, and weaker than that of positive drugs (5-FU). To summarize, triterpenoids found in eucalyptus leaf residue exhibit moderate antitumor activity and hold potential for further development. The data from this study provided insights and theoretical support for pharmaceutical research on the effective components of eucalyptus leaves.

3. Experimental Section

3.1. Instruments and Apparatus

Ultra performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF MS), BEH C18 column (2.1 × 100 mm, 1.7 μm): Waters corporation, Milford, MA, USA; DK-98-2 water bath: Tianjin test company, Tianjin, China; MM400 retsch grinder: Leica company, Wetzlar, Germany.

3.2. Materials and Reagents

Eucalyptus leaves were collected from Yunnan. The eucalyptus leaf residue was obtained after the extraction of essential oil from eucalyptus leaves by the method of steam distillation. The residue was dried, ground, sifted through a 40–60 mesh sieve, and stored for extraction.
Ethanol and n-hexane from Yongda Reagent (Tianjin, China); Acetonitrile from Fisher (Waltham, MA, USA); Deionized water (18.2 MΩ·cm) from Millipore Milli-Q plus (Haverhill, MA, USA) system. Solvents were of analytical grade for extraction purposes and LC/MS grade for UPLC-Q/TOF MS.

3.3. Sample Preparation and Enrichment

Preparation of Analysis Samples: Firstly, 10 g eucalyptus leaf residue was weighed and transferred to a conical flask, followed by the addition of 60 mL of n-hexane for extraction at 50 °C for 3 h. The extract was then filtered and concentrated to obtain an n-hexane concentrate with a yield of 4.38%. Subsequently, the remaining residue was further extracted with 60 mL of 70% ethanol at 80.0 °C for 3 h, after which the extract was filtered and concentrated to obtain a 70% ethanol concentrate with a yield of 10.56%. Then, the n-hexane and 70% ethanol extracts were redissolved, diluted to 3.0 mg/mL, and filtered through a 0.22 μm membrane into the liquid phase injection vial for UPLC-Q/TOF MS analysis.
Isolation of Triterpenoid Components: Firstly, 10 g eucalyptus leaf residue was weighed and transferred to a conical flask, followed by the addition of 60 mL of 70% ethanol for extraction at 85 °C for 3 h. The extract was then filtered and concentrated to obtain ethanol extract. Subsequently, the triterpenoids, with high purity (content > 80%), were obtained from ethanol extract by silica gel column chromatography using a mixture of n-hexane and ethyl acetate (v:v, 2:1) as eluent, and were used for in vitro antitumor experiments.

3.4. Experimental Methods

3.4.1. Chromatographic Conditions

Chromatographic column: Waters Acquity UPLC BEH C18 (2.1 × 150 mm, 1.7 μm); Mobile phase: A was 0.1% formic acid in water, and B was acetonitrile or isopropanol: acetonitrile (v:v, 1:1); Elution conditions: 0–30 min (25–100%B), 30–35 min (100%B), 35–35.5 min (100–25%B), 35.5–38 min (25%B); Flow rate: 0.3 mL/min; Column temperature: 40 °C; Injection volume: 1.0 μL.
Due to the different polarities of n-hexane and 70% ethanol extracts, different elution solvents were used to ensure the complete elution of components. n-hexane extract was eluted and separated using 0.1% formic acid in water and isopropanol: acetonitrile (v:v, 1:1) as the mobile phase, and 70% ethanol extract was eluted and separated using 0.1% formic acid in water and acetonitrile as the mobile phase.

3.4.2. Mass Spectrometry Conditions

Electrospray ionization source (ESI) in negative ion mode was used to collect MSE data. Calibration solutions were 200 pg/μL leucine enkephalin solution and 0.5 mmol/L sodium formate solution. The scan range was m/z 50–1200, with a scan time of 0.5 s. In negative ion mode, the capillary voltage was 2.5 kV, cone voltage was 40 V, ion source temperature was 120 °C, and high-purity N2 was used as the auxiliary spray ionization and desolvation gas. The desolvation gas temperature was 400 °C, and the flow rate was 800 L/h [27,28].

3.4.3. Data Analysis

Mass spectrometry data were collected and processed using Masslynx V4.1 software. The UNIFI scientific information system was used for data browsing, storage, and comprehensive analysis. Component identification was performed by extracting MS spectra and related MS/MS information, based on built-in mass spectrometry analysis platforms, including ChemSpider online databases (LIPID, Hmbd.ca, etc.) and traditional Chinese medicine databases (TCM Chinese [UNIFI 1.7]), combined with literature information [29,30,31].

3.4.4. Quantitative Methods

Total sugar, total phenol, total triterpenoid, and moisture content in the extracts were determined using the phenol-sulfuric acid method, Folin-phenol method, vanillin-aluminum chloride method, and 105 °C oven method, respectively. Crude fiber, crude protein, crude fat, and crude ash content in eucalyptus leaf residue were determined according to [GB/T 5009.10-1985] [32], [GB/T 5009.5-2016] [33], Soxhlet extraction method, [GB/T 23742-2009] [34], and [GB/T 5009.124-2016] [35].

3.4.5. Efficacy Experiments

In Vitro Antitumor Activity Experiments

Cancer cells (human breast cancer cells MDA-MB-231, human gastric adenocarcinoma cells SGC-7901, and human cervical cancer cells Hela) were cultured in DMEM medium at 37 °C and 5% CO2 until logarithmic growth phase. Cells in logarithmic growth phase were digested with trypsin, and 10 μL of cell suspension was transferred to a 96-well plate. After 24 h of adherent culture, 100 μL of different concentrations of triterpenoid solution (mass concentrations of 0, 6.25, 12.5, 25, 50, and 100 μg/mL) was added. After 48 h of incubation, 100 μL of CCK-8 reaction reagent (10:1) was added, mixed, and incubated for an additional 1.5 h at 37 °C and 5% CO2. The liquid in the wells was then removed, and 200 μL of 0.1% dimethyl sulfoxide (DMSO) was added to each well, followed by oscillation for 10 min to dissolve the crystals [36,37]. Absorbance values (OD values) were detected at 450 nm using a microplate reader. The negative control group was the DMSO, and each experimental group was set up in 5 replicate wells. The inhibition rate of triterpenoids on cancer cells was calculated using Formula (1):
I n h i b i t i o n r a t e = ( 1 O D S a m p l e c o n t r o l O D N e g a t i v e c o n t r o l ) × 100 %

Statistical Analysis

GraphPad Prism 8 statistical software was used for statistical analysis, and Microsoft Excel 2010 was used to assist in calculating the half-maximal inhibitory concentration (IC50) values.

4. Conclusions

This study employed UPLC-Q/TOF MS to isolate and identify the chemical components in eucalyptus leaf residue. A total of 55 chemical components were preliminarily identified, including phloroglucinol derivatives, terpenoids, flavonoids, polyphenols, organic acids, amino acids, and 77% of the chemical components of eucalyptus leaf residue were revealed. Furthermore, the residue was abundant in phloroglucinols and triterpenoid compounds, with contents of 1.93% and 2.84%, respectively, making it a valuable natural source for these compounds. Additionally, the triterpenoid compounds in eucalyptus leaf residue exhibited moderate inhibitory effects on MDA-MB-231, SGC-7901, and HeLa cells, indicating their potential as promising candidates for anticancer drug development. This research offers a framework for pharmaceutical studies on the active components of eucalyptus leaves, thereby establishing a theoretical foundation for the rational development and utilization of eucalyptus leaf residue resources.

Author Contributions

Conceptualization, J.W. and D.W.; methodology, Z.W.; software, X.C.; validation, Z.W. and X.C.; formal analysis, Y.L.; investigation, X.A.; resources, X.A.; data curation, Z.W.; writing—original draft preparation, J.W.; writing—review and editing, D.W.; visualization, J.W.; supervision, D.W.; project administration, Y.L.; funding acquisition, Y.L. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by the Key Research and Development Program of China (2022YFE0124900).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

Authors Juan-Juan Wu, Zi-Xuan Wang, Yun-He Lian, Xiao-Dong An and Di Wu were employed by the company Chenguang Biotech Group Co., Ltd. Author Xin-Ying Cheng was employed by the company Chenguang Biotech Group HanDan Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Fensham, R.J.; Laffineur, B.; Collingwood, T.D.; Beech, E.; Bell, S.; Hopper, S.D.; Phillips, G.; Rivers, M.C.; Walsh, N.; White, M. Rarity or decline: Key concepts for the Red List of Australian eucalypts. Biol. Conserv. 2020, 243, 108455. [Google Scholar] [CrossRef]
  2. Zhu, F.L.; Ren, S.X.; Qiu, B.L.; Huang, Z.; Peng, Z.Q. The abundance and population dynamics of Leptocybe invasa (Hymenoptera: Eulophidae) galls on Eucalyptus spp. in China. J. Integr. Agric. 2012, 11, 2116–2123. [Google Scholar] [CrossRef]
  3. Zhou, X.D.; Wingfield, M.J. Eucalypt diseases and their management in China: Keynote paper APPS 2011. Australas. Plant Pathol. 2011, 40, 339–345. [Google Scholar] [CrossRef]
  4. Salehi, B.; Sharifi-Rad, J.; Quispe, C.; Llaique, H.; Villalobos, M.; Smeriglio, A.; Martins, N. Insights into Eucalyptus genus chemical constituents, biological activities and health-promoting effects. Trends Food Sci. Tech. 2019, 91, 609–624. [Google Scholar] [CrossRef]
  5. Dhakad, A.K.; Pandey, V.V.; Beg, S.; Rawat, J.M.; Singh, A. Biological, medicinal and toxicological significance of Eucalyptus leaf essential oil: A review. J. Sci. Food Agric. 2018, 98, 833–848. [Google Scholar] [CrossRef] [PubMed]
  6. Heidari, A.; Khaki, E.; Younesi, H.; Lu, H.R. Evaluation of fast and slow pyrolysis methods for bio-oil and activated carbon production from eucalyptus wastes using a life cycle assessment approach. J. Clean. Prod. 2019, 241, 118394. [Google Scholar] [CrossRef]
  7. Okba, M.M.; El Gedaily, R.A.; Ashour, R.M. UPLC-PDA-ESI-qTOF-MS profiling and potent anti-HSV-II activity of Eucalyptus sideroxylon leaves. J. Chromatogr. B 2017, 1068, 335–342. [Google Scholar] [CrossRef]
  8. Ashour, R.M.S.; Okba, M.M.; Menze, E.T.; Gedaily, R.A.E. Eucalyptus sideroxylon bark anti-inflammatory potential, its UPLC-PDA-ESI-qTOF-MS profiling, and isolation of a new phloroglucinol. J. Chromatogr. Sci. 2019, 57, 565–574. [Google Scholar] [CrossRef]
  9. Tsiri, D.; Aligiannis, N.; Graikou, K.; Spyropoulos, C.; Chinou, I. Triterpenoids from Eucalyptus camaldulensis DEHNH. tissue cultures. Helv. Chim. Acta 2008, 91, 2110–2114. [Google Scholar] [CrossRef]
  10. Eyles, A.; Davies, N.W.; Mohammed, C. Novel detection of formylated phloroglucinol compounds (FPCs) in the wound wood of Eucalyptus globulus and E. nitens. J. Chem. Ecol. 2003, 29, 881–898. [Google Scholar] [CrossRef]
  11. Santos, B.M.; Zibrandtsen, J.F.S.; Gunbilig, D.; Sørensen, M.; Cozzi, F.; Boughton, B.A.; Heskes, A.M.; Neilson, E.H.J. Quantification and localization of formylated phloroglucinol compounds (FPCs) in Eucalyptus species. Front. Plant Sci. 2019, 10, 186. [Google Scholar] [CrossRef]
  12. Zhou, X.F.; Gao, Z.P. Plant origin and structural classification of phloroglucinols. J. Beijing Univ. Tradit. Chin. Med. 2012, 35, 399–405. [Google Scholar]
  13. Guo, Q.Y.; Huang, X.J.; Zhao, B.X.; Jian, Y.Q.; Luo, S.L.; Wang, Y.; Ye, W.C. Five new acylphloroglucinol glycosides from the leaves of Eucalyptus robusta. Nat. Prod. Commun. 2014, 9, 1934578X1400900218. [Google Scholar] [CrossRef]
  14. Amakura, Y.; Umino, Y.; Tsuji, S.; Ito, H.; Hatano, T.; Yoshida, T.; Tonogai, Y. Constituents and their antioxidative effects in eucalyptus leaf extract used as a natural food additive. Food Chem. 2002, 77, 47–56. [Google Scholar] [CrossRef]
  15. Marsh, K.J.; Saraf, I.; Hocart, C.H.; Youngento, K.; Singh, I.P.; Foleya, W.J. Occurrence and distribution of unsubstituted B-ring flavanones in Eucalyptus foliage. Phytochemistry 2019, 160, 31–39. [Google Scholar] [CrossRef]
  16. Saraf, I.; Marsh, K.J.; Vir, S.; Foley, W.J.; Singh, I.P. Quantitative analysis of various B-ring unsubstituted and substituted flavonoids in ten Australian species of Eucalyptus. Nat. Prod. Commun. 2017, 12, 1695–1699. [Google Scholar] [CrossRef]
  17. Chen, Z.; Huang, K.Y.; Ling, Y.; Goto, M.; Duan, H.Q.; Tong, X.H.; Liu, Y.L.; Cheng, Y.Y.; Natschke, S.L.M.; Yang, P.C.; et al. Discovery of an oleanolic acid/hederagenin-nitric oxide donor hybrid as an EGFR tyrosine kinase inhibitor for non-small-cell lung cancer. J. Nat. Prod. 2019, 82, 3065–3073. [Google Scholar] [CrossRef]
  18. Wang, H.; Zhong, W.; Zhao, J.; Zhang, H.; Zhang, Q.; Liang, Y.; Chen, S.; Liu, H.J.; Zong, S.M.; Tian, Y.X.; et al. Oleanolic acid inhibits epithelial-mesenchymal transition of hepatocellular carcinoma by promoting iNOS dimerization. Mol. Cancer Ther. 2019, 18, 62–74. [Google Scholar] [CrossRef]
  19. Kayouka, M.; Hamade, A.; Saliba, E.; Najjar, F.; Landy, D.; Gergesa, H.G. P-glycoprotein modulates oleanolic acid effects in hepatocytes cancer cells and zebrafish embryos. Chem.-Biol. Interact. 2020, 315, 108892. [Google Scholar] [CrossRef]
  20. Shopit, A.; Li, X.; Tang, Z.; Awsh, M.; Shobet, L.; Niu, M.Y.; Wang, H.Y.; Mousa, H.; Alshwmi, M.; Tesfaldet, T.; et al. MiR-421 up-regulation by the oleanolic acid derivative K73-03 regulates epigenetically SPINK1 transcription in pancreatic cancer cells leading to metabolic changes and enhanced apoptosis. Pharmacol. Res. 2020, 161, 105130. [Google Scholar] [CrossRef]
  21. Wang, Y.; Zhang, Q.; Chen, Y.; Liang, C.L.; Liu, H.; Qiu, F.; Dai, Z. Antitumor effects of immunity-enhancing traditional Chinese medicine. Biomed. Pharmacother. 2020, 121, 109570. [Google Scholar] [CrossRef] [PubMed]
  22. Berillo, D.; Kozhahmetova, M.; Lebedeva, L. Overview of the biological activity of anthraquinons and flavanoids of the plant rumex species. Molecules 2022, 27, 1204. [Google Scholar] [CrossRef] [PubMed]
  23. Kumar, V.; Kaur, R.; Aggarwal, P.; Singh, G. Underutilized citrus species: An insight of their nutraceutical potential and importance for the development of functional food. Sci. Hortic. 2022, 296, 110909. [Google Scholar] [CrossRef]
  24. Wang, R.Q.; Guo, C.; Wang, Z.W.; Liu, D.L.; Niu, T.H.; Wu, G.T. Effects of Cremanthodium humile total terpenes on Th1/Th2 drift and Bax/Bcl-2 balance in H22 tumor-bearing mice. Immunol. J. 2018, 34, 101–108. [Google Scholar]
  25. Yang, Q.W.; Liang, H.P.; Chen, X.P.; Zhang, Z.F.; Liu, Y.Q. Effects of Celastrus orbiculatus extract on Eca-109 cell prolifeeration and invasion and metastasis of esophageal cancer. Heilongjiang Med. Pharmacy 2020, 43, 27–29. [Google Scholar]
  26. Liang, H.; He, J.; Zhang, S.C.; Dong, C.J.; Ma, A.G. Antitumor activities and its immunologic functions of Laurencia terpenoids. Chin. J. Mar. Drugs 2005, 24, 6–9. [Google Scholar]
  27. Vlčková, H.K.; Catapano, M.C.; Mitašík, L.; Kotland, O.; Nejmanová, I.; Pourová, J.; Nováková, L. Featuring ultimate sensitivity of high-resolution LC-MS analysis of phenolics in rat plasma. J. Sep. Sci. 2021, 44, 1893–1903. [Google Scholar] [CrossRef]
  28. Zandonadi, F.S.; Silva, A.A.R.; Melo, A.A.S.; Ignarro, R.S.; Matos, T.S.; Santos, E.A.; Sussulini, A. Understanding ayahuasca effects in major depressive disorder treatment through in vitro metabolomics and bioinformatics. Anal. Bioanal. Chem. 2023, 415, 4367–4384. [Google Scholar] [CrossRef]
  29. Zhu, H.; Lin, H.; Tan, J.; Wang, C.; Wang, H.; Wu, F.; Liu, J. UPLC-QTOF/MS-based nontargeted metabolomic analysis of mountain-and garden-cultivated Ginseng of different ages in Northeast China. Molecules 2018, 24, 33. [Google Scholar] [CrossRef]
  30. Wei, W.; Yu, Y.; Wang, X.; Yang, L.; Zhang, H.; Ji, H.; Guo, D. Simultaneous determination of bufalin and its nine metabolites in rat plasma for characterization of metabolic profiles and pharmacokinetic study by LC-MS/MS. Molecules 2019, 24, 1662. [Google Scholar] [CrossRef]
  31. Wei, L.; Gu, A.; Guo, Z.; Ding, J.; Jin, G.; Lei, Y. An integrated study on the fading mechanism of malachite green industrial dye for the marquisette curtain in the studio of cleansing fragrance, the palace museum (Beijing). Molecules 2022, 27, 4411. [Google Scholar] [CrossRef] [PubMed]
  32. GB/T 5009.10-1985; Determination of Crude Fiber in Vegetable Foods. Ministry of Health of the People Republic of China: Beijing, China, 1985.
  33. GB 5009.5-2016; Determination of Protein in Food of the People’s Republic of China. National Medical Products Administration: Beijing, China, 2016; p. 12.
  34. GB/T 23742-2009; Animal Feeding Stuffs-Determination of Ash Insoluble in Hydrochloric Acid. Chinese Academy of Agricultural Sciences: Beijing, China, 2009.
  35. GB 5009.124-2016; Determination of Amino Acid in Food. The National Standard of China: Beijing, China, 2016.
  36. Sun, F.; Liu, J.Y.; He, F.; Liu, Z.; Wang, D.M.; Wang, Y.F.; Yang, D.P. In-vitro antitumor activity evaluation of hyperforin derivatives. J. Asian Nat. Prod. Res. 2011, 13, 688–699. [Google Scholar] [CrossRef] [PubMed]
  37. Sun, J.; Gao, G.; Gao, Y.L.; Xiong, L.; Li, X.; Guo, J.; Zhang, Y. Experimental research on the in vitro antitumor effects of Crataegus sanguinea. Cell Biochem. Biophys. 2013, 67, 207–213. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Total ion chromatography (TIC) of n-hexane extracts of Eucalyptus leaf residue in negative modes.
Figure 1. Total ion chromatography (TIC) of n-hexane extracts of Eucalyptus leaf residue in negative modes.
Molecules 29 00280 g001
Figure 2. Total ion chromatography (TIC) of 70% ethanol extracts of Eucalyptus leaf residue in negative modes.
Figure 2. Total ion chromatography (TIC) of 70% ethanol extracts of Eucalyptus leaf residue in negative modes.
Molecules 29 00280 g002
Figure 3. Fragmentation pathway of oleanolic acid or ursolic acid.
Figure 3. Fragmentation pathway of oleanolic acid or ursolic acid.
Molecules 29 00280 g003
Figure 4. Fragmentation pathway of 3-O-trans-p-Coumaroyltormentic acid.
Figure 4. Fragmentation pathway of 3-O-trans-p-Coumaroyltormentic acid.
Molecules 29 00280 g004
Figure 5. Fragmentation pathway of Sideroxylonal A (a) and Macrocarpal A (b).
Figure 5. Fragmentation pathway of Sideroxylonal A (a) and Macrocarpal A (b).
Molecules 29 00280 g005
Figure 6. Fragmentation pathway of Eucalmaidin D.
Figure 6. Fragmentation pathway of Eucalmaidin D.
Molecules 29 00280 g006
Table 1. Compounds identified from Eucalyptus leaf n-hexane extracts by UPLC-Q/TOF MS.
Table 1. Compounds identified from Eucalyptus leaf n-hexane extracts by UPLC-Q/TOF MS.
No.T (min)[M − H]− (m/z)Molecular FormulaTheoretical MolecularRelative Error (ppm)MSn (m/z)CompoundsClassification
Weight
18.55251.1372C14H20O4251.128933MS2: 207.1435; 153.0010Tetradeca-trienedioic acidFatty acids
28.66251.1372C14H20O4251.128933MS2: 207.1435; 153.0010Tetradeca-trienedioic acid isomerFatty acids
310.18251.1372C14H20O4251.128933MS2: 207.1435; 153.0010Tetradeca-trienedioic acid isomerFatty acids
410.39325.1252C19H18O5326.115430MS2: 325.1252, 310.0999, 309.0922, 295.0750, 282.1047, 267.0828, 239.0883, 177.0358, 150.0489, 133.0463EucalyptolMonoterpene
510.52265.1544C15H22O4266.151710-LeptospermoneSesquiterpenketone
610.74489.3018C28H42O7490.29318MS2: 461.2899, 207.0286, 250.0839Macrocarpal J/ISesquiterpene
712.49485.2709C28H38O7486.261719MS2: 207.0345, 183.0167Eucalyptone IsomerPhloroglucinol sesquiterpenoids
812.75485.2709C28H38O7486.261719MS2: 453.3354, 439.2457, 251.0918, 250.0839, 207.028EucalyptonePhloroglucinol sesquiterpenoids
913.29485.2709C28H38O7486.261719MS1: 403.2620, 325.1956; MS2: 207.0345, 183.0167Eucalyptone IsomerPhloroglucinol sesquiterpenoids
1013.46455.3518C30H46O3456.360319MS2: 455.3518, 407.3403, 363.3391, 248.9815Ursolic/Oleanolic/Betulinic AcidTriterpene
1114.07471.2937C28H40O6472.282524MS1: 453.2439, 401.2091, 339.2095; MS2: 469.2737, 453.2439, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/EPhloroglucinol sesquiterpenoids
1214.36471.2937C28H40O6472.282524MS2: 469.2737, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
1315.14499.1778C26H28O10500.168219MS1: 471.2937, 249.0845; MS2: 471.2937, 453.2814, 249.0845Sideroxylonal A/B/CPhloroglucinol sesquiterpenoids
1415.46471.2937C28H40O6472.282524MS1: 339.2095; MS2: 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
1515.85487.343C30H48O5488.350115-Arjunolic/Asiatic AcidTriterpene
1616.31471.2937C28H40O6472.282524MS2: 469.2737, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
1716.61471.2937C28H40O6472.282524MS2: 469.2737, 453.3529, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
1816.82471.2937C28H40O6472.282524MS2: 469.2737, 453.3529, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
1917.65471.2892C28H40O6472.282514MS1: 401.2091, 385.2156, 325.0000; MS2: 469.2737, 249.0845, 207.0345Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
2018.12471.347C30H48O4472.355217MS2: 469.2737, 249.0845, 207.0374Hydroxyursolic acid/HederageninTriterpene
2118.33385.2156C23H30O5386.209316-EuglobalPhloroglucinol sesquiterpenoids
2218.56497.3773C32H50O4498.370913-Oleanolic acid 3-acetateTriterpene
2319.16467.2581C28H36O6468.251115MS2: 471.2892, 207.0374, 249.9943UnknownPhloroglucinol sesquiterpenoids
2419.8469.2737C28H36O6470.266815MS2: 325.0000, 265.1544 (423.0046)Withanolide A/Eucalrobusone OPhloroglucinol sesquiterpenoids
2520.24385.2156C23H30O5386.209316-Euglobal IsomerPhloroglucinol sesquiterpenoids
2620.45453.3011C29H42O4454.308316MS1: 385.2156, 311.1812, 249.9896Unknown-
2720.7453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal CPhloroglucinol sesquiterpenoids
2821.06453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
2921.28453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3021.43453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3121.74453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3221.98453.2788C28H38O=5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3322.19453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3422.32453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3522.81453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3623.03453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3723.62633.3807C39H54O7634.386910MS1: 568.3217, 525.3054, 485.2456, MS2: 471.2018, 248.98323-O-trans-p-Coumaroyltormentic acidTriterpene
3824703.3736C41H52O10704.35625MS2: 453.2788, 249.0845UnknownDiterpene Phenol
3924.13689.6077 MS2: 393.326, 207.0345UnknownDiterpene Phenol Sesquiterpene
4025.23775.5295C51H66O10 -UnknownDimethylated Diterpene Olefin
4126.4599.3742C39H52O5600.381412-GarcinialiptoneTriterpene
4227.21281.2483C18H34O2282.255827MS2: 249.2125, 181.2023Oleic AcidFatty Acid
4328.27485.2536C28H38O7486.261717MS2: 325.1920, 207.0374Eucalyptone IsomerFormyl Sesquiterpene Phenol
4428.92469.2584C28H38O6470.266818MS2: 423.2351Withanolide A/Eucalrobusone O IsomerFormyl Phloroglucinol Meroterpenoids
4529741.5692C50H32O10 -UnknownPhloroglucinol sesquiterpenoids
Note: Relative error (ppm) = |actual value − theoretical value|/theoretical value × 106.
Table 2. Compounds identified from Eucalyptus leaf 70% ethanol extracts by UPLC-Q/TOF MS.
Table 2. Compounds identified from Eucalyptus leaf 70% ethanol extracts by UPLC-Q/TOF MS.
No.RT (min)[M − H] (m/z)Molecular FormulaTheoretical Molecular WeightRelative Error (ppm)MSn (m/z)CompoundClassification
10.51353.1057C16H18O9354.095030MS2: 165.0710, 203.0526, 233.0618, 259.0286, 275.1095, 335.0879Chlorogenic AcidPolyphenol
20.66247.1153C14H16O4248.104842MS2: 147.0990, 159.1346IsohistidineAmino Acid
30.98497.1765C23H30O12498.17376-EucaglobulinMonoterpene
41.43629.2004C31H34O14630.19489MS2: 301.0416, 300.0337, 463.0988Eucalmaidin D/Cypellogin A/BFlavonoid
52.78561.2329C26H42O13562.262553MS2: 285.0383, 257.0797, 183.102619-Hydroxycinnzeylanol 19-GlucosideDiterpene Glycoside
63.79301.0025C14H6O8302.006212275.0965, 273.0799Ellagic acidPolyphenol
77.80487.3430C30H48O5488.350115-Arjunolic/Asiatic AcidTriterpene
89.11311.0909C18H16O5312.099728-Sideroxylin Flavonoid
910.79489.2883C28H42O7490.293010MS2: 461.2899, 207.0286, 250.0839Macrocarpal J/IPhloroglucinol sesquiterpenoids
1010.95489.2883C28H42O7490.293010MS2: 461.2899, 207.0286, 250.0839Macrocarpal J/I IsomerPhloroglucinol sesquiterpenoids
1111.13471.3470C30H48O4472.355217MS2: 453.3354, 249.0748, 207.0286Hydroxyursolic acid/HederageninTriterpene
1212.12489.2838C28H42O7490.293019MS2: 461.2899, 457.2581, 443.2794, 250.0839, 207.0286Macrocarpal J/I IsomerPhloroglucinol sesquiterpenoids
1312.37471.3470C30H48O4472.355217MS2: 453.3354, 249.0748, 207.0286Hydroxyursolic acid/Hederagenin IsomerTriterpene
1413.34489.2883C28H42O7490.293010MS2: 461.2899, 207.0286, 250.0839Macrocarpal J/I IsomerPhloroglucinol sesquiterpenoids
1514.53485.2528C28H38O7486.261718MS2: 453.3354, 439.2457, 251.0918, 250.0839, 207.028EucalyptonePhloroglucinol sesquiterpenoids
1614.75485.2528C28H38O7486.261718MS2: 453.3354, 439.2457, 251.0918, 250.0839, 207.028Eucalyptone IsomerPhloroglucinol sesquiterpenoids
1715.71617.3841C39H54O6618.392013MS1: 499.1595, 485.2528, 471.2759, 455.2471, 325.1800, 161.9348; MS2: 497.3271, 451.2456O-p coumaroyl maslinic/alphitolic acidTriterpene
1816.12471.3470C30H48O4472.355217MS2: 453.3354, 249.0748, 207.0286Hydroxyursolic acid/Hederagenin IsomerTriterpene
1916.24471.3470C30H48O4472.355217MS2: 453.3354, 249.0748, 207.0286Hydroxyursolic acid/Hederagenin IsomerTriterpene
2016.39471.3470C30H48O4472.355217MS2: 453.3354, 249.0748, 207.0286Hydroxyursolic acid/Hederagenin IsomerTriterpene
2116.87471.2759C28H40O6472.282514MS2: 469.2737, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/EPhloroglucinol sesquiterpenoids
2217.07455.3514C30H48O3456.360320MS1: 369.8584, 339.1982, 311.1667Ursolic/Oleanolic/Betulinic AcidTriterpene
2317.31499.1778C26H28O10500.168219MS1: 471.2937, 249.0845; MS2: 471.2937, 453.2814, 249.0845Sideroxylonal A/B/CPhloroglucinol sesquiterpenoids
2417.68471.2937C28H40O6472.282524MS1: 453.2439, 401.2091, 339.2095; MS2: 469.2737, 453.2439, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
2517.86471.2937C28H40O6472.282524MS1: 453.2439, 401.2091, 339.2095; MS2: 469.2737, 453.2439, 443.2924, 249.0845, 207.0374Isomer of Eucalyptal A/B/D/EPhloroglucinol sesquiterpenoids
2618.07471.2937C28H40O6472.282524MS1: 453.2439, 401.2091, 339.2095; MS2: 469.2737, 453.2439, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
2718.6453.3354C30H46O3454.344620MS1: 325.1846Dehydroxyursolic LactoneTriterpene
2818.94471.2937C28H40O6472.282524MS1: 453.2439, 401.2091, 339.2095; MS2: 469.2737, 453.2439, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
2919.42471.2937C28H40O6472.282524MS1: 453.2439, 401.2091, 339.2095; MS2: 469.2737, 453.2439, 443.2924, 249.0845, 207.0374Macrocarpal A/B/D/E IsomerPhloroglucinol sesquiterpenoids
3020.12499.1778C26H28O10500.168219MS1: 471.2937, 249.0845; MS2: 471.2937, 453.2814, 249.0845Sideroxylonal A/B/C IsomerPhloroglucinol sesquiterpenoids
3120.28469.2560C28H38O6470.266823MS1: 443.0080, 325.1809, 265.1477; MS2: 425.2687Withanolide A/Eucalrobusone OFormyl Phloroglucinol Meroterpenoids
3220.4599.3732C39H52O5600.381414MS1: 455.2475GarcinialiptoneTriterpene
3320.78629.2004C31H34O14630.19489MS2: 301.0416, 300.0337, 463.0988Eucalmaidin D/Cypellogin A/BFlavonoid
3421.06453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal CPhloroglucinol sesquiterpenoids
3522.57453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3624.11453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3724.6453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3825.16453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
3925.42453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
4025.83453.2788C28H38O5454.271915MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
4126.69607.3989C38H56O6608.407714MS2: 249.0845, 207.0374Sesquiterpene Alcohol EsterSesquiterpene Phenol Alcohol
4226.95453.2657C28H38O5454.271914MS2: 207.0374, 250.0936Macrocarpal C IsomerPhloroglucinol sesquiterpenoids
Note: relative error (ppm) = |actual value − theoretical value|/theoretical value × 106.
Table 3. Summary of the proportions of components in eucalyptus leaves.
Table 3. Summary of the proportions of components in eucalyptus leaves.
CompositionContent/%Summation/%
Crude Fiber27.6076.99
Crude Protein5.64
Crude Fat4.85
Crude Ash4.10
Moisture18.40
Total Sugars6.86
Total Polyphenols4.77
Ursolic Acid0.586
Total Terpenes2.84
Phloroglucinols1.93
Table 4. Inhibitory effect of the triterpenoids and positive control drug against human cancer cell MDA-MB-231, SGC-7901 and Hela.
Table 4. Inhibitory effect of the triterpenoids and positive control drug against human cancer cell MDA-MB-231, SGC-7901 and Hela.
CompoundConcentration
/μg∙mL−1
Inhibition Ratio/%IC50/μg∙mL−1
MDA-MB-
231
SGC-
7901
HelaMDA-MB-
231
SGC-
7901
Hela
Triterpenoids0.000.000.000.0050.6743.1242.65
6.2516.154.625.17
12.5032.014.249.83
25.0029.5228.9016.73
50.0046.6362.8757.97
100.0064.0168.9677.13
5-FU0.000.000.000.002.3612.6122.81
6.2560.9535.0517.47
12.5069.7135.9922.89
25.0073.2638.7234.07
50.0073.1241.4836.25
100.0073.0450.2640.07
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

Wu, J.; Wang, Z.; Cheng, X.; Lian, Y.; An, X.; Wu, D. Preliminary Study on Total Component Analysis and In Vitro Antitumor Activity of Eucalyptus Leaf Residues. Molecules 2024, 29, 280. https://doi.org/10.3390/molecules29020280

AMA Style

Wu J, Wang Z, Cheng X, Lian Y, An X, Wu D. Preliminary Study on Total Component Analysis and In Vitro Antitumor Activity of Eucalyptus Leaf Residues. Molecules. 2024; 29(2):280. https://doi.org/10.3390/molecules29020280

Chicago/Turabian Style

Wu, Juanjuan, Zixuan Wang, Xinying Cheng, Yunhe Lian, Xiaodong An, and Di Wu. 2024. "Preliminary Study on Total Component Analysis and In Vitro Antitumor Activity of Eucalyptus Leaf Residues" Molecules 29, no. 2: 280. https://doi.org/10.3390/molecules29020280

Article Metrics

Back to TopTop