Next Article in Journal
Innovative Fermentation Approach Employing Lachancea thermotolerans for the Selective Production of High-Acidity Wines, Designed for Blending with Low-Acidity Counterparts to Achieve Chemically and Organoleptically Balanced Final Compositions
Previous Article in Journal
Biodegradable Quercetin-Incorporated Poly(Lactic Acid)/Chitosan Functional Films: A Study of the Properties and Application in Enhancing Fish Preservation
Previous Article in Special Issue
Health Benefits of Traditional Sage and Peppermint Juices: Simple Solutions for Antioxidant and Antidiabetic Support
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Ultrasound-Optimized Extraction and Multi-Target Mechanistic Analysis of Antioxidant and Hypoglycemic Effects of Amomum villosum Essential Oil

1
Guangxi Key Laboratory of Research on Medical Engineering Integration and Innovation, Medical College, Guangxi University of Science and Technology, Liuzhou 545006, China
2
Guangxi Industrial Technology Research Institute on Karst Rocky Desertification Control Co., Ltd., Nanning 530012, China
*
Author to whom correspondence should be addressed.
Foods 2025, 14(16), 2772; https://doi.org/10.3390/foods14162772 (registering DOI)
Submission received: 24 June 2025 / Revised: 26 July 2025 / Accepted: 6 August 2025 / Published: 9 August 2025

Abstract

Amomum villosum, a medicinal and edible plant, has shown promise in improving digestive health; however, the mechanisms underlying its antioxidant and hypoglycemic effects remain unclear. This study aimed to optimize the extraction of A. villosum essential oil (AVEO) and elucidate its bioactive potential. Ultrasound-assisted extraction yielded 3.84% AVEO under optimal conditions. Gas chromatography–mass spectrometry combined with SwissADME analysis identified nine active components, including bornyl acetate, (−)-Spathulenol, and (−)-Pogostol. In vitro assays demonstrated potent α-glucosidase inhibition (IC50: 0.99 mg/mL) and strong free radical scavenging activities against 1,1-diphenyl-2-picrylhydrazyl (IC50: 0.87 mg/mL), hydroxyl (IC50: 0.18 mg/mL), and superoxide anion radicals (IC50: 0.01 mg/mL). A significant positive correlation was observed between its antioxidant and hypoglycemic activities. Network pharmacology identified 11 core targets involved in oxidative stress and glucose metabolism, with functional enrichment pointing to the PPAR and steroid hormone signaling pathways. Molecular docking confirmed stable binding affinities of bornyl acetate, (−)-spathulenol, and (−)-pogostol to JAK2, NCOA2, and PPARA via hydrogen bonding and hydrophobic interactions. These findings provide a mechanistic basis for the dual antioxidant–hypoglycemic effects of AVEO and support its potential application in the development of functional foods and natural therapeutics targeting metabolic disorders.

1. Introduction

Amomum villosum Lour., a medicinal food homologous plant native to tropical Asia, has traditionally been used in folk medicine across southern China and Southeast Asia to improve digestion, alleviate vomiting, and reduce abdominal distension [1,2,3]. Currently, it is also used as a natural ingredient and flavoring agent in wines, teas, candies, and cosmetics [4]. The primary constituents of A. villosum Lour include monoterpenes, sesquiterpenes, diterpenes, flavonoids, phenolic compounds, and polysaccharides [1]. The essential oil of A. villosum (AVEO), with bornyl acetate, camphor, and other terpenoids as its core components, is regarded as the primary material basis for its pharmacological activities [5]. This essential oil is listed as a quality control marker in the Chinese Pharmacopoeia, and its high content is generally regarded as an indicator of good quality [5,6].
Given its therapeutic value and significance in functional food development, the enhancement of AVEO yield represents a critical research priority. In terms of extraction technologies, traditional steam distillation offers low yields (~3% according to the Chinese Pharmacopoeia 2020, or 1.50–2.02% v/w in some studies) [5,7], and the high temperatures involved risk degrading thermolabile compounds such as bornyl acetate [5], thereby reducing bioactivity. Among emerging extraction technologies, ultrasound-assisted extraction (UAE) has shown promise as a highly efficient and selective method [8,9,10,11]. The ultrasonic cavitation theory posits that transient high pressure and localized heat generated by ultrasound selectively disrupt plant cell walls, releasing intracellular bioactive compounds while minimizing thermal degradation [12,13]. UAE aligns well with green chemistry principles and is considered a sustainable alternative to conventional methods [8]. However, optimizing ultrasound parameters is highly dependent on the properties of the plant matrix. For example, Gavahian et al. [14] demonstrated that cellulase pretreatment enhanced extraction uniformity in citrus peels; however, its applicability to lipid-rich Zingiberaceae species like A. villosum remains unclear. Additionally, the composition of AVEO varies markedly across different geographical origins [15], largely due to differences in terpene synthase gene expression [4]. These variations demand the development of more universal and adaptable UAE protocols. Therefore, tailored UAE optimization accounting for species-specific factors represents a critical unmet need in AVEO extraction.
Regarding the bioactivities of AVEO, partial experimental evidence has been accumulated by existing studies. Antimicrobial [5,16], antioxidant [17,18], anti-obesity [19], anti-hyperuricemic, and anti-gout arthritic activities [20] have been demonstrated for A. villosum extracts (not limited to essential oils). Recent in vitro and murine model studies have highlighted AVEO’s antioxidant, hypoglycemic, and anti-inflammatory characteristics [21,22], with demonstrated effects against gastric cancer [23], non-alcoholic fatty liver disease [22], alcoholic liver disease [24], and allergic rhinitis [25]. These findings underscore AVEO’s potential as a functional food additive and pharmaceutical ingredient [26]. However, the regulatory pathways through which AVEO exerts these activities remain insufficiently understood.
The multicomponent synergistic effects of plant volatile oils and their cross-pathway regulatory networks require deeper mechanistic investigation. Network pharmacology and molecular docking have recently emerged as powerful tools for analyzing the multi-target effects of plant-derived compounds [26,27]. Emerging evidence indicates that antioxidant effects in A. villosum stem–leaf extracts may involve novel bioactive compounds that stabilize PGAM5 binding through residue-specific hydrogen bonds (Lys93/Arg214) [26]. Reference pathways regulating oxidative stress in other essential oils include γ-aminobutyric acidergic synapses [28], AGE-RAGE [29], calcium signaling [30], and PI3K/AKT/mTOR cascades [31], with the latter recognized as a conserved hypoglycemic pathway [31]. While multicomponent bioactivities attract growing interest, synergistic mechanisms remain less explored than single-compound effects. It has been reported that activation of the Nrf2-Keap1 pathway constitutes an established antioxidant mechanism with a demonstrated capacity to ameliorate diabetes and its complications [32]. Nevertheless, it remains unclear whether AVEO exerts its effects via this pathway. Furthermore, the precise mechanisms underlying the synergistic actions of AVEO components in oxidative stress and glucose metabolism are still poorly understood.
To address these knowledge limitations, this study proposes an integrated “process optimization-functional validation-mechanistic elucidation” framework. First, a Box–Behnken response surface methodology was used to optimize ultrasound parameters (extraction time, ultrasonic power, and solvent ratio) for maximum yield and component retention, with validation across samples of different origins and harvest years. Second, α-glucosidase inhibition and radical scavenging assays were conducted to quantify the hypoglycemic and antioxidant capacities of AVEO. Finally, gas chromatography–mass spectrometry (GC-MS) analysis, network pharmacology, and molecular docking were used to construct a “component–target–pathway” network to identify key bioactive compounds and their molecular targets, elucidating AVEO’s metabolic regulatory mechanisms. We hypothesized that (1) UAE enhances AVEO extraction efficiency; (2) AVEO exhibits potent hypoglycemic and antioxidant activities; and (3) the hypoglycemic–antioxidant synergy may be mediated by metabolic or oxidative stress-related pathways such as PI3K/AKT and Nrf2-Keap1. The novel integration of process optimization, bioactive profiling, and network pharmacology-based mechanistic investigation represents the core innovation of this study, addressing both extraction applicability and underlying mechanistic insights. The findings of this study provide a scalable technical solution for AVEO extraction and a mechanistic basis for the functional development of A. villosum as a natural food additive.

2. Materials and Methods

2.1. Plant Material

Fruits of Amomum villosum Lour. were collected in 2023 and 2022 from cultivation sites in Baise (23.90° N, 106.62° E) and Long’an (23.18° N, 107.45° E), Guangxi, China, respectively. The samples were provided by Guangxi Botanical Garden of Medicinal Plants. Specimens were authenticated by Dr. Hu Renchuan (Guangxi Institute of Chinese Medicine) and Dr. Huang Yuan (Guangxi Botanical Garden of Medicinal Plants). Each sample consisted of mixed fruits collected from three randomly 1 × 1 m quadrats at the cultivation base. The samples were freeze-dried for 72 h until constant weight, ground, sieved through a 40-mesh screen, vacuum-sealed, and stored at –20 °C until analysis. They were labeled as Amomum1, Amomum2, Amomum3, and Amomum4 (Table 1).

2.2. Chemicals and Reagents

α-Glucosidase (EC 3.2.1.20, activity ≥ 10 U/mg), 1,1-diphenyl-2-picrylhydrazyl (DPPH), and salicylic acid were purchased from Sigma-Aldrich, St. Louis, MO, USA. Acarbose (purity ≥ 98%) was obtained from Shanghai Yuanye Bio-Technology Co., Ltd., Shanghai, China.

2.3. Experimental Methods

2.3.1. Optimization of Essential Oil Extraction

  • Extraction procedure: Essential oil was extracted using a customized ultrasound-essential oil co-extraction system (Model KQ5200DV, Shanghai Precision Instrument Co., Shanghai, China) following Method A of the Chinese Pharmacopoeia (2020 Edition, Part IV). Briefly, 15.0 g of A. villosum powder was mixed with ultrapure water at a preset solvent-to-material ratio and subjected to UAE at 25 ± 1 °C. The extraction yield (Y, %) was calculated using the formula:
    Y ( % ) = V m × 100
    where V is the volume of extracted essential oil (mL), and m is the mass of the powder sample (g).
  • Single-factor experiments: To identify optimal conditions, the following parameters were evaluated individually:
    • Ultrasound time: 5, 10, 15, 20, and 25 min under fixed conditions (200 W, 1:20 g/mL).
    • Solvent-to-material ratio: 1:15, 1:20, 1:25, 1:30, and 1:35 g/mL after, optimizing the ultrasound time (15 min).
    • Ultrasound power: 120, 160, 200, 240, and 280 W (using the optimal time and ratio).
    • Each experiment was performed in triplicate.
  • Box–Behnken response surface design: Three independent variables—ultrasound time (A: 10, 15, 20 min), solvent-to-material ratio (B: 1:20, 1:25, 1:30 g/mL), and ultrasound power (C: 160, 200, 240 W)—were optimized using Design-Expert 13.0 software (Stat-Ease, Inc., Minneapolis, MN, USA). The design included 17 runs with 5 center points. A quadratic polynomial model was used for data fitting.

2.3.2. Hypoglycemic and Antioxidant Activity Assays

The residual moisture in the extracted AVEO was initially dehydrated using anhydrous sodium sulfate, followed by filtration through a 0.22 μm membrane for subsequent use.
  • α-Glucosidase Inhibition:
Adapted from Yang et al. [33], 25 μL of essential oil ethanol solution (0.5, 1.0, 1.5, 2.0, 2.5 mg/mL) and 25 μL of α-glucosidase (1 U/mL in phosphate-buffered saline, pH 6.8) were preincubated in a 96-well plate at 37 °C for 10 min. Then, 50 μL of 2.5 mM p-nitrophenyl-α-D-glucopyranoside was added, and the mixture was incubated for 30 min. The reaction was terminated by adding 100 μL of 0.2 M Na2CO3. Absorbance was measured at 405 nm (Multiskan GO, Thermo Fisher Scientific Co., Ltd., Waltham, MA, USA). The inhibition rate was calculated as follows:
I n h i b i t i o n ( % ) = ( 1 A 1 A 3 A 2 A 4 ) × 100
where A1 is the absorbance of the sample (AVEO) with enzyme; A2 is the control (buffer + enzyme); A3 is the background (AVEO + buffer); and A4 is the blank (buffer only). Acarbose was used as the positive control to validate the assay system and provide a reference for α-glucosidase inhibitory activity.
  • Free Radical Scavenging Assays:
    • DPPH Assay: 1 mL of AVEO solution (0.2–1.0 mg/mL) was mixed with 1 mL of 0.1 mM DPPH ethanol solution and incubated in the dark for 30 min. Absorbance was measured at 517 nm [34].
    • Hydroxyl Radical (·OH) Assay: 1 mL of AVEO solution was mixed with 0.5 mL of 6 mM FeSO4 and 0.5 mL of 8.8 mM H2O2. After 10 min, 0.5 mL of 6 mM salicylic acid was added, and absorbance was measured at 510 nm after 30 min at 37 °C [35].
    • Superoxide Anion (O2·) Assay: 0.5 mL of AVEO solution was added to 1.5 mL of 50 mM Tris-HCl (pH 8.2) and incubated at 25 °C for 20 min. Then, 0.5 mL of 3 mM pyrogallol (in 10 mM HCl) was added. After 5 min, the reaction was terminated with HCl, and absorbance was measured at 320 nm [36].
Scavenging activity (%) was calculated as follows:
S c a v e n g i n g ( % ) = ( 1 A 1 A 2 A 0 ) × 100
where A1 is the sample; A2 is the background (radical source replaced with buffer); and A0 is the blank (sample replaced with buffer). Ascorbic acid (vitamin C) was used as the positive control to validate the radical scavenging assay system and serve as a reference for antioxidant activity.

2.3.3. GC-MS Analysis of AVEO Components

The samples were dried with anhydrous sodium sulfate and filtered through a 0.22 μm filter membrane. GC-MS was conducted using an Agilent 7890A–5975C system (Agilent Technologies, Inc., Santa Clara, CA, USA) equipped with an HP-1ms capillary column (60.0 m × 250 μm × 0.25 μm). The initial oven temperature was set to 60 °C (held for 1 min), ramped to 130 °C at 5 °C/min (held for 1 min), then to 160 °C at 2 °C/min (no hold, i.e., 0 min), and finally to 230 °C at 5 °C/min. Helium was used as the carrier gas at a flow rate of 1.2 mL/min. Mass spectrometry conditions: EI source (70 eV), ion source temperature 230 °C, scan range 35–500 m/z. Compounds were identified by matching mass spectra against the NIST 20 library (match factor > 85). Relative content (Ci) was determined by peak area normalization and was retrieved from the system-generated report.
C i = A i A 1 + A 2 + A i + + A n × 100 %
where the following definitions apply: Ci—percentage content of the i-th identified compound (%); Ai—chromatographic peak area of the i-th compound; n—total number of identified compounds.

2.4. Data Analysis

2.4.1. Validation of Response Surface Model

A quadratic regression model was developed using Design-Expert 13.0 (Stat-Ease, Inc., Minneapolis, MN, USA) to relate extraction yield (Y) to ultrasound time (A), solvent-to-material ratio (B), and ultrasound power (C). Model significance was assessed using analysis of variance (ANOVA), including p-values, lack of fit, coefficient of determination (R2), and adjusted R2. Residual normality was verified using the Shapiro–Wilk test (SPSS 25.0, IBM Corp., Armonk, NY, USA). All experiments were conducted independently in triplicate.

2.4.2. Statistical Analysis of Bioactivity Data

Data from α-glucosidase inhibition and radical scavenging assays were analyzed using one-way ANOVA, followed by Tukey’s post hoc test (p < 0.05). Dose–response curves and IC50 values (95% confidence interval [CI]) were generated using a four-parameter logistic regression model (GraphPad Prism 10.0, GraphPad Software, LLC, San Diego, CA, USA). Pearson correlation and heat maps were used to visualize associations between antioxidant and hypoglycemic activities.

2.4.3. Network Pharmacology Analysis

Candidate compounds identified via GC-MS were screened for high gastrointestinal absorption—a parameter indicating favorable pharmacokinetic properties, using SwissADME analysis (http://www.swissadme.ch/index.php, accessed on 30 May 2025) [36,37]. Target prediction was conducted using SwissTargetPrediction (http://www.swisstargetprediction.ch/, accessed on 30 May 2025) [38], and gene names were standardized using UniProt (https://www.uniprot.org/, accessed on 30 May 2025) [39]. Disease-related genes for oxidative stress and diabetic nephropathy were obtained from GeneCards (https://www.genecards.org/, accessed on 31 May 2025) [40] and OMIM (https://www.omim.org/, accessed on 31 May 2025) [41].
Common targets between A. villosum components and antioxidant–hypoglycemic genes were identified using Venny 2.1.0. A “compound–target–disease” network was constructed in Cytoscape 3.9.1 (Cytoscape Consortium, San Diego, CA, USA), with node degree (Degree) used to identify core targets. Higher degree values indicated greater functional importance within the network [42].
Pathway Enrichment and Protein–Protein Interaction Network: DAVID (https://david.ncifcrf.gov/, accessed on 2 June 2025) [43] was used to perform gene ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses on intersection targets. The top 10 pathways (p < 0.05) related to antioxidant and hypoglycemic activities were visualized. A protein–protein interaction (PPI) network for the compound-disease intersection was generated using STRING (https://cn.string-db.org/, accessed on 3 June 2025) [44], and core modules were identified using the MCODE plugin.
Molecular Docking Validation: Key bioactive compounds and target proteins were subjected to molecular docking. Receptor structures with a resolution ≤ 2.5 Å were retrieved from the PDB database (https://www.rcsb.org/, accessed on 5 June 2025) [45], and three-dimensional of ligands structures were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/, accessed on 6 June 2025) [46]. Ligands and water molecules were removed using PyMOL (Schrödinger, LLC, New York, NY, USA). Docking simulations were performed using AutoDockTools-1.5.7 (The Scripps Research Institute, La Jolla, CA, USA), with docking boxes set to enclose the entire binding pocket. Lower binding energy is generally associated with strong ligand–receptor interactions [47]. A binding affinity ≤ −5.0 kcal/mol is considered indicative of strong binding potential [48,49], representing as an effective interaction between the ligand and the target compound [50]. The results were visualized using Discovery Studio 4.5 (Dassault Systèmes BIOVIA, San Diego, CA, USA).

3. Results

3.1. Optimization of AVEO

Under single-factor experimental conditions, the optimal parameters for essential oil extraction were identified as follows: ultrasound time of 15 min, solvent-to-material ratio of 1:25 g/mL, and ultrasound power of 200 W (Figure 1).
A quadratic polynomial regression model was developed based on 17 Box–Behnken experimental runs (Table S1), describing the relationship among ultrasound time (A), solvent-to-material ratio (B), ultrasound power (C), and essential oil yield (Y), as follows:
Y = 3.77 − 0.0075A + 0.1075B + 0.0325C − 0.1175AB + 0.1025AC − 0.0675BC − 0.3808A2 − 0.2457B2 − 0.4257C2
ANOVA (Table 2) indicated that the model was statistically significant (p = 0.0011), with a non-significant lack-of-fit term (p = 0.9379), confirming that the model errors primarily originated from random factors rather than systematic bias. The high correlation coefficient (R2 = 0.95) validated the reliability of the experimental design and the accuracy of the model.
Standardized effect analysis (Figure 2) ranked the key influencing factors in the following order: solvent-to-material ratio > ultrasound power > ultrasound time.
The predicted optimal conditions from the model were 15.00 min ultrasound time, 1:26.13 g/mL solvent-to-material ratio, and 200.59 W ultrasound power, yielding a theoretical essential oil output of 3.79%.
A validation experiment using the adjusted ultrasound power (200 W) achieved an actual oil yield of 3.84 ± 0.08% (RSD = 1.95%, n = 3), representing only a 1.3% deviation from the predicted value. Additional validation with Amomum 2–4 yielded consistent results, with an average essential oil yield of 3.47 ± 0.10%, confirming the model’s applicability and stability across samples.

3.2. Hypoglycemic and Antioxidant Properties of AVEO

3.2.1. α-Glucosidase Inhibitory Activity

AVEO exhibited a dose-dependent inhibitory effect on α-glucosidase (Figure 3A). Among the tested samples, Amomum3 demonstrated the strongest inhibitory activity, with an IC50 value of 0.99 ± 0.05 mg/mL, significantly outperforming the other congeners (IC50 = 1.10–1.87 mg/mL). Notably, Amomum1 achieved the highest inhibition rate of 87.07 ± 1.23% at a concentration of 2.5 mg/mL, comparable to that of the clinical hypoglycemic drug acarbose. These findings suggest that AVEO, particularly Amomum1 and Amomum3, holds promise as a natural sugar-lowering ingredient for functional food applications.

3.2.2. Antioxidant Capacity

AVEO exhibited strong antioxidant activity by scavenging DPPH radicals, hydroxyl radicals (·OH), and superoxide anions (·O2) (Figure 3B–D). At a concentration of 1 mg/mL, the scavenging rates were 57.79%, 84.59%, and 65.61% for DPPH, ·OH, and ·O2, respectively. Amomum1 exhibited the strongest DPPH radical scavenging activity (IC50 = 0.78 ± 0.02 mg/mL), while Amomum3 showed the highest ·O2 scavenging activity (IC50 = 0.012 ± 0.01 mg/mL). Conversely, Amomum4 demonstrated superior ·OH scavenging ability (IC50 = 0.18 ± 0.04 mg/mL), comparable to the standard antioxidant ascorbic acid (vitamin C). Overall, Amomum3 was particularly effective in scavenging both ·OH and ·O2.

3.2.3. Functional Synergy Analysis

Spearman correlation analysis (Figure 3E) revealed a significant positive correlation between α-glucosidase inhibitory activity and DPPH scavenging capacity (r = 0.82, p < 0.01), as well as a significant negative correlation with ·OH scavenging capacity (r = −0.65, p < 0.05). These relationships suggest that the inhibitory and antioxidant effects of AVEO are mediated by overlapping bioactive compounds responsible for both α-glucosidase inhibition and DPPH radical scavenging. The negative correlation between its α-glucosidase inhibitory activity and ·OH scavenging capacity may indicate substrate competition or spatial antagonism at shared binding sites. Collectively, these findings support the potential of AVEO as a dual-functional food additive possessing both antioxidant and glucose-regulating properties.

3.3. Composition Analysis of AVEO

GC-MS analysis of Amomum3 AVEO (Figure 4) identified 15 characteristic components (Table 3), with their relative contents quantified using chromatographic peak area normalization. The predominant compounds included bornyl acetate (52.1%), (+)-2-bornanone (15.91%), cyclohexane-1-butenylidene (5.21%), and 5,5-dimethyl-1,3-hexadiene (3.52%).

3.4. Network Pharmacology Analysis of the Antioxidant and Hypoglycemic Mechanisms of AVEO

3.4.1. Screening of Candidate Compounds and Potential Targets

Nine candidate compounds with high gastrointestinal absorption were identified from AVEO using GC-MS and SwissADME screening (Table 3, A1–A9). These included primarily monoterpenoid—(+)-2-bornanone, bornyl acetate, linalool, isoborneol, terpinen-4-ol, and α-terpineol—and sesquiterpenoids, such as (–)-spathulenol, (–)-pogostol, and α-bisabolol.
Following UniProt standardization, 127 well-characterized target genes were mapped to these compounds. Through multi-database integration (see Methods), 5014 antioxidant-related and 4049 hypoglycemic-related genes were identified.
A total of 56 overlapping targets were identified among the component-related, antioxidant, and hypoglycemic gene sets (Figure 5A). An “A. villosum-Active Component-Disease-Target” network was constructed using Cytoscape 3.9.0 (Figure 5B,C). All nine compounds exhibited a high degree of centrality, indicating their central roles as regulators of antioxidant and hypoglycemic pathways.
A PPI network of shared targets was established (Figure 5D), consisting of 56 proteins interconnected by 199 edges (average node degree: 7.11; average local clustering coefficient: 0.50). Enrichment analysis showed statistically significant functional involvement (p < 1.0 × 10−16), suggesting critical functions in antioxidant and hypoglycemic processes. Core genes identified via topological analysis (Betweenness unDir, Closeness unDir, Degree metrics exceeding mean values) included CYP3A4, PTGS2, IL6, PTPN1, NR3C1, PARP1, CYP19A1, ESR1, NCOA2, JAK2, HMGCR, PPARA, and CYP2C19 (Figure 5E), underscoring their pivotal regulatory roles in both antioxidant and hypoglycemic responses. Node size was scaled according to the degree value. Key values of the core targets are shown in Figure 5F.

3.4.2. Functional Enrichment

Functional enrichment analyses of the 56 intersecting target genes associated with the antioxidant and hypoglycemic activities of AVEO identified 123 significantly enriched biological processes, 25 cellular components, and 53 molecular functions (p < 0.05). The top 10 terms most closely associated with oxidative stress and glucose regulation in each category were visualized (Figure 6A–C). GO biological process analysis (Figure 6A) revealed significant enrichment in nuclear receptor-mediated steroid hormone signaling and intracellular receptor signaling pathways. GO cellular component analysis (Figure 6B) identified the chromatin, endoplasmic reticulum membrane, and cytosol as the primary locations. GO molecular function analysis (Figure 6C) showed significant enrichment in nuclear receptor activity, enzyme binding, and protein binding.
KEGG pathway analysis (Figure 6D) identified 18 significantly enriched pathways (p < 0.05), with the peroxisome proliferator-activated receptor (PPAR) signaling pathway, arachidonic acid metabolism, and general metabolic pathways being the most critical. These findings provide molecular-level evidence that the essential oil exerts dual antioxidant and hypoglycemic effects through synergistic regulation of nuclear hormone signaling, lipid metabolism, and oxidative stress homeostasis.

3.5. Molecular Docking Validation

Molecular docking studies revealed that compounds A4–A9 in AVEO (e.g., terpinen-4-ol, α-terpineol, bornyl acetate, (−)-spathulenol, (−)-pogostol, α-bisabolol) exhibited significant multi-target binding activities against core targets involved in both antioxidant and hypoglycemic pathways (e.g., CYP3A4, CYP2C19, PTGS2, NR3C1, PPARA, NCOA2, JAK2, ESR1) (Figure 7A). Most compounds exhibited binding energies below −6.0 kcal/mol across all targets, indicating relatively stable binding energies. The lowest binding energy was observed for (−)-spathulenol binding to JAK2 (−9.0 kcal/mol), stabilized by hydrogen bonds (e.g., Ser936) and hydrophobic interactions (e.g., Tyr930, Leu855, Leu982, Leu983, Ala880, and Val863) (Figure 7B). Similarly, (−)-pogostol showed stable binding to NCOA2 (−8.8 kcal/mol), primarily mediated through hydrophobic interactions (e.g., Leu346, Leu349, Leu384, Leu387, Leu525, Ala350, Trp383, and Phe404) (Figure 7E). In addition to NCOA2 and JAK2, both (−)-spathulenol and (−)-pogostol also stably bound to CYP3A4 and CYP2C19 via hydrophobic interactions, with binding energies below −8.0 kcal/mol (Figure 7C,D,F–H). Notably, bornyl acetate (52.10% by GC-MS) bound stably to all core targets with binding energies below −5.2 kcal/mol. It showed the strongest interaction with PPARA (−6.9 kcal/mol) through hydrogen bonds (e.g., Asn219, Met220) and hydrophobic contacts (e.g., Leu321, Val324) (Figure 7I). Collectively, these computational findings suggest that the key bioactive constituents’ AVEO may synergistically interact with multiple targets through strong binding affinities, providing preliminary mechanistic support for its potential dual antioxidant–hypoglycemic effects.

4. Discussion

4.1. Significant Optimization of AVEO Ultrasonic Extraction Process

The UAE protocol established in this study achieved synergistic optimization of yield (3.84%) and retention of bioactive compounds (52.10%) in AVEO (Figure 1 and Figure 4). Compared with previous reports stating that A. villosum contains 1.7–3.0% essential oil with bornyl acetate (its major component) accounting for 5–47% [4,51], our results show superior performance in both yield and bioactive compound retention. These findings not only strongly support the first hypothesis of this study but also fully validate the significant value of UAE technology in plant essential oil extraction. Model predictions closely matched the experimental outcomes with only a 1.3% deviation, indicating strong model reliability. Regression analysis revealed nonlinear regulatory effects of ultrasonic parameters (time, power, solvent-to-material ratio) on extraction yield, with the solvent-to-material ratio (parameter B) identified as the critical factor. This aligns with previous research demonstrating that an optimized solvent-to-material ratio enhances essential oil yield by improving solvent penetration efficiency [52], while ultrasonic cavitation-induced microjets accelerate cell wall disruption to release lipophilic compounds [12,53]. Despite known variations in plant metabolite profiles due to geographical and environmental factors [54,55], the optimized UAE protocol consistently yields 3.47 ± 0.10% across multi-year and multi-region samples, indicating its robustness against raw material variability. Compared to traditional steam distillation (4–6 h) and supercritical CO2 extraction [51], UAE reduced extraction time to just 1 h [12] while increasing bornyl acetate retention by 5.10% (Figure 4). These advantages likely result from two key ultrasonic mechanisms: (1) enhanced mass transfer efficiency through cavitation [56,57] and (2) localized high-temperature/high-pressure microenvironments that selectively protect thermolabile terpenes [58,59]. Notably, excessive ultrasonic power triggered a “threshold ultrasound” phenomenon, where reaction efficiency decreased despite increased energy input [60]. Our study observed this effect, and optimized parameters (200 W, 15 min) were selected to maximize yield without compromising compound stability. The high predictive accuracy (R2 = 0.95) confirms the reliability of response surface methodology for complex process optimization [61], providing a paradigm for the green extraction of natural products.

4.2. AVEO Exhibited Excellent Hypoglycemic and Antioxidant Activities

In vitro assays demonstrated that AVEO (Amomum3) exhibited potent α-glucosidase inhibitory activity (IC50 = 0.99 ± 0.05 mg/mL), with 87.07% maximum inhibition at high concentrations (Figure 3A). Although significantly less potent than that of the clinical drug acarbose, its activity was still comparable. At 1 mg/mL, ·OH and O2· scavenging capacities reached 84.59% and 65.61%, respectively (Figure 3C,D), with O2· scavenging approaching the reference levels of vitamin C. These findings support the second hypothesis regarding AVEO’s dual bioactivity.
Regarding the hypoglycemic activity of AVEO, our findings are consistent with previous studies, although the inhibition rate observed in our experiments was higher than the range (31.99–62.58%) reported by Kim [62]. This enhanced activity may be attributed to the high bornyl acetate content (52.1%) in AVEO, as terpene-rich essential oils are increasingly recognized as potential antidiabetic agents targeting carbohydrate-metabolizing enzymes [63]. Consistent with our findings, in vivo studies have documented pancreatic β-cell protection and amelioration of ultrastructural changes in diabetic models [62,64,65], while reviews have acknowledged its hypoglycemic potential [1]. Meanwhile, we found that the α-glucosidase inhibitory activity of AVEO was superior to that of several reported natural hypoglycemic plants. These include cinnamon extract (IC50 = 1.15 mg/mL [66]), bitter melon terpenoids (IC50 = 1.60 mg/mL [67]), hot-pressed peanut meal protein hydrolysates (IC50 = 5.63 mg/mL [68]), Dianthus basuticus saponins (IC50 = 3.80 mg/mL [69]), and Citrus paradisi peel extract (IC50 = 1.23 mg/mL [70]). Here, the IC50 value of Amomum3 was comparable to that of acarbose (IC50 = 0.75 mg/mL), a commonly used clinical α-glucosidase inhibitor (with literature-reported values up to 1.04 mg/mL [71]). Given the excellent glucose-lowering effect of AVEO, it can serve as a natural alternative to hypoglycemic drugs.
Previous studies have examined the antioxidant capacity of water and extracts of A. villosum, particularly phenolic acids and flavonoids [17,18,72]. While plant phenolics are classical antioxidants [35], our in vitro experiments confirmed that AVEO also has good antioxidant capacity, with a maximum broad-spectrum scavenging rate of 84.59% against DPPH, ·OH, and ·O2. Among them, the hydroxyl radical scavenging activity of Amomum4 (IC50 = 0.18 mg/mL) showed no statistical difference from reference standard vitamin C (IC50 = 0.14 mg/mL; p > 0.05), suggesting that AVEO could serve as a natural antioxidant to replace synthetic antioxidants.
Notably, correlation analysis showed a significant positive correlation between α-glucosidase inhibitory activity and DPPH scavenging capacity (r = 0.82), but a significant negative correlation with ·OH scavenging capacity (r = −0.65). This differential association may be related to the differences in the scavenging mechanisms of the two types of free radicals and their effects on enzyme active sites. DPPH clearance relies on electron transfer from hydroxyl or amino groups, potentially enhancing enzyme inhibition via active site interactions [73]. Conversely, ·OH scavenging depends on hydrogen atom donation [35], and strong ·OH scavengers may generate oxidative byproducts that impair enzyme activity [74]. These correlations highlight the functional synergy between hypoglycemic and antioxidant activities of AVEO; however, the mechanism underlying this functional synergistic effect requires further in-depth analysis.

4.3. Potential Hypoglycemic–Antioxidant Synergistic Mechanisms of AVEO

Network pharmacology and molecular docking identified AVEO components A4-A9 as key regulators of oxidative stress and glucose metabolism via multi-target synergy, with major targets and hub pathways mediating its hypoglycemic–antioxidant synergy through multi-dimensional cross-regulation (Figure 5, Figure 6 and Figure 7). Molecular docking further verified the high-affinity binding of (−)-spathulenol and (−)-pogostol to JAK2, NCOA2, and PTGS2, as well as the favorable affinity of bornyl acetate for PPARA (Figure 7). The PPAR, nuclear receptor-mediated steroid hormone, and JAK-STAT signaling pathways were identified as core regulators of AVEO antioxidant and hypoglycemic mechanisms in this study. For PPAR signaling pathway, it aligns with previously reported improvements in anti-diabetic efficacy linked to enhanced PPAR expression [75]. Type 2 diabetes mellitus (T2DM) rat models have demonstrated that hypoglycemic effects, insulin sensitization, antioxidant activity, and PPARA upregulation are associated [76,77,78]. In the present study, bornyl acetate, the most abundant component in AVEO (Figure 4), was also shown to have a favorable affinity for PPARA (−6.9 kcal/mol). While existing studies confirm its potent in vitro and in vivo antioxidant and anti-inflammatory activities [79,80], bornyl acetate’s direct role in hypoglycemia requires further validation. Given its dominance in composition and PPARA binding capability, bornyl acetate is hypothesized to be a primary contributor to AVEO’s observed antioxidant effects and potentially participates in hypoglycemic regulation through PPAR pathway modulation. Other components (e.g., (−)-spathulenol, (−)-pogostol, α-bisabolol) exhibited strong binding (binding energies < −7.2 kcal/mol with PPARA). These results suggest that AVEO may enhance insulin sensitivity or exert antioxidant effects through the activation of PPARα. Inhibition of the JAK-STAT pathway should be another core of the synergistic mechanism. As the inhibitory effect is recognized as one of the key mechanisms underlying metformin, a first-line anti-diabetic drug [81], previous studies have indicated that normalization or downregulation of the JAK-STAT pathway improves diabetic phenotypes and increases anti-inflammatory cytokine levels [82]. The significant enrichment of AVEO’s antioxidant–hypoglycemic intersection targets in the JAK-STAT pathway further supports the theory of multi-target synergistic regulation through this pathway. All components A1–A9 showed potential for stable binding to JAK2, with (−)-spathulenol (binding energy: −9.0 kcal/mol) and (−)-pogostol (binding energy: −8.2 kcal/mol) showing the most stable binding. For other pathways, the PI3K/AKT pathway is considered a critical therapeutic target for obesity and T2DM [76,77,78]. However, neither GO nor KEGG analyses revealed significant enrichment of this pathway, offering no direct support for Hypothesis 3. Nevertheless, several identified targets may still modulate PI3K/AKT activity. Nonetheless, other target proteins could directly affect the PI3K/AKT pathway. For instance, JAK2 phosphorylation [83,84], improvement in PTPN1 (PTP1B) [85,86], or through nuclear receptor-mediated steroid hormone signaling [87] could influence the pathway. In this study, most active components (bornyl acetate, (−)-spathulenol, (−)-pogostol, α-bisabolol) were found to have the potential for stable binding to targets JAK2 and PTPN1 (Figure 7), with the nuclear receptor-mediated steroid hormone signaling pathway also identified among the major enriched pathways. These findings collectively indicate that AVEO modulates the PI3K/AKT pathway through parallel mechanisms. Thus, the results substantiate the mechanism whereby these core components (e.g., bornyl acetate, (−)-spathulenol, and (−)-pogostol) mitigate hyperglycemia and oxidative stress by regulating pivotal targets (e.g., CYP3A4, PPARA, JAK2) to activate a multidimensional network involving (e.g., the PPAR signaling pathway, nuclear receptor-mediated steroid hormone pathway, and JAK-STAT signaling pathway).
Elucidation of binding sites provides a structural basis for the aforementioned mechanisms. Research on the stable binding bonds between AVEO and core targets related to hypoglycemic and oxidative stress effects is limited. A previous study reported that a compound from the stems and leaves of A. villosum [(7S-9′R)-(4,5-dihydroxy-3-methoxyphenyl)-7-methoxyacetoxy)-7′-(1′-hydroxyphenyl)butanoic acid] formed hydrogen bond interactions with Lys93 and Agr214 to maintain binding with the antioxidant target protein PGAM5, thereby inhibiting the upregulation of intracellular reactive oxygen species levels under oxidative stress stimulation [26]. Conversely, using the fruit extract, we identified distinct targets and binding modes. The most stably bound compounds, (−)-spathulenol and (−)-pogostol targeted NCOA2, JAK2, CYP3A4, and CYP2C19 through differential hydrogen bonds (e.g., Ser936) and hydrophobic interactions (e.g., Tyr930, LEU855, LEU982, LEU983, ALA880, VAL863, LEU346, LEU349, LEU384, LEU387, LEU525, ALA350, TRP383, PHE404). Components with relatively high content targeted PPARA through hydrogen bonds (e.g., Asn219, Met220) and hydrophobic contacts (e.g., Leu321, Val324). To some extent, this provides new binding site insights for the regulation of hypoglycemic and antioxidant effects by AVEO.

4.4. Limitations and Future Perspectives

Our study elucidates the material basis and molecular network underlying the synergistic antioxidant and hypoglycemic effects of AVEO through an “extraction optimization–functional validation–mechanistic elucidation” framework. Predictive conclusions from network pharmacology and molecular docking require experimental validation, representing an essential progression for in-depth mechanistic exploration. Future work will proceed from computational prediction to experimental verification to functional food development continuum, comprehensively evaluating AVEO’s hypoglycemic and antioxidant effects through integrated cellular and animal studies. This will provide more robust experimental evidence for applying A. villosum in metabolic disorder management and functional foods.

5. Conclusions

An optimized ultrasound-assisted extraction protocol for AVEO was established in this study. The actual extraction conditions (15 min ultrasonication, 1:26.13 g/mL solvent-to-material ratio, and 200 W power) were determined using response surface methodology, yielding a high extraction rate of 3.84%. AVEO exhibited concentration-dependent α-glucosidase inhibitory activity, with efficacy comparable to the reference substance, acarbose. AVEO also demonstrated potent scavenging activity against DPPH, ·OH, and ·O2 radicals. Mechanistic studies indicated that the dual bioactivities involve shared bioactive constituents. Network pharmacology analysis identified terpinen-4-ol, α-terpineol, bornyl acetate, (‒)-spathulenol, (‒)-pogostol, and α-bisabolol as core components that regulate a multi-target network encompassing CYP3A4, CYP2C19, PTGS2, NR3C1, PPARA, NCOA2, JAK2, and ESR1. PPI analysis revealed that these targets are significantly enriched in the PPAR signaling pathway, steroid hormone pathway, and JAK-STAT cascade. Molecular docking validated the strong binding abilities of (‒)-spathulenol and (‒)-pogostol to JAK2 and NCOA2 (binding energy < −8.2 kcal/mol), with stable binding involving hydrogen bonds and hydrophobic interactions. Additionally, the strong binding potential of the main component, bornyl acetate, to PPARA via hydrogen bonds and hydrophobic interactions was confirmed. Collectively, these results elucidate the synergistic multi-target mechanisms underlying the antioxidant and hypoglycemic efficacy of AVEO, laying a foundation for the development of nutraceuticals and therapeutic agents derived from Amomum villosum. However, further cellular and in vivo studies are needed to assess its therapeutic efficacy and safety profile fully.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/foods14162772/s1: Table S1: Box–Behnken design and results for response surface optimization. Design matrix and corresponding outcomes for ultrasonic extraction parameters affecting essential oil yield from AVEO.

Author Contributions

Conceptualization, W.W.; Methodology, W.W., Y.L., L.W., X.F., Y.D., Q.L. and S.F.; Software, W.W., X.F., Y.D. and Q.L.; Formal Analysis, W.W. and L.W.; Investigation, L.W., Y.D. and Q.L.; Writing—Original Draft Preparation, W.W.; Writing—Review and Editing, W.W. and S.F.; Supervision, S.F. and X.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Natural Science Foundation of Guangxi Province (2025GXNSFHA069148, “Changes and Regulation of Phosphorus Component Characteristics in Three-Dimensional Cultivation Patterns of Multi-Species Medicinal Herbs in Karst Areas Based on Metagenomics”; 2023GXNSFBA026053, “Effects of Nitrogen Addition on Carbon Accumulation of Microbial Residues Driven by Lignin Carbon in Karst Upland”), the Doctoral Scientific Research Project of Guangxi University of Science and Technology (XKB21Z09), the National Natural Science Foundation of China (32460318, 32001208), and the Guangxi Institute of Industrial Technology Research (CYY-HT2023-JSJJ-0038), and was supported by the Guangxi College Key Laboratory of Innovation Research on Medical and Engineering Integration.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author.

Acknowledgments

We gratefully acknowledge the support provided by the Guangxi Botanical Garden of Medicinal Plants for supplying the samples used in this study.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AVEOAmomum villosum essential oil
α-GIα-Glucosidase inhibition
DPPH2,2-Diphenyl-1-picrylhydrazyl
GC-MSGas chromatography–mass spectrometry
GOGene ontology
KEGGKyoto Encyclopedia of Genes and Genomes
PPIProtein-protein interaction
RNARibonucleic acid
DNADeoxyribonucleic acid
PPARPeroxisome proliferator-activated receptor
JAK-STATJanus kinase–signal transducer and activator of transcription
TICTotal ion chromatogram

References

  1. Feng, L.; Wang, Z.; Lei, Z.; Zhang, X.; Zhai, B.; Sun, J.; Guo, D.; Wang, D.; Luan, F.; Zou, J.; et al. Amomum villosum Lour.: An insight into ethnopharmacological, phytochemical, and pharmacological overview. J. Ethnopharmacol. 2024, 335, 118615. [Google Scholar] [CrossRef]
  2. Xu, Y.; Sun, D.; Xiong, L.; Zhang, Z.; Li, Y.; Liu, K.; Li, H.; Chen, L. Phenolics and terpenoids with good anti-inflammatory activity from the fruits of Amomum villosum and the anti-inflammatory mechanism of active diterpene. Bioorg. Chem. 2024, 145, 107190. [Google Scholar] [CrossRef] [PubMed]
  3. Peng, J.M.; Zhang, L.X.; Ma, J.; Guan, Z.B. The review of Amomum villosum in Xishuangbanna. China J. Chin. Mater. Med. 2006, 31, 97–101. [Google Scholar]
  4. Chen, L.; Lai, Y.; Zhang, W.; Cai, J.; Hu, H.; Wang, Y.; Zhao, J.; Li, S. Comparison of volatile compounds in different parts of fresh Amomum villosum Lour. from different geographical areas using cryogenic grinding combined HS–SPME–GC–MS. Chin. Med. 2020, 15, 97. [Google Scholar] [CrossRef]
  5. Thinh, B.B.; Chac, L.D.; Hanh, D.H.; Korneeva, A.A.; Hung, N.; Igoli, J.O. Effect of extraction method on yield, chemical composition and antimicrobial activity of essential oil from the fruits of Amomum villosum var. xanthioides. J. Essent. Oil-Bear. Plants 2022, 25, 28–37. [Google Scholar] [CrossRef]
  6. Ao, H.; Wang, J.; Chen, L.; Li, S.; Dai, C. Comparison of volatile oil between the fruits of Amomum villosum Lour. and Amomum villosum Lour. var. xanthioides T. L. Wu et senjen based on GC-MS and chemometric techniques. Molecules 2019, 24, 1663. [Google Scholar] [CrossRef]
  7. Chen, Z.; Guo, S.; Cao, J.; Pang, X.; Geng, Z.; Wang, Y.; Zhang, Z.; Du, S. Insecticidal and repellent activity of essential oil from Amomum villosum Lour. and its main compounds against two stored-product insects. Int. J. Food Prop. 2018, 21, 2265–2275. [Google Scholar] [CrossRef]
  8. Barik, P.; Dagar, K.; Makhija, R.; Singh, A.; Asati, V. A green technology for the extraction of essential oil using microwave and ultrasound-assisted techniques. Curr. Anal. Chem. 2025, 21, 79–105. [Google Scholar] [CrossRef]
  9. Li, G.; Liu, S.; Zhou, Q.; Han, J.; Qian, C.; Li, Y.; Meng, X.; Gao, X.; Zhou, T.; Li, P.; et al. Effect of response surface methodology-optimized ultrasound-assisted pretreatment extraction on the composition of essential oil released from tribute citrus peels. Front. Nutr. 2022, 9, 840780. [Google Scholar] [CrossRef]
  10. Yang, Y.; Wang, C.; Wei, M. Development and validation of an ultrasound-assisted supercritical carbon-dioxide procedure for the production of essential oils from Perilla frutescens. LWT 2020, 128, 109503. [Google Scholar] [CrossRef]
  11. Kazemi, M.; Niazi, A.; Yazdanipour, A. Extraction of Satureja rechingeri volatile components through ultrasound-assisted and microwave-assisted extractions and comparison of the chemical composition with headspace solid-phase microextraction. J. Essent. Oil Res. 2022, 34, 21–35. [Google Scholar] [CrossRef]
  12. Chemat, F.; Rombaut, N.; Sicaire, A.G.; Meullemiestre, A.; Fabiano-Tixier, A.; Abert-Vian, M. Ultrasound assisted extraction of food and natural products. Mechanisms, techniques, combinations, protocols and applications. A review. Ultrason. Sonochem. 2017, 34, 540–560. [Google Scholar] [CrossRef]
  13. Suslick, K.S.; Flannigan, D.J. Inside a collapsing bubble: Sonoluminescence and the conditions during cavitation. Annu. Rev. Phys. Chem. 2008, 59, 659–683. [Google Scholar] [CrossRef] [PubMed]
  14. Gavahian, M.; Chu, Y.H.; Mousavi Khaneghah, A. Recent advances in orange oil extraction: An opportunity for the valorisation of orange peel waste a review. Int. J. Food Sci. Technol. 2019, 54, 925–932. [Google Scholar] [CrossRef]
  15. Wang, B.; Chen, H.; Qu, P.; Lin, R.; He, S.; Li, W.; Zhang, C.; Shi, X.; Liu, Y.; Du, H.; et al. Effect of different cultivation patterns on Amomum villosum yield and quality parameters, rhizosphere soil properties, and rhizosphere soil microbes. Horticulturae 2023, 9, 306. [Google Scholar] [CrossRef]
  16. Zhang, K.; Cao, F.; Zhao, Y.; Wang, H.; Chen, L. Antibacterial Ingredients and modes of the methanol-phase extract from the fruit of Amomum villosum Lour. Plants 2024, 13, 834. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, F.; Li, X.; Lan, L.; Wang, J.; Guo, P.; Sun, G. Simultaneous determination of eight components in Amomum villosum and its overall qualityconsistency evaluation by four-dimensional fingerprints assisted with antioxidant activity. J. Chromatogr. A 2022, 1674, 463135. [Google Scholar] [CrossRef]
  18. Ai, Z.; Mowafy, S.; Liu, Y. Comparative analyses of five drying techniques on drying attributes, physicochemical aspects, and flavor components of Amomum villosum fruits. LWT 2022, 154, 112879. [Google Scholar] [CrossRef]
  19. Lee, J.H.; Kim, H.R.; Antonisamy, P.; Kim, Y.S.; Ryu, D.G.; Lee, G.; Kwon, K.B. Efficacy and safety of Amomum villosum extracts in obese adults: A randomized, double-blind, placebo-controlled trial. J. King Saud Univ. Sci. 2023, 35, 102580. [Google Scholar] [CrossRef]
  20. Dong, L.; Zhang, S.; Chen, L.; Lu, J.; Zhao, F.; Long, T.; Wen, J.; Huang, J.; Mao, Y.; Qi, Z.; et al. In vivo anti-hyperuricemia and anti-gouty arthritis effects of the ethanol extract from Amomumvillosum Lour. Biomed. Pharmacother. 2023, 161, 114532. [Google Scholar] [CrossRef]
  21. Luo, D.; Zeng, J.; Guan, J.; Xu, Y.; Jia, R.B.; Chen, J.; Jiang, G.; Zhou, C. Dietary supplement of Amomum villosum Lour. polysaccharide attenuates ulcerative colitis in BALB/c mice. Foods 2022, 11, 3737. [Google Scholar] [CrossRef]
  22. Cho, J.H.; Lee, J.S.; Kim, H.G.; Lee, H.W.; Fang, Z.; Kwon, H.H.; Kim, D.W.; Lee, C.M.; Jeong, J.W. Ethyl acetate fraction of Amomum villosum var. xanthioides attenuates hepatic endoplasmic reticulum stress-induced non-alcoholic steatohepatitis via improvement of antioxidant capacities. Antioxidants 2021, 10, 998. [Google Scholar] [CrossRef]
  23. Yue, J.; Zhang, S.; Zheng, B.; Raza, F.; Luo, Z.; Li, X.; Zhang, Y.; Nie, Q.; Qiu, M. Efficacy and mechanism of active fractions in fruit of Amomum villosum Lour. for gastric cancer. J. Cancer 2021, 12, 5991–5998. [Google Scholar] [CrossRef]
  24. Wei, J.; Wang, S.; Huang, J.; Zhou, X.; Qian, Z.; Wu, T.; Fan, Q.; Liang, Y.; Cui, G. Network medicine-based analysis of the hepatoprotective effects of Amomum villosum Lour. on alcoholic liver disease in rats. Food Sci. Nutr. 2024, 12, 3759–3773. [Google Scholar] [CrossRef] [PubMed]
  25. Fan, Y.; Nguyen, T.V.; Piao, C.H.; Shin, H.S.; Song, C.H.; Chai, O.H. Fructus amomi extract attenuates nasal inflammation by restoring Th1/Th2 balance and down-regulation of NF-κB phosphorylation in OVA-induced allergic rhinitis. Biosci. Rep. 2022, 42, BSR20212681. [Google Scholar] [CrossRef] [PubMed]
  26. Liang, J.; Xu, Y.; Chen, J.; Zhang, Z.; Wang, H.; Liu, K.; Sun, D.; Li, H.; Chen, L. Antioxidant aromatic compounds from Amomum villosum and target prediction of active ingredients. Bioorg. Chem. 2024, 147, 107375. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, M.; Yuan, G.; Luo, G.; Guo, X.; Chen, M.; Yang, H.; He, F.; Yang, T.; Zhang, X.; Wu, Q.; et al. Network pharmacology analysis and experimental verification strategies reveal the action mechanism of Danshen decoction in treating ischemic cardiomyopathy. Evid. Based Compl. Alt. Med. 2022, 2022, 7578055. [Google Scholar] [CrossRef]
  28. Zhu, L.Y.; Liu, X.L.; Zheng, Q.; Kang, Y.J.; Li, W.J.; Xiao, S.; Xiong, Y.F.; Cai, K.Z.; Wu, M.Q.; Yang, M. Prediction of Q-markers of Citri Reticulatae Pericarpium volatile oil and GC-MS based quantitative analysis. China J. Chin. Mater. Med. 2021, 46, 6403–6409. [Google Scholar]
  29. Zhou, P.; Wang, X.; Zhao, Y.; She, X.; Jia, Y.; Wang, W.; Li, J.; Luo, X. Evaluation of the mechanism of action of rosemary volatile oil in the treatment of Alzheimer’s disease using gas chromatography -mass spectrometry analysis and network pharmacology. Comb. Chem. High Throughput Screen. 2023, 26, 2321–2332. [Google Scholar] [CrossRef]
  30. Li, S.; Huang, Y.; Liu, L.; Zhang, F.; Ao, H.; Luo, Y. Mechanism of magnolia volatile oil in the treatment of acute pancreatitis based on GC-MS, network pharmacology, and molecular docking. Evid. Based Complement Altern. Med. 2023, 2023, 3503888. [Google Scholar] [CrossRef]
  31. Yang, J.J.; Yu, H.; Wu, K.; He, D.; Zhang, H.; Cui, Z.X.; Chai, X.; Duan, X. Potential anti-gouty arthritis of citronella essential oil and nutmeg essential oil through reducing oxidative stress and inhibiting PI3K/Akt/mTOR activation-induced NLRP3 activity. Chem. Biodivers. 2024, 21, e202400448. [Google Scholar] [CrossRef]
  32. Bhakkiyalakshmi, E.; Sireesh, D.; Rajaguru, P.; Paulmurugan, R.; Ramkumar, K.M. The emerging role of redox-sensitive Nrf2–Keap1 pathway in diabetes. Pharmacol. Res. 2015, 91, 104–114. [Google Scholar] [CrossRef]
  33. Yang, Y.; Zhang, P.; Huang, Z.; Zhao, Z. Phenolics from Sterculia nobilis Smith pericarp by-products delay carbohydrate digestion by uncompetitively inhibiting α-glucosidase and α-amylase. Food Sci. Technol. 2023, 173, 114339. [Google Scholar] [CrossRef]
  34. Gómez-Mejía, E.; Rosales-Conrado, N.; León-González, M.E.; Madrid, Y. Citrus peels waste as a source of value-added compounds: Extraction and quantification of bioactive polyphenols. LWT-Food Chem. 2019, 295, 289–299. [Google Scholar] [CrossRef]
  35. Gulcin, İ. Antioxidants and antioxidant methods: An updated overview. Arch. Toxicol. 2020, 94, 651–715. [Google Scholar] [CrossRef] [PubMed]
  36. Yin, T.; Wang, H.; Zou, Y. Application of network pharmacology, bioinformatics, computational molecular docking, and experimental validation to study the anticancer effects of oleanolic acid in oral squamous carcinoma cells. Acta Pharm. 2025, 75, 41–68. [Google Scholar] [CrossRef] [PubMed]
  37. Daina, A.; Michielin, O.; Zoete, V. Swissadme: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [PubMed]
  38. Daina, A.; Michielin, O.; Zoete, V. Swisstargetprediction: Updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019, 47, W357–W364. [Google Scholar] [CrossRef]
  39. Bateman, A.; Martin, M.; Orchard, S.; Magrane, M.; Ahmad, S.; Alpi, E.; Bowler-Barnett, E.H.; Britto, R.; Bye-A-Jee, H.; Cukura, A.; et al. UniProt: The universal protein knowledgebase in 2023. Nucleic Acids Res. 2023, 51, D523–D531. [Google Scholar]
  40. Safran, M.; Dalah, I.; Alexander, J.; Rosen, N.; Iny Stein, T.; Shmoish, M.; Nativ, N.; Bahir, I.; Doniger, T.; Krug, H.; et al. Genecards version 3: The human gene integrator. Database 2010, 2010, baq020. [Google Scholar] [CrossRef]
  41. Amberger, J.S.; Bocchini, C.A.; Schiettecatte, F.; Scott, A.F.; Hamosh, A. OMIM.org: Online Mendelian Inheritance in Man (OMIM®), an online catalog of human genes and genetic disorders. Nucleic Acids Res. 2015, 43, D789–D798. [Google Scholar] [CrossRef]
  42. Wu, F.; Lin, B.; Chen, J.; Zheng, F.; Yang, Y.; Rasheed, U.; Chen, G. Mechanistic insights into the antioxidant potential of sugarcane vinegar polyphenols: A combined approach of DPPH-UPLC-MS, network pharmacology and molecular docking. Foods 2024, 13, 3379. [Google Scholar] [CrossRef]
  43. Sherman, B.T.; Hao, M.; Qiu, J.; Jiao, X.; Baseler, M.W.; Lane, H.C.; Imamichi, T.; Chang, W. David: A web server for functional enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res. 2022, 50, W216–W221. [Google Scholar] [CrossRef]
  44. Szklarczyk, D.; Kirsch, R.; Koutrouli, M.; Nastou, K.; Mehryary, F.; Hachilif, R.; Gable, A.L.; Fang, T.; Doncheva, N.T.; Pyysalo, S.; et al. The string database in 2023: Protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 2023, 51, D638–D646. [Google Scholar] [CrossRef]
  45. Flachsenberg, F.; Ehrt, C.; Gutermuth, T.; Rarey, M. Redocking the pdb. J. Chem. Inf. Model. 2024, 64, 219–237. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B.A.; Thiessen, P.A.; Yu, B.; et al. Pubchem 2025 update. Nucleic Acids Res. 2025, 53, D1516–D1525. [Google Scholar] [CrossRef] [PubMed]
  47. Paggi, J.M.; Pandit, A.; Dror, R.O. The art and science of molecular docking. Annu. Rev. Biochem. 2024, 93, 389–410. [Google Scholar] [CrossRef] [PubMed]
  48. Suleman, M.; Murshed, A.; Sayaf, A.M.; Khan, A.; Khan, S.A.; Tricarico, P.M.; Moltrasio, C.; Agouni, A.; Yeoh, K.K.; Marzano, A.V.; et al. Exploring global natural product databases for NLRP3 inhibition: Unveiling novel combinatorial therapeutic strategy for hidradenitis suppurativa. J. Infect. Public Health 2025, 18, 102697. [Google Scholar] [CrossRef]
  49. Zhang, H.; Wu, B.; Ke, L.; Fan, X.; Huang, L.; Peng, Z. Exploring the antitumor potential of cucurbitacin B in hepatocellular carcinoma through network pharmacology, molecular docking, and molecular dynamics simulations. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2025, 1–19. [Google Scholar] [CrossRef]
  50. Shady, N.H.; Zayed, A.; Alaaeldin, R.; Hisham, M.; Gawesh, M.; Mohammed, R.; Elrehany, M.A.; Abdelmohsen, U.R. Plant and endophyte-derived anti-hyperlipidemics: A comprehensive review with in silico studies. South Afr. J. Bot. 2023, 163, 105–120. [Google Scholar] [CrossRef]
  51. Yang, D.; Zhang, Y.; Shu, Y.; Cao, H. Research progress on chemical constituents and pharmacological effects of Amomum villosum. Guangdong Chem. Ind. 2022, 49, 111–114. [Google Scholar]
  52. Ramanathan, A.; Thangarasu, V. Effect of high-frequency microwave irradiation on Aegle Marmelos Correa oil extraction: Kinetic and thermodynamic study. Energy Procedia 2019, 158, 1046–1051. [Google Scholar] [CrossRef]
  53. Hussein, F.; Antonescu, C.; Karshafian, R. Ultrasound and microbubble induced release from intracellular compartments. BMC Biotechnol. 2017, 17, 45. [Google Scholar] [CrossRef] [PubMed]
  54. Guo, X.; Xun, Z.; Xu, L.; Li, Y.; Wang, Y.; Guo, S. Constructed the metabolic compound accumulation profiles of daylily at different main producing areas using multi-omics techniques. Food Biosci. 2024, 59, 104128. [Google Scholar] [CrossRef]
  55. Wen, B.; Ren, S.; Zhang, Y.; Duan, Y.; Shen, J.; Zhu, X.; Wang, Y.; Ma, Y.; Zou, Z.; Fang, W. Effects of geographic locations and topographical factors on secondary metabolites distribution in green tea at a regional scale. Food Control 2020, 110, 106979. [Google Scholar] [CrossRef]
  56. Suslick, K.S. Sonochemistry. Science 1990, 247, 1439–1445. [Google Scholar] [CrossRef]
  57. Chen, X.; Bayanheshig; Jiao, Q.; Tan, X.; Wang, W. Numerical simulation of ultrasonic enhancement by acoustic streaming and thermal effect on mass transfer through a new computation model. Int. J. Heat Mass Transf. 2021, 171, 121074. [Google Scholar] [CrossRef]
  58. Kimbaris, A.C.; Siatis, N.G.; Daferera, D.J.; Tarantilis, P.A.; Pappas, C.S.; Polissiou, M.G. Comparison of distillation and ultrasound-assisted extraction methods for the isolation of sensitive aroma compounds from garlic (Allium sativum). Ultrason. Sonochem. 2006, 13, 54–60. [Google Scholar] [CrossRef]
  59. Zabot, G.L.; Viganó, J.; Silva, E.K. Low-frequency ultrasound coupled with high-pressure technologies: Impact of hybridized techniques on the recovery of phytochemical compounds. Molecules 2021, 26, 5117. [Google Scholar] [CrossRef]
  60. Asakura, Y.; Yasuda, K. Frequency and power dependence of the sonochemical reaction. Ultrason. Sonochem. 2021, 81, 105858. [Google Scholar] [CrossRef]
  61. Bezerra, M.A.; Santelli, R.E.; Oliveira, E.P.; Villar, L.S.; Escaleira, L.A. Response surface methodology (RSM) as a tool for optimization in analytical chemistry. Talanta 2008, 76, 965–977. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, H.R.; Antonisamy, P.; Kim, Y.S.; Lee, G.; Ham, H.D.; Kwon, K.B. Inhibitory effect of Amomum villosum water extracts on α-glucosidase activity. Physiol. Mol. Plant Pathol. 2022, 117, 101779. [Google Scholar] [CrossRef]
  63. Khatib, S.; Mahdi, I.; Drissi, B.; Fahsi, N.; Bouissane, L.; Sobeh, M. Tetraclinis articulata (Vahl) Mast.: Volatile constituents, antioxidant, antidiabetic and wound healing activities of its essential oil. Heliyon 2024, 10, e24563. [Google Scholar] [CrossRef] [PubMed]
  64. Ding, M.; Wu, S.L.; Hu, J.; He, X.F.; Huang, X.Y.; Li, T.Z.; Ma, Y.B.; Zhang, X.M.; Geng, C.A. Norlignans as potent GLP-1 secretagogues from the fruits of Amomum villosum. Phytochemistry 2022, 199, 113204. [Google Scholar] [CrossRef]
  65. Rongjie, Z.; Zhenglin, Z.; Meihong, J.; Sangchan, K.; Zaixia, Y. Hypoglycemic effects of extract of Amomum xanthoides on experimental diabetes in rats. J. Yanbian Med. Coll. 2006, 29, 97–99. [Google Scholar]
  66. Sriramavaratharajan, V.; Murugan, R. Screening of chemical composition, in vitro antioxidant, α-amylase and α-glucosidase inhibitory activities of the leaf essential oils of Cinnamomum wightii from different populations. Nat. Prod. Commun. 2018, 13, 1539–1542. [Google Scholar] [CrossRef]
  67. Sallau, A.B.; Yakubu, R.N.; Aliyu, S.M.; Salihu, A.; Boniface, B.Y. In vitro effect of terpenoids-rich extract of Momordica charantia on alpha glucosidase activity. Vitae 2018, 25, 148–153. [Google Scholar] [CrossRef]
  68. Yang, X.; Wang, D.; Dai, Y.; Zhao, L.; Wang, W.; Ding, X. Identification and molecular binding mechanism of novel α-glucosidase inhibitory peptides from hot-pressed peanut meal protein hydrolysates. Foods 2023, 12, 663. [Google Scholar] [CrossRef]
  69. Nafiu, M.; Tom Ashafa, A. Antioxidant and inhibitory effects of saponin extracts from Dianthus basuticus Burtt Davy on key enzymes implicated in type 2 diabetes in vitro. Pharmacogn. Mag. 2017, 13, 576. [Google Scholar] [CrossRef]
  70. Mehmood, M.F.U.; Marvi, H.A.; Abuelizz, H.A.; Aziz, N.; Bano, R.; Wazir, A.; Ahmad, I.; Abbas, K.; Ishtiaq, S.; Amin, A. Advance glycation end products inhibition by citrus paradisi peel extract; characterization, LCMS-QTOF analysis, and biological evaluation. Food Sci. Nutr. 2024, 12, 10655–10665. [Google Scholar] [CrossRef]
  71. Hao, W.; Wang, M.; Lv, M. The inhibitory effects of Yixing black tea extracts on α-glucosidase. J. Food Biochem. 2017, 41, e12269. [Google Scholar] [CrossRef]
  72. Zhang, M.; Shuai, X.X.; Wei, Z.; Dai, T.T.; Wei, C.B.; Li, Y.; He, J.J.; Du, L.Q. Characterization, antioxidant and antitumor activities of phenolic compounds from Amomum villosum Lour. Front. Nutr. 2024, 11, 1327164. [Google Scholar] [CrossRef]
  73. Gowd, V.; Jia, Z.; Chen, W. Anthocyanins as promising molecules and dietary bioactive components against diabetes–A review of recent advances. Trends Food Sci. Technol. 2017, 68, 1–13. [Google Scholar] [CrossRef]
  74. Alam, F.; Shafique, Z.; Amjad, S.T.; Bin Asad, M.H.H. Enzymes inhibitors from natural sources with antidiabetic activity: A review. Phytother. Res. 2019, 33, 41–54. [Google Scholar] [CrossRef] [PubMed]
  75. Mohamed, W.A.; Schaalan, M.F. Antidiabetic efficacy of lactoferrin in type 2 diabetic pediatrics; controlling impact on PPAR-γ, SIRT-1, and TLR4 downstream signaling pathway. Diabetol. Metab. Syndr. 2018, 10, 89. [Google Scholar] [CrossRef] [PubMed]
  76. Al-Harbi, L.N.; Alshammari, G.M.; Al-Dossari, A.M.; Subash-Babu, P.; Binobead, M.A.; Alhussain, M.H.; AlSedairy, S.A.; Al-Nouri, D.M.; Shamlan, G. Beta vulgaris L. (Beetroot) methanolic extract prevents hepatic steatosis and liver damage in T2DM rats by hypoglycemic, insulin-sensitizing, antioxidant effects, and upregulation of PPARα. Biology 2021, 10, 1306. [Google Scholar] [CrossRef] [PubMed]
  77. Alshareef, N.S.; AlSedairy, S.A.; Al-Harbi, L.N.; Alshammari, G.M.; Yahya, M.A. Carthamus tinctorius L. (safflower) flower extract attenuates hepatic injury and steatosis in a rat model of type 2 diabetes mellitus via nrf2-dependent hypoglycemic, antioxidant, and hypolipidemic effects. Antioxidants 2024, 13, 1098. [Google Scholar] [CrossRef]
  78. Alwadani, A.H.; Almasri, S.A.; Aloud, A.A.; Albadr, N.A.; Alshammari, G.M.; Yahya, M.A. The synergistic protective effect of γ-oryzanol (OZ) and N-acetylcysteine (NAC) against experimentally induced NAFLD in rats entails hypoglycemic, antioxidant, and PPARα stimulatory effects. Nutrients 2023, 15, 106. [Google Scholar] [CrossRef]
  79. Zhao, Z.; Sun, Y.; Ruan, X. Bornyl acetate: A promising agent in phytomedicine for inflammation and immune modulation. Phytomedicine 2023, 114, 154781. [Google Scholar] [CrossRef]
  80. Kim, S.H.; Lee, S.Y.; Hong, C.Y.; Gwak, K.S.; Park, M.J.; Smith, D.; Choi, I.G. Whitening and antioxidant activities of bornyl acetate and nezukol fractionated fromCryptomeria japonica essential oil. Int. J. Cosmet. Sci. 2013, 35, 484–490. [Google Scholar] [CrossRef]
  81. Abdelsamia, E.M.; Khaleel, S.A.; Balah, A.; Abdel Baky, N.A. Curcumin augments the cardioprotective effect of metformin in an experimental model of type I diabetes mellitus; Impact of Nrf2/HO-1 and JAK/STAT pathways. Biomed. Pharmacother. 2019, 109, 2136–2144. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, J.; Meng, X. Aronia melanocarpa anthocyanin extracts improve hepatic structure and function in high-fat diet-/streptozotocin-induced T2DM mice. J. Agric. Food Chem. 2022, 70, 11531–11543. [Google Scholar] [CrossRef] [PubMed]
  83. Yoshioka, Y.; Zhang, Q.; Wang, X.; Kitakaze, T.; Yamashita, Y.; Kohno, M.; Ashida, H. Mung bean peptides promote glucose uptake via Jak2 activation in L6 myotubes. Food Funct. 2023, 14, 5375–5390. [Google Scholar] [CrossRef] [PubMed]
  84. Jiang, H.; Yamashita, Y.; Nakamura, A.; Croft, K.; Ashida, H. Quercetin and its metabolite isorhamnetin promote glucose uptake through different signalling pathways in myotubes. Sci. Rep. 2019, 9, 2690. [Google Scholar] [CrossRef]
  85. Gao, J.; Cang, X.; Liu, L.; Lin, J.; Zhu, S.; Liu, L.; Liu, X.; Zhu, J.; Xu, C. Farrerol alleviates insulin resistance and hepatic steatosis of metabolic associated fatty liver disease by targeting PTPN1. J. Cell. Mol. Med. 2024, 28, e70096. [Google Scholar] [CrossRef]
  86. Du, R.; Wu, N.; Bai, Y.; Tang, L.; Li, L. circMAP3K4 regulates insulin resistance in trophoblast cells during gestational diabetes mellitus by modulating the miR-6795-5p/PTPN1 axis. J. Transl. Med. 2022, 20, 180. [Google Scholar] [CrossRef]
  87. Wu, J.; Williams, D.; Walter, G.A.; Thompson, W.E.; Sidell, N. Estrogen increases Nrf2 activity through activation of the PI3K pathway in MCF-7 breast cancer cells. Exp. Cell Res. 2014, 328, 351–360. [Google Scholar] [CrossRef]
Figure 1. Results of single-factor experiments on AVEO (A. villosum essential oil). Influence of ultrasonic time, solvent-to-material ratio, and ultrasonic power on the extraction yield of AVEO.
Figure 1. Results of single-factor experiments on AVEO (A. villosum essential oil). Influence of ultrasonic time, solvent-to-material ratio, and ultrasonic power on the extraction yield of AVEO.
Foods 14 02772 g001
Figure 2. Response surface plots for extraction optimization. Three-dimensional surface plots showing interaction effects among ultrasonic time, solvent-to-material ratio, and ultrasonic power on AVEO yield, based on the Box–Behnken design.
Figure 2. Response surface plots for extraction optimization. Three-dimensional surface plots showing interaction effects among ultrasonic time, solvent-to-material ratio, and ultrasonic power on AVEO yield, based on the Box–Behnken design.
Foods 14 02772 g002
Figure 3. Antioxidant and hypoglycemic activities of AVEO, including: (A) α-glucosidase inhibition; (B) 1,1-diphenyl-2-picrylhydrazyl (DPPH) scavenging activity; (C) superoxide anion (·O2) scavenging activity; (D) hydroxyl radical (·OH) scavenging activity; (E) heatmap showing the correlation between free radical scavenging capacity and hypoglycemic activity. Different lowercase letters indicate statistically significant differences between samples at the same concentration (LSD test, p < 0.05), while identical letters indicate no significant difference.
Figure 3. Antioxidant and hypoglycemic activities of AVEO, including: (A) α-glucosidase inhibition; (B) 1,1-diphenyl-2-picrylhydrazyl (DPPH) scavenging activity; (C) superoxide anion (·O2) scavenging activity; (D) hydroxyl radical (·OH) scavenging activity; (E) heatmap showing the correlation between free radical scavenging capacity and hypoglycemic activity. Different lowercase letters indicate statistically significant differences between samples at the same concentration (LSD test, p < 0.05), while identical letters indicate no significant difference.
Foods 14 02772 g003
Figure 4. Gas chromatography–mass spectrometry (GC-MS) total ion chromatogram (TIC) of AVEO. The chromatographic profile shows the total ion counts of volatile compounds in AVEO, as identified using GC–MS. Peaks labeled with numbers correspond to the compounds listed in Table 3, which account for 81.94% of the total relative content. Peaks marked as “Unmatched” represent components with a match factor < 85 (based on the NIST 20 library), which are not commonly reported in existing literature for Amomum species. Unmarked peaks represent minor constituents (<0.1% relative content) that were excluded from analytical consideration.
Figure 4. Gas chromatography–mass spectrometry (GC-MS) total ion chromatogram (TIC) of AVEO. The chromatographic profile shows the total ion counts of volatile compounds in AVEO, as identified using GC–MS. Peaks labeled with numbers correspond to the compounds listed in Table 3, which account for 81.94% of the total relative content. Peaks marked as “Unmatched” represent components with a match factor < 85 (based on the NIST 20 library), which are not commonly reported in existing literature for Amomum species. Unmarked peaks represent minor constituents (<0.1% relative content) that were excluded from analytical consideration.
Foods 14 02772 g004
Figure 5. Network pharmacology of AVEO: (A) targets of active components in Amomum villosum and antioxidant–hypoglycemic-related genes; (B) “compound–disease–target gene” network; (C) degree values of each compound in the “compound–disease–target gene” network; (D) protein–protein interaction (PPI) network; (E) core targets; (F) data related to core targets.
Figure 5. Network pharmacology of AVEO: (A) targets of active components in Amomum villosum and antioxidant–hypoglycemic-related genes; (B) “compound–disease–target gene” network; (C) degree values of each compound in the “compound–disease–target gene” network; (D) protein–protein interaction (PPI) network; (E) core targets; (F) data related to core targets.
Foods 14 02772 g005
Figure 6. Functional enrichment analysis of AVEO target genes with (A) the gene ontology (GO) biological process, (B) the GO cellular component, (C) the GO molecular function, and (D) the KEGG pathway. Abbreviations: PPAR, peroxisome proliferator-activated receptor; JAK-STAT, Janus kinase–signal transducer and activator of transcription; GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; RNA, ribonucleic Acid; DNA, deoxyribonucleic Acid.
Figure 6. Functional enrichment analysis of AVEO target genes with (A) the gene ontology (GO) biological process, (B) the GO cellular component, (C) the GO molecular function, and (D) the KEGG pathway. Abbreviations: PPAR, peroxisome proliferator-activated receptor; JAK-STAT, Janus kinase–signal transducer and activator of transcription; GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; RNA, ribonucleic Acid; DNA, deoxyribonucleic Acid.
Foods 14 02772 g006
Figure 7. Molecular docking results of AVEO compounds with core targets: (A) heatmap of binding energies between AVEO active components and protein targets; (BH) representative docking poses showing binding interactions (hydrogen bonding and hydrophobic forces) with energy values < −8 kcal/mol; (I) binding conformation of bornyl acetate (dominant constituent, 52.10%) with PPARA.
Figure 7. Molecular docking results of AVEO compounds with core targets: (A) heatmap of binding energies between AVEO active components and protein targets; (BH) representative docking poses showing binding interactions (hydrogen bonding and hydrophobic forces) with energy values < −8 kcal/mol; (I) binding conformation of bornyl acetate (dominant constituent, 52.10%) with PPARA.
Foods 14 02772 g007
Table 1. Amomum villosum sample information.
Table 1. Amomum villosum sample information.
SampleCollection SitePlanting YearCollection YearPlanting Density
(Plants/m2)
Annual Output
(kg/ha)
Amomum1Leyi Village, Wangdian Yao Ethnic Township, Youjiang District, Baise City, Guangxi, China201920238.60 ± 0.55 a1233.30 ± 60.00 a
Amomum2201920228.20 ± 0.45 a1224.60 ± 149.90 a
Amomum3Shangmeng Village, Pingshan Township, Long’an County, Nanning City, Guangxi, China201920238.80 ± 0.84 a1248.00 ± 106.00 a
Amomum4201920228.60 ± 1.14 a1319.40 ± 143.20 a
The letter “a” indicates no significant differences (p > 0.05) among Amomum1, Amomum2, Amomum3, and Amomum4 groups based on the LSD test.
Table 2. Analysis of variance results from the response surface test. Statistical significance of linear, quadratic, and interaction terms for extraction variables affecting AVEO (A. villosum essential oil) yield.
Table 2. Analysis of variance results from the response surface test. Statistical significance of linear, quadratic, and interaction terms for extraction variables affecting AVEO (A. villosum essential oil) yield.
Source of VariationSum of SquaresDegrees of Freedom (df)Mean SquareFp
Model2.0290.224913.890.0011 **
A—Ultrasound Time0.000510.00050.02780.8723
B—Solvent-to-Material Ratio0.092510.09255.710.0482 *
C—Ultrasound Power0.008510.00850.5220.4934
AB0.055210.05523.410.1072
AC0.04210.0422.60.1512
BC0.018210.01821.130.3239
A20.610410.610437.710.0005 ***
B20.254310.254315.710.0054 **
C20.763210.763247.150.0002 ***
Residual0.113370.0162  
Lack of Fit0.0130.00330.1290.9379
Pure Error0.103340.0258  
Total2.1416   
R2 = 0.9470R2adj = 0.8788    
* p < 0.05, ** p < 0.01, *** p < 0.001.
Table 3. Gas chromatography–mass spectrometry (GC-MS) profile of AVEO. Identified chemical constituents are listed with their retention times and relative contents of key active ingredients based on GC–MS analysis.
Table 3. Gas chromatography–mass spectrometry (GC-MS) profile of AVEO. Identified chemical constituents are listed with their retention times and relative contents of key active ingredients based on GC–MS analysis.
No.Retention Time (min)ComponentMolecular FormulaCASMatch Factor (NIST20.L,%)Relative Peak Area (%)GI AbsorptionNo. of Active Ingredients
18.457p-CymeneC10H1499-87-687.060.35Low/
28.74Cyclohexane-1-butenylidene-C10H1636144-40-886.495.21Low/
310.42LinaloolC10H18O78-70-687.370.59HighA1
411.463(+)-2-BornanoneC10H16O464-49-388.9315.91HighA2
511.907IsoborneolC10H18O124-76-586.130.16HighA3
612.1695,5-Dimethyl-1,3-hexadieneC8H141515-79-386.783.52Low/
712.495Terpinen-4-olC10H18O562-74-385.850.43HighA4
812.811α-TerpineolC10H18O98-55-586.150.19HighA5
915.71Bornyl acetateC12H20O25655-61-887.6852.1HighA6
1018.785(+)-CyclosativeneC15H2422469-52-988.061.43Low/
1122.422α-CubebeneC15 H2417699-14-891.850.25Low/
1224.075β-SesquiphellandreneC15H2420307-83-986.660.66Low/
1325.974(−)-SpathulenolC15H24O77171-55-286.820.8HighA7
1426.691(−)-PogostolC15H26 O21698-41-993.130.18HighA8
1530.681α-BisabololC15H26 O515-69-585.830.16HighA9
The query results for gastrointestinal (GI) absorption were sourced from the SwissADME database (http://www.swissadme.ch/index.php, accessed on 30 May 2025). Components marked as “high” for GI absorption were selected as active ingredients.
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, W.; Liao, Y.; Wei, L.; Feng, X.; Dai, Y.; Liu, Q.; Feng, S. Ultrasound-Optimized Extraction and Multi-Target Mechanistic Analysis of Antioxidant and Hypoglycemic Effects of Amomum villosum Essential Oil. Foods 2025, 14, 2772. https://doi.org/10.3390/foods14162772

AMA Style

Wu W, Liao Y, Wei L, Feng X, Dai Y, Liu Q, Feng S. Ultrasound-Optimized Extraction and Multi-Target Mechanistic Analysis of Antioxidant and Hypoglycemic Effects of Amomum villosum Essential Oil. Foods. 2025; 14(16):2772. https://doi.org/10.3390/foods14162772

Chicago/Turabian Style

Wu, Wenxiang, Yining Liao, Lixia Wei, Xuezhen Feng, Yan Dai, Qingrong Liu, and Shuzhen Feng. 2025. "Ultrasound-Optimized Extraction and Multi-Target Mechanistic Analysis of Antioxidant and Hypoglycemic Effects of Amomum villosum Essential Oil" Foods 14, no. 16: 2772. https://doi.org/10.3390/foods14162772

APA Style

Wu, W., Liao, Y., Wei, L., Feng, X., Dai, Y., Liu, Q., & Feng, S. (2025). Ultrasound-Optimized Extraction and Multi-Target Mechanistic Analysis of Antioxidant and Hypoglycemic Effects of Amomum villosum Essential Oil. Foods, 14(16), 2772. https://doi.org/10.3390/foods14162772

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

Article Metrics

Back to TopTop