Next Article in Journal
Genetic Risk Profiles for Atherosclerosis and Venous Thromboembolism in Azorean and Mainland Portuguese Populations: A Comparative Analysis
Previous Article in Journal
Fyn Kinase: A Potential Target in Glucolipid Metabolism and Diabetes Mellitus
Previous Article in Special Issue
Analysis of Endoplasmic Reticulum Stress Proteins in Spermatogenic Cells After Paclitaxel Administration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of Bioactive Compounds, Antioxidant Activity, and Anticancer Potential of Wild Ganoderma lucidum Extracts from High-Altitude Regions of Nepal

1
Department of Biotechnology, SANN International College, Purbanchal University, Biratnagar 56600, Nepal
2
Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
3
School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2025, 47(8), 624; https://doi.org/10.3390/cimb47080624
Submission received: 3 July 2025 / Revised: 29 July 2025 / Accepted: 1 August 2025 / Published: 5 August 2025

Abstract

Wild Ganoderma lucidum from Nepal’s high-altitude regions was studied to identify key bioactive compounds and assess the influence of solvent type—water, ethanol, methanol, and acetone—on extraction efficiency and biological activity. Extracts were evaluated for antioxidant potential, cytotoxicity against HeLa cells, and phytochemical composition via gas chromatography–mass spectrometry (GC-MS). Solvent type significantly affected both yield and bioactivity. Acetone yielded the highest crude extract (5.01%), while ethanol extract exhibited the highest total phenolic (376.5 ± 9.3 mg PG/g) and flavonoid content (30.3 ± 0.5 mg QE/g). Methanol extract was richest in lycopene (0.07 ± 0.00 mg/g) and β-carotene (0.45 ± 0.02 mg/g). Ethanol extract demonstrated consistently strong DPPH, superoxide, hydroxyl, and nitric oxide radical scavenging activity, along with high reducing power. All extracts showed dose-dependent cytotoxicity against HeLa cells, with ethanol and water extracts showing the greatest inhibition (>65% at 1000 µg/mL). GC-MS profiling identified solvent-specific bioactive compounds including sterols, terpenoids, polyphenols, and fatty acids. Notably, pharmacologically relevant compounds such as hinokione, ferruginol, ergosterol, and geranylgeraniol were detected. These findings demonstrate the therapeutic potential of G. lucidum, underscore the importance of solvent selection, and suggest that high-altitude ecological conditions may influence its bioactive metabolite profile.

Graphical Abstract

1. Introduction

Ganoderma lucidum, commonly known as “Lingzhi” in China, “Reishi” in Japan, and “Dadu chyau” in Nepal, is a polypore mushroom with a long and storied history of use in East Asian medicine for promoting health and longevity [1]. It is often referred to as the “King of Herbs” or the “Mushroom of Immortality”. Its medicinal use dates back over 2000 years, with its effects documented in ancient scripts like the Shen Nong Ben Cao Jing from China’s Eastern Han dynasty (25–220 AD) [2]. Traditionally, it has been used to treat a variety of ailments and is believed to enhance stamina, increase brain power, improve circulation, and strengthen the immune system [3].
Modern research has begun to validate these traditional claims, attributing the mushroom’s medicinal properties to its rich and varied chemical composition. Bioactive compounds such as polysaccharides, triterpenes, adenosine, organic germanium, phenolic compounds, flavonoids, and ergosterol contribute to its therapeutic effects, which include antioxidant, anticancer, anti-inflammatory, and antimicrobial activities [4,5,6,7,8]. For instance, triterpenic acids have demonstrated significant anticancer effects, while polysaccharides have shown anti-diabetic and antibiotic properties [9,10,11,12]. The antioxidant properties of G. lucidum are particularly noteworthy, as they neutralize reactive oxygen species (ROS) and reactive nitrogen species (RNS), which are implicated in the development of chronic diseases such as cancer, aging, diabetes, cardiovascular disorders, and neurodegeneration [4,13]. Studies have shown that polysaccharides in G. lucidum exhibit potent ROS-scavenging activity and can inhibit tumor growth through immunomodulation and the induction of apoptosis [6,9,10]. Furthermore, its anti-inflammatory effects have been observed in animal models, suggesting significant therapeutic potential [14].
Nepal, with its diverse geography ranging from the Terai plains to the high Himalayas, provides a unique environment for a wide variety of mushrooms [15,16,17,18,19]. While approximately 1300 mushroom species have been reported in Nepal, with 73 identified as having medicinal value, the therapeutic potential of most, including native G. lucidum, remains largely unexplored [20,21]. Research on Nepalese mushrooms, initiated by the Nepal Agricultural Research Council (NARC) in 1974, has historically focused more on cultivation and taxonomy rather than on the detailed analysis of their bioactive properties.
The unique environmental conditions of Nepal’s high-altitude regions—characterized by lower oxygen levels, intense UV radiation, and distinct soil compositions—may lead to the production of G. lucidum strains with enhanced or novel bioactive profiles [22,23]. Despite Nepal’s rich biodiversity, research on high-altitude G. lucidum is scarce, leaving a gap in our understanding of its full potential. This study, therefore, investigated the bioactive constituents, antioxidant activities, and cytotoxic effects of wild G. lucidum collected from the high altitudes of Nepal (Figure 1), utilizing various solvent extracts to comprehensively characterize its mycochemical composition and therapeutic promise.

2. Materials and Methods

2.1. Materials

2,2-Diphenyl-1-picrylhydrazyl (DPPH, ≥95%), pyrogallol (≥98%), Folin–Ciocalteu reagent (2M), sodium carbonate (Na2CO3, ≥99.5%), aluminum chloride (≥99%), potassium acetate (≥99%), quercetin (≥95%), ferrous sulfate (FeSO4, ≥99%), salicylic acid (≥99%), potassium ferricyanide (K3Fe(CN)6, ≥99%), and ferric chloride (FeCl3, ≥98%) were obtained from Sigma-Aldrich Co. (Burlington, MA, USA). Ascorbic acid (≥99%), trichloroacetic acid (99%), NP-40, and isopropanol (≥99%) were purchased from Thermo Scientific Chemicals (Waltham, MA, USA). The Griess reagent (≥98%), hydrochloric acid (HCl, >36.5%), hydrogen peroxide (H2O2), methanol, ethanol, acetone, and hexane were sourced from Fisher Scientific (Waltham, MA, USA) and were of analytical grade. Sodium nitroprusside (≥99%) and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide, ≥98%] were obtained from Merck Millipore (Darmstadt, Germany).
For solvent removal, a Buchi R-215 rotary vacuum evaporator equipped with a V-850 vacuum controller (BUCHI Corporation, New Castle, DE, USA) and a Multi-Vapor P-6 heating bath (BUCHI Corporation, New Castle, DE, USA) was used for simultaneous solvent evaporation under reduced pressure. Absorbance measurements for antioxidant assays were performed using a Labindia Analytical Double Beam UV 3200 Spectrophotometer (Labindia Analytical, Maharashtra, India). Cell culture reagents, including Dulbecco’s Modified Eagle Medium (DMEM), fetal bovine serum (FBS), and penicillin–streptomycin, were obtained from Gibco (Thermo Fisher Scientific, Waltham, MA, USA).

2.2. Collection and Identification

Fresh fruiting bodies of wild G. lucidum were collected from a dead trunk of Quercus lanata on Chandragiri Hill, Nepal (elevation 7482 ft; latitude 27.67402; longitude 85.19874) (Figure 2). The specimens were identified based on detailed macro- and micro-morphological characteristics (Table 1).

2.3. Sample Preparation and Extraction

The samples were cleaned and oven-dried gradually from 45 °C to 60 °C over 3 days until a constant weight was attained. The dried mushrooms were milled into a fine powder and stored in airtight containers for future use. For extraction, 10 g of dried G. lucidum powder, pooled from multiple wild-collected fruiting bodies, was used. Solvent extraction was performed for 10 h in a Soxhlet apparatus with 250 mL each of water (GWE, 100 °C), 70% ethanol (GEE, 60 °C), 80% methanol (GME, 70 °C), and 50% acetone (GAE, 50 °C). Extracts were concentrated under vacuum in a rotary evaporator (50 °C) as shown in the Supplementary Figure S1 and stored in dark vials at 4 °C. The % yield for each solvent was calculated using the following formula:
% Y i e l d = w e i g h t o f t h e G .   l u c i d u m p o w d e r b e f o r e e x t r a c t i o n ( g m ) w e i g h t o f t h e o b t a i n e d e x t r a c t ( g m ) × 100

2.4. Estimation of Total Phenolic, Flavonoid, β-Carotene, and Lycopene

2.4.1. Total Phenolic Content

Total phenolic content was determined using the Folin–Ciocalteu method with pyrogallol as the standard [24]. A calibration curve was established by measuring the absorbance of pyrogallol standards (10–100 µg/mL) at 760 nm (Supplementary Figure S2). Sample solutions (1 mL) were mixed with 5 mL of 10% Folin–Ciocalteu reagent for 5 min at room temperature. Subsequently, 4 mL of 15% Na2CO3 solution was added, and samples were vigorously mixed and incubated in the dark for 2 h at room temperature. Absorbance was read at 760 nm, and results were expressed as pyrogallol equivalents (mg/g dry extract).

2.4.2. Total Flavonoid Content

Total flavonoid content was determined using the aluminum chloride colorimetric method, as adapted from Shraim et al., 2021 [25]. A quercetin standard curve was prepared by diluting a 10 mg quercetin stock in 50% methanol to concentrations ranging from 10 to 100 µg/mL, with absorbance measured at 415 nm (Supplementary Figure S3). Sample aliquots (1 mL) were mixed with 0.5 mL of 1.2% aluminum chloride, 0.5 mL of 120 mM potassium acetate, and 1 mL of distilled water. After 30 min of incubation at room temperature, absorbance was read at 415 nm. Results were expressed as mg quercetin equivalents per g of dry extract (mg QE/g dry weight).

2.4.3. Estimation of β-Carotene and Lycopene Content

β-carotene and lycopene were determined as per Prakash et al., 2016 [26]. Dried extracts (100 mg) were extracted with 10 mL of acetone-hexane (4:6) for 1 min, then filtered. Filtrate absorbance was measured at 453, 505, 645, and 663 nm. Carotenoid concentrations were calculated using the following equations:
Lycopene = (−0.0458 × A663) + (0.372 × A505) + (0.0806 × A453)
β-carotene = (0.216 × A663) − (0.304 × A505) + (0.452 × A453)
Results are presented as mg carotenoid per g of dry extract (mg carotenoid/g dry extract).

2.5. Determination of In Vitro Antioxidant Activities

2.5.1. DPPH (2,2-Diphenyl-1-Picryl-Hydrazyl) Assay

Different concentrations of extracts (20–100 µg/mL) were prepared. To 1 mL of each extract concentration, 2 mL of ice-cold 0.1 mM DPPH solution was added. The mixtures were incubated in the dark at room temperature for 30 min. Absorbance was then measured at 517 nm against a methanol blank [27]. A 3 mL DPPH solution was used as the control. Percentage inhibition was calculated using the formula:
% inhibition = [(A0 − A1)/(A0)] × 100
where A0 is the absorbance of the control, and A1 is the absorbance of the sample.

2.5.2. Superoxide Radical Scavenging Assay

Superoxide radical scavenging activity was assessed by a modified pyrogallol auto-oxidation method [28]. The reaction mixture contained 4.5 mL of 50 mM Tris-HCl buffer (pH 8.2), 0.4 mL of 25 mM pyrogallol, and 1 mL of sample (0.1–0.5 mg/mL). After 5 min incubation at 25 °C, the reaction was terminated by adding 1 mL of 8 mM HCl. Absorbance was measured at 420 nm. Ascorbic acid served as the positive control. Superoxide radical scavenging activity was calculated using the formula:
% inhibition = [(A0 − A1)/(A0)] × 100
where A0 is the absorbance of the control, and A1 is the absorbance of the sample.

2.5.3. Hydroxyl Radical Scavenging Assay

Hydroxyl radical scavenging activity was assessed by measuring the inhibition of salicylic acid hydroxylation [28]. The 7 mL reaction mixture contained 1 mL of sample/standard (100–500 µg/mL), 2 mL of 6 mM FeSO4, 2 mL of 6 mM H2O2, and 2 mL of 6 mM salicylic acid. After incubation at 37 °C for 1 h, absorbance was measured at 510 nm due to the color change of salicylic acid. Scavenging activity was calculated as follows:
% inhibition = [(A0 − A1)/(A0)] × 100
where A0 is the absorbance of the control, and A1 is the absorbance of the sample.

2.5.4. Nitric Oxide Radical Scavenging Assay

Nitric oxide radical scavenging activity was determined with slight modification from Alam et al., 2013 [29]. Samples or ascorbic acid (20–100 µg/mL, 1 mL) were mixed with 2 mL of 10 mM sodium nitroprusside in phosphate buffer and incubated at 25 °C for 2.5 h. To 3 mL of the incubated solution, 3 mL of Griess reagent (1% sulfanilamide, 0.1% naphthylethylenediamine dihydrochloride in 2% H3PO3) was added. Absorbance of the pink color was measured at 540 nm against a blank. Ascorbic acid served as a positive control. Percentage inhibition was calculated using the following equation:
% inhibition = [(A0 − A1)/(A0)] × 100
where A0 is the absorbance of the control, and A1 is the absorbance of the sample.

2.5.5. Reducing Power Assay

The reducing power of samples was assessed via the ferric reducing antioxidant power (FRAP) assay [30]. One mL of sample or standard (20–100 µg/mL) was combined with 2.5 mL of 0.2 M phosphate buffer (pH 6.6) and 2.5 mL of 1% (w/v) K3Fe(CN)6. After 20 min incubation at 50 °C in a water bath, 2.5 mL of 10% (w/v) trichloroacetic acid was added to terminate the reaction. The mixture was centrifuged at 3000 rpm for 10 min. A 2.5 mL aliquot of the supernatant was then mixed with 2.5 mL distilled water and 0.5 mL of 0.1% (w/v) FeCl3. Absorbance was measured at 700 nm against a blank.

2.6. MTT Cell Viability Assay

HeLa cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA; Catalog no. CCL-2). Cells were maintained in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin at 37 °C in a humidified atmosphere containing 5% CO2. For the MTT assay, approximately 5 × 103 cells were seeded per well in a 96-well plate. After 24 h, cells were treated with dimethyl sulfoxide or various extract concentrations for 48 h. Post-treatment, media were removed and replaced with 100 µL fresh medium containing the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) reagent (final concentration 0.4 mg/mL). Plates were incubated at 37 °C for 3 h, during which intracellular purple formazan was observed. Next, 100 µL of solubilization solution (4 mM HCl, 0.1% NP-40 in isopropanol) was added, and plates were kept in the dark for 15 min at room temperature. Absorbance was measured at 570 nm using a microplate reader [31]. Percentage inhibition was calculated as follows:
% inhibition = 100 − [(A1/A0) × 100]
where A0 is the absorbance of the control, and A1 is the absorbance of sample.

2.7. Gas Chromatography–Mass Spectrometry (GC-MS) Analysis

GC-MS analysis of G. lucidum extracts was conducted using a GCMS-QP2010 Ultra (Shimadzu, Kyoto, Japan) equipped with an Rtx-5 M5 capillary column (30 m × 0.25 mm, 0.25 µm film thickness; Restek, Bellefonte, PA, USA). The operating conditions, including solvent cut-off, temperature program, and MS scan parameters, were identical to those described by Tiwari et al., 2023 [32]. Detailed information on the GC-MS run-time procedures and analytical parameters are provided in the Supplementary Materials. Compounds were identified using NIST libraries (NIST 14, Gaithersburg, MD, USA).

2.8. Statistical Analysis

Statistical analysis was performed using GraphPad Prism 10 (GraphPad Software, San Diego, CA, USA). All experiments were conducted in three biological replicates, expressed as mean ± standard deviation (SD). One-way ANOVA with Tukey’s post hoc test was performed to test the statistical significance between control and experimental groups (p < 0.05). IC50 values were calculated using the built-in “Inhibitor vs. Response” nonlinear regression model in GraphPad Prism. Exact p-values for total phenolic and flavonoid content, β-carotene and lycopene content, antioxidant assays, and cytotoxicity assays are provided in Supplementary Figures S4–S11.

3. Results

3.1. Extraction Yield

Extraction efficiency is affected by the chemical nature of bioactive compounds, the extraction method used, sample particle size, the solvent used, as well as the presence of interfering substances [33,34]. The yield of extraction depends on the solvent with varying polarity, temperature, pH, extraction time, and composition of the sample [33,34]. Extraction efficiency (% yield) varied significantly (p < 0.0001) among solvents, with acetone yielding the highest crude extract (GAE; 5.01%), followed by ethanol (GEE; 3.43%), methanol (GME; 2.98%), and water (GWE; 2.29%) (Table 2).

3.2. Estimation of Total Phenolic and Flavonoid Content

Phenolic compounds are recognized as potent chain-breaking antioxidants due to the radical-scavenging capabilities of their hydroxyl groups [35]. The total phenolic content (TPC) exhibited significant variation among the tested solvents (Figure 3A). Ethanol extract (GEE) demonstrated the highest TPC (376.5 ± 9.3 mg PG/g), which was significantly greater (p < 0.0001) than that of methanol extract (GME; 97.3 ± 2.8 mg PG/g), water extract (GWE; 96.6 ± 2.6 mg PG/g), and acetone extract (GAE; 60.5 ± 7.4 mg PG/g). However, no significant difference was observed between GWE and GME (p = 0.9987), indicating comparable phenolic content in these two extracts (see Supplementary Figure S4).
Flavonoid content was quantified using the aluminum chloride colorimetric method and also showed significant variation among extracts (Figure 3B). GEE (30.3 ± 0.5 mg QE/g extract) and GWE (26.7 ± 0.6 mg QE/g extract) had the highest total flavonoid content, significantly (p < 0.0001) exceeding those of GME (6.3 ± 0.4 mg QE/g extract) and GAE (7.9 ± 0.2 mg QE/g extract). Notably, GEE contained approximately 4.7-fold higher total flavonoids compared to GME. All comparisons for TFC were statistically significant (p < 0.01), confirming solvent-dependent differences in flavonoid recovery (Supplementary Figure S4).

3.3. Estimation of β-Carotene and Lycopene

The concentrations of lycopene and β-carotene in G. lucidum extracts were estimated spectrophotometrically. Carotenoid analysis demonstrated limited solvent efficacy and significant variation in extraction efficiency among solvents (Figure 4A,B). β-carotene content was highest in GME (0.45 ± 0.02 mg/g), followed by GEE (0.20 ± 0.01 mg/g), GWE (0.16 ± 0.00 mg/g), and GAE (0.09 ± 0.01 mg/g) (Figure 4A). Statistical analysis confirmed significant differences across most pairwise comparisons (p < 0.0001), with lesser degree between GWE and GEE (p = 0.0174) (see Supplementary Figure S4).
Similarly, lycopene content varied considerably among the extracts, ranging from 0.016 ± 0.000 to 0.067 ± 0.001 mg/g of dry extract (Figure 4B). The highest lycopene content was found in the methanolic extract (GME; 0.067 ± 0.001 mg/g), followed by the ethanolic extract (GEE; 0.031 ± 0.004 mg/g), water extract (GWE; 0.018 ± 0.002 mg/g), and acetone extract (GAE; 0.016 ± 0.000 mg/g). All pairwise comparisons showed statistically significant differences (p < 0.0001), except between GWE and GAE.

3.4. Comparative In Vitro Antioxidant Activities

Antioxidant activity cannot be definitively concluded from a single assay due to the diverse mechanisms involved and variations between in vitro test models. These diverse mechanisms include free radical scavenging, metal ion chelation, reducing power, single electron transfer, and others [29,36]. Therefore, this study employed multiple in vitro antioxidant assays (DPPH radical scavenging, superoxide radical scavenging, hydroxyl radical scavenging, nitric oxide radical scavenging, and reducing power) to comprehensively evaluate and compare the antioxidant potential of G. lucidum solvent extracts.

3.4.1. DPPH Radical Scavenging Activity

The DPPH radical scavenging activity of the extracts (at 100 µg/mL) ranged from 87 ± 5% to 96 ± 0.2% (Figure 5). Methanolic extract (GME) exhibited the highest activity (96 ± 0.2%), followed by acetone extract (GAE; 92 ± 0%), ethanolic extract (GEE; 92 ± 0%), and water extract (GWE; 87 ± 5%). Ascorbic acid, as a standard, showed 97 ± 1% inhibition. All extracts demonstrated strong radical scavenging activity in a dose-dependent manner, with scavenging over 80% of the DPPH radical even at 80 µg/mL. The regression analysis yielded strong R2 values for all extracts, ranging from 0.951 to 0.990, indicating high linearity in the dose–response relationships.

3.4.2. Superoxide Radical Scavenging Activity

Superoxide radical scavenging activity increased significantly with extract concentration (p < 0.05), demonstrating a dose-dependent response (Figure 6). At 500 µg/mL, GEE exhibited the highest scavenging activity (72 ± 1%), followed by GME (55 ± 2%), GAE (40 ± 2%), and GWE (39 ± 1%). Ascorbic acid showed 99 ± 0% scavenging. GEE was significantly more active than GWE, GME, and GAE (p < 0.0001), while GWE and GAE showed similar scavenging patterns, particularly at higher concentrations, and were significantly less active than GEE and GME (p < 0.0001). The scavenging efficiency followed the order: GEE > GME > GAE > GWE. Regression analysis of dose–response curves showed moderate to strong linearity, with R2 values ranging from 0.726 (ascorbic acid) to 0.895 (GEE). Among all extracts, GEE (R2 = 0.895) showed the best-fitting curve, followed by GAE (0.877), GME (0.856), and GWE (0.764).

3.4.3. Hydroxyl Radical Scavenging Activity

All G. lucidum extracts exhibited significant, dose-dependent hydroxyl radical scavenging activity (p < 0.001) (Figure 7). At the tested concentrations, GME showed the highest activity (78 ± 1.5%), closely followed by GEE (77 ± 2.8%), and GWE (66 ± 1.3%), with GAE showing comparatively lower activity (42 ± 1.59%). The standard, ascorbic acid, achieved 48 ± 1.3% inhibition under same conditions, which was notably lower than that of GME, GEE, and GWE. Importantly, the IC50 values for GWE, GEE, and GME were lower than that of ascorbic acid, indicating their superior hydroxyl radical scavenging potential. Although GWE had a relatively low R2 value (0.0649), it displayed robust activity, particularly at lower concentrations, whereas GME, GEE, and GAE showed stronger linearity in their dose–response curves, with R2 values of 0.966, 0.937, and 0.952, respectively.

3.4.4. Nitric Oxide Radical Scavenging Activity

The G. lucidum extracts demonstrated good inhibition of nitric oxide radicals in a concentration-dependent manner (Figure 8). Among them, GEE was significantly more active than GME, GAE, and GWE (p < 0.0001). At 100 µg/mL, GEE exhibited the highest scavenging potential (82 ± 1.2%), followed by GME (64 ± 3.4%), while GAE (42 ± 2.8%), and GWE (42 ± 2.9%) showed statistically similar lower activities (p > 0.999). Ascorbic acid showed 92 ± 6.9% inhibition. GEE and ascorbic acid showed no significant difference (p = 0.0523) at 100 µg/mL despite the numerical difference in inhibition. The order of activity was GEE > GME > GAE ≈ GWE. Regression analysis of the dose–response curve revealed strong linearity for all extracts. Among the extracts, GEE, GME, GWE, and GAE showed consistent trends, supporting the reliability of the observed dose-dependent activity.

3.4.5. Reducing Power Assay

The reducing power assay, which reflects the electron-donating capacity of antioxidants, further confirmed the strong antioxidant potential of G. lucidum extracts (Figure 9). At 100 µg/mL, GEE demonstrated the highest reducing power (0.353 ± 0.003), followed by GME (0.176 ± 0.002), GWE (0.164 ± 0.005), and GAE (0.158 ± 0.001). Standard ascorbic acid showed a reducing power of 0.493 ± 0.001 at the same concentration. GEE had significantly higher reducing power than all other extracts at all concentrations (p < 0.0001). GWE and GME were significantly different (p = 0.0018 at 100 µg/mL), while GWE and GAE were not significantly different at higher dose (p > 0.1). Regression analysis showed strong linearity across all samples, with R2 values ranging from 0.981 to 0.995, indicating highly consistent dose–response behavior.

3.5. MTT-Based Viability Assay in HeLa Cells

Following the characterization of bioactive compounds and antioxidant activities, the cytotoxic potential of G. lucidum extracts was evaluated against human cervical cancer (HeLa) cells via MTT assay (Figure 10). This colorimetric assay measures cellular metabolic activity based on the reduction of MTT reagent to insoluble formazan crystals by mitochondrial dehydrogenases in viable cells (see Supplementary Figure S10). Extracts were tested at concentrations of 100, 500, and 1000 µg/mL, and dose-dependent inhibition of cell viability was observed (Figure 10). At the highest concentration tested (1000 µg/mL), GEE and GWE extracts demonstrated significantly higher cytotoxicity (p < 0.0001), suppressing cell proliferation by >65%. Specifically, GEE achieved 83 ± 1% inhibition and GWE 67 ± 1% inhibition. In comparison, GME and GAE showed more moderate inhibition at 1000 µg/mL, with 52 ± 2% and 35 ± 5% inhibition, respectively. DMSO, used as a vehicle control, showed minimal cytotoxicity. The regression analysis showed high R2 values for GWE (0.970), GME (0.933), and GEE (0.918), indicating a strong dose–response relationship, whereas GAE showed a weak correlation (R2 = 0.750).

3.6. IC50 Comparison of Extraction Solvents for Antioxidant and Cytotoxicity Activities

The extraction efficiency and bioactive potential of G. lucidum varied significantly depending on the solvent used (Figure 11). GEE showed the highest total phenolic (377 ± 9.32 PG/g) and flavonoid (30 ± 1 QE/g) content, while GME showed higher carotenoids (lycopene: 0.067 ± 0.001 mg/g; β-carotene: 0.454 ± 0.000 mg/g) (Figure 11). Antioxidant assays revealed solvent-specific efficacy. All extracts (GWE, GEE, GME, GAE) demonstrated strong DPPH radical scavenging ability, with IC50 values ranging between 5.82 and 19.13 µg/mL, comparable to that of ascorbic acid. Notably, GEE was the most potent in superoxide radical scavenging (IC50: 328.95 µg/mL), nitric oxide radical scavenging (IC50: 57.67 µg/mL), and reducing power (IC50: 78.04 µg/mL) assays (Figure 12). Hydroxyl radical inhibition was strongest in GWE (IC50: 237.89 µg/mL), closely followed by GEE (274.34 µg/mL).
Cytotoxicity analysis using MTT assay further supported solvent-specific trends. GEE and GWE showed dose-dependent inhibition of HeLa cell viability, with IC50 values of 520.19 µg/mL and 702.41 µg/mL, exhibiting moderate activity (Figure 11). GME also showed notable cytotoxicity (951.61 µg/mL), while GAE had the highest IC50 (1513.92 µg/mL), indicating lower potency.
Collectively, ethanol was found to be the most effective extraction solvent, capable of extracting phenolic- and flavonoid-rich fractions with broad-spectrum antioxidant and cytotoxic activities, likely due to ethanol extracting a diverse range of polar bioactive compounds [37,38,39]. These findings underscore the strong correlation between ethanol’s high phenolic/flavonoid content and its multi-target bioactivity, positioning it as the optimal solvent for extracting compounds with therapeutic potential against oxidative stress and cancer.

3.7. Solvent-Dependent Variation in Bioactive Compounds via GC-MS Profiling

GC-MS analysis revealed distinct solvent-dependent chemical profiles in G. lucidum extracts. The GC-MS chromatograms of GEE, GME, and GAE are shown in Figure 12, and individual mass spectra with their chemical structure of key pharmacologically active compounds are presented in Figure 13. Detailed information on the identified compounds detected by GC-MS is provided in the Supplementary Materials (Tables S1–S3). The extracts demonstrated unique distributions of sterols, triterpenoids, terpenoids, fatty acids, and polyphenols, highlighting the influence of solvent polarity on bioactive compound composition (Table 3).
Fatty acids dominated ethanol (53.18%) and methanol (48.57%) extracts, with 9,12-octadecadienoic acid (linoleic derivative: 20.60% in ethanol, 14.05% in methanol) and pentadecanoic acid (14.52% in ethanol) as major constituents. Acetone exhibited the lowest fatty acid content (5.64%) but uniquely contained ergosterol (Vitamin D2 precursor) and retinoic acid. Polar protic solvents (ethanol, methanol) efficiently extracted free fatty acids and esters, including (E)-9-octadecenoic acid ethyl ester (oleic derivative: 3.86% in ethanol), while acetone’s mid-polarity favored sterols (7,22-ergostadienone, 9(11)-dehydroergosteryl benzoate) and triterpenoids (ergosta-4,6,8(14),22-tetraen-3-one). Similarly, Hinokione, an abietene diterpene, was detected across solvent extracts (0.9% in GAE, 2.9% in GEE, and 5.5% in GME) (Table 3).
Pharmacologically significant compounds included ferruginol (exclusive to ethanol), nerolidol acetate (methanol-specific), geranylgeraniol (anti-inflammatory terpenoid), and Hinokione (anti-inflammatory and anticancer) (Table 3). Ethanol and methanol extracts had the highest polyphenol, diterpenoid, and fatty acid content, whereas acetone had higher sterols and triterpenoids, demonstrating solvent polarity as a critical determinant of bioactive compound selectivity. These findings show G. lucidum’s diverse phytochemical composition and the impact of solvent choice in optimizing targeted metabolite extraction.

4. Discussion

This study demonstrated the solvent-dependent extraction of bioactive compounds from G. lucidum collected from high-altitude regions of Nepal, supporting our initial hypothesis that unique environmental factors at these altitudes influence the mushrooms’ secondary metabolite profile. The observed variability in extraction yields across different solvents, with acetone yielding the highest, followed by ethanol, methanol, and water, demonstrated the role of solvent polarity in determining extraction efficiency. This observation aligns with established principles of phytochemistry, where solvent polarity dictates the solubilization and subsequent extraction of specific compound classes [33,79,80,81,82,83,84,85].
GEE displayed the highest TPC and TFC, correlating strongly with its superior antioxidant and cytotoxic performance (Figure 4, Figure 5, Figure 6, Figure 7, Figure 8, Figure 9, Figure 10 and Figure 11). GEE’s TPC exceeded that of 62 wild mushrooms previously reported in Nepal and outperformed other Ganoderma species and various commercial mushrooms [16,17,86]. GEE also contained approximately 4.7 times more flavonoids than GME, with flavonoid levels surpassing those reported in G. applanatum and G. resinaceum [86,87]. This suggests that flavonoids likely constitute a significant portion of the total phenolic content in the tested extracts. Collectively, these findings support the efficacy of ethanol in extracting phenolic- and flavonoid-rich fractions with potential nutraceutical value.
Carotenoid extraction with methanol proved to be most effective among the tested solvents due to its polar nature, disrupting the cellular matrix to release hydrophobic carotenoids, consistent with previous reports of carotenoid content in G. lucidum [79]. Nevertheless, overall carotenoid yields were relatively low compared to the phenolic and flavonoid content (Figure 3 and Figure 4). The values obtained for GME were higher than those previously reported in Turkish mushrooms [88] and some Indian strains of G. lucidum but still lower than those observed in wild Portuguese mushrooms [89]. This reinforces the role of solvent polarity in selective compound recovery. Nonetheless, both β-carotene and lycopene contents were comparatively low when contrasted with the phenolic and flavonoid contents of the same extracts.
Multiple in vitro antioxidant assays, including DPPH, superoxide, hydroxyl, nitric oxide radical scavenging, and reducing power assays, confirmed the strong antioxidant potential of the extracts. All extracts scavenged over 80% of DPPH radicals at 80–100 µg/mL, with GEE and GME showing the strongest activity, likely due to their higher phenolic content. These results indicate better scavenging activity compared to some previously reported wild mushrooms from Nepal, including G. lucidum [16,17,86]. Among the assays, superoxide radical scavenging was highest in GEE (72 ± 1%), suggesting strong potential to neutralize ROS via electron donation, primarily attributed to phenolic hydroxyl groups [90]. Similarly, hydroxyl radical scavenging showed potent activity, with IC50 values for GWE, GEE, and GME lower than that of ascorbic acid, indicating a potent ability to counter lipid peroxidation and protect against DNA damage, mutagenesis, and oxidative cytotoxicity [91,92,93,94]. GEE also demonstrated strong nitric oxide scavenging (82 ± 1.2%), implying its ability to mitigate nitrosative stress and inhibit peroxynitrite formation, which is implicated in nitrosamine-mediated carcinogenesis within the digestive tract [95]. Reducing power, another key antioxidant indicator, was highest in GEE and significantly exceeded values for Boletus edulis and Pleurotus ostreatus [89,96,97,98]. This capacity is linked to the hydrogen-donating ability of flavonoids and phenolics [99,100]. Reducing powers of the ethanolic extracts were notably higher than those reported for other G. lucidum, Boletus edulis, and Pleurotus ostreatus [89,96,97,98]. This reducing capacity is likely due to the hydrogen-donating ability of the compounds present in the extracts, which can halt peroxide formation and terminate radical chain reactions [99,100].
The cytotoxicity of the extracts was assessed against HeLa cells using the MTT assay. All extracts demonstrated dose-dependent inhibition of cell viability. At 1000 µg/mL, GEE exhibited the strongest cytotoxic effect (82.53 ± 1.46%), followed by GWE (Figure 11). These results are consistent with earlier reports on the anticancer effects of G. lucidum, suggesting that ethanol and water extracts contain compounds that may induce apoptosis, modulate immune responses, and arrest cell cycle progression [79,98,101]. The anti-proliferative effects of G. lucidum extracts are well documented in the literature and have been reported in a variety of cancer cell lines, including HeLa (cervical cancer), A549 (lung cancer), LS174 (colon cancer), and MCF-7 (breast cancer) cells [101,102]. As described in recent studies, including the work by Mousavi et al. (2023), these cytotoxic effects are largely attributed to the presence of bioactive compounds such as pentadecanoic acid, 14-methyl ester; hexanoic acid; (Z,Z)-9,12-octadecadienoic acid methyl ester; ergosta-4,6,8(14),22-tetraen-3-one (ergosta-tetraenone); 7,22-ergostadienone; and various Ganoderma-derived polysaccharides [101]. Notably, our GC-MS profiling confirmed the presence of these compounds in the solvent extracts of G. lucidum, providing mechanistic support for the observed cytotoxicity in HeLa cells and reinforcing their potential therapeutic relevance in cancer treatment.
When IC50 values were compared across assays, ethanol emerged as the most effective solvent for extracting multifunctional bioactives (Figure 12). GEE had the lowest IC50 values in superoxide (328.95 µg/mL), nitric oxide (57.67 µg/mL), and reducing power (78.04 µg/mL) assays. Although GWE had stronger hydroxyl radical inhibition, GEE consistently performed across multiple assays and demonstrated superior cytotoxicity (IC50: 520.19 µg/mL). These findings highlight ethanol’s extraction of polar antioxidant and anticancer agents with broad-spectrum activity. Interestingly, although acetone yielded the highest crude extract mass, it performed poorly in both antioxidant and cytotoxic assays. This apparent discrepancy highlights that a high extraction yield does not necessarily translate to high biological activity. GC-MS analysis revealed that acetone preferentially extracted sterols and retinoids which are structurally large or less bioactive compounds, which, while pharmacologically interesting, may not exert immediate antioxidant or cytotoxic effects at the concentration tested. These findings indicate that extract mass alone is not a reliable indicator of bioactivity.
GC-MS analysis confirmed solvent-specific extraction efficiency, identifying steroids, terpenoids, diterpenoids, triterpenoids, polyphenols, and fatty acids (Table 3 and Supplementary Tables S1–S3). Polyunsaturated fatty acids were most abundant in ethanol and methanol. One of the major bioactive constituents gaining a lot of attention recently and found in all three extracts is hinokione, an abietane-type diterpene known for its significant anticancer and anti-inflammatory activities [70]. Hinokione has been shown to exhibit cytotoxicity against MV-3 and MIAPaCa-2 human cancer cell lines with IC50 values of 34.1 and 17.9 µM, respectively, and has demonstrated β-cell regeneration and hypoglycemic effects in zebrafish [70,103,104]. Ferruginol, another abietane diterpenoid with neuroprotective and anticancer activity, was exclusively present in GEE. It has shown antiproliferative activity in melanoma (Sk-MEL28) and various cancer cell lines, including prostate, lung, gastric, and breast cancers, as well as efficacy in CL1-5 xenograft mouse models [59,61,63]. Methanol extract contained nerolidol acetate, a sesquiterpene with antioxidant, antibacterial, anti-biofilm, antifungal, and anticancer properties [73,74,75]. Geranylgeraniol, an anti-inflammatory isoprenoid, was also detected in methanol and ethanol extracts, likely contributing to their antioxidant activity [66,67,68,69]. GAE was rich in ergosterol and retinoic acid, with ergosterol comprising more than two-thirds of the total extracted compounds. As a vitamin D precursor, ergosterol has potential for addressing vitamin D deficiency-associated diseases, including cancers, rheumatoid arthritis, and multiple sclerosis [105]. Estrogenic derivatives such as 7,22-ergostadienone and 9(11)-dehydroergosteryl benzoate, known for their therapeutic applications, were found across all extracts (Table 3). Collectively, the GC-MS dataset underscores the profound impact of solvent choice on the chemical profile of mushroom extracts and the types of bioactive molecules recovered.
The observed solvent-dependent variation in extracted compounds across solvents can be explained by their fundamental physiochemical principles. Ethanol and methanol, both polar protic solvents, can form hydrogen bonds and penetrate cell walls easily, allowing them to extract a broad range of polar bioactives such as phenolics, flavonoids, and fatty acids. Acetone, with its intermediate polarity and aprotic nature, is better at extracting more hydrophobic and structurally rigid compounds. Water, while highly polar, has limited ability to solubilize non-polar or moderately polar compounds. As a result, it primarily extracts hydrophilic constituents, such as polysaccharides, simple phenolics, and certain proteins. These solvent-dependent metabolic signatures not only explain the variation in antioxidant and cytotoxic activities observed across assays but also provide mechanistic insight into the functional contributions of specific compound classes. The selective enrichment of fatty acids, sterols, and terpenoids by distinct solvents offers a strategic basis for tailoring extraction protocols to maximize therapeutic yield.
While our findings provide a strong preliminary basis for the therapeutic potential of G. lucidum, several limitations should be addressed in future studies. The cytotoxicity effects were evaluated solely using the MTT assay on a single cancer cell line, without comparison to normal cells. This limits our understanding of extract selectivity and potential off-target effects in normal cells. Follow-up studies should expand cytotoxicity screening to include additional cancer cell lines, such as MCF-7, A549, HT-29, as well as non-cancerous cell lines like HEK293 to assess therapeutic selectivity and safety profiles. Investigating apoptosis-related pathways, cell cycle arrest, or mitochondrial dysfunction will be essential to validate the anticancer properties observed. Building upon these findings, future investigations should also focus on isolating and functionally characterizing the specific bioactive compounds responsible for the observed activities through selective extraction and purification of bioactive candidate compounds. Testing these isolated compounds will provide a clearer understanding of their therapeutic potential.

5. Conclusions

Our study aimed to investigate the therapeutic potential of G. lucidum from Nepal’s high-altitude regions, and our findings strongly confirm it as a key source of bioactive compounds. Through this work, we have shown that the choice of extraction solvent is critical, significantly impacting not only the yield but also the specific bioactive compounds obtained and, consequently, their biological activities. While acetone yielded the highest amount of crude extract, ethanol and methanol extracts showed higher phenolic and flavonoid content, correlated with high antioxidant activity across a spectrum of in vitro assays. We also showed that high extraction yield does not necessarily translate to high biological activity. The ethanol and water extracts also demonstrated moderate ability to inhibit HeLa cell growth. GC-MS analysis identified a diverse array of beneficial compounds, including fatty acids, sterols like ergosterol, and various terpenoids (diterpenoids, triterpenoids). The specific distribution of these compounds varied depending on the extraction solvents, and they collectively contribute to the observed health benefits. Future research should focus on optimizing extraction methods and characterizing these individual compounds to maximize specific bioactivities, which will be critical for bridging the gap between traditional use and modern applications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cimb47080624/s1.

Author Contributions

Conceptualization, I.T., A.P., S.T., and S.C.A.; methodology, I.T., A.P., and S.C.A.; software, I.T. and S.T.; validation, I.T., A.P., S.T., and S.C.A.; formal analysis, I.T.; investigation, I.T. and A.P.; resources, S.C.A.; data curation, I.T. and A.P.; writing—original draft preparation, I.T. and S.T.; writing—review and editing, I.T., A.P., S.T., and S.C.A.; visualization, I.T.; supervision, S.C.A.; project administration, I.T. and S.T.; funding acquisition, I.T. and S.C.A. All authors have read and agreed to the published version of the manuscript.

Funding

The research involved received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

Authors of this paper are highly grateful to Nabina Maharjan and Dilip Bhattrai. We are also thankful to the Nepal Academy of Science and Technology and SANN International College for providing the funding and laboratory facilities.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
GWEGanoderma lucidum water extract
GEEGanoderma lucidum ethanol extract
GMEGanoderma lucidum methanol extract
GAEGanoderma lucidum acetone extract
TPCTotal phenolic content
TFPTotal flavonoid content
DPPH2,2-diphenyl-1-picryl-hydrazyl
MTT3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide
GC-MSGas chromatography–mass spectrometry
LTPLong-term potentiation
MRSAMethicillin-resistant Staphylococcus aureus
HeLaHuman cervical carcinoma cell lines

References

  1. Zhou, L.W.; Cao, Y.; Wu, S.H.; Vlasák, J.; Li, D.W.; Li, M.J.; Dai, Y.C. Global Diversity of the Ganoderma lucidum Complex (Ganodermataceae, Polyporales) Inferred from Morphology and Multilocus Phylogeny. Phytochemistry 2015, 114, 7–15. [Google Scholar] [CrossRef] [PubMed]
  2. Bishop, K.S.; Kao, C.H.J.; Xu, Y.; Glucina, M.P.; Paterson, R.R.M.; Ferguson, L.R. From 2000 Years of Ganoderma lucidum to Recent Developments in Nutraceuticals. Phytochemistry 2015, 114, 56–65. [Google Scholar] [CrossRef]
  3. Ekiz, E.; Oz, E.; Abd El-Aty, A.M.; Proestos, C.; Brennan, C.; Zeng, M.; Tomasevic, I.; Elobeid, T.; Çadırcı, K.; Bayrak, M.; et al. Exploring the Potential Medicinal Benefits of Ganoderma lucidum: From Metabolic Disorders to Coronavirus Infections. Foods 2023, 12, 1512. [Google Scholar] [CrossRef] [PubMed]
  4. Du, Y.; Tian, L.; Wang, Y.; Li, Z.; Xu, Z. Chemodiversity, Pharmacological Activity, and Biosynthesis of Specialized Metabolites from Medicinal Model Fungi Ganoderma lucidum. Chin. Med. 2024, 19, 51. [Google Scholar] [CrossRef]
  5. Baby, S.; Johnson, A.J.; Govindan, B. Secondary Metabolites from Ganoderma. Phytochemistry 2015, 114, 66–101. [Google Scholar] [CrossRef]
  6. Zhu, M.; Chang, Q.; Wong, L.K.; Chong, F.S.; Li, R.C. Triterpene Antioxidants from Ganoderma lucidum. Phytother. Res. 1999, 531, 529–531. [Google Scholar] [CrossRef]
  7. Yuen, J.W.M.; Gohel, M.D.I.; Yuen, J.W.M.; Gohel, M.D.I. Anticancer Effects of Ganoderma lucidum: A Review of Scientific Evidence. Nutr. Cancer 2005, 53, 11–17. [Google Scholar] [CrossRef]
  8. Acharya, K.; Bera, M.; Tarafder, E.; Dasgupta, A. Pharmacognostic Standardization of Ganoderma lucidum: A Commercially Explored Medicinal Mushroom. Der Pharmacia Lettre 2015, 7, 175–181. [Google Scholar]
  9. Ferreira, I.C.F.R.; Heleno, S.A.; Reis, F.S.; Stojkovic, D.; Queiroz, M.J.R.P.; Vasconcelos, M.H.; Sokovic, M. Chemical Features of Ganoderma Polysaccharides with Antioxidant, Antitumor and Antimicrobial Activities. Phytochemistry 2015, 114, 38–55. [Google Scholar] [CrossRef]
  10. Mohan, K.; Padmanaban, M.; Uthayakumar, V. Isolation, Structural Characterization and Antioxidant Activities of Polysaccharide from Ganoderma lucidum (Higher Basidiomycetes). Am. J. Biomed. Life Sci. 2015, 3, 168–175. [Google Scholar]
  11. Heleno, S.A.; Barros, L.; Martins, A.; João, M.; Queiroz, R.P. Fruiting Body, Spores and in Vitro Produced Mycelium of Ganoderma lucidum from Northeast Portugal: A Comparative Study of the Antioxidant Potential of Phenolic and Polysaccharidic Extracts. Food Res. Int. 2012, 46, 135–140. [Google Scholar] [CrossRef]
  12. Niedermeyer, T.H.J.; Lindequist, U.; Mentel, R.; Go, D.; Schmidt, E.; Thurow, K.; Lalk, M. Antiviral Terpenoid Constituents of Ganoderma pfeifferi. J. Nat. Prod. 2005, 68, 1728–1731. [Google Scholar] [CrossRef] [PubMed]
  13. Martínez-Montemayor, M.M.; Ling, T.; Suárez-Arroyo, I.J.; Ortiz-Soto, G.; Santiago-Negrón, C.L.; Lacourt-Ventura, M.Y.; Valentín-Acevedo, A.; Lang, W.H.; Rivas, F. Identification of Biologically Active Ganoderma lucidum Compounds and Synthesis of Improved Derivatives That Confer Anti-Cancer Activities in Vitro. Front. Pharmacol. 2019, 10, 115. [Google Scholar] [CrossRef]
  14. Hasnat, M.A.; Pervin, M.; Cha, K.M.; Kim, S.K.; Lim, B.O. Anti-Inflammatory Activity on Mice of Extract of Ganoderma lucidum Grown on Rice via Modulation of MAPK and NF-ΚB Pathways. Phytochemistry 2015, 114, 125–136. [Google Scholar] [CrossRef]
  15. Uddin, K.; Shrestha, H.L.; Murthy, M.S.R.; Bajracharya, B.; Shrestha, B.; Gilani, H.; Pradhan, S.; Dangol, B. Development of 2010 National Land Cover Database for the Nepal. J. Environ. Manag. 2015, 148, 82–90. [Google Scholar] [CrossRef]
  16. Hai Bang, T.; Suhara, H.; Doi, K.; Ishikawa, H.; Fukami, K.; Parajuli, G.P.; Katakura, Y.; Yamashita, S.; Watanabe, K.; Adhikari, M.K.; et al. Wild Mushrooms in Nepal: Some Potential Candidates as Antioxidant and ACE-Inhibition Sources. Evid.-Based Complement. Altern. Med. 2014, 2014, 195305. [Google Scholar] [CrossRef]
  17. Tamrakar, S.; Tran, H.B.; Nishida, M.; Kaifuchi, S.; Suhara, H.; Doi, K.; Fukami, K.; Parajuli, G.P.; Shimizu, K. Antioxidative Activities of 62 Wild Mushrooms from Nepal and the Phenolic Profile of Some Selected Species. J. Nat. Med. 2016, 70, 769–779. [Google Scholar] [CrossRef] [PubMed]
  18. Christensen, M.; Bhattarai, S.; Devkota, S.; Larsen, H.O. Collection and Use of Wild Edible Fungi in Nepal. Econ. Bot. 2008, 62, 12–23. [Google Scholar] [CrossRef]
  19. Devkota, S.; Fang, W.; Arunachalam, K.; Phyo, K.M.M.; Shakya, B. Systematic Review of Fungi, Their Diversity and Role in Ecosystem Services from the Far Eastern Himalayan Landscape (FHL). Heliyon 2023, 9, e12756. [Google Scholar] [CrossRef]
  20. Adhikari, M.K. Mushrooms of Nepal; Durrieu, G., Cotter, H.V.T., Eds.; Self-Published: Kathmandu, Nepal, 2014. [Google Scholar]
  21. Paudel, P.K.; Bhattarai, B.P.; Kindlmann, P. An Overview of the Biodiversity in Nepal. In Himalayan Biodiversity in the Changing World; Springer: Berlin/Heidelberg, Germany, 2012; pp. 1–40. ISBN 9789400718029. [Google Scholar]
  22. Pan, L.; Yang, N.; Sui, Y.; Li, Y.; Zhao, W.; Zhang, L.; Mu, L.; Tang, Z. Altitudinal Variation on Metabolites, Elements, and Antioxidant Activities of Medicinal Plant. Asarum. Metab. 2023, 13, 1193. [Google Scholar] [CrossRef]
  23. Rodríguez-Hernández, D. Secondary Metabolites as a Survival Strategy in Plants of High Mountain Habitats. Bol. Latinoam. Caribe Plantas Med. Aromat. 2019, 18, 444–458. [Google Scholar] [CrossRef]
  24. Jadhav, A.P.; Kareparamban, J.A.; Nikam, P.H.; Kadam, V.J. Spectrophotometric Estimation of Ferulic Acid from Ferula Asafoetida by Folin-Ciocalteu’s Reagent. Der Pharm. Sin. 2012, 3, 680–684. [Google Scholar]
  25. Shraim, A.M.; Ahmed, T.A.; Rahman, M.M.; Hijji, Y.M. Determination of Total Flavonoid Content by Aluminum Chloride Assay: A Critical Evaluation. LWT 2021, 150, 111932. [Google Scholar] [CrossRef]
  26. Kumari, K.; Prakash, V.; Rana, S.; Sagar, A. In Vitro Antioxidant Activity of Methanolic Extract of Ganoderma lucidum (Curt) P. Karst. Int. J. Adv. Sci. Res. 2016, 1, 51–54. [Google Scholar]
  27. Chang, S.S.T.; Wu, J.H.; Wang, S.Y.; Kang, P.L.; Yang, N.S.; Shyur, L.F. Antioxidant Activity of Extracts from Acacia confusa Bark and Heartwood. J. Agric. Food Chem. 2001, 49, 3420–3424. [Google Scholar] [CrossRef]
  28. Liu, J.; Jia, L.; Kan, J.; Jin, C.H. In Vitro and in Vivo Antioxidant Activity of Ethanolic Extract of White Button Mushroom (Agaricus bisporus). Food Chem. Toxicol. 2013, 51, 310–316. [Google Scholar] [CrossRef] [PubMed]
  29. Alam, N.; Bristi, N.J. Review on in Vivo and in Vitro Methods Evaluation of Antioxidant Activity. Saudi Pharm. J. 2013, 21, 143–152. [Google Scholar] [CrossRef] [PubMed]
  30. El Jemli, M.; Kamal, R.; Marmouzi, I.; Zerrouki, A.; Cherrah, Y.; Alaoui, K. Radical-Scavenging Activity and Ferric Reducing Ability of Juniperus Thurifera (L.), J. Oxycedrus (L.), J. Phoenicea (L.) and Tetraclinis Articulata (L.). Adv. Pharmacol. Sci. 2016, 2016, 6392656. [Google Scholar] [CrossRef]
  31. Li, X.M.; Luo, X.G.; He, J.F.; Wang, N.; Zhou, H.; Yang, P.L.; Zhang, T.C. Induction of Apoptosis in Human Cervical Carcinoma Hela Cells by Active Compounds from Hypericum ascyron L. Oncol. Lett. 2018, 15, 3944–3950. [Google Scholar] [CrossRef]
  32. Tiwari, S.; Dhakal, N. Analysis of Variations in Biomolecules during Various Growth Phases of Freshwater Microalgae Chlorella sp. Appl. Food Biotechnol. 2023, 10, 73–84. [Google Scholar] [CrossRef]
  33. Do, Q.D.; Angkawijaya, A.E.; Tran-Nguyen, P.L.; Huynh, L.H.; Soetaredjo, F.E.; Ismadji, S.; Ju, Y.H. Effect of Extraction Solvent on Total Phenol Content, Total Flavonoid Content, and Antioxidant Activity of Limnophila aromatica. J. Food Drug Anal. 2014, 22, 296–302. [Google Scholar] [CrossRef]
  34. Gil-Martín, E.; Forbes-Hernández, T.; Romero, A.; Cianciosi, D.; Giampieri, F.; Battino, M. Influence of the Extraction Method on the Recovery of Bioactive Phenolic Compounds from Food Industry By-Products. Food Chem. 2022, 378, 131918. [Google Scholar] [CrossRef] [PubMed]
  35. Platzer, M.; Kiese, S.; Tybussek, T.; Herfellner, T.; Schneider, F.; Schweiggert-Weisz, U.; Eisner, P. Radical Scavenging Mechanisms of Phenolic Compounds: A Quantitative Structure-Property Relationship (QSPR) Study. Front. Nutr. 2022, 9, 882458. [Google Scholar] [CrossRef] [PubMed]
  36. Apak, R.; Özyürek, M.; Güçlü, K.; Çapanoʇlu, E. Antioxidant Activity/Capacity Measurement. 1. Classification, Physicochemical Principles, Mechanisms, and Electron Transfer (ET)-Based Assays. J. Agric. Food Chem. 2016, 64, 997–1027. [Google Scholar] [CrossRef]
  37. Kozhantayeva, A.; Tursynova, N.; Kolpek, A.; Aibuldinov, Y.; Tursynova, A.; Mashan, T.; Mukazhanova, Z.; Ibrayeva, M.; Zeinuldina, A.; Nurlybayeva, A.; et al. Phytochemical Profiling, Antioxidant and Antimicrobial Potentials of Ethanol and Ethyl Acetate Extracts of Chamaenerion latifolium. Pharmaceuticals 2024, 17, 996. [Google Scholar] [CrossRef] [PubMed]
  38. Dai, J.; Mumper, R.J. Plant Phenolics: Extraction, Analysis and Their Antioxidant and Anticancer Properties. Molecules 2010, 15, 7313–7352. [Google Scholar] [CrossRef]
  39. Lefebvre, T.; Destandau, E.; Lesellier, E. Selective Extraction of Bioactive Compounds from Plants Using Recent Extraction Techniques. J. Chromatogr. A 2021, 1635, 461770. [Google Scholar] [CrossRef]
  40. Thang, T.D.; Kuo, P.C.; Hwang, T.L.; Yang, M.L.; Ngoc, N.T.B.; Han, T.T.N.; Lin, C.W.; Wu, T.S. Triterpenoids and Steroids from Ganoderma mastoporum and Their Inhibitory Effects on Superoxide Anion Generation and Elastase Release. Molecules 2013, 18, 14285–14292. [Google Scholar] [CrossRef]
  41. Gao, J.; Wang, L.W.; Zheng, H.C.; Damirin, A.; Ma, C.M. Cytotoxic Constituents of Lasiosphaera fenzlii on Different Cell Lines and the Synergistic Effects with Paclitaxel. Nat. Prod. Res. 2016, 30, 1862–1865. [Google Scholar] [CrossRef]
  42. Ramos-Ligonio, A.; López-Monteon, A.; de la Soledad Lagunes-Castro, M.; Suárez-Medellín, J.; Espinoza, C.; Mendoza, G.; Trigos, Á. In Vitro Expression of Toll-like Receptors and Proinflammatory Molecules Induced by Ergosta-7,22-Dien-3-One Isolated from a Wild Mexican Strain of Ganoderma oerstedii (Agaricomycetes). Int. J. Med. Mushrooms 2017, 19, 203–211. [Google Scholar] [CrossRef]
  43. Krivošija, S.; Nastić, N.; Karadžić Banjac, M.; Kovačević, S.; Podunavac-Kuzmanović, S.; Vidović, S. Supercritical Extraction and Compound Profiling of Diverse Edible Mushroom Species. Foods 2025, 14, 107. [Google Scholar] [CrossRef]
  44. Parmar, R.; Kumar, D. Study of Chemical Composition in Wild Edible Mushroom Pleurotus cornucopiae (Paulet) from Himachal Pradesh, India by Using Fourier Transforms Infrared Spectrometry (FTIR), Gas Chromatography-Mass Spectrometry (GCMS) and X-Ray Fluorescence (XRF). Biol. Forum–An. Int. J. 2015, 7, 1057–1066. [Google Scholar]
  45. Akwu, N.A.; Naidoo, Y.; Singh, M.; Lin, J.; Aribisala, J.O.; Sabiu, S.; Lekhooa, M.; Aremu, A.O. Phytochemistry, Antibacterial and Antioxidant Activities of Grewia lasiocarpa E. Mey. Ex Harv. Fungal Endophytes: A Computational and Experimental Validation Study. Chem. Biodivers. 2025, 22, e202402908. [Google Scholar] [CrossRef]
  46. Das Gupta, S.; Suh, N. Tocopherols in Cancer: An Update. Mol. Nutr. Food Res. 2016, 60, 1354–1363. [Google Scholar] [CrossRef] [PubMed]
  47. Huang, H.; He, Y.; Cui, X.X.; Goodin, S.; Wang, H.; Du, Z.Y.; Li, D.; Zhang, K.; Tony Kong, A.N.; Dipaola, R.S.; et al. Potent Inhibitory Effect of β-Tocopherol on Prostate Cancer Cells Cultured in Vitro and Grown as Xenograft Tumors in Vivo. J. Agric. Food Chem. 2014, 62, 10752–10758. [Google Scholar] [CrossRef]
  48. Biernacki, K.; Daśko, M.; Ciupak, O.; Kubiński, K.; Rachon, J.; Demkowicz, S. Novel 1,2,4-Oxadiazole Derivatives in Drug Discovery. Pharmaceuticals 2020, 13, 111. [Google Scholar] [CrossRef] [PubMed]
  49. Glomb, T.; Świątek, P. Antimicrobial Activity of 1,3,4-Oxadiazole Derivatives. Int. J. Mol. Sci. 2021, 22, 6979. [Google Scholar] [CrossRef] [PubMed]
  50. Siwach, A.; Verma, P.K. Therapeutic Potential of Oxadiazole or Furadiazole Containing Compounds. BMC Chem. 2020, 14, 70. [Google Scholar] [CrossRef]
  51. Ehrsam, D.; Porta, F.; Mori, M.; Meyer Zu Schwabedissen, H.E.; Via, L.D.; Garcia-Argaez, A.N.; Basile, L.; Meneghetti, F.; Villa, S.; Gelain, A. Unravelling the Antiproliferative Activity of 1,2,5-Oxadiazole Derivatives. Anticancer Res. 2019, 39, 3453–3461. [Google Scholar] [CrossRef]
  52. Luczynski, M.; Kudelko, A. Synthesis and Biological Activity of 1,3,4-Oxadiazoles Used in Medicine and Agriculture. Appl. Sci. 2022, 12, 3756. [Google Scholar] [CrossRef]
  53. Su, J.C.; Pan, X.X.; Wei, X.; Lei, X.; Zhang, P. Structurally Diverse Steroids from an Endophyte of Aspergillus tennesseensis 1022LEF Attenuates LPS-Induced Inflammatory Response through the Cholinergic Anti-Inflammatory Pathway. Chem. Biol. Interact. 2022, 362, 109998. [Google Scholar] [CrossRef]
  54. Zhao, Y.Y.; Cheng, X.L.; Cui, J.H.; Yan, X.R.; Wei, F.; Bai, X.; Lin, R.C. Effect of Ergosta-4,6,8(14),22-Tetraen-3-One (Ergone) on Adenine-Induced Chronic Renal Failure Rat: A Serum Metabonomic Study Based on Ultra Performance Liquid Chromatography/High-Sensitivity Mass Spectrometry Coupled with MassLynx i-FIT Algorithm. Clin. Chim. Acta 2012, 413, 1438–1445. [Google Scholar] [CrossRef]
  55. Zhao, Y.Y.; Shen, X.; Chao, X.; Ho, C.C.; Cheng, X.L.; Zhang, Y.; Lin, R.C.; Du, K.J.; Luo, W.J.; Chen, J.Y.; et al. Ergosta-4,6,8(14),22-Tetraen-3-One Induces G2/M Cell Cycle Arrest and Apoptosis in Human Hepatocellular Carcinoma HepG2 Cells. Biochim. Biophys. Acta Gen. Subj. 2011, 1810, 384–390. [Google Scholar] [CrossRef]
  56. Nilkhet, S.; Vongthip, W.; Lertpatipanpong, P.; Prasansuklab, A.; Tencomnao, T.; Chuchawankul, S.; Baek, S.J. Ergosterol Inhibits the Proliferation of Breast Cancer Cells by Suppressing AKT/GSK-3beta/Beta-Catenin Pathway. Sci. Rep. 2024, 14, 19664. [Google Scholar] [CrossRef]
  57. Dupont, S.; Fleurat-Lessard, P.; Cruz, R.G.; Lafarge, C.; Grangeteau, C.; Yahou, F.; Gerbeau-Pissot, P.; Abrahão Júnior, O.; Gervais, P.; Simon-Plas, F.; et al. Antioxidant Properties of Ergosterol and Its Role in Yeast Resistance to Oxidation. Antioxidants 2021, 10, 1024. [Google Scholar] [CrossRef]
  58. Rangsinth, P.; Sharika, R.; Pattarachotanant, N.; Duangjan, C.; Wongwan, C.; Sillapachaiyaporn, C.; Nilkhet, S.; Wongsirojkul, N.; Prasansuklab, A.; Tencomnao, T.; et al. Potential Beneficial Effects and Pharmacological Properties of Ergosterol, a Common Bioactive Compound in Edible Mushrooms. Foods 2023, 12, 2529. [Google Scholar] [CrossRef]
  59. Li, W.; Cao, J.; Wang, X.; Zhang, Y.; Sun, Q.; Jiang, Y.; Yao, J.; Li, C.; Wang, Y.; Wang, W. Ferruginol Restores SIRT1-PGC-1α-Mediated Mitochondrial Biogenesis and Fatty Acid Oxidation for the Treatment of DOX-Induced Cardiotoxicity. Front. Pharmacol. 2021, 12, 773834. [Google Scholar] [CrossRef] [PubMed]
  60. Varbanov, M.; Philippot, S.; González-Cardenete, M.A. Anticoronavirus Evaluation of Antimicrobial Diterpenoids: Application of New Ferruginol Analogues. Viruses 2023, 15, 1342. [Google Scholar] [CrossRef] [PubMed]
  61. Bispo de Jesus, M.; Zambuzzi, W.F.; Ruela de Sousa, R.R.; Areche, C.; Santos de Souza, A.C.; Aoyama, H.; Schmeda-Hirschmann, G.; Rodríguez, J.A.; Monteiro de Souza Brito, A.R.; Peppelenbosch, M.P.; et al. Ferruginol Suppresses Survival Signaling Pathways in Androgen-Independent Human Prostate Cancer Cells. Biochimie 2008, 90, 843–854. [Google Scholar] [CrossRef] [PubMed]
  62. Salih, A.M.; Al-Qurainy, F.; Tarroum, M.; Khan, S.; Nadeem, M.; Shaikhaldein, H.O.; Alansi, S. Phytochemical Compound Profile and the Estimation of the Ferruginol Compound in Different Parts (Roots, Leaves, and Seeds) of Juniperus Procera. Separations 2022, 9, 352. [Google Scholar] [CrossRef]
  63. Shao, L.; González-Cardenete, M.A.; Prieto-Garcia, J.M. In Vitro Cytotoxic Effects of Ferruginol Analogues in Sk-MEL28 Human Melanoma Cells. Int. J. Mol. Sci. 2023, 24, 16322. [Google Scholar] [CrossRef]
  64. Ho, S.T.; Tung, Y.T.; Kuo, Y.H.; Lin, C.C.; Wu, J.H. Ferruginol Inhibits Non-Small Cell Lung Cancer Growth by Inducing Caspase-Associated Apoptosis. Integr. Cancer Ther. 2015, 14, 86–97. [Google Scholar] [CrossRef]
  65. Wang, X.; Cao, G.; Ding, D.; Li, F.; Zhao, X.; Wang, J.; Yang, Y. Ferruginol Prevents Degeneration of Dopaminergic Neurons by Enhancing Clearance of α-Synuclein in Neuronal Cells. Fitoterapia 2022, 156, 105066. [Google Scholar] [CrossRef]
  66. Sediva, A.; Orlicky, M.; Vrabcova, P.; Klocperk, A.; Kalina, T.; Fujiwara, H.; Hsu, F.-F.; Bambouskova, M. Geranylgeraniol Supplementation Leads to an Improvement in Inflammatory Parameters and Reversal of the Disease Specific Protein Signature in Patients with Hyper-IgD Syndr. medRxiv 2024. [Google Scholar] [CrossRef]
  67. Gheith, R.; Sharp, M.; Stefan, M.; Ottinger, C.; Lowery, R.; Wilson, J. The Effects of Geranylgeraniol on Blood Safety and Sex Hormone Profiles in Healthy Adults: A Dose-Escalation, Randomized, Placebo-Controlled Trial. Nutraceuticals 2023, 3, 605–618. [Google Scholar] [CrossRef]
  68. Chung, E.; Elmassry, M.M.; Cao, J.J.; Kaur, G.; Dufour, J.M.; Hamood, A.N.; Shen, C.L. Beneficial Effect of Dietary Geranylgeraniol on Glucose Homeostasis and Bone Microstructure in Obese Mice Is Associated with Suppression of Proinflammation and Modification of Gut Microbiome. Nutr. Res. 2021, 93, 27–37. [Google Scholar] [CrossRef]
  69. Chin, K.Y.; Ekeuku, S.O.; Trias, A. The Role of Geranylgeraniol in Managing Bisphosphonate-Related Osteonecrosis of the Jaw. Front. Pharmacol. 2022, 13, 878556. [Google Scholar] [CrossRef]
  70. Wang, Z.; Yu, Z.W.; Zhang, Y.; Wang, W.H.; Wu, X.Y.; Liu, S.Z.; Bin, Y.L.; Cai, B.P.; Huang, S.Y.; Fang, M.J.; et al. Hinokione: An Abietene Diterpene with Pancreatic β Cells Regeneration and Hypoglycemic Activity, and Other Derivatives with Novel Structures from the Woods of Agathis dammara. J. Nat. Med. 2024, 78, 849–862. [Google Scholar] [CrossRef]
  71. Gáborová, M.; Šmejkal, K.; Kubínová, R. Abietane Diterpenes of the Genus Plectranthus sensu lato. Molecules 2022, 27, 166. [Google Scholar] [CrossRef] [PubMed]
  72. Ulubelen, A.; Topcu, G.; Johansson, C.B. Norditerpenoids and Diterpenoids from Salvia Multicaulis with Antituberculous Activity. J. Nat. Prod. 1997, 60, 1275–1280. [Google Scholar] [CrossRef]
  73. Dong, J.R.; Chang, W.W.; Chen, S.M. Nerolidol Inhibits Proliferation of Leiomyoma Cells via Reactive Oxygen Species-Induced DNA Damage and Downregulation of the ATM/Akt Pathway. Phytochemistry 2021, 191, 112901. [Google Scholar] [CrossRef]
  74. Judzentiene, A.; Butkiene, R.; Budiene, J.; Tomi, F.; Casanova, J. Composition of Seed Essential Oils of Rhododendron tomentosu. Nat. Prod. Commun. 2012, 7, 227–230. [Google Scholar] [PubMed]
  75. Chan, W.K.; Tan, L.T.H.; Chan, K.G.; Lee, L.H.; Goh, B.H. Nerolidol: A Sesquiterpene Alcohol with Multi-Faceted Pharmacological and Biological Activities. Molecules 2016, 21, 529. [Google Scholar] [CrossRef]
  76. Pouso, M.R.; Cairrao, E. Effect of Retinoic Acid on the Neurovascular Unit: A Review. Brain Res. Bull. 2022, 184, 34–45. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, H.P.; Casadesus, G.; Zhu, X.; Lee, H.G.; Perry, G.; Smith, M.A.; Gustaw-Rothenberg, K.; Lerner, A. All-Trans Retinoic Acid as a Novel Therapeutic Strategy for Alzheimer’s Disease. Expert Rev. Neurother. 2009, 9, 1615–1621. [Google Scholar] [CrossRef]
  78. Pino-Lagos, K.; Benson, M.J.; Noelle, R.J. Retinoic Acid in the Immune System. Ann. N. Y. Acad. Sci. 2008, 1143, 170–187. [Google Scholar] [CrossRef]
  79. Erbiai, E.H.; Amina, B.; Kaoutar, A.; Saidi, R.; Lamrani, Z.; Pinto, E.; Esteves da Silva, J.C.G.; Maouni, A.; Pinto da Silva, L. Chemical Characterization and Evaluation of Antimicrobial Properties of the Wild Medicinal Mushroom Ganoderma lucidum Growing in Northern Moroccan Forests. Life 2023, 13, 1217. [Google Scholar] [CrossRef]
  80. Zhou, K.; Yu, L. Effects of Extraction Solvent on Wheat Bran Antioxidant Activity Estimation. LWT–Food Sci. Technol. 2004, 37, 717–721. [Google Scholar] [CrossRef]
  81. Yousuf, B.; Panesar, P.S.; Chopra, H.K.; Gul, K. Characterization of Secondary Metabolites from Various Solvent Extracts of Saffron Floral Waste. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2015, 87, 89–100. [Google Scholar] [CrossRef]
  82. Packialakshmi, N.; Naziya, S. Fourier Transform Infrared Spectroscopy Analysis of Various Solvent Extracts of Caralluma Fimbriyata. Asian J. Biomed. Pharm. Sci. 2014, 4, 20–25. [Google Scholar]
  83. Tomsone, L.; Kruma, Z.; Galoburda, R. Comparison of Different Solvents and Extraction Methods for Isolation of Phenolic Compounds from Horseradish Roots. Int. Sch. Sci. Res. Innov. 2012, 6, 1164–1169. [Google Scholar]
  84. Pham, H.; Nguyen, V.; Vuong, Q.; Bowyer, M.; Scarlett, C. Effect of Extraction Solvents and Drying Methods on the Physicochemical and Antioxidant Properties of Helicteres hirsuta lour Leaves. Technologies 2015, 3, 285–301. [Google Scholar] [CrossRef]
  85. Avcı, E.; Avcı, G.A.; Kose, D.A. Determination of Antioxidant and Antimicrobial Activities of Medically Important Mushrooms Using Different Solvents and Chemical Composition via GC / MS Analyses. J. Food Nutr. Res. 2014, 2, 429–434. [Google Scholar] [CrossRef]
  86. Zengin, G.; Sarikurkcu, C.; Gunes, E.; Uysal, A.; Ceylan, R.; Uysal, S.; Gungor, H.; Aktumsek, A. Two Ganoderma Species: Profiling of Phenolic Compounds by HPLC-DAD, Antioxidant, Antimicrobial and Inhibitory Activities on Key Enzymes Linked to Diabetes Mellitus, Alzheimer’s Disease and Skin Disorders. Food Funct. 2015, 6, 2794–2802. [Google Scholar] [CrossRef]
  87. Nagaraj, K.; Mallikarjun, N.; Naika, R.; Venugopal, T.M. Antioxdative Activities of Wild Macro Fungi Ganoderma applanatum (PERS.) PAT. Asian J. Pharm. Clin. Res. 2014, 7, 166–171. [Google Scholar]
  88. Tel, G.; Ozturk, M.; Duru, M.E.; Turkoglu, A. Antioxidant and Anticholinesterase Activities of Five Wild Mushroom Species with Total Bioactive Contents. Pharm. Biol. 2015, 53, 824–830. [Google Scholar] [CrossRef]
  89. Barros, L.; Ferreira, M.J.; Queirós, B.; Ferreira, I.C.F.R.; Baptista, P. Total Phenols, Ascorbic Acid, B-Carotene and Lycopene in Portuguese Wild Edible Mushrooms and Their Antioxidant Activities. Food Chem. 2007, 103, 413–419. [Google Scholar] [CrossRef]
  90. Bravo, L.; Sources, D.; Significance, N. Polyphenols: Chemistry, Dietary Sources, Metabolism, and Nutritional Significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef]
  91. Esmaeili, M.A.; Sonboli, A. Antioxidant, Free Radical Scavenging Activities of Salvia brachyantha and Its Protective Effect against Oxidative Cardiac Cell Injury. Food Chem. Toxicol. 2010, 48, 846–853. [Google Scholar] [CrossRef]
  92. Kim, K.C.; Kim, I.G. Ganoderma lucidum Extract Protects DNA from Strand Breakage Caused by Hydroxyl Radical and UV Irradiation. Int. J. Mol. Med. 1999, 4, 273–277. [Google Scholar] [CrossRef]
  93. Lakshmi, B.; Ajith, T.A.; Sheena, N.; Gunapalan, N.; Janardhanan, K.K. Antiperoxidative, Anti-Inflammatory, and Antimutagenic Activities of Ethanol Extract of the Mycelium of Ganoderma lucidum Occurring in South India. Teratog. Carcinog. Mutagen. 2003, 23 (Suppl. 1), 85–97. [Google Scholar] [CrossRef]
  94. Lee, J.M.; Kwon, H.; Jeong, H.; Lee, J.W.; Lee, S.Y.; Baek, S.J.; Surh, Y.J. Inhibition of Lipid Peroxidation and Oxidative DNA Damage by Ganoderma lucidum. Phytother. Res. 2001, 15, 245–249. [Google Scholar] [CrossRef]
  95. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric Oxide and Peroxynitrite in Health and Disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef]
  96. Vamanu, E.; Nita, S.S. Antioxidant Capacity and the Correlation with Major Phenolic Compounds, Anthocyanin, and Tocopherol Content in Various Extracts from the Wild Edible Boletus edulis Mushroom. BioMed Res. Int. 2013, 2013, 313905. [Google Scholar] [CrossRef]
  97. Jayakumar, T.; Thomas, P.A.; Geraldine, P. In-Vitro Antioxidant Activities of an Ethanolic Extract of the Oyster Mushroom, Pleurotus ostreatus. Innov. Food Sci. Emerg. Technol. 2009, 10, 228–234. [Google Scholar] [CrossRef]
  98. Rajasekaran, M.; Rajasekaran, M.; Kalaimagal, C. In Vitro Antioxidant Activity of Ethanolic Extract of a Medicinal Mushroom, Ganoderma Lucidum. J. Pharm. Sci. Res. 2011, 3, 1427–1433. [Google Scholar]
  99. Abdullah, N.; Ismail, S.M.; Aminudin, N.; Shuib, A.S.; Lau, B.F. Evaluation of Selected Culinary-Medicinal Mushrooms for Antioxidant and ACE Inhibitory Activities. Evid.-Based Complement. Altern. Med. 2012, 2012, 464238. [Google Scholar] [CrossRef]
  100. Lim, C.S.H.; Lim, S.L. Ferric Reducing Capacity Versus Ferric Reducing Antioxidant Power for Measuring Total Antioxidant Capacity. Lab. Med. 2013, 44, 51–55. [Google Scholar] [CrossRef]
  101. Mousavi, S.M.; Hashemi, S.A.; Gholami, A.; Omidifar, N.; Chiang, W.H.; Neralla, V.R.; Yousefi, K.; Shokripour, M. Ganoderma lucidum Methanolic Extract as a Potent Phytoconstituent: Characterization, In-Vitro Antimicrobial and Cytotoxic Activity. Sci. Rep. 2023, 13, 17326. [Google Scholar] [CrossRef]
  102. Veljović, S.; Veljović, M.; Nikićević, N.; Despotović, S.; Radulović, S.; Nikšić, M.; Filipović, L. Chemical Composition, Antiproliferative and Antioxidant Activity of Differently Processed Ganoderma lucidum Ethanol Extracts. J. Food Sci. Technol. 2017, 54, 1312–1320. [Google Scholar] [CrossRef]
  103. Fronza, M.; Murillo, R.; Ślusarczyk, S.; Adams, M.; Hamburger, M.; Heinzmann, B.; Laufer, S.; Merfort, I. In Vitro Cytotoxic Activity of Abietane Diterpenes from Peltodon Longipes as well as Salvia miltiorrhiza and Salvia sahendica. Bioorg. Med. Chem. 2011, 19, 4876–4881. [Google Scholar] [CrossRef]
  104. Li, S.; Wang, P.; Deng, G.; Yuan, W.; Su, Z. Cytotoxic Compounds from Invasive Giant Salvinia (Salvinia molesta) against Human Tumor Cells. Bioorg. Med. Chem. Lett. 2013, 23, 6682–6687. [Google Scholar] [CrossRef]
  105. Cardwell, G.; Bornman, J.F.; James, A.P.; Black, L.J. A Review of Mushrooms as a Potential Source of Dietary Vitamin, D. Nutrients 2018, 10, 1498. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the study workflow. Ganoderma lucidum fruiting bodies were collected from a high-altitude site (~2500 m) and taxonomically identified (I). Crude extracts were prepared using four solvents: water, ethanol, methanol, and acetone (II). Bioactive compounds were screened by quantifying total phenolics (TPC), flavonoids (TFC), and carotenoids (III). Antioxidant and cytotoxic assays were conducted to evaluate bioactivity (IV). Chemical constituents of the extracts were further identified using gas chromatography–mass spectrometry (GC–MS) (V).
Figure 1. Overview of the study workflow. Ganoderma lucidum fruiting bodies were collected from a high-altitude site (~2500 m) and taxonomically identified (I). Crude extracts were prepared using four solvents: water, ethanol, methanol, and acetone (II). Bioactive compounds were screened by quantifying total phenolics (TPC), flavonoids (TFC), and carotenoids (III). Antioxidant and cytotoxic assays were conducted to evaluate bioactivity (IV). Chemical constituents of the extracts were further identified using gas chromatography–mass spectrometry (GC–MS) (V).
Cimb 47 00624 g001
Figure 2. Map of Nepal in a global context (A), G. lucidum collection site (B), and its fruiting body growing on Quercus lanata (C).
Figure 2. Map of Nepal in a global context (A), G. lucidum collection site (B), and its fruiting body growing on Quercus lanata (C).
Cimb 47 00624 g002
Figure 3. Total phenolic content (A) and total flavonoid content (B) expressed as pyrogallol and quercetin equivalents (mg/g dry extract), respectively. Data are mean ± SD from three biological replicates. Statistical analysis was performed using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Bars with the same letter (a–d) are not significantly different; different letters indicate significant differences between solvents.
Figure 3. Total phenolic content (A) and total flavonoid content (B) expressed as pyrogallol and quercetin equivalents (mg/g dry extract), respectively. Data are mean ± SD from three biological replicates. Statistical analysis was performed using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Bars with the same letter (a–d) are not significantly different; different letters indicate significant differences between solvents.
Cimb 47 00624 g003
Figure 4. β-carotene (A) and lycopene (B) content as mg carotenoid per g of dry extract in various solvent extract. Data are mean ± SD from three biological replicates. Statistical analysis was performed using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Bars with the same letter (a–d) are not significantly different; different letters indicate significant differences between solvents.
Figure 4. β-carotene (A) and lycopene (B) content as mg carotenoid per g of dry extract in various solvent extract. Data are mean ± SD from three biological replicates. Statistical analysis was performed using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Bars with the same letter (a–d) are not significantly different; different letters indicate significant differences between solvents.
Cimb 47 00624 g004
Figure 5. DPPH radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S5.
Figure 5. DPPH radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S5.
Cimb 47 00624 g005
Figure 6. Superoxide radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S6.
Figure 6. Superoxide radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S6.
Cimb 47 00624 g006
Figure 7. Hydroxyl radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S7.
Figure 7. Hydroxyl radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S7.
Cimb 47 00624 g007
Figure 8. Nitric oxide radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S8.
Figure 8. Nitric oxide radical scavenging activity (%) of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S8.
Cimb 47 00624 g008
Figure 9. Reducing power assay of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S9.
Figure 9. Reducing power assay of G. lucidum extracts prepared with four different solvents, compared to the standard antioxidant ascorbic acid. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S9.
Cimb 47 00624 g009
Figure 10. Cytotoxic activity of G. lucidum extracts against HeLa cervical cancer cell lines at varying concentration (100, 500, and 1000 µg/mL) with DMSO control. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S11.
Figure 10. Cytotoxic activity of G. lucidum extracts against HeLa cervical cancer cell lines at varying concentration (100, 500, and 1000 µg/mL) with DMSO control. Dose–response regression lines with corresponding R2 values are shown. Data are mean ± SD from three biological replicates. Statistical analysis was conducted using one-way ANOVA with Tukey’s multiple comparisons test (p < 0.05). Exact p-values indicating significant difference between extracts at each concentration are provided in Supplementary Figure S11.
Cimb 47 00624 g010
Figure 11. Heatmap of IC50 values obtained from various antioxidant and cytotoxic assays. Values are expressed in µg/mL. Within each assay, values with the same letter (a–e) are not significantly different; however, different letters indicate significant differences between solvents (p < 0.05). Ascorbic acid: AA.
Figure 11. Heatmap of IC50 values obtained from various antioxidant and cytotoxic assays. Values are expressed in µg/mL. Within each assay, values with the same letter (a–e) are not significantly different; however, different letters indicate significant differences between solvents (p < 0.05). Ascorbic acid: AA.
Cimb 47 00624 g011
Figure 12. GC-MS chromatograms of G. lucidum extracts obtained using different solvents, GEE (A), GME (B), and GAE (C), showing the presence of distinct compounds at various retention times.
Figure 12. GC-MS chromatograms of G. lucidum extracts obtained using different solvents, GEE (A), GME (B), and GAE (C), showing the presence of distinct compounds at various retention times.
Cimb 47 00624 g012
Figure 13. Mass spectra of selected bioactive compounds identified in G. lucidum (GEE, GME, and GAE) extracts, along with their chemical structures, molecular formulas, and molecular weights.
Figure 13. Mass spectra of selected bioactive compounds identified in G. lucidum (GEE, GME, and GAE) extracts, along with their chemical structures, molecular formulas, and molecular weights.
Cimb 47 00624 g013
Table 1. Summary of the ecological, morphological, anatomical, and taxonomic characteristics of G. lucidum.
Table 1. Summary of the ecological, morphological, anatomical, and taxonomic characteristics of G. lucidum.
ParameterDescription
Collection monthSeptember–October
LocationChandragiri Hill, Kathmandu, Nepal
Elevation7482 ft (2280 m) above sea level
CoordinatesLatitude: 27.67402° N; Longitude: 85.19874° E
Ecosystem typeSolitary
SubstrateWood, stump, log, stick, base of tree, bark
Host treeQuercus lanata
Rot typeWhite-rot
Surrounding treesPredominantly hardwoods
Basidiocarp size
Texture
6–10 cm × 4–16 cm × 1.3 cm
Woody to corky
StipeSub-sessile to laterally stipitate, 2–3 cm
Pileus shapeReniform
Upper surfaceLaccate, dark reddish to purplish, yellowish at margins; brittle, soft
MarginBlunt, rounded, brown-white
Pore surfaceCreamy to milky coffee; ~5 pores/mm
Tube layer2–9 mm long, white turning brown when brushed or aged
Context9 mm thick, brown, without horny deposition
Cutis typeThick-walled claviform with diverticula; 35–42 × 6–8.5 µm
Hyphal systemTrimitic: Generative (3.3 µm, hyaline, thin-walled, with clamp); Skeletal (5.8–7.5 µm, brown, thick); Binding (5–7.5 µm, brown)
Basidiospores8.3–10 × 6.6 µm; yellowish-brown
Identification authorityProf. Mahesh Kumar Adhikari, Dept. of Plant Resources, Kathmandu
Table 2. Percentage yield of various solvent extracts.
Table 2. Percentage yield of various solvent extracts.
ExtractWeight of Sample Before Extraction (gm)Weight Obtained After Extraction (gm)% Yield Value
Water100.2292.29 d
Ethanol100.3433.43 b
Methanol100.2982.98 c
Acetone100.5015.01 a
Values with the same letter (a–d) are not significantly different; different letters indicate significant differences between solvents (p < 0.05).
Table 3. Summary of key compounds detected by the GC-MS analysis in various solvents with reported pharmacological relevance.
Table 3. Summary of key compounds detected by the GC-MS analysis in various solvents with reported pharmacological relevance.
Compound NameSolvent Extracts (% Area)Compound ClassKey Pharmacological RelevanceReference
GEEGMEGAE
7,22-Ergostadienone3.542.902.56SterolAntithrombotic activity with cardiovascular benefit; antidiabetic, anticancer, and neuroprotective effects; pro-inflammatory properties (activating Toll-like receptors, cytokines, and chemokines)[37,40,41,42,43]
9(11)-Dehydroergosteryl 3,5-dinitrobenzoate2.903.132.70Sterol conjugateAnti-inflammatory; antibacterial (MRSA and S. aureus); and cytotoxic properties[44,45]
δ-Tocopherol2.133.910.75TocopherolAntioxidant; anti-inflammatory (primarily Via inhibiting protein kinase C and reducing eicosanoid production); anticancer (both in vitro and in vivo prostate xenograft models); cardioprotective and neuroprotective[46,47]
4-[5-(2-bromophenyl)-1,2,4-oxadiazol-3-yl]-1,2,5-oxadiazol-3-amine--0.35Synthetic heterocycleAnticancer (potentially via targeting topoisomerase II relaxation activity); antibacterial; anti-inflammatory; analgesic properties; antioxidant [48,49,50,51,52]
Ergosta-tetraenone3.86-1.67Sterol derivativeAnticancer (Via G2/M arrest and apoptosis induction); nephroprotection (mitigation of renal damage in mouse model); anti-inflammatory[53,54,55]
Ergosterol--73.99SterolVitamin D2 precursor; lipid soluble antioxidant; anticancer effects (cell cycle arrest and modulates Wnt/β-catenin signaling pathway); antimicrobial; antidiabetic; immunomodulatory effects [56,57,58]
Ferruginol3.18--Abietane diterpeneAnticancer (apoptosis induction in melanoma, prostate, lung, and ovarian cancer cells); neuroprotective (reduces α-synuclein toxicity and restores LTP in Alzheimer’s models); cardioprotective (both in vitro and in vivo models); antimicrobial and antiviral [59,60,61,62,63,64,65]
Geranylgeraniol5.26-0.89Diterpenoid alcoholAnti-inflammatory (NF-κB inhibition; ↓ IL-1β, TNF-α, IL-6, COX-2); pain relief; bone and muscle support (muscle regeneration and prevents bisphosphonate-related bone damage); antimicrobial activity; hormonal balance; glucose homeostasis [66,67,68,69]
Hinokione2.95.50.9Abietane diterpeneAnticancer; anti-inflammatory; hypoglycemic and β-Cell regenerative properties (promotes β-cell differentiation and improved glycemia in zebrafish); antibacterial; antioxidant[70,71,72]
Nerolidol acetate-1.70-Sesquiterpene esterAnticancer; anti-inflammatory; neuroprotective; antimicrobial; antifungal; antioxidant[73,74,75]
Retinoic acid--0.50RetinoidAcne and photoaging (promotes cell differentiation and skin repair); anticancer (induces differentiation of malignant promyelocytes in acute promyeloid leukemia); neuroprotective[76,77,78]
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

Thapa, I.; Pandey, A.; Tiwari, S.; Awal, S.C. Evaluation of Bioactive Compounds, Antioxidant Activity, and Anticancer Potential of Wild Ganoderma lucidum Extracts from High-Altitude Regions of Nepal. Curr. Issues Mol. Biol. 2025, 47, 624. https://doi.org/10.3390/cimb47080624

AMA Style

Thapa I, Pandey A, Tiwari S, Awal SC. Evaluation of Bioactive Compounds, Antioxidant Activity, and Anticancer Potential of Wild Ganoderma lucidum Extracts from High-Altitude Regions of Nepal. Current Issues in Molecular Biology. 2025; 47(8):624. https://doi.org/10.3390/cimb47080624

Chicago/Turabian Style

Thapa, Ishor, Ashmita Pandey, Sunil Tiwari, and Suvash Chandra Awal. 2025. "Evaluation of Bioactive Compounds, Antioxidant Activity, and Anticancer Potential of Wild Ganoderma lucidum Extracts from High-Altitude Regions of Nepal" Current Issues in Molecular Biology 47, no. 8: 624. https://doi.org/10.3390/cimb47080624

APA Style

Thapa, I., Pandey, A., Tiwari, S., & Awal, S. C. (2025). Evaluation of Bioactive Compounds, Antioxidant Activity, and Anticancer Potential of Wild Ganoderma lucidum Extracts from High-Altitude Regions of Nepal. Current Issues in Molecular Biology, 47(8), 624. https://doi.org/10.3390/cimb47080624

Article Metrics

Back to TopTop