Next Article in Journal
Synthesis and Properties of Novel Alkyl-Substituted Hexaazacyclophanes and Their Diradical Dications
Next Article in Special Issue
Teadenol B as a Component of Microorganism-Fermented Tea Extract Inhibited Breast Cancers by Promoting Autophagy
Previous Article in Journal
Characteristic Evaluation of Gas Chromatography with Different Detectors for Accurate Determination of Sulfur Hexafluoride
Previous Article in Special Issue
An Overview on the Hallucinogenic Peyote and Its Alkaloid Mescaline: The Importance of Context, Ceremony and Culture
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Non-Volatile Component and Antioxidant Activity: A Comparative Analysis between Litsea cubeba Branches and Leaves

1
Teaching and Experimental Center, Guangdong Pharmaceutical University, Guangzhou 510006, China
2
School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
3
Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China
*
Authors to whom correspondence should be addressed.
Molecules 2024, 29(4), 788; https://doi.org/10.3390/molecules29040788
Submission received: 4 December 2023 / Revised: 2 February 2024 / Accepted: 6 February 2024 / Published: 8 February 2024
(This article belongs to the Special Issue Research on Chemical Composition and Activity of Natural Products)

Abstract

:
Litsea cubeba, which is found widely distributed across the Asian region, functions as both an economic tree and a medicinal plant with a rich historical background. Previous investigations into its chemical composition and biological activity have predominantly centered on volatile components, leaving the study of non-volatile components relatively unexplored. In this study, we employed UPLC-HRMS technology to analyze the non-volatile components of L. cubeba branches and leaves, which successfully resulted in identifying 72 constituents. Comparative analysis between branches and leaves unveiled alkaloids, organic acids, and flavonoids as the major components. However, noteworthy differences in the distribution of these components between branches and leaves were observed, with only eight shared constituents, indicating substantial chemical variations in different parts of L. cubeba. Particularly, 24 compounds were identified for the first time from this plant. The assessment of antioxidant activity using four methods (ABTS, DPPH, FRAP, and CUPRAC) demonstrated remarkable antioxidant capabilities in both branches and leaves, with slightly higher efficacy observed in branches. This suggests that L. cubeba may act as a potential natural antioxidant with applications in health and therapeutic interventions. In conclusion, the chemical composition and antioxidant activity of L. cubeba provides a scientific foundation for its development and utilization in medicine and health products, offering promising avenues for the rational exploitation of L. cubeba resources in the future.

1. Introduction

Litsea cubeba, a member of the Lauraceae family in the Litsea genus, thrives primarily in China, India, Thailand, and various Asian countries [1]. Despite occupying a mere 0.04% of China’s wild forest area, the L. cubeba tree stands out for its rich plant resources [2]. Recognized for its economic significance, L. cubeba has been an integral part of traditional Chinese folk medicine for generations. Its roots, stems, leaves, flowers, and fruits have been employed for medicinal purposes, offering remedies for dispelling wind and cold, reducing swelling, and alleviating pain [1,3].
A considerable amount of research has delved into unraveling the chemical constituents and biological activities of L. cubeba, with a predominant focus on its volatile components, especially the essential oil. The fruit and leaves of L. cubeba, rich in essential oils, exhibit a diverse composition of monoterpenes and sesquiterpenes [4]. Notably, citral predominates, widely used in toothpaste, soap, perfumes, and various pharmaceutical and chemical products [5,6,7]. While volatile components have received significant attention, there remains a noteworthy gap in exploring non-volatile components. Over 150 non-volatile components, including alkaloids, lignans, flavonoids, and terpenoids, have been identified in L. cubeba. Alkaloids and lignans, constituting over half of these components, emerge as prominent contributors with significant pharmacological activities [8,9,10,11]. Aporphine alkaloids, in particular, have demonstrated hypoglycemic, anti-inflammatory, and anti-cancer effects [12]. Despite these promising findings, research on non-volatile components remains comparatively limited, overshadowed by the predominant focus on essential oils. The imperative for an in-depth exploration of L. cubeba’s active ingredients and their potential pharmacological effects is evident.
Oxidative stress, implicated in the onset and progression of numerous diseases, poses a significant threat to human health, particularly in the context of neurodegenerative and cardiovascular diseases [13]. Addressing this challenge involves exploring exogenous antioxidants or enhancing endogenous antioxidant defenses. Plants, recognized as valuable repositories of secondary metabolites, emerge as promising sources of natural antioxidants. Studies underscore the potential of L. cubeba as a natural antioxidant, with its essential oil showcasing robust antioxidant activity [14,15]. Extracts from L. cubeba bark, including methanol, chloroform, n-butanol, and water extracts, also exhibit antioxidant activity [16].
Given the abundant resources of L. cubeba, and the relatively limited research on its non-volatile components, there is a need for a more comprehensive exploration to facilitate the rational development and utilization of this plant resource. This is particularly crucial for uncovering its broader medicinal value, with a specific focus on discovering its potential as a robust antioxidant. This study endeavors to bridge this knowledge gap, focusing on the branches and leaves of L. cubeba. Employing UPLC-HRMS, we aim to comprehensively identify non-volatile components in the methanol extract. The antioxidant activity will be assessed through standardized assays, including ABTS (2,2′-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid)), DPPH (1,1-diphenyl-2-trinitrophenylhydrazine), FRAP (ferric reducing antioxidant power), and CUPRAC (cupric reducing antioxidant capacity). By meticulously comparing and analyzing differences in non-volatile components and antioxidant activity, this study aspires to provide a fresh perspective for the rational development and utilization of L. cubeba resources, potentially unlocking novel avenues in medicinal research.

2. Results

2.1. UPLC-HRMS Analysis of Non-Volatile Components from L. cubeba Branches and Leaves

The chemical composition of the methanol extracts from L. cubeba branches and leaves was analyzed using UPLC-Q-Orbitrap HRMS in both positive and negative ion modes. Figures S1–S4 in the Supplementary Materials present the total ion chromatograms of mass spectrometry analysis for the branches and leaves of L. cubeba. Detailed identification information for the compounds is provided in Table 1. A total of 72 compounds were identified in L. cubeba, comprising 23 alkaloids, 15 flavonoids, 14 organic acids, five amino acids, four alcohols, three carbohydrates, two ketones, two phenols, two esters, one ether, and one phenylpropanoid. Alkaloids, flavonoids, and organic acids emerged as the major chemical constituents of L. cubeba. Specifically, 36 and 44 compounds were identified in the branches and leaves of L. cubeba, respectively, with 24 compounds being identified for the first time from this plant. The discovery of these compounds significantly contributes to the chemical information available for L. cubeba.

2.1.1. Analysis of Non-Volatile Components in Branches

Within the L. cubeba branches, a total of 36 non-volatile compounds were identified, encompassing 16 alkaloids (No. 15, 23, 24, 26, 27, 28, 30, 32, 33, 35, 39, 41, 53, 54, 58, and 59). These alkaloids comprised 11 isoquinoline alkaloids (No. 15, 23, 24, 28, 30, 32, 33, 35, 39, 41, and 53), two indole alkaloids (No. 26 and 27), two amide alkaloids (No. 54 and 59), and one other alkaloid (No. 59). Additionally, seven flavonoids (No. 36, 37, 44, 46, 47, 48, and 49), five organic acids (No. 56, 65, 66, 67, and 68), four alcohols (No. 60, 64, 69, and 70), two esters (No. 61 and 62), one ether (No. 14), and one phenylpropanoid (No. 52) were identified. Alkaloids constituted the major component, accounting for nearly 45% of the identified compounds. The sum of flavonoids, organic acids, and alcohol compounds also represented approximately 45% of the total identified compounds in the branches.

2.1.2. Analysis of Non-Volatile Components in Leaves

In L. cubeba leaves, a total of 44 non-volatile compounds were identified, including 12 alkaloids (No. 1, 12, 17, 18, 30, 32, 33, 45, 54, 58, 63, and 72). Among the alkaloids, four were isoquinoline alkaloids (No. 30, 32, 33, and 45), two were amide alkaloids (No. 1 and 54), two were pyridine alkaloids (No. 17 and 18), one was a purine alkaloid (No. 12), one was a pyrrolidine alkaloid (No. 72), one was a quinoline alkaloid (63), and one was classified as other (No. 58). Additionally, 10 organic acids (No. 3, 5, 8, 9, 16, 19, 20, 25, 31, and 56), nine flavonoids (No. 34, 36, 38, 42, 43, 50, 51, 55, and 57), five amino acid compounds (No. 6, 10, 11, 21, and 22), three carbohydrates (No. 2, 4, and 7), two phenols (No. 29 and 40), two ketones (No. 29 and 40), and one alcohol were identified. Alkaloids, organic acids, and flavonoids were the major constituents of L. cubeba leaves, constituting over 70% of the total identified compounds.

2.2. Comparative Analysis of Non-Volatile Components in Branches and Leaves

The comparative analysis of non-volatile components in the methanol extracts of L. cubeba branches and leaves revealed substantial differences in their chemical compositions (Figure 1). Out of the 72 identified compounds in L. cubeba, only eight (No. 30, 32, 33, 36, 54, 56, 58, and 64) were common to both branches and leaves. Leaves displayed 36 unique compounds (No. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 16, 17, 18, 19, 20, 21, 22, 25, 29, 31, 34, 38, 40, 42, 43, 45, 50, 51, 55, 57, 63, 71, and 72), while branches had 28 unique compounds (No. 14, 15, 23, 24, 26, 27, 28, 35, 37, 39, 41, 44, 46, 47, 48, 49, 52, 53, 59, 60, 61, 62, 65, 66, 67, 68, 69, and 70). These distinctive compounds underscore pronounced differences in their chemical profiles.
Alkaloids constituted a major component in both branches and leaves, sharing several compounds such as No. 30, 32, 33, 54, and 58. However, variations exist in the types of alkaloids, with branches primarily featuring isoquinoline alkaloids and leaves encompassing isoquinoline, amide, and other alkaloid categories. Organic acids were prevalent in both branches and leaves, with compound 56 being the sole shared compound (15-Hexadecynoic acid). Nevertheless, other organic acid components exhibited significant differences. Flavonoids, another major class, were identified in both parts, with only Rutin (compound 36) being a shared compound. Other flavonoids were unique to either branches or leaves. Notable distinctions were observed in alcohol and ester content, with branches featuring a higher representation of compounds (No. 60, 64, 69, 70, 61, and 62). Amino acids were exclusively found in leaves (No. 6, 10, 11, 21, and 22). Phenols and ketones were unique to leaves (No. 29 and 40).
In conclusion, while alkaloids, organic acids, and flavonoids were major components in both branches and leaves, the number of shared components was limited. The presence of unique compounds in each part contributed significantly to their distinct chemical profiles, enhancing our understanding of the chemical diversity of L. cubeba and the potential biological activities associated with its various components.

2.3. Antioxidant Activity

Results of Four In Vitro Antioxidant Activity Tests

The evaluation of in vitro antioxidant activity involves various methods based on different principles, with the most commonly utilized ones being ABTS, DPPH, FRAP, and CUPRAC. ABTS and DPPH assess the antioxidant capacity of samples through measuring absorbance change resulting from the reduction of their respective free radicals under the influence of oxidants. In contrast, FRAP and CUPRAC characterize the activity of antioxidants through reducing metal ions to lower oxidation states and measuring their generation. The comprehensive application of these methods provides a nuanced understanding of antioxidant potential within samples. In this study, we assessed the in vitro antioxidant activity of methanol extracts from both branches and leaves of L. cubeba using ABTS, DPPH, FRAP, and CUPRAC methods. For the ABTS, DPPH, and FRAP experiments, antioxidant capacity was expressed as TEAC µmol/g, where higher values signify greater antioxidant capability. For the FRAP method, results were indicated as Fe (Ⅱ) µmol/g, with higher values reflecting stronger reducing power and thus greater antioxidant potential. The methanol extracts from L. cubeba branches exhibited the following results for the four antioxidant assays, as shown in Table 2. These antioxidant activity data indicate that both branches and leaves of L. cubeba possess considerable antioxidant capabilities, with a slight predominance in the antioxidant capacity of the branches over the leaves.

3. Discussion

In this investigation, UPLC-HRMS technology facilitated the rapid and precise analysis of non-volatile components within L. cubeba branches and leaves. The established analytical method for non-volatile components offered insights into the diverse compounds present in these plant parts, contributing detailed information to enhance our understanding of the plant’s chemical characteristics. The identification of 72 compounds, including alkaloids, flavonoids, and organic acids, underscored the chemical diversity of L. cubeba. Moreover, methanol extracts from both branches and leaves exhibited noteworthy antioxidant activity in vitro.
While the results align with some previously reported chemical compositions, this study uncovered 24 compounds not documented before, enriching our comprehensive understanding of L. cubeba compounds and supporting its chemical taxonomy, development, and utilization [74]. Alkaloids, especially isoquinoline alkaloids, emerged as significant components in the non-volatile fraction of L. cubeba, aligning with prior research [74]. Intriguingly, a higher abundance of isoquinoline alkaloids in branches compared to leaves was observed. Further analysis of alkaloid differences identified eight shared compounds, five of which belonged to the alkaloid category (compounds 30, 32, 33, 54, and 58). The predominance of isoquinoline alkaloids in branches, and the greater diversity of alkaloid types in leaves, suggest potential variations in pharmacological activities. Known for their diverse biological activities, especially in pharmaceutical research, the abundance of isoquinoline alkaloids in L. cubeba implies their essential role in its medicinal effects.
Flavonoids, renowned for their antioxidant properties, were identified as major components in both branches and leaves of L. cubeba. The presence of the shared compound rutin (compound 36) suggests common antioxidant capabilities. However, unique flavonoid compositions in each part may contribute to subtle differences in antioxidant activity. The study also confirmed the presence of organic acids, a less explored aspect of L. cubeba chemistry. The only shared organic acid between the branches and leaves was 15-hexadecynoic acid (compound 56), indicating potential variations in their health-promoting properties. Beyond the primary compound categories, differences in alcohols, esters, amino acids, and phenols between the branches and leaves add layers to the plant’s chemical diversity, potentially influencing related biological activities and the potential medicinal value of L. cubeba.
The assessment of antioxidant potential using ABTS, DPPH, FRAP, and CUPRAC assays demonstrated significant antioxidant capabilities in both the branch and leaf extracts, with a slightly higher antioxidant capacity observed in the branches. In comparison with antioxidant capacities of traditional medicinal plants, such as Angelica dahurica, Fritillaria cirrhosa, and Perilla frutescens, L. cubeba branches and leaves exhibited superior or comparable antioxidant abilities [75]. The plant’s rich content of alkaloids, flavonoids, and organic acids, known for their antioxidant properties, likely contributes to its robust antioxidant activity, emphasizing the potential health benefits of L. cubeba in traditional herbal applications.
In summary, this comparative analysis highlights the diverse chemical characteristics and antioxidant capabilities of L. cubeba branches and leaves. As a botanical resource, L. cubeba exhibits unique features in terms of chemical composition and antioxidant activity, providing a theoretical foundation for its applications in medicine and the food industry. Future research could delve deeper into the biological activities and pharmacological effects of L. cubeba compounds to explore its potential health benefits. This study offers valuable information for the development and utilization of L. cubeba, laying a scientific foundation for its diverse applications.

4. Materials and Methods

4.1. Plant Material and Extraction

4.1.1. Plant Material

L. cubeba was sourced from Yunfu, China, in April 2023, and Dr. Xinger Ye, affiliated with the College of Traditional Chinese Medicine Resources at Guangdong Pharmaceutical University, authenticated the plant materials.

4.1.2. Preparation of Liquid-Sample Solution for Mass Spectrometry

The dried leaves and stems of L. cubeba were individually pulverized using the DFY–300C Swing Crusher from Wenling Linda Machinery Co., Ltd, Wenling, China. Approximately 1.0 g of each sample powder was precisely weighed into a 100 mL conical flask. Analytical methanol (50 mL) was added accurately, and the mixture was re-weighed. The Q-500DE ultrasound equipment (Dongguan Keqiao Ultrasonic Equipment Co., Ltd., Dongguan, China) was used for ultrasonic extraction at 50 °C with 500 W power and 40 kHz frequency for 30 min. After cooling, the sample was re-weighed, and any weight loss was compensated by adding methanol. Ten milliliters of the upper clear liquid were transferred to a centrifuge tube, and centrifuged at 2200× g for 15 min using a high-speed centrifuge. Two hundred microliters of the supernatant were extracted, mixed with 800 µL of chromatographic methanol, diluted to 1 mL, thoroughly mixed, and then filtered through a 0.22 µm microporous membrane. The resultant filtered solution served as the liquid–sample solution for mass spectrometry.

4.1.3. Preparation of Samples for Antioxidant Activity Testing

For antioxidant activity testing, 1.0 g of L. cubeba leaves and branches powder was precisely weighed and combined with 40 mL of methanol. Ultrasonic extraction was conducted at 50 °C using an ultrasonic power of 500 W (frequency, 40 kHz) for 50 min, followed by filtration and rotary evaporation to obtain the respective extracts. The yield was 117.7 mg for leaf extract and 61.4 mg for branch extract. The extracts were dissolved in 15 mL of methanol. Based on preliminary experiments, the leaf methanol extract was diluted 10 times, and the branch methanol extract was diluted 5 times to produce the test solutions.

4.2. The Main Chemicals and Reagents

Analytical grade methanol from Da Mao Chemical Reagent Co., Ltd., Tianjin, China; chromatography-grade acetonitrile from Honeywell Trading (Shanghai) Co., Ltd., Shanghai, China; and distilled water from Watsons, Hong Kong, China. The total antioxidant capacity assay kits (ABTS, DPPH, FRAP methods) were obtained from Shanghai Macklin Biochemical Technology Co., Ltd., Shanghai, China, and the liquid sample total antioxidant capacity assay kit (Cu2+ method) was sourced from Beijing Applygen Technology Co., Ltd., Beijing, China.

4.3. UPLC-HRMS Analysis

4.3.1. Instrumentation and Conditions

The analysis employed the Vanquish Flex UPLC system, coupled with the Orbitrap Exploris 120 quadrupole electrostatic field orbital well high-resolution mass spectrometer for UPLC-Q-Orbitrap HRMS, sourced from Thermo Fisher Scientific (Waltham, MA, USA). A Hypersil GOLD C18 column (100 mm × 2.1 mm, 1.9 μm) was used for chromatographic analysis with a flow rate of 0.3 mL·min−1, maintaining the column temperature at 35 °C. A 2.00 μL sample was injected into the system. The mobile phase comprised acetonitrile (A) and a 0.1% formic acid solution (B). The gradient elution profile was programmed as follows: starting at 95% B for 5 min, gradually decreasing to 80% B from 5 to 8 min, further decreasing to 75% B from 8 to 20 min, reducing to 5% B from 20 to 22 min, holding at 5% B from 22 to 22.001 min, and finally increasing to 95% B from 22.001 to 25 min.
The mass spectrometry analysis utilized positive and negative ion switching scan modes (Full scan/dd-MS2). H-ESI ionization was employed with an electrospray voltage of 3.5 kV (+)/2.8 kV (−). The ion transfer tube temperature was set at 325 °C, and the auxiliary gas temperature was maintained at 350 °C, RF Lens at 70%. Nitrogen served as nebulizing gas, auxiliary gas, and sheath gas. The flow rates for sheath gas, auxiliary gas, and sheath gas were 50 Arb, 8 Arb, and 1 Arb, respectively. Primary and secondary resolutions were set at 60,000 (MS) and 15,000 (MS2), respectively. The ion scan range (m/z) was 100–1500, and the collision energy was normalized. The gradient for collision energy was set at 20%, 40%, and 60%.

4.3.2. Data Analysis and Identification of Compounds

The original data files were processed using Compound Discoverer 3.3 software provided by Thermo Fisher Scientific (Waltham, MA, USA), employing a compound identification method template. This involved peak area extraction, alignment, and matching secondary fragment spectra against the mzCloud network database and the local database mzVault. Filtering criteria included mass deviation control, optimal scores, and comparison with compound information in the compound library. Subsequent analysis of compounds included cross-referencing with literature and online databases (PubChem, CNKI, PubMed) for enhanced understanding.

4.4. Antioxidant Activity

4.4.1. ABTS Radical Scavenging Assay

The ABTS assay followed the Total Antioxidant Capacity Assay Kit. ABTS solution was combined with an oxidizing agent, and the resulting mixture served as the working solution. After 12–16 h of dark storage at room temperature, the ABTS working solution underwent dilution with 80% ethanol, and the absorbance at 734 nm was measured using an Agilent Synergy H1 microplate reader (Agilent Technologies, Inc., VT, USA) until reaching 0.7 ± 0.05 absorbance units. Test and standard solutions were mixed with the ABTS working solution in a 96-well plate, and absorbance at 734 nm was recorded after incubation. Trolox served as the standard, and a series of Trolox standard solutions (0.15, 0.3, 0.6, 0.9, 1.2, 1.5 mM) were prepared. The total antioxidant capacity of each sample was expressed as µmol Trolox/g of herbal medicine dry weight [76].

4.4.2. DPPH Radical Scavenging Assay

The DPPH assay followed the Total Antioxidant Capacity Assay Kit. Test and standard solutions were mixed with the reagent, and after a 20-min dark reaction at room temperature, absorbance was measured at 515 nm using a microplate reader. Trolox served as the standard, and a series of Trolox standard solutions (0.18, 0.15, 0.12, 0.09, 0.06, 0.03 mM) were prepared. The total antioxidant capacity of each sample was expressed as µmol Trolox/g of herbal medicine dry weight [77].

4.4.3. Ferric Reducing Ability of Plasma (FRAP) Assay

The FRAP assay followed the Total Antioxidant Capacity Assay Kit. The FRAP working solution was prepared by mixing TPTZ (2,4,6-tris(2-pyridyl)-s-triazine) diluent, TPTZ solution, and detection buffer solution at a ratio of 10:1:1 (v/v/v). After incubation at 37 °C and using within 1–2 h, test and standard solutions were mixed with the FRAP working solution in a 96-well plate and incubated at 37 °C for 3–5 min. After cooling to room temperature, the absorbance of the mixture was recorded at 593 nm using a microplate reader. FeSO4 served as the standard, and a series of FeSO4 standard solutions (0.15, 0.3, 0.6, 0.9, 1.2, 1.5 mM) were prepared. The total antioxidant capacity of each sample was expressed as µmol Fe(Ⅱ)/g of herbal medicine dry weight [78].

4.4.4. Cupric Reducing Antioxidant Capacity (CUPRAC) Assay

The CUPRAC assay followed the Total Antioxidant Capacity Assay Kit (Cu2+ method). The Cu2+ working solution was prepared by mixing Cu2+ chelating agent solution and Cu2+ solution at a ratio of 50:1 (v/v). Test and standard solutions were mixed with the Cu2+ working solution in a 96-well plate, incubated at room temperature for 30 min, and absorbance was measured at 570 nm using a microplate reader. Trolox served as the standard, and a series of Trolox standard solutions (1, 0.5, 0.25, 0.125, 0.062, 0.031 mM) were prepared. The total antioxidant capacity of each sample was expressed as µmol Trolox/g of herbal medicine dry weight [79].

4.4.5. Construction of Standard Curves

In this study, standard curves were constructed for the ABTS, DPPH, FRAP, and CUPRAC assays. The ABTS assay’s standard curve correlated absorbance values with known Trolox (6-hydroxy-2,5,7,8-etramethylchroman-2-carboxylic acid) concentrations, facilitating the calculation of Trolox Equivalent Antioxidant Capacity (TEAC) using a linear regression equation. Similarly, the DPPH assay utilized absorbance values correlated with various Trolox concentrations to generate a standard curve, enabling the conversion of experimental sample absorbance readings to TEAC values through a linear equation. For the FRAP assay, we created a standard curve by plotting absorbance against different concentrations of Fe(Ⅱ). The resulting linear regression equation was then applied to compute the FRAP of the tested samples. Likewise, the CUPRAC assay employed a standard curve established with known Trolox concentrations, utilizing the linear equation to quantify the CUPRAC of the samples. These standard curves not only formed the foundation for quantification and data analysis but also enhanced the accuracy and reliability of antioxidant activity assessments. Refer to Figure 2 for the standard curves, and Table 3 for the corresponding equations. The R2 values, ranging from 0.99133 to 0.99987, indicate excellent linearity, meeting the required standards.

4.4.6. Statistical Analysis

The experiments were conducted in triplicate, and the results were reported as the mean ± standard deviation.

5. Conclusions

Through a comparative analysis of the non-volatile components and antioxidant activity in the branches and leaves of the traditional medicinal plant L. cubeba, we unveiled its diverse chemical composition, encompassing alkaloids, flavonoids, and organic acids across various categories. This underscores its potential as a valuable resource in traditional herbal medicine and the food industry. The distinct variations in alkaloids, organic acids, and flavonoids between the branches and leaves suggest potential differences in their suitability for various applications. Supporting our observations, UPLC-HRMS technology identified 72 constituents, unveiling significant chemical heterogeneity between the branches and leaves, with only eight components shared. This comprehensive characterization contributes to our understanding of L. cubeba’s chemical diversity. Regarding antioxidant activity, both the branches and leaves exhibited significant capabilities, with the branches demonstrating slightly higher efficacy. Consistent support from ABTS, DPPH, FRAP, and CUPRAC methods underscores the robust antioxidant potential within L. cubeba. This suggests L. cubeba’s promising role as a natural antioxidant with implications for health and therapeutic interventions. In summary, L. cubeba stands out as a botanical resource with unique chemical composition and potent antioxidant properties, providing theoretical support for its applications in medicine and the food industry. The identified chemical diversity and robust antioxidant activity lay a solid scientific foundation for further exploration. Future research endeavors could delve deeper into the biological activities and pharmacological effects of L. cubeba compounds, enhancing our understanding of its potential health benefits. This study contributes valuable insights for the ongoing exploration and utilization of L. cubeba in various fields.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules29040788/s1, Figures S1–S4: UPLC-Q-Orbitrap HRMS chromatograms of Litsea cubeba.

Author Contributions

Conceptualization, W.D., Q.W. and G.Q.; methodology, L.D. and Q.W.; software, Y.T. and B.L.; validation, W.D. and Y.T.; formal analysis, L.Z. and Y.X.; investigation, Y.T. and L.Z.; resources, W.D.; data curation, Y.T. and L.Z.; writing—original draft preparation, W.D. and B.L.; writing—review and editing, G.Q. and Q.W.; funding acquisition, W.D. and G.Q. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the following: Guangzhou Basic Research Program Fund Project (No. 202102020630); Scientific research project of Guangdong Provincial Administration of Traditional Chinese Medicine (No. 20231208); Guangdong Medical Research Fund (No. B2021324).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and supplementary materials.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Editorial Committee of Flora of China, Chinese Academy of Science. Flora of China, 1st ed.; Science Press: Beijing, China, 1993; Volume 31, pp. 271–272. [Google Scholar]
  2. Ru, H.; Liu, M.; Liao, T.; Zhu, J.J. Analysis on technological innovation ability and competitive situation of Litsea cubeba industry from the perspective of patent, Hunan. For. Sci. Tec. 2022, 49, 89–94. [Google Scholar]
  3. Yu, L.Y.; Jia, D.; Shen, H.; Sun, X.L.; Qin, L.P.; Han, T. Anti-inflammatory study on 9,9′-O-di-(E)-feruloyl-meso-5,5′-dimethoxysecoisolariciresinol (LCA), an active ingredient in Litsea cubeba (Lour.) Pers. J. Pha. Pra. 2020, 38, 216–220. [Google Scholar]
  4. Zhang, L.; Chen, X.S.; Fan, G.R.; Liao, S.L.; Wang, Z.D. Research progress in application and functions of Litsea cubeba essential oil components. Agric. Univ. Jiangxiensis 2021, 43, 355–363. [Google Scholar]
  5. Li, X.Y.; Gou, Y.H.; Shen, H.J.; Li, Y.H.; Wang, T.; Yin, L.Z. Antibacterial mechanism of Litsea cubeba essential oil on Salmonella. J. Sci. Agric. Uni. 2021, 39, 385–390. [Google Scholar]
  6. Liu, T.; Yang, T.S. Antimicrobial impact of the components of essential oil of Litsea cubeba from Taiwan and antimicrobial activity of the oil in food systems. Int. J. Food. Mic. 2012, 156, 68–75. [Google Scholar] [CrossRef]
  7. Xiang, Y.J.; Wang, H.H.; Sun, Y.D. Advances in the application of Litsea cubeba oil. Chin. J. Cer. 2020, 35, 186–195. [Google Scholar]
  8. Zhang, S.Y.; Guo, Q.; Gao, X.L.; Guo, Z.Q.; Zhao, Y.F.; Chai, X.Y.; Tu, O.F. A phytochemical and pharmacological advance on medicinal plant Litsea cubeba (Lauraceae). Chin. Herb. Med. 2014, 39, 769–776. [Google Scholar]
  9. Feng, T.; Xu, Y.; Cai, X.H.; Du, Z.Z.; Luo, X.D. Antimicrobially Active Isoquinoline Alkaloids from Litsea cubeba. Planta Med. 2009, 75, 76–79. [Google Scholar] [CrossRef]
  10. Guo, Q.; Bai, R.F.; Su, G.Z.; Zhu, Z.X.; Zeng, K.W.; Tu, P.F.; Chai, X.Y. Chemical Constituents from the Roots and Stems of Litsea cubeba. J. Asian Nat. Prod. Res. 2016, 18, 51–58. [Google Scholar] [CrossRef]
  11. Lee, S.S.; Chen, C.K.; Huang, F.M.; Chen, C.H. Two dibenzopyrrocoline alkaloids from Litsea cubeba. J. Nat. Prod. 1996, 59, 80–82. [Google Scholar] [CrossRef]
  12. Zhang, S.S.; Tan, Q.W.; Guan, L.P. Antioxidant, anti-inflammatory, antibacterial, and analgesic activities and mechanisms of quinolines, indoles and related derivatives. Mini Rev. Med. Chem. 2021, 21, 2261–2275. [Google Scholar] [CrossRef] [PubMed]
  13. Diniz Vilela, D.; Gomes Peixoto, L.; Teixeira, R.R.; Belele Baptista, N.; Carvalho Caixeta, D.; de Souza, A.V.; Machado, H.L.; Pereira, M.N.; Sabino–Silva, R.; Espindola, F.S. The Role of Metformin in Controlling Oxidative Stress in Muscle of Diabetic Rats. Oxid. Med. Cell Longev. 2016, 2016, 6978625. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, S.; Zhao, C.; Cao, Y.; Li, Y.; Zhang, Z.; Nie, D.; Tang, W.; Li, Y. Comparison of Chemical Compositions and Antioxidant Activity of Essential Oils from Litsea cubeba, Cinnamon, Anise, and Eucalyptus. Molecules 2023, 28, 5051. [Google Scholar] [CrossRef] [PubMed]
  15. She, Q.H.; Li, W.S.; Jiang, Y.Y.; Wu, Y.C.; Zhou, Y.H.; Zhang, L. Chemical Composition, Antimicrobial Activity, and Antioxidant Activity of Litsea cubeba Essential Oils in Different Months. Nat. Prod. Res. 2020, 34, 3285–3288. [Google Scholar] [CrossRef]
  16. Hwang, J.K.; Choi, E.M.; Lee, J.H. Antioxidant activity of Litsea cubeba. Fitoterapia 2005, 76, 684–686. [Google Scholar] [CrossRef]
  17. Zhang, Y.; Fang, H.; Zhang, Y. Study on formation of acrylamide in asparagine-sugar microwave heating systems using UPLC–MS/MS analytical method. Food Chem. 2008, 108, 542–550. [Google Scholar] [CrossRef]
  18. Zhang, L.; Wang, X.Y.; Guo, J.Z.; Xia, Q.L.; Zhao, G.; Zhou, H.N.; Xie, F.W. Metabolic profiling of Chinese tobacco leaf of different geographical origins by GC–MS. J. Agric. Food Chem. 2013, 61, 2597–2605. [Google Scholar] [CrossRef]
  19. Shen, Q.Y.; Yang, R.J.; Hua, X.; Ye, F.Y.; Wang, H.; Zhao, W.; Wang, K. Enzymatic synthesis and identification of oligosaccharides obtained by transgalactosylation of lactose in the presence of fructose using β-galactosidase from Kluyveromyces lactis. Food Chem. 2012, 135, 1547–1554. [Google Scholar] [CrossRef]
  20. Clifford, M.N.; Knight, S.; Kuhnert, N. Discriminating between the six isomers of dicaffeoylquinic acid by LC-MSn. J. Agric. Food Chem. 2005, 53, 3821–3832. [Google Scholar] [CrossRef]
  21. Gale, T.V.; Schieffelin, J.S.; Branco, L.M.; Garry, R.F.; Grant, D.S. Elevated L-threonine is a biomarker for Lassa fever and Ebola. Virol. J. 2020, 17, 188. [Google Scholar] [CrossRef]
  22. Song, M.; Hang, T.J.; Wang, C.; Yang, L.; Wen, A.D. Precolumn derivatization LC-MS/MS method for the determination and pharmacokinetic study of glucosamine in human plasma and urine. J. Pharm. Anal. 2012, 2, 19–28. [Google Scholar] [CrossRef] [PubMed]
  23. Ma, J.S.; Xu, R.Y.; Wang, B.K.; Qi, Q.; Zhang, C.N. Metabolomics analysis reveals the protective effect of fructooligosaccharide on abnormal metabolism of liver in Megalobrama amblycephala induced by Aeromonas hydrophila. Aquacult. Int. 2023, 2023, 1–18. [Google Scholar] [CrossRef]
  24. Fernandez-Fernandez, R.; Lopez-Martinez, J.C.; Romero-Gonzalez, R.; Martinez-Vidal, J.L.; Flores, M.I.A.; Garrido Frenich, A. Simple LC–MS determination of citric and malic acids in fruits and vegetables. Chromatographia 2010, 72, 55–62. [Google Scholar] [CrossRef]
  25. Moussa, S.; Van Horn, M.R.; Shah, A.; Pollegioni, L.; Thibodeaux, C.J.; Ruthazer, E.S.; Mauzeroll, J. Editors’ choice—A miniaturized enzymatic biosensor for detection of sensory–evoked D–serine release in the brain. J. Electrochem. Soc. 2021, 168, 025502. [Google Scholar] [CrossRef]
  26. Klupczynska, A.; Misiura, M.; Miltyk, W.; Oscilowska, I.; Palka, J.; Kokot, Z.; Matysiak, J. Development of an LC-MS targeted metabolomics methodology to study proline metabolism in mammalian cell cultures. Molecules 2020, 25, 4639. [Google Scholar] [CrossRef] [PubMed]
  27. Barry, C.G.; Day, C.S.; Bierbach, U. Duplex–Promoted Platination of Adenine–N3 in the Minor Groove of DNA: Challenging a Longstanding Bioinorganic Paradigm. J. Am. Chem. Soc. 2005, 127, 1160–1169. [Google Scholar] [CrossRef]
  28. Zhang, J.; Shi, L.y.; Ding, L.P.; Liu, Y.; Liang, H.; Tu, P.F.; Li, L.; Zhang, Q.Y. Condensation derivatives of 4-isopropylbenzaldehyde with acetophenone from the red alga Laurencia tristicha. Phytochemistry. 2021, 192, 112960. [Google Scholar] [CrossRef]
  29. Wang, R.; Xiong, X.P.; Yang, M.; He, S.R.; Xu, X.P. A pharmacokinetics study of orally administered higenamine in rats using LC–MS/MS for doping control analysis. Drug Test Anal. 2020, 12, 485–495. [Google Scholar] [CrossRef]
  30. Larsen, J.; Cornett, C.; Jaroszewski, J.W.; Hansen, S.H. Reaction between drug substances and pharmaceutical excipients: Formation of citric acid esters and amides of carvedilol in the solid state. J. Pharmaceut. Biomed. 2009, 49, 11–17. [Google Scholar] [CrossRef]
  31. Liu, M.; Zhang, D.; Wang, X.L.; Zhang, L.N.; Han, J.; Yang, M.; Xiao, X.; Zhang, Y.N.; Liu, H.C. Simultaneous quantification of niacin and its three main metabolites in human plasma by LC–MS/MS. J. Chromatogr. B 2012, 904, 107–114. [Google Scholar] [CrossRef]
  32. Sadilkova, K.; Gospe, S.M.; Hahn, S.H. Simultaneous determination of alpha–aminoadipic semialdehyde, piperideine-6-carboxylate and pipecolic acid by LC–MS/MS for pyridoxine–dependent seizures and folinic acid–responsive seizures. J. Neurosci. 2009, 184, 136–141. [Google Scholar] [CrossRef] [PubMed]
  33. Magera, M.J.; Helgeson, J.K.; Matern, D.; Rinaldo, P. Methylmalonic acid measured in plasma and urine by stable-isotope dilution and electrospray tandem mass spectrometry. Clin. Chem. 2000, 46, 1804–1810. [Google Scholar] [CrossRef] [PubMed]
  34. Rostami–Yazdi, M.; Clement, B.; Schmidt, T.J.; Schinor, D.; Mrowietz, U. Detection of Metabolites of Fumaric Acid Esters in Human Urine: Implications for Their Mode of Action. J. Invest. Dermatol. 2009, 129, 231–234. [Google Scholar] [CrossRef] [PubMed]
  35. Xiao, Y.S.; Vecchi, M.M.; Wen, D.Y. Distinguishing between Leucine and Isoleucine by Integrated LC–MS Analysis Using an Orbitrap Fusion Mass Spectrometer. Anal. Chem. 2016, 88, 10757–10766. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, J.L.; Eris, T.; Lauren, S.L.; Stearns, G.W.; Westcott, K.R.; Lu, H. Use of LC/MS peptide mapping for characterization of isoforms in 14N–labeled recombinant human leptin. Tech. Protein Chem. 1996, 8, 155–163. [Google Scholar]
  37. Chen, Q.F.; Zhang, B.C.; Hicks, L.M.; Wang, S.P.; Jez, J.M. A liquid chromatography–tandem mass spectrometry-based assay for indole–3–acetic acid–amido synthetase. Anal. Chem. 2009, 390, 149–154. [Google Scholar] [CrossRef]
  38. Reed, J.D.; Krueger, C.G.; Vestling, M.M. MALDI-TOF mass spectrometry of oligomeric food polyphenols. Phytochemistry 2005, 66, 2248–2263. [Google Scholar] [CrossRef] [PubMed]
  39. Xu, F.F.; Qi, R.; Jiang, S.W.; Wang, W.; Chen, Z.Q.; Guo, D.A.; Liu, B. Simultaneous determination of ten compounds in Xiangbin Fang by LC-MRM-MS. Zhongguo Zhong Yao Za Zhi 2017, 32, 2226–2229. [Google Scholar]
  40. Pan, H.F.; Lundgren, L.N. Phenolic extractives from root bark of Picea abies. Phytochemistry 1995, 39, 1423–1428. [Google Scholar] [CrossRef]
  41. Mollataghi, A.; Hadi, A.H.A.; Awang, K.; Mohamad, J.; Litaudon, M.; Mukhtar, M. (+)–Kunstlerone, a new antioxidant neolignan from the leaves of Beilschmiedia kunstleri Gamble. Molecules 2011, 16, 6582–6590. [Google Scholar] [CrossRef]
  42. Chiwocha, S.D.S.; Abrams, S.R.; Ambrose, S.J.; Cutler, A.J.; Loewen, M.; Ross, A.R.S.; Kermode, A.R. A method for profiling classes of plant hormones and their metabolites using liquid chromatography-electrospray ionization tandem mass spectrometry: An analysis of hormone regulation of thermodormancy of lettuce (Lactuca sativa L.) seeds. Plant Physiol. 2003, 35, 405–417. [Google Scholar] [CrossRef]
  43. Husna, H.; Muhammad, H.; Sivasothy, Y.; Khaw, K.; Nafiah, M.A.; Hazni, H.; Litaudon, M.; Wan, R.; Wan, A.; Liew, S.Y. N-Methyl Costaricine and Costaricine, Two Potent Butyrylcholinesterase Inhibitors from Alseodaphne pendulifolia Gamb. Int. J. Mol. Sci. 2023, 24, 10699. [Google Scholar] [CrossRef]
  44. Takemura, T.; Takatsu, Y.; Kasumi, M.; Marubashi, W.; Iwashina, T.H. Flavonoids and their distribution patterns in the flowers of Gladiolus cultivars. Acta Hortic. 2005, 673, 487–493. [Google Scholar] [CrossRef]
  45. Choi, S.J.; Kim, J.K.; Jang, J.M.; Shin, K.H.; Lim, S.S. Rapid identification of the α–glucosidase inhibitory compounds from Thunberg’s Geranium (Geranium thunbergii Sieb. et Zucc.). Food Sci. Biotechnol. 2012, 21, 987–996. [Google Scholar] [CrossRef]
  46. Li, Y.Q.; Zhou, F.C.; Gao, F.; Bian, J.S.; Shan, F. Comparative Evaluation of Quercetin, Isoquercetin and Rutin as Inhibitors of α-Glucosidase. J. Agric. Food Chem. 2009, 57, 11463–11468. [Google Scholar] [CrossRef] [PubMed]
  47. Jorge, A.P.; Horst, H.; Sousa, E.; Pizzolatti, M.G.; Silva, F.R.M.B. Insulinomimetic effects of kaempferitrin on glycemia and on 14C–glucose uptake in rat soleus muscle. Chem. Biol. Interact. 2004, 149, 89–96. [Google Scholar] [CrossRef] [PubMed]
  48. Li, S.Y.; Zhang, Z.Z.; Cain, A.; Wang, B.; Long, M.; Taylor, J. Antifungal Activity of Camptothecin, Trifolin, and Hyperoside Isolated from Camptotheca acuminata. J. Agric. Food Chem. 2005, 53, 32–37. [Google Scholar] [CrossRef]
  49. Yin, R.; Han, F.; Tang, Z.; Liu, R.; Zhao, X.; Chen, X.H.; Bi, K.S. UFLC–MS/MS method for simultaneous determination of luteolin-7-O-gentiobioside, luteolin-7-O-β-D-glucoside and luteolin-7-O-β-D-glucuronide in beagle dog plasma and its application to a pharmacokinetic study after administration of traditional Chinese medicinal preparation: Kudiezi injection. J. Pharmaceut. Biomed. 2013, 72, 127–133. [Google Scholar]
  50. Uppugundla, N.; Engelberth, A.; Vandhana, R.S.; Clausen, E.C.; Lay, J.O.; Gidden, J.; Carrier, D.J. Switchgrass Water Extracts: Extraction, Separation and Biological Activity of Rutin and Quercitrin. J. Agric. Food Chem. 2009, 57, 7763–7770. [Google Scholar] [CrossRef] [PubMed]
  51. Cova, D.; De, A.L.; Giavarini, F.; Palladini, G.; Perego, R. Pharmacokinetics and metabolism of oral diosmin in healthy volunteers. Int J. Clin Pharm Ther. 1992, 30, 29–33. [Google Scholar]
  52. Zhang, J.K.; Sun, C.D.; Yan, Y.Y.; Chen, Q.J.; Luo, F.L.; Zhu, X.Y.; Li, X.; Chen, K.S. Purification of naringin and neohesperidin from Huyou (Citrus changshanensis) fruit and their effects on glucose consumption in human HepG2 cells. Food Chem. 2012, 135, 1471–1478. [Google Scholar] [CrossRef]
  53. Takumi, H.; Nakamura, H.; Simizu, T.; Harada, R.; Kometani, T.; Nadamoto, T.; Mukai, R.; Murota, K.; Kawai, Y.; Terao, J. Bioavailability of orally administered water-dispersible hesperetin and its effect on peripheral vasodilatation in human subjects: Implication of endothelial functions of plasma conjugated metabolites. Food Funct. 2012, 3, 389–398. [Google Scholar] [CrossRef] [PubMed]
  54. Nectoux, A.M.; Abe, C.; Huang, S.W.; Ohno, N.; Tabata, J.; Miyata, Y.; Tanaka, K.; Tanaka, T.; Yamamura, H.; Matsui, T. Absorption and Metabolic Behavior of Hesperidin (Rutinosylated Hesperetin) after Single Oral Administration to Sprague–Dawley Rats. J. Agric. Food Chem. 2019, 67, 9812–9819. [Google Scholar] [CrossRef] [PubMed]
  55. Yang, J.H.; Kondratyuk, T.P.; Marler, L.E.; Qiu, X.; Choi, Y.; Cao, H.m.; Yu, R.; Sturdy, M.; Pegan, S.; Liu, Y. Isolation and evaluation of kaempferol glycosides from the fern Neocheiropteris palmatopedata. Phytochemistry 2010, 71, 641–647. [Google Scholar] [CrossRef] [PubMed]
  56. Imran, M.; Salehi, B.; Sharifi–Rad, J.; Gondal, T.A.; Saeed, F.; Imran, A.; Shahbaz, M.; Fokou, P.V.T.; Arshad, M.U.; Khan, H. Kaempferol: A key emphasis to its anticancer potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef]
  57. Nishioka, T.; Watanabe, J.; Kawabata, J.; Niki, R. Isolation and activity of N-p-coumaroyltyramine, an α-glucosidase inhibitor in Welsh onion (Allium fistulosum). Biosci. Biotech. Biochem. 1997, 61, 1138–1141. [Google Scholar] [CrossRef]
  58. Le, P.M.; Srivastava, V.; Nguyen, T.T.; Pradines, B.; Madamet, M.; Mosnier, J.; Trinh, T.T.; Lee, H. Stephanine from Stephania venosa (Blume) Spreng Showed Effective Antiplasmodial and Anticancer Activities, the Latter by Inducing Apoptosis through the Reverse of Mitotic Exit. Phytother Res. 2017, 31, 1357–1368. [Google Scholar] [CrossRef]
  59. Li, J.; Wang, Z.; Fan, M.X.; Hu, G.W.; Guo, M.Q. Potential Antioxidative and Anti-Hyperuricemic Components Targeting Superoxide Dismutase and Xanthine Oxidase Explored from Polygonatum Sibiricum Red. Antioxidants 2022, 11, 1651. [Google Scholar] [CrossRef]
  60. Qin, N.; Li, C.B.; Jin, M.N.; Shi, L.H.; Duan, H.Q.; Niu, W.Y. Synthesis and biological activity of novel tiliroside derivatives. Eur. J. Med. Chem. 2011, 46, 5189–5195. [Google Scholar] [CrossRef] [PubMed]
  61. Fiorini, C.; David, B.; Fouraste, I.; Vercauteren, J. Acylated kaempferol glycosides from Laurus nobilis leaves. Phytochemistry 1998, 47, 821–824. [Google Scholar] [CrossRef]
  62. Lotscher, H.R.; DeJong, C.; Capaldi, R.A. Interconversion of high and low ATPase activity forms of ECF1 by the detergent lauryldimethylamine oxide. Biochemistry 1984, 23, 4140–4143. [Google Scholar] [CrossRef]
  63. Cank, K.B.; Henkin, J.M.; Cook, A.G.; Oberlies, N.H. Droplet probe: A non–destructive residue analysis of Wari ceramics from the imperial heartland. J. Archaeol. Sci. 2021, 134, 105468. [Google Scholar] [CrossRef]
  64. Hu, Y.W.; Wu, Q.; Qiao, Y.; Zhang, P.; Dai, W.T.; Tao, H.Y.; Chen, S. Disturbances in Metabolic Pathways and the Identification of a Potential Biomarker Panel for Early Cartilage Degeneration in a Rabbit Anterior Cruciate Ligament Transection Model. Cartilage 2021, 13, 1376–1387. [Google Scholar] [CrossRef]
  65. Caviglioli, G.; Valeria, P.; Brunella, P.; Sergio, C.; Attilia, A.; Gaetano, B. Identification of degradation products of Ibuprofen arising from oxidative and thermal treatments. J. Pharm. Biomed. 2002, 30, 499–509. [Google Scholar] [CrossRef] [PubMed]
  66. Valente, J.J.; Peddicord, M.B.; Rinaldi, F.A.; Kelly, K.A.; Bolgar, M.S. Identification of leachables observed in the size exclusion chromatograms of a low concentration product stored in prefilled syringes. J. Pharmceut. Biomed. 2017, 139, 133–142. [Google Scholar] [CrossRef]
  67. Su, C.H.; Cheng, Y.C.; Chang, Y.C.; Kung, T.H.; Chen, Y.L.; Lai, K.H.; Hsieh, H.L.; Chen, C.Y.; Hwang, T.L.; Yang, Y.L. Untargeted LC-MS/MS-Based Multi-Informative Molecular Networking for Targeting the Antiproliferative Ingredients in Tetradium ruticarpum Fruit. Molecules 2022, 27, 4462. [Google Scholar] [CrossRef] [PubMed]
  68. Hu, H.; Wang, Y.Y.; Zhu, H.Q.; Dong, J.; Qiao, J.T.; Kong, L.J.; Zhang, H.C. Two novel markers to discriminate poplar–type propolis from poplar bud extracts: 9-oxo-ODE and 9-oxo-ODA. J. Food Compos. Anal. 2022, 105, 104196. [Google Scholar] [CrossRef]
  69. Kato, T.; Yamaguchi, Y.; Hirano, T.; Yokoyama, T.; Uyehara, T.; Namai, T.; Yamanaka, S.; Harada, N. Unsaturated hydroxy fatty acids, the self defensive substances in rice plant against rice blast disease. Chem. Lett. 1984, 3, 409–412. [Google Scholar] [CrossRef]
  70. Zhao, B.; Tomoda, Y.; Mizukami, H.; Makino, T. 9-Oxo-(10E,12E)-octadecadienoic acid, a cytotoxic fatty acid ketodiene isolated from eggplant calyx, induces apoptosis in human ovarian cancer (HRA) cells. J. Nat. Med. Tokyo 2015, 69, 296–302. [Google Scholar] [CrossRef] [PubMed]
  71. Li, Z.X.; Dong, F.; Sun, Y.Z.; Sun, Z.H.; Song, X.Y.; Dong, Y.G.; Huang, X.C.; Zhong, J.Y.; Zhang, R.; Wang, M.Q. Qualitative and quantitative analysis of six fatty acid amides in 11 edible vegetable oils using liquid chromatography-masss pectrometry. Front Nutr. 2022, 9, 857858. [Google Scholar] [CrossRef] [PubMed]
  72. Colsch, B.; Afonso, C.; Popa, I.; Portoukalian, J.; Fournier, F.; Tabet, J.; Baumann, N. Characterization of the ceramide moieties of sphingoglycolipids from mouse brain by ESI–MS/MS: Identification of ceramides containing sphingadienine. J. Lipid Res. 2004, 45, 281–286. [Google Scholar] [CrossRef] [PubMed]
  73. Lin, D.D.; Chang, H.C.; Huang, S.H. Characterization of allegedly musk-containing medicinal products in Taiwan. J. Forensic Sci. 2004, 49, 1187–1193. [Google Scholar] [CrossRef]
  74. Zhang, Y.; Pan, Z.; Wu, K.; Yan, G.; Xu, H.; Gong, Q.; Ni, L. Research Progress on the Chemical Constituents and Pharmacological Activity of Litsea cubeba (Lour) Pers. Rec. Nat. Prod. 2023, 17, 577–594. [Google Scholar]
  75. Song, F.L.; Gan, R.Y.; Zhang, Y.; Xiao, Q.; Kuang, L.; Li, H.B. Total Phenolic Contents and Antioxidant Capacities of Selected Chinese Medicinal Plants. Int. J. Mol. Sci. 2010, 11, 2362–2372. [Google Scholar] [CrossRef] [PubMed]
  76. Zhu, H.L.; Zhang, J.C.; Li, C.F.; Liu, S.X.; Wang, L. Morinda citrifolia L. leaves extracts obtained by traditional and eco–friendly extraction solvents: Relation between phenolic compositions and biological properties by multivariate analysis. Ind. Crop. Prod. 2020, 153, 112586. [Google Scholar] [CrossRef]
  77. Qin, Y.; Wang, L.; Liu, Y.; Zhang, Q.; Li, Y.; Wu, Z.Q. Release of phenolics compounds from Rubus idaeus L. dried fruits and seeds during simulated in vitro digestion and their bio-activities. J. Funct. Foods 2018, 46, 57–65. [Google Scholar] [CrossRef]
  78. Xia, Q.; Wang, L.P.; Xu, C.C.; Mei, J.; Li, Y.F. Effects of germination and high hydrostatic pressure processing on mineral elements, amino acids and antioxidants in vitro bioaccessibility, as well as starch digestibility in brown rice (Oryza sativa L.). Food Chem. 2017, 214, 533–542. [Google Scholar] [CrossRef]
  79. Saravanakumar, K.; Sarikurkcu, C.; Sarikurkcu, R.T.; Wang, M.H. A comparative study on the phenolic composition, antioxidant and enzyme inhibition activities of two Endemic Onosma species. Ind. Crop. Prod. 2019, 142, 111878. [Google Scholar] [CrossRef]
Figure 1. Venn diagram of the non-volatile components of L. cubeba branches and leaves.
Figure 1. Venn diagram of the non-volatile components of L. cubeba branches and leaves.
Molecules 29 00788 g001
Figure 2. Standard curves for four antioxidant activity methods. ((A) Standard curve for Trolox in the ABTS method; (B) Standard curve for Trolox in the DPPH method; (C) Standard curve for Fe (Ⅱ) in the FRAP method; (D) Standard curve for Trolox in the CUPRAC method).
Figure 2. Standard curves for four antioxidant activity methods. ((A) Standard curve for Trolox in the ABTS method; (B) Standard curve for Trolox in the DPPH method; (C) Standard curve for Fe (Ⅱ) in the FRAP method; (D) Standard curve for Trolox in the CUPRAC method).
Molecules 29 00788 g002aMolecules 29 00788 g002b
Table 1. Compounds identified in the methanol extract of L. cubeba branches and leaves by UPLC-HRMS.
Table 1. Compounds identified in the methanol extract of L. cubeba branches and leaves by UPLC-HRMS.
No.RT/minCompoundsMolecular
Formula
Error/ppmm/zIon ModeCompound TypesReferences
LeavesBranches
10.8512-Aminosuccinamic acidC4H8N2O3−1.54131.04599[M − H]alkaloids[17]
20.88Gluconic acidC6H12O7−1.22195.05079[M − H]carbohydrates
30.89L-Threonic acidC4H8O5−1.26135.02972[M − H]organic acids[18]
40.893D-(−)-FructoseC6H12O6−0.76179.05597[M − H]carbohydrates[19]
50.904D-(-)-Quinic acidC7H12O6−0.89191.05595[M − H]organic acids[20]
60.908L-ThreonineC4H9NO3−0.3120.06559[M + H]+amino acids[21]
70.915D-GlucosamineC6H13NO50180.0867[M + H]+carbohydrates[22]
80.918(2R)-2,3-Dihydroxypropanoic acid C3H6O4−1.37105.01919[M − H]organic acids[23]
90.940DL-Malic acidC4H6O5−1.53133.01403[M − H]organic acids[24]
100.950D-Serine C3H7NO3−0.06106.04986[M + H]+amino acids[25]
110.982ProlineC5H9NO20.5116.07066[M + H]+amino acids[26]
120.998AdenineC5H5N50.91136.06189[M + H]+alkaloids[27]
131.019AcetophenoneC8H8O0.54121.06486[M + H]+ketones[28]
141.0244-MethoxycinnamaldehydeC10H10O2−0.18180.10186[M + H]+ethers
151.140Higenamine C16H17NO30.31272.1282[M + H]+alkaloids[29]
161.219Citric acidC6H8O7−0.32191.01967[M − H]organic acids[30]
171.232Nicotinic acidC6H5NO20.14124.03932[M + NH4]+alkaloids[31]
181.238Pipecolic acidC6H11NO20.45130.08631[M + H]+alkaloids[32]
191.331Methylmalonic acidC4H6O4−1.03117.01921[M − H]organic acids[33]
201.335Fumaric acidC4H4O4−1.05115.00356[M − H]organic acids[34]
211.373IsoleucineC6H13NO20.35132.10195[M + H]+amino acids[35]
221.456L-NorleucineC6H13NO20.41132.10196[M + H]+amino acids[36]
231.615L-N-Acetylphenylalaninol C11H15NO2−0.04194.11755[M + H]+alkaloids
241.989(-)-SalsolineC11H15NO2−0.17194.11752[M + H]+alkaloids
252.089Gentisic acid 5-O-β-glucoside C13H16O9−0.85315.07189[M − H]organic acids
262.098Indole-3-acetic acidC10H9NO2−0.03176.07061[M + H]+alkaloids[37]
273.396trans-3-Indoleacrylic acid C11H9NO2−0.39188.07054[M + H]+alkaloids[38]
285.066NorisoboldineC18H19NO40.42314.13882[M + H]+alkaloids[39]
295.0913-(4-Hydroxyphenyl)-3-oxopropyl β-D-glucopyranosideC15H20O8−0.82327.10827[M − H]phenols[40]
305.8885.261BoldineC19H21NO4−0.07328.15432[M + H]+alkaloids[41]
315.486Dihydrophaseic acid C15H22O5−0.41281.13933[M − H]organic acids[42]
326.2136.072(S)-BoldineC19H21NO40.26328.15442[M + H]+alkaloids[41]
336.6366.487GlaufinineC19H21NO40.07328.15436[M + H]+alkaloids[43]
346.604Kaempferol 3-sophoroside C27H30O16−0.28609.14594[M − H]flavonoids[44]
356.765IsocorydineC20H23NO40.26342.17007[M + H]+alkaloids[45]
367.5267.817RutinC27H30O16−0.69609.14558[M − H]flavonoids[41]
377.117IsoquercetinC21H20O12−0.28463.08796[M − H]flavonoids[46]
387.316KaempferitrinC27H30O14−0.31577.15588[M − H]flavonoids[47]
397.551laurotetanineC19H21NO40.07328.15436[M + H]+alkaloids[41]
407.5575-(4-Hydroxypentyl)-1,3-benzenediol C11H16O3−0.11197.1172[M + H]+phenols
417.589Corydine C20H23NO4−0.21342.16991[M + H]+alkaloids[43]
427.631Trifolin C21H20O11−0.34447.09298[M − H]flavonoids[48]
437.951CynarosideC21H20O11−0.26447.09299[M − H]flavonoids[49]
448.048QuercitrinC21H20O11−0.36447.09312[M − H]flavonoids[41]
458.246CassythicinC19H19NO40.33326.13879[M + H]+alkaloids[50]
468.349Diosmin C28H32O15−0.77607.16638[M − H]flavonoids[51]
478.503Neohesperidin C28H34O15−0.78609.18202[M − H]flavonoids[52]
488.644HesperetinC16H14O60.57303.08649[M + H]+flavonoids[53]
498.648HesperidinC28H34O150.65611.19739[M + H]+flavonoids[54]
509.196AfzelinC21H20O10−0.63431.0981[M − H]flavonoids[55]
519.443Kaempferol C15H10O60.05287.05503[M + H]+flavonoids[56]
529.930N-p-CoumaroyltyramineC17H17NO30.32284.12821[M + H]+phenylpropanoids[57]
5310.391CrebanineC20H21NO40.3340.15444[M + H]+alkaloids[58]
5410.57610.454MoupinamideC18H19NO40.24314.13876[M + H]+alkaloids[59]
5510.869Tiliroside C30H26O13−0.92593.12952[M − H]flavonoids[60]
5614.44714.02915-Hexadecynoic acid C16H28O2−0.2253.21616[M + H]+organic acids
5714.5083-[[6-Deoxy-3,4-bis-O-[(2E)-3-(4-hydroxyphenyl)-1-oxo-2-propen-1-yl]-α-L-mannopyranosyl]oxy]-5,7-dihydroxy-2-(4-hydroxyphenyl)-4H-1-benzopyran-4-oneC39H32O14−0.9723.17128[M − H]flavonoids[61]
5815.60915.177Lauryldimethylamine oxide C14H31NO−0.09230.24782[M + H]+alkaloids[62]
5916.099Aurantiamide acetateC27H28N2O40.05445.21222[M + H]+alkaloids[63]
6016.2862-Amino-1,3,4-octadecanetriol C18H39NO30.55318.30045[M + H]+alcohols[64]
6116.2904-EthylbenzaldehydeC9H10O0.59135.08052[M + H]+esters[65]
6216.2974-Ethoxy ethylbenzoateC11H14O3−0.12195.10155[M + H]+esters[66]
6316.529Schinifoline C17H23NO−0.02258.18524[M + H]+alkaloids[67]
6416.88216.9191,3:2,4-Di(p-ethylbenzylidene)sorbitol C24H30O60.51415.2117[M + H]+alcohols
6517.0979-Oxo-ODEC18H30O30.09295.2268[M + H]+organic acids[68]
6617.36813(S)-HOTrEC18H30O30.08293.21224[M − H]organic acids[69]
6718.3959-Oxo-10(E),12(E)-octadecadienoic acidC18H30O30.4295.22687[M + H]+organic acids[70]
6819.14712-Oxo-10,15(Z)-phytodienoic acid C18H28O30.45293.21124[M + H]+organic acids
6919.440Linoleoyl ethanolamide C20H37NO20.28324.28979[M + H]+alcohols[71]
7020.267Sphingosine (d18:1) C18H37NO20.6300.28989[M + H]+alcohols[72]
7121.806MusconeC16H30O−0.21239.23689[M + H]+ketones[73]
7221.829Pheophorbide A C35H36N4O50.14593.27594[M + H]+alkaloids
–: not detected, : indicates compounds identified for the first time from L. cubeba.
Table 2. Antioxidant activity of methanol extracts from branches and leaves of L. cubeba.
Table 2. Antioxidant activity of methanol extracts from branches and leaves of L. cubeba.
PartsABTS
(TEAC µmol/g)
DPPH
(TEAC µmol/g)
FRAP
(Fe(Ⅱ) µmol/g)
CUPRAC
(TEAC µmol/g)
Branches42.75 ± 3.947.55 ± 0.4322.21 ± 0.5342.65 ± 0.35
Leaves57.78 ± 1.879.29 ± 0.3928.68 ± 0.7845.89 ± 0.27
Table 3. Standard curve equations for four antioxidant activity methods.
Table 3. Standard curve equations for four antioxidant activity methods.
MethodEquationR2
ABTSy = −0.32338x + 0.767450.99755
DPPHy = −1.23810x + 0.570000.99133
FRAPy = 0.20124x + 0.108700.99987
CUPRACy = 0.43161x + 0.180270.99903
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

Dai, W.; Li, B.; Xiong, Y.; Dai, L.; Tian, Y.; Zhang, L.; Wang, Q.; Qian, G. Non-Volatile Component and Antioxidant Activity: A Comparative Analysis between Litsea cubeba Branches and Leaves. Molecules 2024, 29, 788. https://doi.org/10.3390/molecules29040788

AMA Style

Dai W, Li B, Xiong Y, Dai L, Tian Y, Zhang L, Wang Q, Qian G. Non-Volatile Component and Antioxidant Activity: A Comparative Analysis between Litsea cubeba Branches and Leaves. Molecules. 2024; 29(4):788. https://doi.org/10.3390/molecules29040788

Chicago/Turabian Style

Dai, Wei, Boyi Li, Yanli Xiong, Liping Dai, Yuan Tian, Liangqian Zhang, Qi Wang, and Guoqiang Qian. 2024. "Non-Volatile Component and Antioxidant Activity: A Comparative Analysis between Litsea cubeba Branches and Leaves" Molecules 29, no. 4: 788. https://doi.org/10.3390/molecules29040788

Article Metrics

Back to TopTop